9%x%01! 2007 Status Report Laboratory Medicine A National From The Lewin Group 2

2015-05-08

: Pdf 2007 Status Report Laboratory Medicine - A National Status Report From The Lewin Group-2 2007_status_report_laboratory_medicine_-_a_national_status_report_from_the_lewin_group-2 02 2013 clpmag uploads wp-content

Open the PDF directly: View PDF PDF.
Page Count: 385

Download9%x%01!  2007 Status Report Laboratory Medicine - A National From The Lewin Group-2
Open PDF In BrowserView PDF
Laboratory Medicine:
A National Status Report

Prepared for:

Division of Laboratory Systems
National Center for Preparedness, Detection, and
Control of Infectious Diseases
Centers for Disease Control and Prevention

Prepared by:

The Lewin Group

Under subcontract to Battelle Memorial Institute

May 2008

Laboratory Medicine:
A National Status Report

Prepared for:

Division of Laboratory Systems
National Center for Preparedness, Detection, and
Control of Infectious Diseases
Centers for Disease Control and Prevention

Prepared by:

The Lewin Group
Julie Wolcott, MA
Amanda Schwartz
Clifford Goodman, PhD

May 2008

Laboratory Medicine: A National Status Report

Executive Summary

TABLE OF CONTENTS
ACKNOWLEDGMENTS

v

EXECUTIVE SUMMARY

1

The Value of Laboratory Medicine to Health Care ....................................................................... 2
Market Profile of the Laboratory Medicine Sector ........................................................................ 3
Laboratory Medicine Workforce...................................................................................................... 4
Quality and the Total Testing Process............................................................................................. 4
Quality Systems and Performance Measurement ......................................................................... 6
Laboratory Information Systems ..................................................................................................... 6
Federal Regulatory Oversight Of Laboratory Medicine............................................................... 7
Reimbursement for Laboratory Medicine ...................................................................................... 8
INTRODUCTION

11

Context of the Report....................................................................................................................... 12
Scope of Report................................................................................................................................. 12
Reference List.................................................................................................................................... 14
METHODS AND LIMITATIONS

15

Methods ............................................................................................................................................. 15
Limitations ........................................................................................................................................ 17
CHAPTER I - THE VALUE OF LABORATORY MEDICINE TO HEALTH CARE

19

Value to the Quality of Patient Care.............................................................................................. 19
Value to Evidence-based Medicine................................................................................................ 22
Value to Clinical Practice Guidelines for Patient Care ............................................................... 22
Value of Laboratory Tests and Services Across Patient Care Continuum ............................... 24
Protecting the Blood Supply and Transplant Recipients............................................................ 33
Detecting Exposure to Illegal or Toxic Drugs .............................................................................. 34
Value to the Measurement of Quality of Care ............................................................................. 35
Value to Public Health and Surveillance ...................................................................................... 38
Quantifying Value Using Cost-effectiveness Analysis ............................................................... 42
Conclusions ....................................................................................................................................... 48
Reference List.................................................................................................................................... 50

May 2008

i

Laboratory Medicine: A National Status Report

Executive Summary

CHAPTER II - MARKET PROFILE OF THE LABORATORY MEDICINE SECTOR

67

U.S. Market Size................................................................................................................................ 67
Market by Laboratory Type and Setting....................................................................................... 69
Market for Laboratory Tests ........................................................................................................... 81
Research-support Testing ............................................................................................................... 94
Conclusions ....................................................................................................................................... 95
Reference List.................................................................................................................................... 97
CHAPTER III - LABORATORY MEDICINE WORKFORCE

105

Types of Professionals ................................................................................................................... 105
Workforce Demographics ............................................................................................................. 109
Vacancy Rates ................................................................................................................................. 111
Wages............................................................................................................................................... 113
Education, Training, and Recruitment........................................................................................ 116
Licensing and Certification ........................................................................................................... 124
Conclusions ..................................................................................................................................... 129
Reference List.................................................................................................................................. 131
CHAPTER IV - QUALITY AND THE TOTAL TESTING PROCESS

139

Definition of the Total Testing Process ....................................................................................... 139
Quality and Errors ......................................................................................................................... 141
Preanalytic Phase ........................................................................................................................... 142
Clinical Pathology Transitional Preanalytic ............................................................................... 151
Clinical Pathology Analytic Phase............................................................................................... 153
Anatomic Pathology Transitional Preanalyic............................................................................. 157
Anatomic Pathology Analytic Phase ........................................................................................... 159
Postanalytic Phase.......................................................................................................................... 162
Point-of-Care Testing..................................................................................................................... 168
Preanalytic Phase ........................................................................................................................... 169
Analytic Phase ................................................................................................................................ 171
Postanalytic Phase.......................................................................................................................... 175
Conclusions ..................................................................................................................................... 177
Reference List.................................................................................................................................. 180

May 2008

ii

Laboratory Medicine: A National Status Report

Executive Summary

CHAPTER V - QUALITY SYSTEMS AND PERFORMANCE MEASUREMENT

197

Approaches to Quality Systems in Laboratory Medicine ........................................................ 197
Performance Measurement........................................................................................................... 210
Conclusions ..................................................................................................................................... 228
Reference List.................................................................................................................................. 230
CHAPTER VI - LABORATORY INFORMATION SYSTEMS

243

Structure of LIS............................................................................................................................... 243
Advanced Applications for Comprehensive Systems .............................................................. 250
Unresolved Issues Related to Advanced Applications............................................................. 250
Data Standards for Comprehensive Systems ............................................................................. 259
Conclusions ..................................................................................................................................... 261
Reference List.................................................................................................................................. 263
CHAPTER VII - FEDERAL REGULATORY OVERSIGHT OF LABORATORY
MEDICINE

271

Regulatory Oversight Systems ..................................................................................................... 271
CLIA Program ................................................................................................................................ 273
Waived Tests................................................................................................................................... 275
Outstanding Issues in Waived Testing ....................................................................................... 276
Non-waived Tests .......................................................................................................................... 279
Outstanding Issues Associated with CLIA Standards.............................................................. 280
Genetic Testing ............................................................................................................................... 291
Outstanding Issues in Genetic Testing........................................................................................ 291
Transfusion-related Services......................................................................................................... 296
Outstanding Issues for Transfusion-related Services................................................................ 299
Conclusions ..................................................................................................................................... 300
Reference List.................................................................................................................................. 303

May 2008

iii

Laboratory Medicine: A National Status Report

Executive Summary

CHAPTER VIII - REIMBURSEMENT FOR LABORATORY MEDICINE

313

Public and Private Sector Payers.................................................................................................. 313
Coverage Decisions........................................................................................................................ 314
Payment Methodologies................................................................................................................ 322
Coding ............................................................................................................................................. 343
Conclusions ..................................................................................................................................... 348
Reference List.................................................................................................................................. 350
APPENDIX A – DESIRABLE CHARACTERISTICS FOR PERFORMANCE
MEASURES

A-1

APPENDIX B – SUMMARY OF SELECTED PERFORMANCE INDICATORS
USED BY STAKEHOLDERS

B-1

APPENDIX C – DEVELOPMENT OF THE MEDICARE PAYMENT SYSTEM

C-1

May 2008

iv

Laboratory Medicine: A National Status Report

Executive Summary

ACKNOWLEDGMENTS
This report was prepared by The Lewin Group, Inc. Staff contributing to the report include: Julie
Wolcott, Amanda Schwartz, Clifford Goodman, Palak Parikh, Charlene Chen, Christel Villarivera,
and Debbie Faulk.
The Lewin Group wishes to acknowledge the many people who contributed to this report. This
report was crafted with the guidance of a Technical Experts Committee whose members shared
their expertise and perspectives, including: Raj Behal, Rush University; Robert Christensen,
University of Maryland; D. Robert Dufour, Veterans Affairs Medical Center; Julie Gayken,
Regions Hospital; Lee Hilborne, The RAND Corporation; Michael Laposata, Massachusetts
General Hospital; Stephen Raab, University of Colorado; Ana Stankovic, Becton, Dickinson and
Company; Judith Yost, Centers for Medicare and Medicaid Services (CMS). Lewin also thanks
Robert Black of The Battelle Memorial Institute, primary contractor for this effort, for his guidance
in meeting the report objectives and contractual requirements.
Several officials at the Centers for Disease Control and Prevention (CDC) participated in a thorough
review of the report to ensure that its content meets institutional standards for objectivity and
evidence. Their constructive comments and suggestions were instrumental in shaping the final
report. In particular, we thank D. Joe Boone, Nancy Anderson, Devery Howerton, Anne Pollock,
and Susan Snyder for their leadership; and Fred Angulo, Diane Bosse, Debbie Deppe, Maribeth
Gagnon, Sharon Granade, Scott Grosse, Bin Chen, Ira Lubin, Adam Manasterski, Daniel Pollock,
Shahram Shahangian, Darshan Singh, Fred Tenover, Eric Thompson, Pamela Thompson, and
Glennis Westbrook for their helpful comments.
Others providing important information and insights include: Scott Becker, Association of Public
Health Laboratories; Lynn Boyd, College of American Pathologists; Peter Cholakis, Avitar,
Incorporated; Greg Countryman, Oregon Health and Science University; Laura Creswell,
Organization of Economic Cooperation and Development; Rachel DeSoignie, National
Credentialing Agency for Laboratory Personnel; Jody DeVoll, Association of Public Health
Laboratories; Helena Duncan, Commission on Laboratory Accreditation; Karen Edrington, CMS;
Sylvia Etzel, American Medical Association; Andrea Ferreira-Gonzalez, Virginia Commonwealth
University; Glen Fine, Clinical Laboratory Standards Institute; Daniel Fink, New York Presbyterian
Hospital; Steven Ford, Commonwealth of Virginia; Susan Fridy, Organization of Economic
Cooperation and Development; Jonah Frohlich, California HealthCare Foundation; Gloria Gantt,
CMS; Tara Goldman, Food and Drug Administration (FDA); Anita Greenberg, CMS; Alberto
Gutierrez, FDA; Steven Gutman, FDA; Gary Hoffman, Wisconsin State Laboratory of Hygiene; E.
Blair Holladay, American Society for Clinical Pathology; Olga Ioffe, University of Maryland; Dina
Itkin, Bureau of Labor Statistics; Gail Javitt, Johns Hopkins University; Penelope Jones, American
Association for Clinical Chemistry; David Kennell, Kennell and Associates; Kurt Kroemer,
American Red Cross; Sam Lee, Misys Healthcare Systems; Sonya Lemmerbrock, MedPlus,
Incorporated; Norman Marks, FDA; Melissa McElroy, American Society for Histocompatibility and
Immunogenetics; Charles Meader, DiagnosisPro; David Mongillo, American Clinical Laboratory
Association; Georgina Morris, Quintiles Transnational; Geri Mulcahy, American Medical
Technologists; Cheryl Murray, American Academy of Family Physicians; Robert Murray, Lutheran
General Hospital; Joyce Obradovic, American Osteopathic Association; Perry Patterson, IOMA;
Margaret Peck, The Joint Commission; Esther Reagan, Health Management Associates; William

May 2008

v

Laboratory Medicine: A National Status Report

Executive Summary

Riley, University of Minnesota; Ahren Rittershaus, Medical University of South Carolina; Charles
Root, CodeMap LLC; Joseph San Filippo, Becton, Dickinson and Company; Jayme Sellers, American
Medical Association; Barb Short, Quest Diagnostics; David Smalley, American Association of
Bioanalysts; Derek Smart, American Medical Association; Thomas Sodeman, College of American
Pathologists; Roberta Spiro, Genetests.org; Mary Stevenson, CMS; Vince Stine, American
Association for Clinical Chemistry; Robin Stombler, Auburn Health Strategies; Susan Stramer,
American Red Cross; Judy Sullivan, AABB; Mark Terry, IOMA; Dan Tice, National Accrediting
Agency for Clinical Laboratory Sciences; Lorrie Van Akkeren, American Association of Colleges of
Osteopathic Medicine; Susan Walker, Office of Laboratory Quality Assurance, Washington State;
Audrey Watson, Bureau of Labor Statistics; Mike Zaranek, Quintiles Transnational; Carol Zeller,
CMS; and Rebecca Zeller, CMS.
Other individuals that provided comments on the report outline early in the development process
include: Sousan Altaie, FDA; Rex Astles, CDC; Michael Astion, University of Washington;
Patricia Charache, Johns Hopkins Medical Institute; John Counts, Foundation for Health Quality;
Kimberly Glenn, American Society for Histocompatibility and Immunogenetics; Renata
Greenspan, Armed Forces Institute of Pathology; Shirley McKenzie, American Society for Clinical
Laboratory Science; J. Michael Miller, CDC; Elissa Passiment, American Society for Clinical
Laboratory Science; Mary Paton, College of American Pathologists; Howard Thompson, CDC.

May 2008

vi

Laboratory Medicine: A National Status Report

Executive Summary

EXECUTIVE SUMMARY
Although the U.S. ranks highest in per capita health care spending, there is overwhelming
evidence of gaps between well-founded standards of care and health care practice. The Institute
of Medicine reports, To Err is Human: Building a Safer Health System (1999) and Crossing the Quality
Chasm: A New Health System for the 21st Century (2001), and other sentinel studies have focused
national attention on improving the quality and safety of health care. Stakeholders agree that the
quality of care delivered in the U.S. is inadequate and that the organization and delivery of health
care must be improved.
Given the shortfalls in quality and continued escalation in costs, health care must be assessed
continually to inform decision-making, and redesign delivery and incentives as needed, to yield
appropriate, high quality care. An integral component of care is laboratory medicine, which
extends across research; screening, diagnosis, and treatment; and public health. Appropriate use
of laboratory testing is essential for achieving safe, effective, and efficient care to patients.
Health care must be informed by data derived from scientific assessment of efficacy and
effectiveness of procedures, and must adapt to ongoing changes in science, technology, and
practice.9 Laboratory medicine is not only responding to these changes, but is contributing to
them in an environment of demographic, social, and economic change.
The Centers for Disease Control and Prevention (CDC) has commissioned this report to
contribute to the groundwork for transforming laboratory medicine over the next decade. CDC
charged The Lewin Group, under subcontract to Battelle Memorial Institute, with drafting this
document, Laboratory Medicine: A National Status Report. The report examines in detail the key
factors affecting the laboratory medicine sector, and is organized into chapters on the following
main topics:

ƒ

Value of laboratory medicine

ƒ

Market profile of the laboratory medicine sector

ƒ

Laboratory medicine workforce

ƒ

Quality and the total testing process

ƒ

Quality systems and performance measurement

ƒ

Laboratory information systems

ƒ

Federal regulatory oversight of laboratory medicine

ƒ

Reimbursement for laboratory medicine

May 2008

1

Laboratory Medicine: A National Status Report

Executive Summary

THE VALUE OF LABORATORY MEDICINE TO HEALTH CARE
Laboratory testing has a major effect on clinical decisions, providing physicians, nurses, and other
health care providers with information that aids in the prevention, diagnosis, treatment, and
management of disease. Despite this scope of influence, spending on laboratory services accounts
for only 2.3% of U.S. health care expenditures and 2%of Medicare expenditures.

ƒ

Laboratory tests provide objective data about patient health that enable screening for
risk factors, accurate and early diagnosis, determination of disease severity and
likelihood of recovery, selection and monitoring of treatment, and evaluation of
potential adverse outcomes. Some laboratory tests are vital to patient self
management of chronic conditions.

ƒ

Information provided by laboratory testing is critical for maintaining quality and safety,
including the prevention of adverse reactions. For managing medication, testing
provides information for maintaining optimum drug levels, helps to detect and recover
from medication errors, and enables use of genetic information to guide personalized
prescribing. Laboratories protect the blood supply from pathogens and accurately
match patients and blood products.

ƒ

Services provided by clinical laboratories are critical to public health at the individual and
population levels by identifying nosocomial infections, antimicrobial resistance, infectious
disease outbreaks, exposure to toxic substances, and chemical and biological threats.
Laboratories also help to mitigate the effects of natural disasters by enabling rapid
turnaround of tests used during triage and emergency care of individual patients as well
as tests to confirm the presence of communicable diseases that threaten the population.

ƒ

Laboratory medicine supports the practice of evidence-based medicine and is being
incorporated into clinical practice guidelines, which assist practitioners and patients in
making decisions about individuals’ health care in specific circumstances.

ƒ

Laboratory testing is one of several important indicators for assessing quality of care,
particularly for national priority health conditions such as diabetes, heart failure, and
colon cancer. Laboratory data can be used in support of value-based purchasing.

ƒ

Greater attention by providers and payers to evidence-based medicine, practice
guidelines, and quality indicators is contributing to more appropriate use of laboratory
tests, diminishing both overuse and underuse of tests.

ƒ

The evidence base for the cost-effectiveness of laboratory tests, and the broader therapeutic
regimens and other interventions of which they are a part, is growing. This evidence is
helping to inform appropriateness of test selection and sequencing, technology acquisition
decisions, formulary design (including for pharmacogenomic-mediated therapies), and
screening and other population-based interventions. It is also being considered in selected
coverage and payment policies of some health plans and other third-party payers.

May 2008

2

Laboratory Medicine: A National Status Report

Executive Summary

MARKET PROFILE OF THE LABORATORY MEDICINE SECTOR
The revenue, spending, and test volume of the U.S. clinical laboratory testing market has grown
steadily over the past decade. Market expansion is attributed to changes in demographic factors
and burden of disease; scientific, medical, and technological advances; and increased consumer
awareness and self care.

ƒ

Based on 2007 data from the Centers for Medicare and Medicaid (CMS) Online Survey,
Certification, and Reporting (OSCAR) database, CDC estimates that approximately 6.8
billion laboratory tests are performed annually in the U.S. Other data from 2007
evaluations indicate the following regarding the extent of the market for laboratory tests.
•

Laboratory testing revenues were a projected $52 billion in 2007.

•

Clinical pathology comprises 66% of all laboratory tests and $32 billion in
revenue.

•

Anatomic pathology and cytology account for 23% of laboratory tests and $11
billion in revenue.

•

Molecular and esoteric (e.g., low volume tests such as those for rare diseases)
testing account for 8% of laboratory tests and $4 billion in revenue.

•

Drugs of abuse testing accounts for 3% of laboratory tests and $1.5 billion in
revenue.

ƒ

More than 4,000 laboratory tests are available for clinical use. Of the 1,162 tests that are
reimbursed by Medicare, about 500 are performed regularly.

ƒ

The number of genetic tests is growing. An estimated 1,430 diseases are now detectable
using genetic testing; of these, an estimated 287 are tested only in research settings.

ƒ

The number of Clinical Laboratory Improvement Amendment (CLIA)-certified
laboratories has grown to exceed 200,000 in 2007. Physician office laboratories represent
54% of clinical laboratories in this sector, four out of five of which are certified to
perform only waived and/or provider-performed microscopy tests (e.g., rapid
streptococcal detection, wet mount examination).

ƒ

Hospital-based laboratories account for the largest proportion of total testing volume
(55%) and generate the highest proportion of total testing revenue (54%), projected at
$28.4 billion for 2007. From 1999 to 2006, the average annual growth rate of both test
volume and revenue was approximately 6-7%. In 2006, privately-owned laboratories
generated revenues of $15.5 billion (32% of total laboratory testing revenue that year).

ƒ

Consumer directed testing is a key area for market growth. In 2004, 10-15% of hospital
and commercial clinical laboratories offered some form of direct access testing.
Laboratories should be prepared to assume a greater advisory role and provide other
support to promote informed self care by consumers.

ƒ

Publicly available information about the economic status and quality of the laboratory
medicine sector remains limited. The main current sources are CMS’ OSCAR database,
ad hoc surveys, and commercial market reports used for investment purposes. As a

May 2008

3

Laboratory Medicine: A National Status Report

Executive Summary

group, these leave certain gaps in covering the laboratory market, including reliable
estimates of market revenues, spending, test volume, and laboratory testing trends.

LABORATORY MEDICINE WORKFORCE
Comprising pathologists, doctoral-level laboratory scientists, technologists/scientists, and
technicians, the laboratory medicine workforce has a vital role in the health care system,
managing and applying evidence-based, scientific testing techniques to support patient care and
protect against public health threats. However, there is growing concern regarding shortages in
the number of laboratory professionals entering the workforce. The shortage could become
pronounced with the forthcoming retirement of many laboratorians. At the same time, the
demand for laboratory services continues to increase. Innovative technologies are changing the
practice of laboratory medicine, educational requirements and staff qualifications.

ƒ

In 2005, there were an estimated 19,339 pathologists in the U.S., including 80% in
community practice. Minorities are under-represented in the discipline of pathology,
with 10% identified as Asian, 3% Hispanic, and 1% African American. Slightly more
than half of pathology residents are female.

ƒ

An estimated 160,760 medical technologists/scientists (including cytotechnologists)
and 144,710 technicians were employed in the U.S. in 2006. While nearly three-fourths
of this workforce is female, it is more representative of the diverse ethnic makeup of
the population, i.e., 12% Asian, 11% African American, and 7% Hispanic. By type of
region, 58% of technologists/scientists work in an urban setting, 24% in suburban, and
18% in rural.

ƒ

The number of technologist/scientist and technician education programs has declined
by more than 50% since 1970, with the most dramatic decline in technologist/scientist
programs, 71% of which closed between 1970 and 2007. In contrast, the number of
phlebotomy training programs increased six-fold from 1987 to 2003.

ƒ

Current enrollment in specialized technologist/scientist and technician educational
programs is lowest in blood banking and histotechnology. Recent recruiting efforts
appear to be effective, specifically those targeted at recruiting minorities and males.

ƒ

The shortage of technologists/scientists and technicians is expected to worsen over the
next decade with demographic changes and retirements. Although personnel vacancies
were highest in 2000 (11-22%), they remained steady from 2002 to 2005 at an annual rate
of 4-7%. Vacancies vary according to staff position, laboratory type and size, and
geographic location.

ƒ

Technological advances will change the qualifications required of the next generation of
laboratory professionals. The laboratory sector needs to clearly redefine staffing
qualifications and workforce level requirements accordingly.

QUALITY AND THE TOTAL TESTING PROCESS
The total testing process (TTP) defines the preanalytic, analytic, and postanalytic phases of
laboratory testing, and serves as the basis for designing and implementing interventions,
restrictions, or limits that can reduce or remove the likelihood of errors. Despite continued
May 2008

4

Laboratory Medicine: A National Status Report

Executive Summary

improvements, many sources of error remain to be addressed. Higher rates of error occur in the
preanalytic phase of testing, but the distribution of errors can vary widely among institutions,
settings, and types of tests. Estimated error rates by phase of testing are in the ranges of 32-75% in
the preanalytic phase, 13-32% in the analytic phase, and 9-31% in the postanalytic phase. Some
errors that occur in the analytic phase originate with errors in preanalytic processes.
Chief issues affecting quality include poor communication and insufficient knowledge of tests that
occur most often during test selection/ordering and interpretation of results. Common errors in
clinical and anatomic pathology involve patient and/or specimen misidentification, specimen
collection errors, and specimen contamination. Test turnaround time and notification of critical
values are frequently cited for ratings of below-average to poor in customer satisfaction surveys.
Medical and scientific advances, such as in genetic testing, will raise further quality challenges.

ƒ

Lack of uniformity and standardization of clinical pathology test values among
manufacturers hinders implementation of laboratory-based guidelines, which require
method-dependent decision limits. Heterogeneity of test values also makes it difficult
for clinicians to work in an integrated health system using more than one testing
method, or to address the needs of special patient populations.

ƒ

Laboratorian consultations are standard practice and reimbursed for anatomic pathology,
but this is not always the case in clinical and molecular pathology. The primary barriers to
interpretive consultations in clinical pathology reports are lack of reimbursement for such
consultations and the shortage of subspecialty expertise. Expanded consultation services
to clinicians would contribute to improved patient care and outcomes.

ƒ

Quality control (QC), performance evaluation, and test reproducibility standards to
minimize diagnostic discrepancies and errors have been better defined and applied in
clinical pathology than in anatomic pathology. Such measures should be developed for
anatomic pathology.

ƒ

Standardization of data elements and report formats for all laboratory tests is necessary
to improve physician comprehension and use of results as well as to integrate report
data into clinical practice information technology applications. Better use of graphical
displays in results reports is especially important for new proteomic and genetic tests.

ƒ

Effective technologies and strategies to reduce identification-related errors include use of
barcoded labels for containers and slides, inpatient wristbands, and computerized
physician/practitioner order entry (CPOE). Automated analyzers and results verification
have decreased cognitive-related errors in clinical pathology, while external, secondary
consultation in anatomic pathology can help decrease errors for complex cases.

ƒ

Point-of-care testing (POCT) has the potential to significantly enhance the quality of care,
although additional research is needed to identify the best methods for integrating
POCT into daily clinical processes and improving its accuracy as needed. Operators of
POCT devices must be trained appropriately in testing practices.

May 2008

5

Laboratory Medicine: A National Status Report

Executive Summary

QUALITY SYSTEMS AND PERFORMANCE MEASUREMENT
Achieving consistently high levels of quality in laboratory medicine calls for moving beyond
stand-alone, analytic-focused, QC, quality assurance (QA), and proficiency testing (PT) activities.
It requires more comprehensive quality management systems (QMS), such as those espoused in
ISO 9001:2000 and ISO 15189:2003 standards. Performance measurement is an important
component of QMS and has been a core feature of quality improvement programs across many
industries. In laboratory medicine, the great bulk of effort on formal performance measurement
and improvement to date has focused on the analytic phase, with insufficient attention to the preand postanalytic phases.

ƒ

Continuous quality improvement, Toyota “lean” production, Six Sigma, and failure
mode and effects analysis are strategic tools for implementing QMS that are realizing
benefits among early adopters, from small physician office laboratories to large reference
laboratories. Use of continuous quality improvement and Six Sigma has contributed to
financial savings and decreased turnaround time, lean production has improved test
quality and reduced errors, and failure mode and effects analysis has decreased time to
report critical laboratory values.

ƒ

To date, QMS has been most broadly adopted in transfusion medicine to meet Food and
Drug Administration (FDA) requirements. However, adoption of QMS more broadly
among laboratories should increase as CMS and accreditation organizations incorporate
these standards into their requirements. Obstacles to implementation of QMS that must
be addressed include resistance to culture change, lack of leadership and staff
commitment to QMS, and insufficient funding of QMS activities.

ƒ

Aside from PT, CLIA provisions have emphasized structural policies, procedures, and
documentation requirements as a condition for accreditation and certification. Process
measures to assess quality in the TTP remain relatively underdeveloped. Existing ones
have not been uniformly defined or assessed for generalizability, and are subject to wide
variation in their implementation.

ƒ

Substantial work is needed to standardize indicators for pre- and postanalytic processrelated performance measures. Data collection, analysis, and reporting methods also
need to be standardized.

ƒ

Research on laboratory performance has been limited by its focus on the larger, hospitalbased laboratories. Further research is needed examine the challenges faced by smaller
laboratories and physician office laboratories when implementing process-related
performance measurement and quality improvement programs.

ƒ

A small body of evidence addresses the downstream clinical and economic impacts of
particular tests. The lack of substantive research on the impact of laboratory testing
restrains the demonstration of the value of laboratory medicine.

LABORATORY INFORMATION SYSTEMS
Laboratory information systems (LIS) have evolved over the past 30 years from simple systems
designed to generate accurate reports to complete systems that can link laboratory data “end to
end” across the TTP. Information technology and Web-based applications have enabled dramatic

May 2008

6

Laboratory Medicine: A National Status Report

Executive Summary

improvements in laboratory data management, communications, services, education, and
marketing. Health care organizations have helped to advance the integration and reach of
laboratories by linking the LIS with hospital information systems, pharmacy databases, etc.

ƒ

The extent of LIS adoption and capabilities varies widely. While integrated delivery
systems and large laboratories rely on LISs for many aspects of laboratory testing,
physician office laboratories and smaller laboratories primarily use the LIS to facilitate
compliance with CLIA requirements (e.g., QC, PT, QA, patient test management).

ƒ

Lack of harmonized data standards is the single greatest barrier to laboratories’ ability to
integrate data within the laboratory as well as exchange data with external partners.
Further progress in integrating laboratory data more fully with clinical practice
applications cannot be realized unless laboratories, health care organizations, vendors, and
others stakeholders resolve differences in data interchange and terminology standards.

ƒ

Successful integration of enhanced data management features requires increased
computing power and multidirectional, coordinated communication that links the LIS,
preanalytic processing components, specimen transportation system, analyzers, and
postanalytic archiving system.

ƒ

The volume and complexity of data generated from genetic, proteomic, and
pharmacogenetic testing, especially from high-throughput analyses and increased
reliance on automation, requires that LISs be capable of storing and retrieving large
quantities of data.

ƒ

Enabling CPOE, decision support systems, and electronic health record applications with
laboratory data in real time requires continued development of rule-based algorithms
capable of generating and integrating accurate alerts, reminders, order sets, results reports,
and a list of differential diagnoses based on patient signs, symptoms, and characteristics.

ƒ

Digital pathology systems require further advances in high-power computation, data
storage capacity, image formatting, and processing algorithms to facilitate the shift from
single-field images to whole-tissue-processing.

FEDERAL REGULATORY OVERSIGHT OF LABORATORY MEDICINE
In the U.S. health care system, the purposes of regulation include one or more of the following:
protect personal and public health, advance personal and public health, and ensure that the
public has access to sufficient, accurate information for using regulated products and services to
improve their health. The purpose of oversight by designated agencies and organizations is to
enforce and otherwise achieve adherence to the rules and standards comprising regulation.
CLIA has served as the primary regulatory program governing U.S. clinical laboratory testing
since its implementation in 1992. The CLIA program is administered by CMS, which has primary
oversight of the program, in cooperation with CDC and FDA. Rapid technological advances,
demographic shifts, lower tolerance for error, and higher expectations for personal data security
pose challenges to certain aspects of the current regulatory framework for clinical laboratories.

ƒ

May 2008

Technological advances have made laboratory tests easier to use and less subject to user
error, resulting in considerable growth in the number of waived tests from 9 tests in 1993
to more than 1,600 test systems and 76 analytes in 2007. However, a CMS study found that
7

Laboratory Medicine: A National Status Report

Executive Summary

some certificate of waiver facilities perform tests beyond the approved level of complexity.
CMS has taken measures to support surveys at a percentage of these facilities.

ƒ

For non-waived testing, available evidence on the long-term impact of PT on laboratory
performance is limited, and findings of existing studies are confounded by limited
comparable data from CMS and survey organizations and other methodological
shortcomings. Existing studies indicate generally improved performance in recent years,
although some failure rates remain unacceptably high.

ƒ

While laboratories’ flexibility to self-determine QC procedures is desirable, several
factors may contribute to the inconsistencies in implementing this practice. CMS, the
Clinical and Laboratory and Standards Institute, and other stakeholders are developing
evaluation protocols that will outline principles for validation and provide laboratories
with scientific guidance on the development of QC procedures for specific testing
technologies and environments.

ƒ

CMS has taken several actions to improve the operation of the program, including
aligning CLIA technical requirements for QA, QC, and PT with systems-based
approaches to quality management; issuing guidance documents identifying effective
survey processes; strengthening enforcement of regulatory obligations; and
assembling a working group to assess program expansion opportunities pertaining
to revenues, staffing levels, and data collection capabilities.

ƒ

Only a small number of genetic tests are regulated as in vitro diagnostics subject to FDA
premarket review for safety and efficacy (via the 510(k) or pre-market approval routes).
Most genetic tests are developed in-house by laboratories and are regulated under CLIA
general provisions. This framework may be insufficient for the level of efficacy and
protection sought for many tests by clinicians and patients, and creates incentives for
genetic tests to be categorized as laboratory-developed tests and not be subject to the
510(k) or premarket approval routes associated with FDA-regulated tests. CMS is
working with the Secretary’s Advisory Committee on Genetics, Health, and Society;
CDC; CLIAC; FDA; and other experts to ensure the quality of genetic testing.

ƒ

Recent guidance documents issued by FDA clarify its oversight of in vitro diagnostic
multivariate index assays and analyte specific reagents (the active ingredients used in
some laboratory-developed tests). This guidance indicates a noteworthy assertion of
oversight that exposes the small but growing area of highly complex genetic testing to
greater scrutiny usually associated with premarket review processes.

REIMBURSEMENT FOR LABORATORY MEDICINE
Government and private sector third-party payment has enabled patients to access and benefit
from health care products and services, including laboratory testing. The design and updating of
coverage, coding, and payment systems should strive to enable patient access to medically
necessary care, support delivery of high-quality care, and sustain innovation of new technologies.
Further, these systems should discourage inefficiency, fraud and abuse, and non-competitive
practices. Difficulty in acquiring coverage, appropriate coding, and adequate payment can pose
significant hurdles in the use of laboratory testing and decreased incentives for laboratories and
test manufacturers to engage in further test development.

May 2008

8

Laboratory Medicine: A National Status Report

Executive Summary

Even though private sector insurance accounts for higher total revenues, the Medicare program
exerts the strongest influence on laboratory services payment for all U.S. payers. All public
payers and approximately 67% of private payers use Medicare’s payment methodologies as the
basis for their own and as tools for negotiating discounts with providers. Suboptimal practices
and other shortcomings in the Medicare reimbursement system for laboratory testing affect other
public and private sector payers in the U.S. health system. Redesign of the current Medicare
payment system for laboratory services is needed in order to meet the growing scientific,
technical, clinical, and economic challenges of the U.S. health care system.

ƒ

The Medicare statute restricts payment for screening and other preventive technologies
and services, unless otherwise specified by Congress. Having to add these technologies
and services to Medicare benefits on a case-by-case basis via the legislative route is
cumbersome and impedes access to certain proven, beneficial tests.

ƒ

Continued use of 56 different fee schedules across the U.S. is inefficient and unnecessarily
complex. For certain commonly ordered tests, the multiple schedules result in large
regional variations, while for other tests, national limitation amounts results in inadequate
Medicare payments.

ƒ

There is a notable lack of reliable data on the relationships among historical costs on
which the Medicare Clinical Laboratory Fee Schedule is based, current production
costs, and the effects of economies of scale and other cost-reducing effects of
technological changes.

ƒ

Studies of data-derived methods for evaluating the appropriateness of payment rates
and for designing potential new payment systems, e.g., based on resource-based relative
value and microcosting (e.g., activity-based costing) have not been completed.

ƒ

CMS is proceeding with a competitive bidding demonstration project for laboratory
services, with the expectation of substantial savings. Supporters of the demonstration
believe that current prices on the fee schedule have no substantial relationship to actual
costs. However, the demonstration project model is highly exclusivea and could have
significant detrimental effects on clinical laboratories that lose in the bidding process, as
many depend on Medicare reimbursement for a sizable portion of their revenues.

ƒ

Despite modest improvements in their transparency, the processes for establishing
payment levels for new laboratory tests, including assignment of new and existing Current
Procedural Terminology® codes to tests and related methods of cross-walking and gapfilling, remain archaic and inadequate.

ƒ

Federal government investigations of clinical laboratory-related fraud and abuse resulted
in penalties amounting to more than $1.727 billion from 1992 to 2006. Through a separate
rulemaking, CMS will address contractual joint ventures that enable non-pathologist
physicians and other entities to profit from self-referrals of pathology services.

Approaching the close of the first decade of the 21st century, health care now accounts for one-sixth
of the U.S. economy. Laboratory medicine guides, and is often the pivotal determinant in, decisions
a

Competitive bidding initiatives that rely on exclusive or selecting contracting allow only those laboratories
submitting winning bids to participate; losing laboratories are barred from receiving any payment from contracts
during the time of the procurement.

May 2008

9

Laboratory Medicine: A National Status Report

Executive Summary

that influence the magnitude and allocation of resources in much of this burgeoning sector. Any
efforts to improve the quality of health care, let alone transform it, must engage the vibrant market
of laboratory medicine and its workforce, systems for ensuring quality, systems for managing
information, and scientific and technological advances. Such efforts must also confront complex
and, in certain ways, inadequate regulatory and payment systems that strain to cope with the
extraordinary diversity and volume of this field. Further, it will be incumbent upon those in
laboratory medicine to demonstrate its value continually along the dimensions of access, informed
decisions, patient and provider satisfaction, health care outcomes, and cost-effectiveness.

May 2008

10

Laboratory Medicine: A National Status Report

Introduction

INTRODUCTION
The history of laboratory medicine extends to the first recorded examination of human bodily
fluids during the time of the ancient Greek physician Hippocrates around 300 BC.1 Two thousand
years later, the first true clinical laboratory opened in 1896 at Johns Hopkins Hospital.2 Discovery
of the disease-causing agents of epidemics such as tuberculosis, diphtheria, and cholera and the
development of tests to detect their presence throughout the end of the 19th century propelled the
laboratory to a position of importance by the early 20th century.3 The American Society of Clinical
Pathologists was formed in 1922 as the first professional society supporting physicians
specializing in pathology. In 1926, all hospitals accredited by the American College of Surgeons
were required to establish a clinical laboratory under the direction of a physician.
Today, the clinical laboratory serves a vital role in the health care system, spanning research, clinical
care, and public health surveillance. More than 200,000 clinical laboratories provide testing and
services in the U.S. The Clinical Laboratory Improvement Amendments (CLIA) of 1988,
consolidated regulation of all types of clinical laboratories under one statute and established
standards for quality assurance, record maintenance, and proficiency testing for all laboratories.
CLIA defines a clinical laboratory as:
“…a facility for the biological, microbiological, serological, chemical, immunohematological,
hematological, biophysical, cytological, pathological, or other examination of materials derived
from the human body for the purpose of providing information for the diagnosis, prevention, or
treatment of any disease or impairment of, or the assessment of the health of, human beings. These
examinations also include procedures to determine, measure, or otherwise describe the presence or
absence of various substances or organisms in the body. Facilities only collecting or preparing
specimens (or both) or only serving as a mailing service and not performing testing are not
considered laboratories.”4
While this definition describes components of laboratory medicine and the activities that take place
within a clinical laboratory, it does not fully address the practice of laboratory medicine. The
practice of laboratory medicine implies a broader scope of influence beyond the activities in the
laboratory, such as consultations with clinicians to assist with test ordering and results
interpretation, performance measurement for quality improvement in the delivery of patient care,
and, on a small yet growing scale, direct interactions with patients and the public. During a 1986
meeting of the Center for Disease Control and Prevention’s (CDC) Institute on Critical Issues in
Health Laboratory Practice, the extralaboratory functions were incorporated into the concept of the
total testing process—a framework defined by the activities of three distinct phases―preanalytic,
analytic, and postanalytic―that align with clinical workflow outside and inside the laboratory. In
2003, CLIA provisions were revised to align regulatory requirements correspond with one total
testing process. Thus, for the purposes of this report, laboratory medicine is defined broadly as:
Testing services and associated practices for the assessment, diagnosis, treatment, management, or
prevention of health-related conditions utilized in making patient care decisions and improving
public health.

May 2008

11

Laboratory Medicine: A National Status Report

Introduction

CONTEXT OF THE REPORT
Although the U.S. ranks highest in per capita health care spending, there is overwhelming
evidence of gaps between well-founded standards of care and health care practice.5 The Institute
of Medicine (IOM) reports To Err is Human: Building a Safer Health System (1999) and Crossing the
Quality Chasm: A New Health System for the 21st Century (2001), as well as other sentinel studies,
have focused national attention on improving the quality and safety of care delivery.5-8
Stakeholders agree that the quality of care delivered in the U.S. in inadequate and that the
organization and delivery of health care must be changed.
Given the shortfalls in quality and continued escalation in costs, health care must be assessed
continually to inform decision-making, and redesign delivery and incentives as needed, to yield
appropriate, high quality care. An integral component of care is laboratory medicine, which
extends across research, clinical (i.e., screening, diagnosis, and treatment), and public health
settings. Laboratory services account for only 2.3% of total health care expenditures; however,
they have a significant role in informing health care decisions and spending. Appropriate use of
laboratory testing is essential for achieving safe, effective, and efficient care to patients.

SCOPE OF REPORT
Health care must be informed by data derived from scientific assessment of efficacy and
effectiveness of procedures, and must adapt to rapid changes in science, technology, and
practice.9 Indeed, laboratory medicine is not only responding to these changes, but is contributing
to them in an environment of demographic, social, and economic change. Detailed,
comprehensive information about the laboratory medicine sector, particularly in regard to best
practices and the quality of services provided, is necessary to address current and forthcoming
challenges to this important aspect of health care.
CDC has commissioned this report, among others, to lay the groundwork for transforming
laboratory medicine over the next decade. The Lewin Group, under subcontract to Battelle
Memorial Institute, was charged with drafting this document, Laboratory Medicine: A National
Status Report. The report provides a detailed overview of the key factors affecting the laboratory
medicine sector. It is intended that the report serve as a point of reference for measuring and
improving quality in the future as well as for policy guidance to professional organizations,
government agencies, and others who provide, use, regulate, and pay for laboratory services.
Aside from an executive summary and this introduction, the report is organized as follows:

ƒ

Chapter I describes the value of laboratory medicine in clinical care and the broader
health care system. The chapter addresses the roles of laboratory medicine in
screening, diagnosis, and treatment; evidence-based medicine; clinical practice
guidelines; assessing quality of care; contributing to cost-effective health care; and
protection from threats to public health.

ƒ

Chapter II provides detailed information on the magnitude and composition of the
U.S. clinical laboratory testing market, including data by setting (hospitals, physician
offices, independent laboratories, home), and by clinical discipline (clinical pathology,
including molecular diagnostics; anatomic pathology).

May 2008

12

Laboratory Medicine: A National Status Report

a

Introduction

ƒ

Chapter III provides an overview of the laboratory medicine workforce, including
professional responsibilities, demographic characteristics, vacancy rates, and wages. The
chapter also addresses the status of educational programs and licensing and certification
requirements at the federal and state levels.

ƒ

Chapter IV describes the total testing process and the quality issues and errors most
relevant to each phase of laboratory testing for clinical and anatomic pathology. Also
addressed are communication factors associated with the preanalytic and postanalytic
phases. A section of the chapter examines point-of-care testing and related matters of
quality.

ƒ

Chapter V describes the status and future of quality systems in laboratory medicine,
including the shift away from analytic-focused quality control, quality assurance, and
proficiency testing to more comprehensive, systematic approaches to quality
management. Also included is discussion of the current status of performance
measurement in laboratory medicine.

ƒ

Chapter IV addresses laboratory information systems and automation technology,
including the extent to which they have been adopted into integrated delivery systems
and physician office laboratories. This includes discussion of emerging technologies
(e.g., computerized physician order entry, electronic health records) and health care
system integration requirements.

ƒ

Chapter VII describes federal regulatory oversight of clinical laboratories by the
Centers for Medicare and Medicaid Services (CMS)a and oversight of marketed tests by
the Food and Drug Administration (FDA). The chapter reports on the status of waived
testing, the status of non-waived testing under CLIA (e.g., proficiency testing, quality
control, personnel, and surveys), the respective roles of CMS and FDA in oversight of
laboratory developed tests, and outstanding issues in genetic testing and transfusion
medicine.

ƒ

Chapter VIII describes public and private sector reimbursement systems for clinical
laboratory services, with a focus on coverage decisions, coding and payment
methodologies associated with prospective and fee-for-service systems, and new
initiatives to reduce costs, such as competitive bidding.

Formerly, CMS was the Health Care Financing Administration.

May 2008

13

Laboratory Medicine: A National Status Report

Introduction

REFERENCE LIST
1. Berger D. A brief history of medical diagnosis and the birth of the clinical laboratory.
Part 1—ancient times through the 19th century. MLO Med Lab Obs 1999;31(7):28-40.
2. Lindberg DS, Britt MS, Fisher FW. Williams' introduction to the profession of medical
technology. 4th ed. Philadelphia, PA: Lea & Febiger, 1984.
3. Delwiche FA. Mapping the literature of clinical laboratory science. Journal of the Medical
Library Association 2003;91(3):303-10.
4. Clinical Laboratory Improvement Amendments of 1988. 42 U.S. Code 201. Public law 100-578.
5. Institute of Medicine. Crossing the quality chasm: a new health system for the 21st century.
Washington, DC: National Academy Press, 2001.
6. Institute of Medicine. To err is human: building a safer health system. Washington, DC:
National Academy Press, 2000.
7. McGlynn EA, Asch SM, Adams J, et al. The quality of health care delivered to adults in the
United States. N Engl J Med 2003;348(26):2635-45.
8. Leape LL, Berwick DM. Safe health care: are we up to it? BMJ 2000;320(7237):725-6.
9. Boone DJ, Steindel SJ. Conducting outcomes research: past experience and future directions.
Clin Chem 1995;41(5):795-8.

May 2008

14

Laboratory Medicine: A National Status Report

Methods and Limitations

METHODS AND LIMITATIONS
The process for development, organization, and management of the report content is
summarized in this section. Also briefly discussed are certain limitations of available data and
information used in the report.

METHODS
Report Outline
A kick-off meeting was held in October 2006 in Atlanta to review the purposes and scope of this
report and other tasks associated with the broader contract. CDC, Battelle, and invited experts
provided background information on key aspects and issues in laboratory medicine to be
considered for inclusion in the status report. Drawing from the meeting, Lewin drafted an outline
of the structure and content of the report. The outline was circulated to the representatives of
laboratory medicine stakeholder groups for review and comment. Suggested modifications were
considered and incorporated as appropriate. As Lewin initiated the report, assembled information,
and gained further insights from experts and other stakeholders, the scope of the report was refined
further under the guidance of a Technical Experts Committee.
Technical Experts Committee
In January 2007, nine individuals agreed to serve on the multidisciplinary Technical Experts
Committee. The committee members were selected and approved by CDC on the basis of their
experience, interest, and expertise in one or more of the main topic areas addressed by the
report. They functioned as advisors, providing guidance on the development of specific
chapters to ensure that the report content was comprehensive, appropriate, and accurate.
Members of the committee provided input on specific queries and document drafts via
conference calls and electronic communications.
Data Gathering
In preparing this report, Lewin compiled, analyzed, and synthesized secondary data from
multiple sources, including published and unpublished literature, government databases and
reports, market research reports, Internet searches, and personal communications with industry
experts and government officials.

Literature searches
For a broad environmental scan of the field, Lewin conducted searches of published and
unpublished literature using bibliographic databases and web-based search engines. In the peerreviewed journal literature, searches were conducted predominately in the MEDLINE/PubMed
database. This involved examination of qualitative and quantitative literature covering general and
systematic reviews, guidelines, clinical trials, observational studies, and other analyses. For specific
sections of the report, searches also were conducted in the Cochrane Library, National Guideline
Clearinghouse, and Blue Cross Blue Shield Technology Evaluation Center Assessments database.

May 2008

15

Laboratory Medicine: A National Status Report

Methods and Limitations

Government database searches and reports
The Online Survey, Certification, and Reporting (OSCAR) database of CMS provided information
about laboratory certification, accreditation, and proficiency testing. Queries of the OSCAR
database produced the most recent estimates of test volume for waived and non-waived tests by
laboratory setting and numbers of facilities performing waived and non-waived tests. Searches of
publicly available CMS databases, such as those for the Medicare Physician Fee Schedule, Clinical
Laboratory Fee Schedule database, and Part B Extract Summary System Data File, provided
information about payment rates.
Numerous federal government sources, including public notices, regulations, guidances,
conference proceedings, and other reports, yielded other essential information. Resources were
used from the spectrum of Department of Health and Human Services (DHHS) agencies and
advisory committees, including the Agency for Healthcare Research and Quality (AHRQ)a; CDC;
CMS; Clinical Laboratory Improvement Advisory Committee (CLIAC); Health Research and
Services Administration; FDA; DHHS Office of the Inspector General (OIG); Office of the National
Coordinator for Health Information Technology; and the Secretary’s Advisory Committee on
Genetics, Health, and Society (SACGHS). Federal agencies that provided additional useful
information included the Bureau of the Census, Bureau of Labor Statistics, Government
Accountability Office (GAO)b; Office of Management and Budget; Department of Defense (DoD);
Veterans Administration; Department of Justice; Securities and Exchange Commission; and The
White House. Information at the state level was obtained from state departments of health.

Market research reports
Market research reports and newsletters provided key information about market trends,
workforce, technological advances, and other aspects. Particularly relevant were those from
Washington G-2 Reports, including Lab Industry Strategic Outlook: Market Trends and Analysis 2007
and Laboratory Market Leaders Report 2008, which drew from extensive survey data, CMS and other
agency data, financial reports filed with the SEC, Bureau of the Census data, and expert
interviews. Also useful were Washington G-2 Reports newsletters, e.g., the Laboratory Industry
Report, Diagnostic Testing and Technology Report, G-2 Compliance Report, and National Intelligence
Report. Supplying further data on the laboratory medicine market, regulation, new technology,
and related trends were Knowledge Source Inc.’s Clinical Laboratory Testing Market Overview 2006
and Kalorama Information’s Molecular Diagnostics: Major World Markets (2007).

Internet searches
Along with literature sources, Lewin used Internet search engines to gather web-based
information on particular topics. This included information posted by public and private sector
organizations involved in the laboratory medicine and health care community, such as
government agencies, accreditation organizations, professional societies, industry associations,
standards organizations, research organizations, academic institutions, medical centers,
manufacturers, and international public policy organizations. Resources from these organizations
were evidence-based where applicable, current, and relevant to topics addressed in the report.

a
b

AHRQ was formerly the Agency for Health Care Policy and Research.
GAO was formerly the General Accounting Office.

May 2008

16

Laboratory Medicine: A National Status Report

Methods and Limitations

Personal communications
For clarification of important concepts or to gather information where literature or data sets were
lacking, Lewin relied on personal communications with experts in industry, professional groups,
government agencies, and other public and private sector organizations.

LIMITATIONS
Our extensive search of the sources described above did encounter certain gaps and other
limitations. First, publicly available information about the economic status and quality of the
laboratory medicine sector is patchy, inconsistent, and of uneven quality. Although the OSCAR
database contains the most extensive set of industry data, there is no standardized mechanism for
estimating such important parameters as volume for panel tests. The lack of standardization,
coupled with self-reported, irregular data collection by laboratories, results in gaps and
inconsistencies in estimates of test volumes, spending on services, and market revenue. This
limits determinations of long-term trends and market value for laboratory testing and services.
In addition, the inability of those outside CMS to directly query the OSCAR database further
complicated the research process. In terms of the market research reports, data from surveys was
limited due to low response rates. For example, one of the surveys conducted for the 2007 report
was sent to 12,000 laboratories in their database but only 141 responded. Such data could benefit
from efforts to obtain higher response rates.
Second, lack of data standardization confounds comparability of published study findings
regarding laboratory quality and error rates. Formal measurement and reporting on quality has
been limited largely to analytic-related processes. Data collection in studies examining preanalytic
and postanalytic factors has been insufficiently standardized across participating providers and
laboratories. Many of the quality indicators used for the studies (e.g., specimen
labeling/identification) have not been adequately validated. This contributes to wide ranges in
estimates of errors, and generally inconsistent quality measurement across these institutions.
Third, understanding of the current status of laboratory medicine is constrained by the lack of
research on many important aspects of the field. For example, there are very few studies of
laboratory practice outside the hospital setting (e.g., physician office laboratories), although the
majority of clinical care takes place in the ambulatory care setting. There is a notable lack of
research on the needs of specific populations, such as pediatric patients, frail elderly patients, and
those with multiple chronic conditions, as most published studies involve the non-elderly adult
population and typically focus on a single health issue, e.g., diabetes. Research also is
underdeveloped on the relationship of laboratory services to patient outcomes and cost of care.
Fortunately, there is growing recognition of the importance of such research. Lastly, there is a
notable lack of reliable data on the relationships among historical costs on which payment is based,
costs of providing laboratory testing, what constitutes payment adequacy, and the effects of
economies of scale and other cost-reducing effects of technological changes. These limitations
curtail understanding of important aspects of laboratory medicine related to the quality of services
in physician office laboratories, quality of care for specific patient groups, trends in population
health, and the efficiency and value of laboratory services within the broader health care system.

May 2008

17

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

CHAPTER I
THE VALUE OF LABORATORY MEDICINE TO HEALTH CARE
Laboratory medicine is an essential element of the health care system. It is integral to many clinical
decisions, providing physicians, nurses, and other health care providers with often pivotal
information for prevention, diagnosis, treatment, and management of disease.1 Laboratory tests
and services supply clinicians with information necessary to provide high quality, safe, effective,
and appropriate care to patients. The key role of laboratory testing is reflected in evidence-based
medicine (EBM) and clinical practice guidelines. Health care providers, quality assurance
organizations, and payers are incorporating selected laboratory tests into indicators to objectively
assess quality of care for individual patients and populations and to support payment policies.
Laboratory medicine has an essential role in risk management. Not only can testing help to
prevent certain adverse events, it can facilitate detection and recovery from adverse health events
when they do occur. Laboratory tests help to prevent infectious agents from getting into the
blood supply and ensure the safety of organs and tissues for transplant. Through effective and
timely surveillance, tests can help to mitigate threats to patient and population health (e.g.,
influenza, nosocomial infections, severe acute respiratory syndrome).
The contributions of laboratory tests and services as an essential component and partner in health
systems remain under-recognized. Despite the extensive role of laboratory medicine in informing
medical decision-making, spending on laboratory services accounts for only 2.3% of national health
care spending and 2% of Medicare expenditures.1, 2 As overall expenditures on health care continue
to rise, appropriate use of laboratory tests can facilitate cost-effective care via early detection and
improved management of priority health conditions. Recent and emerging technological advances
gained from mapping the human genome, including applications of genetic testing that enable
personalized medicine, call greater attention to the contributions of laboratory medicine to patient
care as well as the scientific and medical knowledge base. This chapter summarizes the value of
laboratory medicine to health care and patient and population health.

VALUE TO THE QUALITY OF PATIENT CARE
Despite the world’s highest per capita spending on health care, there is overwhelming evidence
in the U.S. of gaps between well-founded standards of care and its actual delivery. Among the
main factors often cited to explain deficiencies in quality are:

ƒ

Growing complexity of science and technology, some of which has advanced more
rapidly than our ability to integrate it into safe, effective, and efficient health care

ƒ

Longer life expectancy, which has significantly increased the chronic disease burden
and the resources being devoted to chronic disease care

ƒ

A highly decentralized health care system that is often bureaucratic, wasteful, and
difficult to navigate

ƒ

Underinvestment and disparities in access to health information technology,
constraining the ability of technology to improve the quality of care3

May 2008

19

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

Health care stakeholders concur that the quality of care delivered in the U.S. is inadequate and that
its organization and delivery must be fundamentally changed. The IOM reports, To Err is Human:
Building a Safer Health System (1999) and Crossing the Quality Chasm: A New Health System for the 21st
Century (2001), and other sentinel studies such as the RAND report on the Quality of Health Care
Delivered to Adults in the U.S. (2003) have helped to mobilize national action on prevention of
medical errors and quality improvement.3-6
Public and private sector efforts to redesign and improve the health care delivery system are
grounded in the six aims of quality identified by the IOM: safety, effectiveness, patientcenteredness, timeliness, efficiency, and equity.5 Using these dimensions of quality, health care
organizations, professional groups, private and public purchasers, and others are developing
specific policies, practices, and measures for each element, with the overarching goals of
diminishing illness, injury, and disability as well as increasing the health and function of the
U.S. population. Examples of how laboratory medicine supports each of the six aims are
provided below.

a

ƒ

Safety refers to protection of patients from harm due to care that is intended to help
them and protection of health care workers from harm while providing care.3
Laboratory medicine contributes to diminishing the risk of harm when patients and
specimens are accurately identified, specimens are collected appropriately, measures
are taken to prevent specimen contamination, process control measures are executed
during analytic processes, and test results are complete and understandable.7

ƒ

Effectiveness refers to measures of how well health care interventions (screening,
diagnosis, treatment, etc.) achieve their intended outcomes or other impacts.3 Laboratory
medicine supports effectiveness when test ordering is evidence-based, specimen collection
follows science-based procedures, specimen analysis and results reporting conform to
well-established standards, and testing results in improved patient outcomes.7

ƒ

Patient-centered care is respectful of and responsive to individual patient values,
preferences, and expressed needs, and ensures that patient values guide decision
making.8 Laboratory medicine supports patient-centered care when test ordering
reflects patient preferences, including end-of-life care, specimen collection is designed
for patient comfort and satisfaction, and test results are understandable to and
actionable by the patient and clinician. These attributes can contribute to favorable
patient experience of the health system and quality of care.

ƒ

Timeliness of care minimizes, unnecessary delays that can result in emotional or
physical harm.3, 9 Timely transport of specimens, decreased turnaround times (TATs)
in routine and stata testing, and timely notification of critical or abnormal values are
primary ways that laboratories support quality of care.7, 10

ƒ

Efficiency refers to using resources to optimize production of desired results.3
Laboratory medicine contributes to health care efficiency when waste is eliminated or
reduced, including that associated with inappropriate test ordering (e.g., underuse,

Stat testing refers to laboratory tests for which results are needed as quickly as possible and which are given
priority by laboratories.

May 2008

20

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

overuse, and misuse), redraws or recollection of specimens, repeating specimen
analysis, and correcting inaccurately documented test results.7

ƒ

Equity of care ensures that quality does not vary because of patient personal
characteristics such as sex, race/ethnicity, geographic location, or socioeconomic
status.3 Laboratories contribute to equitable care when they provide services in a
manner that is unbiased, accommodate the special needs of patients during specimen
collection, use reference intervals that account for population differences, and present
information according to the language and literacy level of the patient.7

Figure 1.1 portrays the dynamic role of laboratory medicine in the health care system. This
diagram has as its premise that value can be expressed in terms of achieving the six aims posed by
the IOM.
Laboratory indicators also provide a means to assess quality of care. Test use as indicated by
clinical practice guidelines and related protocols, as well as test results themselves, are being
incorporated into indicators of provider performance and health care quality. Clinical
laboratories facilitate collection, analysis, and interpretation of data at the individual and
population levels that are necessary for ensuring the public health, such as monitoring rates of
nosocomial infections, development of drug resistance, infectious disease outbreaks, and
biological and chemical threats.
Figure 1.1: Value of Laboratory Medicine to the Health Care System
Six Aims of Quality (IOM)
Safe

Effective

Efficient

Patient-centered

Timely

Biomedical and Health Services Research
Innovation

Clinical Evidence

Economic Evidence

Evidence-based Medicine

Clinical Practice Guidelines

Prevention &
Screening

Diagnosis

Risk & Predictive
Assessment

Prognosis

Monitoring & Management

Intermediate
Outcomes

Outcomes

Laboratory Medicine
Public Health Surveillance

Performance Measurement & Quality Improvement

May 2008

21

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

VALUE TO EVIDENCE-BASED MEDICINE
Though its antecedents extend to the mid-19th century, the widespread practice of EBM and its
broader, yet not universal, acceptance in the U.S. is a more recent development spanning the past
two decades. EBM is commonly defined as the explicit use of best evidence in decision-making
about the medical care of individual patients.11, 12 EBM derives from thorough, well-founded
methods and resources for generating evidence, including the use of randomized controlled trials
and other rigorous study designs as appropriate; use of meta-analyses, systematic reviews, and
other structured approaches for integrating evidence from multiple sources; and standardized
reporting of research results.13 In general, EBM is based on five main principles:

ƒ

Decisions about health should be based on the best patient-, population-, and
laboratory-based evidence

ƒ

The problem determines the most appropriate source of evidence

ƒ

The best evidence is identified by integrating epidemiological, biostatistical, and
pathophysiological methods with personal experience

ƒ

The value of searching for and appraising evidence is derived from translating
evidence into actions that affect patients

ƒ

The ways in which these principles are carried out and performed must be
continuously evaluated14

Laboratory testing plays a major role in supporting EBM in clinical practice. As described in
greater detail below, EBM increasingly informs the development of clinical practice guidelines.13
The hemoglobin A1c (HbA1c) test, which measures the amount of glucose bound to hemoglobin,
allows clinicians to monitor the average amount of glucose in a patient’s blood over the previous
two to three months.15 Based on results of HbA1c testing in clinical trials such as the Diabetes
Control and Complications Trial, a large randomized controlled clinical trial that involved more
than 1,400 type 1 diabetic patients, researchers were able to assess glucose levels and determine
optimal treatment protocols for individuals with this condition.16

VALUE TO CLINICAL PRACTICE GUIDELINES FOR PATIENT CARE
Clinical practice guidelines are systematically-developed statements intended to assist
practitioners and patients in making decisions about health care in specific clinical and personal
circumstances.17 While clinicians have long used such means as treatment recommendations,
immunization schedules, practice bulletins, and algorithms to inform decision-making, clinical
practice guidelines focus on summarizing research and external evidence to develop
recommendations.18 This more rigorous approach typically involves a multidisciplinary team
that reviews some systematic compilation of relevant evidence, working according to explicitly
described methods.19, 20
Among the factors underlying variation in clinical practice are overuse, underuse, and misuse
of health care interventions.13, 21 Clinical practice guidelines are intended to reduce this
variation and improve the quality, safety, efficiency, and effectiveness of clinical care.
Guidelines have been developed for nearly all facets of medical care, spanning prevention,

May 2008

22

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

surveillance, diagnosis, treatment, monitoring, rehabilitation, and palliation; and applying to
drugs, devices, procedures, and systems.13, 19
Quality indicators increasingly are incorporating laboratory testing. For example, 102 (23%) of the
439 quality indicators assessed in the RAND analysis of the quality of care delivered to U.S. adults
involved diagnostic tests.4 The 2006 National Healthcare Quality Report from AHRQ includes eight
clinical conditions, seven of which involve laboratory testing: end-stage renal disease, colorectal
cancer, diabetes, human immunodeficiency virus/acquired immune deficiency syndrome
(HIV/AIDS), mental health/substance abuse, heart disease, and pneumonia.22 In a study
conducted by The Lewin Group in 2005, a search of the National Guideline Clearinghouse and
MEDLINE was undertaken to estimate the extent to which laboratory tests were included as part
of evidence-based clinical practice guidelines across the 23 main condition/disease categories
defined by the National Guideline Clearinghouse. Of 1,230 guidelines identified, 460 (37%)
focused on or involved laboratory tests.
Table 1.1 outlines priority health conditions identified in these sources and corresponding
estimates of the prevalence and spending on each. For each health condition, common laboratory
tests used in screening, diagnosis, and/or monitoring are shown.
Table 1.1: Sample of Priority Health Conditions, Associated Laboratory Tests,
Prevalence, and Cost to the Health System

Health condition

Examples of laboratory tests
used in diagnosis and/or
patient management

Number of Americans
Affected

Spending on Condition
$403 billion (2006)b

Heart disease

Lipid panel, troponin

Respiratory diseaseI

Blood gas test, bacterial culture,
viral culture

15.7 millionII (asthma); 1.3
millionIII (pneumonia)c,d

$144.2 billion (2006)b

Cervical cancer

Pap smear, human papillomavirus
DNA testing

11,150 cervical cancer diagnoses
(2007)e

$1.7 billion (2004)

Colorectal cancer

Fecal-occult blood test

112,340 colon cancer diagnoses
(2007)e

$8.4 billion (2004)f

Diabetes

Glucose, HbA1c

20.8 million (2005)g

$132 billion (2002)g

End-stage renal disease

Creatinine, BUN

HIV/AIDS

Antibody testing, CD4 testing, RNA

Maternal health

79.4 million (2004)

a

472,000 (2004)

h

$32.5 billion (2004)h

1.2 million (2006)i

$21.1 billion (2006)i

Blood and Rh type with antibody
screen

83.9% pregnant women began
prenatal care in first trimester;
3.6% began prenatal care in third
trimester or not at all (2004)j

$26.2 billion (2005)k

Drug tests, liver function

24.6 million adultsIV (classified
with serious psychological
distress) (2005); 22.2 million
peopleV (classified with
substance dependence or abuse)
(2005)l

$104 billion (2001)m

Influenza

Viral culture, serology, rapid
antigen testing

5-20% of the U.S. population is
infected with the influenza virus
each yearn

$200 on treatment per
infected person (2003)o

Health care-associated
infections

Viral culture, molecular typing of
microbial pathogens

1.7 million (2006)p

$10,500-$111,000 per case
(2004)p

(prenatal care)

Mental health/
substance abuse

Based on review of the National Guideline Clearinghouse, Agency for Healthcare Research and Quality, and Medline, National
Library of Medicine. Tests identified from Lab Tests Online.

May 2008

23

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

I

Includes, but is not limited to, severe asthma and pneumonia

II

Indicates number of non-institutionalized adults

III

Indicates number of discharged patients

IV
V

Includes people aged 12 and older

Includes people aged 18 and older

________________________________
a

Rosamond W, Flegal K, Friday G, et al. Heart disease and stroke statistics--2007 update: a report from the American Heart
Association Statistics Committee and Stroke Statistics Subcommittee. Circulation 2007;115(5):e69-171.
b

Fact book fiscal year 2005. Bethesda, MD: National Heart, Lung, and Blood Institute, National Institutes of Health, 2006.

c

Faststats: asthma. Hyattsville, MD: National Center for Health Statistics, 2007.

d

Fastats: pneumonia. Hyattsville, MD: National Center for Health Statistics, 2007.

e

Cancer facts and figures 2007. Atlanta, GA: American Cancer Society, 2007.
f
Cancer trends progress report--2005 update. Bethesda, MD: National Cancer Institute, National Institutes of Health, 2005.
g

National diabetes statistics fact sheet: general information and national estimates on diabetes in the United States, 2005.
Bethesda, MD: National Diabetes Information Clearinghouse, National Institute of Diabetes and Digestive and Kidney Disease,
National Institutes of Health, 2005.
h

U.S. Renal Data System, USRDS 2006 annual data report: atlas of end-stage renal disease in the United States. Bethesda, MD:
National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 2006.

i

HIV/AIDS policy fact sheet. Menlo Park, CA: Kaiser Family Foundation, 2006.

j

Health, United States, 2006 with chartbook on trends in the health of Americans. Hyattsville, MD: National Center for Health
Statistics, 2006.

k

Institute of Medicine. Preterm birth: causes, consequences, and prevention. Washington, DC: National Academy Press, 2006.

l

Results from the 2005 National Survey on Drug Use and Health: national findings. Rockville, MD: Substance Abuse and Mental
health Services Administration, 2006.

m

Mark T, Coffey RM, McKusick T, et al. National expenditures for mental health services and substance abuse treatment, 19912001. Rockville, MD: S Center for Substance Abuse Treatment, Center for Mental Health Services, Substance Abuse and Mental
Health Services Administration, 2005.
n

Key facts about influenza and the influenza vaccine. Atlanta, GA: Centers for Disease Control and Prevention, 2007.

o

Soni A, Hill SC. Average annual health care use and expenses for influenza, 2001-2003. Statistical brief #116. Rockville, MD:
Agency for Healthcare Research and Quality, 2006.
p
Statement by Denise Cardo, MD, Director, Division of Healthcare Quality Promotion, National Center for Infectious Diseases, Centers
for Disease Control and Prevention, on CDC’s role in monitoring and preventing healthcare-associated infections before the
Subcommittee on Oversight and Investigations of the House Committee on Energy and Commerce, 109th Congress, 2nd session. 2006.

VALUE OF LABORATORY TESTS AND SERVICES ACROSS PATIENT CARE
CONTINUUM
Laboratory medicine provides value across the continuum of patient care. In addition to
providing objective data about patient health, laboratory medicine enables early assessment of
disease risk, use of preventive and less invasive treatment, selection of appropriate treatment, and
monitoring treatment. Used appropriately to inform patient management decisions, laboratory
testing can contribute to optimizing use of health care resources and decrease short-, medium-,
and long-term costs of care.23

Screening for Risk Factors of Developing Specific Disorders
Screening tests may be conducted on asymptomatic individuals to check for risk factors and other
indicators of developing or latent disease.24 Especially for children and young adults, such testing
can avoid or diminish the impact of diseases and medical conditions that appear later in life.
Screening tests for adults can detect certain common diseases that, when identified early, can be
more easily treated.25, 26

May 2008

24

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

Access to preventive and screening laboratory testing services in the U.S. varies. Although
Medicare’s authorizing legislation (Title XVIII of the Social Security Act) excludes coverage for
preventive interventions and diagnostic laboratory tests for asymptomatic individuals, Congress
has mandated Medicare coverage of selected screening and diagnostic procedures over the years.
Pursuant to the Medicare Prescription Drug, Improvement, and Modernization Act of 2003 (MMA),
preventive benefits provided by Medicare effective in 2005 include a “Welcome to Medicare”
physical exam and screening for heart disease and diabetes, along with earlier mandated tests for
osteoporosis, glaucoma, and cancers of the colon, breast, cervix, and prostate.27, 28
The disparities between insured and uninsured people in the U.S. in access to care extend to
laboratory testing. Compared to adults with any type of health insurance coverage, uninsured
adults in the U.S. are less likely to receive preventive and screening services at all and are less
likely to receive these services on a timely basis.29 People in the U.S. who are uninsured are more
likely to report duplication of laboratory tests, laboratory test errors, and delays in receiving
laboratory test results.30

Screening to Determine Individual Risk
Genetic testing involves analysis of human deoxyribonucleic acid (DNA), ribonucleic acid
(RNA), chromosomes, proteins, or certain metabolites to detect genetic alterations related to a
heritable medical disorder.31 A universal definition of genetic testing has not been recognized
by regulatory or professional bodies. SACGHS defines a genetic test as one that involves the
analysis of chromosomes, DNA, ribonucleic acid, genes, or gene products to detect heritable or
somatic variations related to genes or health.32 In its draft 2007 report on oversight of genetic
testing, SACGHS calls on relevant agencies to develop an appropriate definition of healthrelated genetic tests.
Genetic testing can be used to determine whether an individual is a carrier for a disease or
condition or has a heightened risk of developing a disease years or decades later.33 It also can be
used for diagnostic or prognostic purposes and as a predictive tool to assess an individual’s drug
metabolism. The main types of genetic testing include the following:

ƒ

Carrier identification is used to determine whether an individual possesses a potentially
harmful gene that can be passed on to progeny. Prenatal/maternal serum screening is
used to determine whether a fetus is at risk of having specific genetic conditions and to
detect open neural tube defects and certain chromosomal abnormalities. Current
prenatal diagnosis is targeted at specific diseases and/or mutations rather than
determining the general genetic make-up of the fetus.

ƒ

Newborn screening is most often used to determine whether a newborn has a medical
condition that requires immediate treatment, e.g., phenylketonuria, congenital
hypothyroidism.

ƒ

Late-onset disorder testing is used to determine an adult’s susceptibility or
predisposition to complex diseases (e.g., cancer, heart disease, Huntington’s disease).

The increasing role of genetic testing is due in great measure to advances in understanding the
role of genetics and molecular biology in disease development. Genetic tests are now available
for more than 1,400 diseases.34 Tests for an estimated 287 of these diseases are being used only in
research settings. Many diseases are known or thought to be caused by inherited or spontaneous
May 2008

25

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

acquired DNA alterations, e.g., Down syndrome, Huntington disease, sickle cell anemia, and
hemophilia, and thought to cause or contribute to many others, including Alzheimer’s disease,
breast cancer, leukemia, and osteoarthritis.35 Genetic testing for early-onset Alzheimer’s disease
detects mutations in three single genes that are understood to lead to the development of the
disease at an early age, typically before the age of 60.36 Breast cancer (BRCA) gene analysis, which
is often performed by sequencing the BRCA1 and BRCA2 genes, identifies mutations in these two
genes that are associated with predisposition to breast and ovarian cancer. BRCA1 and BRCA2
mutations have been identified in 1-2% of women diagnosed with breast cancer and 5-10% of
women diagnosed with ovarian cancer.37

Diagnosing Conditions and Evaluating Prognosis
Early detection
Laboratory tests are critically important for accurately diagnosing a disease in its earliest stages,
determining disease severity, assessing the likelihood of recovery, and evaluating the potential for
adverse outcomes. Accurate and early diagnosis allows clinicians and patients to better evaluate
the benefits and risks of various treatment options, begin treatment promptly and, in the case of
contagious conditions, prevent a disease from spreading to others. Laboratory tests are used by
clinicians and, increasingly, patients, to inform prevention and treatment decisions and related
courses of action.
Early-stage detection via laboratory testing is established for such diseases and conditions as
breast cancer, cervical cancer, skin cancer, thyroid dysfunction, high cholesterol, and diabetes.
Using the Papanicolaou test (Pap test or Pap smear), a pathologist or cytotechnologist can identify
cervical cells that are cancerous or potentially pre-cancerous.38 The American Cancer Society, U.S.
Preventive Services Task Force (USPSTF), and American College of Obstetricians and
Gynecologists recommend that young women receive Pap tests every year beginning no later
than age 21; women over the age of 30 who have no new risk factors and who have had normal
results for three consecutive years are advised to get retested every two-to-three years. Broad use
of the Pap test as a screening tool to detect pre-invasive lesions is credited with reduction in the
annual incidence rate (from 14.8 to 7.0 per 100,000) and mortality rate (from 5.6 to 2.4 per 100,000)
of malignant cervical cancer in the U.S. between 1975 to 2004.39-41
When diseases are identified at an early stage or before symptoms have appeared, patients and
their health care providers can take measures to prevent or reduce the risk of developing the
disease or condition, including increased medical monitoring, lifestyle changes and, when
needed, medical interventions.37 Similarly, early measures may minimize the severity of the
disease and its effects on mortality, morbidity, and quality of life. These measures also can
diminish downstream health care spending that would have been caused by the disease. For
instance, early detection of incipient colorectal cancer (e.g., using fecal-occult blood testing) is
associated with more successful treatment and increased survival.42

Diagnosis
Along with an individual’s signs, symptoms, personal history, and family history, laboratory tests
are used to arrive at or eliminate possible diagnoses.43 A laboratory test used for “ruling in” a
disease or condition indicates that it may be present if the test results are abnormal. “Ruling out”

May 2008

26

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

a disease or a condition allows the clinician to consider alternative diagnoses and make more
efficient use of resources.
Laboratory tests can be used to determine the degree to which the disease has progressed and the
severity of the disease. Laboratory tests, including studies of blood, urine, other bodily fluids,
and bodily tissues play a major role in the “staging” of cancer and other diseases, i.e., describing
the severity of a disease based on the extent to which it has spread throughout the body.44 Tests
to determine the extent and severity of a disease can be the same or different from those for
diagnosis. For example, measurements of HbA1c and glucose are used to diagnose diabetes and
to monitor diabetic individuals.45
Among other examples, laboratory tests can direct a diagnosis in the case of pharyngitis, or
inflammation of the pharynx, which can be caused by a variety of microorganisms.46 Group A
streptococcus (GAS) is a bacterial cause of pharyngitis and can be treated with antibiotics. Only
about 15% of sore throats are caused by GAS; therefore, the results of a streptococcal screen
provide a clinician with information about the underlying cause of the sore throat and inform
determination of the optimal treatment. This also highlights the importance of accurate
laboratory tests in slowing increases in the prevalence of medication-resistant strains of disease
bacteria.47 While antibiotics have little to no effect on the cause of many upper respiratory
infections (including all viral infections) whose symptoms often resemble those cause by GAS,
U.S. clinicians often prescribe antibiotics for these infections.48 As resistance to antibiotics
commonly used to treat GAS has been documented,49 accurate diagnosis of GAS using laboratory
testing is a key component for preventing inappropriate use of antibiotics. Similarly, laboratories
also conduct antimicrobial susceptibility testing to determine the ability of antimicrobial agents to
inhibit the growth of pathogenic bacteria, thereby helping to optimize treatment and reduce the
risk of antibiotic-resistant organisms.50
Brain natriuretic peptide (BNP) is a peptide involved in the control of cardiovascular function;
plasma concentrations of BNP are increased in individuals with heart failure and BNP levels are
correlated with the severity of the symptoms.51, 52 The BNP assay is used by clinicians to
determine whether or not an individual has heart failure. For example, a 2004 study of patients
presenting in the emergency department with acute dyspnea, or shortness of breath, found that,
when used in conjunction with other clinical information, rapid measurement of BNP to diagnose
heart failure improved patient evaluation and treatment and reduced the total time the patient
spent in the emergency department.51 Study results indicated that 75% of patients in the BNP
group were hospitalized, compared to 85% of the control group. Similarly, 15% in the BNP group
required intensive care, compared to 24% in the control group.

Prognosis
Once a diagnosis is made and the severity of the disease has been determined, laboratory test results
can contribute to projecting the course of disease, including estimating the likelihood that an
individual will recover from a disease or medical condition. For instance, certain abnormal results
in a panel of laboratory tests given to women who are suffering from severe pre-eclampsia are
predictive of high risk of maternal morbidity.53, 54 Routine laboratory tests ordered upon hospital
admission of patients following myocardial infarction that measure white blood cell, creatinine,
glucose, lactate dehydrogenase, and platelet counts are predictive of the likelihood of mortality.55

May 2008

27

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

Individuals who have had an unexplained thrombotic episode (i.e., clinical signs and symptoms
associated with a blood clot in a vein or artery) before they are 50 years old or have had certain
abnormal clots are encouraged to have a molecular diagnostic test to determine whether they
have an inherited gene mutation that puts them at increased risk of developing additional blood
clots.56 The test detects factor V Leiden, a variant form of a factor V, a coagulation factor that is
activated following injury of a blood vessel. Factor V Leiden is the most common inherited
predisposition to abnormal clotting in the U.S. and individuals who are found to carry the
mutation are counseled to avoid risk factors that can lead to additional blood clotting, such as oral
contraceptive use.
Although not yet in routine practice, laboratory tests are increasingly used to determine the
potential for future adverse health outcomes following recovery from a disease, such as recurrent
stroke or cancer relapse. In February 2007, FDA cleared for marketing a test (MammaPrint®) that
determines the likelihood of breast cancer returning within 5-10 years after a woman’s initial
cancer.57 The test evaluates 70 genes located in the tumor to determine whether the patient is at
low or high risk for spread of the cancer to another site. Like other predictive tests, the results
from this and others being developed to assess likelihood of disease relapse are not perfectly
accurate and must be used with other information to support management of the disease.58 A
similar laboratory developed test is the Oncotype DX™ that analyzes the expression of a panel of
21 genes to quantify the likelihood of breast cancer recurrence and potential benefit from
chemotherapy in women with newly diagnosed, early stage invasive breast cancer.59 Clinical
practice guidelines of ASCO and NCCN include indications for using the Oncotype DX™ test,
although these organizations await the findings of prospective clinical trials of this and other gene
expression profile tests for offering more definitive guidance. Major U.S. payers cover the cost of
the test for particular indications, sometimes subject to prior authorization.60, 61

Monitoring General Treatment Effectiveness
Laboratory tests play a large role in monitoring and evaluating the efficacy of other medical
treatments.62 They can assist clinicians in deciding whether to modify a specific course of treatment
in order to optimize outcomes, including maximizing therapeutic impact. For instance, tests to
measure viral load, CD4 count, complete blood count, and blood chemistry tests are commonly
used to assess treatment response in patients with HIV.63 Some laboratory tests used to monitor
treatment effectiveness are the same as those used to make the initial disease diagnosis. A very
common instance of this involves tests to measure thyrotropin/thyroid stimulating hormone in the
diagnosis and monitoring of thyroid disease.64 Laboratory testing is also important in monitoring
patients following surgery to gauge the success and effectiveness of a procedure. Laboratory tests
to detect levels of human chorionic gonadotropin in women with trophoblastic disease, which
involves abnormal growth of cells inside a woman’s uterus, are conducted regularly following
surgery to determine whether or not further treatment is required.65

Managing Acute Health Conditions
Acute conditions are those that appear suddenly and follow a short, severe course (i.e., persisting
for several days or weeks) and have the potential to be completely resolved. Most acute care is
relatively short-term. In acute care settings such as the intensive care unit (ICU) or the emergency
department, frequent laboratory testing can be used to monitor quickly and accurately an
individual’s status and response to medical interventions. The five most commonly ordered

May 2008

28

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

types of laboratory tests in ICUs include basic metabolic panels, arterial blood gas profiles,
complete blood counts, partial thromboplastin times, and measures of magnesium levels.66 The
basic metabolic panel, which measures glucose, calcium, electrolytes, and analytes related to
kidney function, is commonly ordered in hospital emergency rooms because its results can
indicate several acute problems, including kidney failure, insulin shock or diabetic coma,
respiratory distress, or heart rhythm changes.67
Particularly in critical care settings, any intervention that shortens or improves the efficiency of care
can dramatically affect patient outcomes and health care costs.68 There is great emphasis on
providing quick and accurate laboratory test results for individuals in critical care settings. One
commonly used method of achieving short TATs is stat testing, which refers to the sequence of
events to obtain urgently needed test results promptly.69 Many hospitals maintain designated stat
laboratory space to meet urgent testing needs, usually located next to operating rooms, critical care
units, or the emergency department.70 Recent research confirms that rapid analysis of cardiac
markers D-dimer (a marker for patients at risk of a pulmonary embolism or deep vein thrombosis)
and serum protein S100 (a marker of brain damage) improves outcomes.71-73
Laboratory tests for certain critical conditions can be conducted using point-of-care testing
(POCT) technology, which allows clinicians to obtain laboratory results in proximity to the
patient.74 Measurements of glucose and oxygen saturation levels at the point-of-care allows
clinicians to determine changes in a patient’s status rapidly and frequently.75 In critical care
settings, nurses often perform POCT.70 Particularly in the emergency room, POCT has the
potential to expedite decision making and allow for more effective triaging of patients.
While acute care is often provided in hospital settings (e.g., emergency department, ICU) it can
also be provided in ambulatory care settings (e.g., physicians’ offices, other primary care sites).
As in critical care, laboratory testing in non-critical primary care is vital to timely and accurate
diagnosis. For instance, testing for urine leukocyte esterase and nitrites to detect urinary tract
infections, testing for H pylori to detect gastrointestinal disorders, and testing for C-reactive
protein to detect bacterial infection are all commonly used in acute care provided in primary care
settings.76-78 POCT is growing in use for non-critical acute care to provide rapid diagnosis and
rule out other tests.79 Whereas urine specimen culture testing requires 24 hours to complete, urine
dip-stick tests provide a means of rapidly detecting the presence of bacteriuria and urinary tract
infections.80 Specimen culture and antimicrobial resistance susceptibility tests are usually
conducted in a laboratory, while dipstick tests contain specially treated plastic strips that change
color when exposed to infected urine and can be conducted in a doctor’s office or at home. In
many cases, negative urine dipstick tests alone can exclude the presence of infection.

Managing Chronic Health Conditions
Laboratory testing is an essential component in managing chronic diseases—health conditions
that persist over an extended period of time and, in many cases, cannot be completely cured (e.g.,
type I diabetes).81 The burden of chronic disease in the U.S. population is high. More than 90
million Americans currently live with chronic illnesses. About half of older adults have at least
two chronic medical conditions.82 Care for chronic illness accounts for more than three-fourths of
U.S. health care costs.83

May 2008

29

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

Laboratory tests are useful tools for clinicians and patients in understanding the status of the
disease(s), informing treatment decisions, determining the urgency of care required, managing
symptoms, educating patients, and incorporating lifestyle changes into the treatment regimen. In
certain instances, regular laboratory testing can prevent the diseases from progressing or
worsening. For example, patients with coronary artery disease are at high risk for myocardial
infarction. Aggressive lipid management in individuals who have heart disease helps to prevent
heart attacks and reduces mortality rates.84 Regular lipid testing of patients with heart disease
helps physicians to tailor disease management regimens and provides a means to motivate some
patients to implement lifestyle changes. Similarly, laboratory tests are a key component in the
management of chronic kidney disease, which can also lead to high blood pressure, anemia, weak
bones, nerve damage, and progression to kidney failure.85 Laboratory tests that measure
glomerular filtration rate are used to assess the severity of chronic kidney disease and to
determine whether to initiate certain treatments.
Disease management refers to the ongoing care associated with chronic conditions. It is defined as a
proactive, multi-component strategy for delivering health care services that aims to reduce adverse
medical events by maximizing patient’s adherence to prescribed treatments and/or lifestyle
changes.86 An important component of the strategy is self management, with patients being
responsible for day-to-day self-monitoring, decision-making, and healthy lifestyle choices.87, 88
Diabetes treatment has been used often as the focus of models for disease management, which
traditionally involves a partnership between physicians, nurses, educators, and the patient.
Physicians monitor patient blood glucose and HbA1c levels during regular office visits and track
trends over the previous two-to-three months. Frequent, daily self-monitoring of blood glucose
levels also is critically important to determine medication and/or dietary changes needed.45
Individuals usually self monitor by using a lancet device to obtain a small blood sample that is then
applied to a reagent strip and inserted into a reflectance photometer for an automated reading of
blood glucose levels.89 Patient education is needed for successful disease management at home.90
Newer technologies support continuous glucose monitoring by measuring interstitial fluid using
minimally invasive methods or by applying noninvasive electromagnetic radiation through the skin
to the body’s blood vessels.91 Continuous monitoring provides clinicians and patients with greater
insight into glucose levels throughout the day than does conventional self-monitoring, thereby
helping to identify and ultimately prevent periods of hypo- and hyperglycemia.

Therapeutic Drug Monitoring
Standard laboratory tests are integral to the management of medication dosages for many
conditions. Therapeutic drug monitoring (TDM) refers to the measurement of specific drugs or
metabolites in the body via blood testing to inform therapeutic regimens that maintain a target
medication concentration in the body.92 Maintaining an appropriate dosage is particularly
important for medications with a narrow therapeutic index, i.e., drugs with smaller dosage ranges
for optimum effectiveness. Medications with narrow therapeutic indexes, such as those used to
treat cardiovascular, kidney, thyroid, and liver disease, typically require precise dosage
modifications to fit the needs of an individual patient as well as close monitoring. Drugs that
have a wider therapeutic index, such as antihypertensives and antibiotics, can usually be
prescribed based on pre-established dosing schedules.

May 2008

30

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

Laboratory tests associated with TDM are instrumental in establishing and maintaining the
medication dosage that will yield the optimum blood level range for a specific individual.92
Calculations are based on optimum therapeutic ranges developed through research and clinical
testing of narrow therapeutic index medications in addition to individual patient testing. TDM
plays an especially vital role in ensuring that treatments are fully effective and that the
individual does not experience any toxicity as a result of treatment. TDM can also be used to
evaluate the extent to which an individual is compliant with or adherent to a clinician’s
prescribed course of treatment.
Most TDM takes place in hospitals or other inpatient settings; however, newer tests are allowing
TDM to be conducted in other settings, such as clinics, physicians’ offices, and at home.93 Certain
medications, such as warfarin, also can be monitored at home between physician’s visits with
portable monitoring devices similar to those used by diabetics to monitor glucose.94 TDM
conducted on-site in physicians’ offices has the benefit of enabling clinicians to adjust therapeutic
drug doses while the patient is still present in the office.95 On-site analyzers enabling TDM in
physicians’ office laboratories (POLs) are available for measuring concentrations of
anticonvulsant and anti-asthmatic medications, among others. POCT available for POL-based
TDM allows clinicians to obtain laboratory results more quickly and cost-effectively than when
specimens are sent to a laboratory. Immunosensor applications that use antibodies to detect drug
concentrations can be plugged into handheld personal digital assistants to allow clinicians to
obtain TDM results in 30 minutes or less in both hospital and outpatient settings.96

Detection and Prevention of Medication Error
Laboratory testing also provides a means to prevent and detect medication errors,b i.e., any error
occurring in the medication-use process, such as wrong dosages prescribed, wrong dosage
administered, failure to administer a medication by the provider, or patient failure to take the
medication as prescribed.100 Computerized physician/practitioner order entry systems, which
allow users to order medications electronically, can compare the medication orders to major
elements of the patient’s history, including laboratory results. Clinicians are alerted when
medication orders conflict with a specific element in the patient’s history.101 Increased networking
of computerized ordering systems, pharmacy information systems, and laboratory information
systems should further increase the ability of laboratory medicine to aid in the prevention of
medication errors. Review of laboratory test results documented in a patient’s medical record
aids in the detection and identification of medication errors.100, 102

b

An adverse drug reaction (ADR) is any adverse effect resulting from the use of a medication in the recommended
manner. An adverse drug event is any injury due to medical harm, including physical harm (e.g., rash), mental
harm (e.g., confusion), or loss of function (e.g., inability to drive a car), whether used in the recommended
manner or not. ADRs and events may be preventable or nonpreventable. A medication error is any error occurring
throughout the medication-use process, including events resulting from use of a medication in both
recommended and non-recommended manners. An error is the failure of a planned action to be completed (i.e.,
error of execution), or the use of a wrong plan to achieve an aim (i.e., error of planning), and may be an act of
commission or omission.97-99

May 2008

31

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

Individualizing Drug Treatments and Reducing Adverse Drug Reactions
As scientists study the human genome and gain understanding of the genetic, behavioral, and
environmental determinants of disease and therapeutic response, laboratory tests can be leveraged
to individualize treatment protocols based on individual patient traits. Pharmacogenomics (PGx)c
uses information from the human genome to understand the spectrum of genes involved in drug
response and studies the relationship between gene-based markers and pharmacology.104, 105 By
assessing factors such as hormone levels and gene expressions that can vary among individuals,
PGx allows clinicians to better understand how an individual is likely to respond to a specific
treatment or therapy and, thereby, to tailor treatment most appropriately. PGx is a key element of
the broader concept of personalized health care.
Molecular laboratory tests provide information used to apply PGx. Tests for estrogen and
progesterone receptors on the surface of cells from tissue biopsies of women with breast cancer can
determine whether the growth of these cancers can be stopped by therapeutic molecules that block
these hormones.106 Trastuzumab (Herceptin), a monoclonal antibody used in the treatment of
breast cancer, is considered to be effective only in women whose breast cells express the HER2/neu
protein, the presence of which is determined by laboratory tests.107 The efficacy of certain drugs for
treating HIV infection is mediated by specific genetic polymorphisms; individuals expressing a
single-nucleotide polymorphism on the MDR1 gene respond more favorably to anti-retroviral
agents.108 Response to warfarin, a commonly prescribed drug for individuals at high risk of
developing blood clots, varies; individuals with the *2 and *3 allelic variants of the cytochrome P450
2C9 enzyme clear the drug much slower than individuals without this allelic variation, and
therefore require lower doses.109, 110 A major goal of determining optimal warfarin dosage is to
reduce adverse reactions and to determine the optimal therapeutic dosage.
PGx testing can also be used to identify individuals who are at risk for developing an adverse drug
reaction.97 When therapeutic drugs are indicated, PGx testing can improve appropriate drug
selection and management of medication dosage, as well as reduce the potential for adverse drug
reactions (ADRs).111,112
Several PGx tests are readily available for preventing ADRs. One example is laboratory testing
using DNA microarray assays to identify variants in cytochrome P450, a group of drugmetabolizing enzymes that exists in more than 50 forms and catalyzes the oxidation of many
drugs, including beta blockers, antiarrhythmics, and antidepressants.d,114, 115 Similarly,
administration of 5-fluorouracil, a chemotherapy used in the treatment of various types of cancer,
can lead to potentially life-threatening toxicity in individuals with dihydropyrimidine
dehydrogenase deficiency.116 Laboratory testing can be used to identify individuals with a defect
in the gene that encodes for the dihydropyrimidine dehydrogenase deficiency enzyme, thereby
allowing clinicians to make more appropriate treatment decisions.

Pharmacogenomics is defined as the general study of all of the genes that influence and determine drug behavior.
The term pharmacogenetics refers to the study of particular inherited differences in drug metabolism and
response. However, the two terms are used interchangeably, and this distinction is considered arbitrary.103
d Research into targeted drug treatments is promising. For instance, a 2007 systematic review prepared on behalf of
CDC’s Evaluation of Genomic Applications in Practice and Prevention Project determined that there is a paucity of
good-quality data addressing whether testing for CYP450 polymorphisms in adults who are about to enter treatment
for non-psychotic depression leads to improved outcomes.113
c

May 2008

32

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

Because PGx makes feasible developing laboratory tests capable of identifying patients and
conditions that will be responsive to highly targeted therapeutic agents, clinical laboratories are
expected to play a greater role in the development of this field over the next several years.117 In
particular, clinical laboratories are expected to provide consumers with access to PGx testing and to
provide clinicians with the evidence required to make decisions regarding medical applications.118
As knowledge builds about the utility of genetic information for tailoring drug therapies, the
number of PGx tests is expected to increase. In order for PGx to make a significant contribution to
personalized health care and have an impact on population health, many more molecular biology
tests than are currently available to detect genetic polymorphisms will have to be validated.119, 120
Laboratories will have important roles in studies to identify and verify polymorphisms and clinical
trials of PGx-guided interventions.

PROTECTING THE BLOOD SUPPLY AND TRANSPLANT RECIPIENTS
Blood and blood products are vital health care resources that are required in a large number of
medical procedures. Availability of safe blood and blood products is essential for millions of
people in the U.S., including accident victims, transplant recipients, and patients undergoing a
wide range of surgeries.121 The health care industry relies on blood donations from volunteers in
order to meet this demand. In 2001, the most recent year for which data are available, U.S.
institutions collected more than 15 million units of whole blood and red blood cells,
approximately 14 million units of which were transfused to 4.9 million patients.122
Through their blood banking responsibilities, clinical laboratories work to guarantee the safety of
the blood supply. Blood collection usually takes place at community blood centers, hospitalbased donor centers, or mobile sites temporarily constructed for blood donations.122 Following
donation, the blood is taken to blood banking laboratories and tested to determine blood type and
detect the presence of antibodies, bacterial contamination, and other agents that could potentially
cause adverse reactions in transfusion recipients. Once testing is completed, donated blood that is
free of infection is stored for future use.e
Screening blood donations for the presence of infectious disease is a high-value service provided
by blood banks and clinical laboratories. Laboratories screen donations for the presence of HIV
and hepatitis B and C viruses, the three transfusion-transmitted viruses of greatest concern to
public health.123 Blood donations have been tested for the presence of HIV-1 infection (the strain
of HIV found most commonly in the U.S.) since 1985.124 Until 1999, testing for HIV-1 required
that the donor’s immune system had already mounted a response against the virus, allowing for a
“window period” in which HIV-contaminated blood would be undetected by laboratory testing
for HIV antigens and antibodies. The creation and implementation of nucleic acid testing (NAT)
in 1999 under an FDA-approved Investigational New Drug application (followed by FDA
approval of a commercial assay in 2002) reduced this window period by almost half, from an
average of 22 days to 12 days. Relative to antibody and antigen tests, which reduced the risk of
HIV infection from a single blood transfusion to approximately 1 in 676,000, NAT reduces the risk

e

Storage methods are determined by the component of blood being stored and how quickly the donation will be used.
Red blood cells can be refrigerated for a maximum of 42 days or frozen for up to 10 years; platelets can be stored at
room temperature for up to 5 days; and fresh frozen plasma can be frozen for up to 1 year.122

May 2008

33

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

of transfusion-transmitted HIV-1 to about 1 in 1.9 million blood units.125, 126 Innovations in
laboratory testing have also allowed donated blood to be screened for human T-lymphotropic
virus types I and II, syphilis, West Nile virus, Chagas’ disease, and other infections.122
Clinical laboratories also conduct blood compatibility testing, or hemocompatibility, to determine
whether a particular unit of blood can be safely transfused into an individual.127 Pretranfusion
hemocompatibility testing includes identifying an individual’s ABO blood group and Rh type,
both of which are determined by the presence or absence of specific antigens on red blood cells, as
well as testing blood for unexpected red cell antibodies.122 Cross-matching determines whether
an individual’s blood has antibodies that will react with the donor’s cells. In the event that a
cross-match indicates a reaction, laboratory professionals identify the specific reacting antibodies
and locate alternate donor blood that lacks the antigen.
Compatibility testing, cross-matching, and other precautions taken by clinical laboratories help
prevent adverse transfusion outcomes, which include acute hemolytic transfusion reactions,
febrile nonhemolytic transfusion reactions, allergic reactions, volume overload, and acute lung
injury caused by transfusing incompatible blood.128 ABO incompatibility is the most common
cause of acute hemolytic transfusion reactions, which usually result from antibodies in the
recipient’s plasma reacting to red blood cell antigens in the donor blood. Technologies such as
portable data terminals that scan patient wristbands at the bedside, bar-coded specimen labels,
and electronic transfer of test results to the laboratory can decrease the risk of a mismatch
between blood recipients and donor blood caused by misidentification and are increasingly the
focus of laboratories, hospitals, and accreditation bodies in the U.S.129, 130

DETECTING EXPOSURE TO ILLEGAL OR TOXIC DRUGS
Exposure to illegal drugs, toxic substances, and incorrect use of therapeutic drugs are major causes
of hospital emergency department visits in the U.S. In 2004, the most recent year for which data are
available, an estimated 1.08 million visits to the emergency department were attributed to poisoning
and toxic effects, representing approximately 1% of all visits.131 As many as 23% of emergency
department visits each year may be attributed to the abuse of alcohol and other substances.132 In
addition to alcohol and drugs of abuse, accidental or inadvertent contact with substances such as
organophosphorous compounds (used in pesticides), rodenticides, heavy metals, and carbon
monoxide can also contribute to significant acute and chronic threats to health.133
Rapid, accurate laboratory testing enables clinicians to identify specific toxic substances to which
individuals have been exposed and to determine appropriate medical care, including what
medications can be prescribed and how quickly treatment must be initiated.134 Toxicology
screenings, most often performed on blood or urine samples and sometimes on gastric contents,
allow clinicians to evaluate the type and approximate amount of legal and illegal drugs an
individual has consumed.135
In its 2005 guideline, the National Academy of Clinical Biochemistry (NACB) divides toxicology
screening into two tiers:

ƒ

May 2008

Tier I testing includes stat testing for selected agents in serum, plasma, and urine.

34

Laboratory Medicine: A National Status Report

ƒ

Chapter I – The Value of Laboratory Medicine to Health Care

Tier II testing is for individuals who have been admitted to the hospital who remain
intoxicated or comatose and for whom tier I testing did not identify the nature of the
problem.134

NACB recommends that hospital emergency departments conduct serum toxicology assays to
detect 14 different substances, including lithium, salicylate, valproic acid, digoxin, methyl alcohol,
and iron, and urine toxicology screenings to detect seven substances, including cocaine,
barbiturates, and opiates.
Due to the nature of acute illness and poisoning and limitations in resources and technology, clinical
laboratories are limited in their ability to provide real-time analyses of a full spectrum of
toxicological screens for patients who appear to be impaired or overdosed. This highlights the
importance of TAT for laboratory tests ordered in the emergency department. For a few tests, rapid
bedside toxicological screening assays allow clinicians to obtain laboratory test results in real time at
the point-of-care. Many emergency departments use breath meters for determining alcohol
concentrations in intoxicated patients at the bedside; these are accurate, precise, and relatively
inexpensive.134 Point-of-care immunoassays are also available for a wide variety of drugs of abuse,
including cocaine, methamphetamine, and antidepressants.136 However, many rapid toxicological
screening tests have not yet been validated in the emergency department setting.137 Specifically,
rapid assays for methanol and ethylene glycol poisoning, which can produce significant morbidity
and mortality, are being tested currently but are not yet in wide use.138
In 2002, 4 million adults in the U.S. diagnosed with serious mental illness also had a substance
abuse disorder.139 Because substance abuse can exacerbate psychiatric symptoms, toxicology
screening is especially important in the emergency treatment of serious mental illness.140
Clinicians are often asked to perform laboratory testing on individuals presenting with acute
behavioral emergencies in order to rule out illnesses that may be causing acute psychiatric
symptoms. The results of laboratory testing, particularly toxicology screening, can identify
substance abusers and help guide treatment of psychiatric patients.

VALUE TO THE MEASUREMENT OF QUALITY OF CARE
The IOM defines quality of care as “the degree to which health care services for individuals and
populations increase the likelihood of desired health outcomes and are consistent with current
professional knowledge.”141 Health care quality measures enable health care decision-makers to
compare quality of specific aspects of health care to relevant reference standards or criteria. They
are increasingly used to measure and assess the performance of the U.S. health care system.142
Quality can be measured in three main dimensions:

ƒ

Structure refers to the environmental factors that support the capacity to achieve quality

ƒ

Process refers to what is done to and for the patient

ƒ

Outcomes refers to changes in patients’ health status as a result of care received143

Most health care quality literature focuses on process of care measures, examining the
appropriateness of the care provided and the adherence of providers to professional standards.144

May 2008

35

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

Laboratories monitor quality in order to ensure that they are providing information that is timely,
accurate, appropriate, and interpretable, and ensures high quality care.145 Quality indicators
within laboratory medicine can also be classified under structure (e.g., staffing benchmarks of
productivity, policies for documenting safe phlebotomy practices), process (e.g., patient
identification errors, analytic accuracy, follow up with clinicians regarding results), or outcomes
(e.g., health outcomes, customer satisfaction, cost-effectiveness).7, 146, 147

Quality Measurement for Priority Health Conditions
Data derived from laboratory testing is a key component in many of the current quality measures,
particularly those that assess health outcomes, the quality measures most relevant to consumers and
many health care plan purchasers.148, 149 Laboratory-based performance is assessed with process
and outcome measures developed for such highly prevalent and/or burdensome conditions as
diabetes, heart failure, ischemic heart disease, stroke, end-stage renal disease, pneumonia, cervical
and colon cancer, and pregnancy and childbirth.7 For instance, the American Quality Alliance
explicitly includes the use of laboratory tests to measure HbA1c and low density lipoprotein
cholesterol as a performance measure to assess the quality of diabetes care.150
Two national sources of validated quality measures include the Health Plan Employer Data and
Information Set (HEDIS), maintained by the National Committee for Quality Assurance (NCQA)
and the National Quality Measures Clearinghouse (NQMC), maintained by AHRQ.151, 152 HEDIS
data can be used by purchasers and patients to make side-by-side comparisons of health care
plans and by health plans to evaluate their own performance. Approximately 90% of managed
care organizations use HEDIS measures to assess provider performance. CMS also requires plans
participating in Medicare programs to report on HEDIS measures.153 The NQMC database
provides information on evidence-based health care quality measures linked to a particular
disease or condition and a particular treatment or intervention. The information can be used by
practitioners, providers, payers, and purchasers to inform health care decisions. A search of the
HEDIS and NQMC reported in 2005 found that 23% of HEDIS measures and 14% of NQMC
measures used to assess the quality of care given by a specific provider or health plan are direct
measures of laboratory test use for measuring health risks, diseases, or medical conditions.154
Performance measurement, including in laboratory testing, can have a direct impact on delivery,
quality, and cost of health care and on the establishment of best practices. According to NCQA, in
2005, more than 70 million Americans benefited from health care improvements facilitated by
quality measurement. Individuals enrolled in health plans that use and publicly report
performance data are more likely to have received preventive care and care for chronic conditions
in accordance with evidence-based clinical guidelines.155 Under-compliance with HEDIS
measures is associated with an increase in the number of avoidable deaths and avoidable hospital
costs. For instance, failure to conduct regular HbA1c testing resulted in an estimated 7,400-15,000
avoidable diabetes-related deaths and $1.35-1.62 billion in avoidable hospital costs in 2005,
according to NCQA.155

Indicators of Quality of Laboratory Processes
Performance measurement is a valuable tool that can improve testing processes, benchmark
progress, and standardize laboratory performance. It also has the potential to decrease waste and
inefficiencies in the laboratory and lead to better health outcomes.156
May 2008

36

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

To date, performance measurement in laboratory medicine has focused mostly on the analytic
processes (i.e., the actual testing of specimens) in order to meet regulatory requirements of the
CLIA. More specifically, regulatory agencies and accrediting organizations have impelled quality
control and quality assurance initiatives during the last 15 years, mainly in the form of proficiency
testing, a quality monitoring approach for evaluating laboratories’ performance of selected tests
and their ability to arrive at the “correct” result.145 Other facets of laboratory performance
associated with the preanalytic and postanalytic phases also are evaluated, such as test
turnaround time and specimen identification errors. As a result of these requirements, the quality
and safety of laboratory testing has improved significantly over the past 20 years.157, 158
Historically, quality measures for preanalytic and postanalytic phases of the testing process have
received less attention, although some accreditation organizations and laboratories have taken the
initiative to assess quality and errors in these areas.159, 160 The quality assurance measures employed
by The Joint Commission, CAP, AABB, and COLA, have targeted certain elements of preanalytic
and postanalytic processes including methods to improve patient identification, specimen
collection, test turnaround time, notification of critical values, and customer satisfaction.161, 162
Outside the context of proficiency testing, formalized performance measurement and public
reporting on key quality indicators in a manner reflecting the broader health system (e.g., HEDIS
measures) has not yet been instituted in the laboratory medicine sector. However, CDC, CMS, other
government agencies, accreditation organizations, and stakeholders have been collaborating to
develop a set of such measures for national reporting and the determination of best practices.7, 163
Studies and programs undertaken by accreditation organizations, academic researchers, and
government agencies will provide an evidence-based foundation for more comprehensive quality
measurement. CDC announced a funding opportunity in 2007 to evaluate clinical laboratory
practice by identifying evidence-based laboratory medicine practices, particularly those associated
with the pre- and postanalytic stages of the total testing process.160 While the quality of laboratory
testing is already high, implementing a comprehensive standardized performance measurement
program will add further value to the overall health system and quality of patient care.

Value-based Purchasing and Pay-for-Performance
Value-based purchasing is among the evolving mechanisms being applied in various sectors of
health care to address rapidly rising health care costs and concerns about shortfalls in quality. It
refers to arrangements in which buyers hold health care providers accountable for cost and
quality, and includes the following attributes:

ƒ

Contracts that specify the responsibilities of employers as purchasers and insurance,
managed care, and hospital and physician groups as suppliers

ƒ

Incentives that reward specific practices by providers and consumers

ƒ

Information that supports purchasing activity management

ƒ

Quality management that leads to continuous improvements in the health care
purchasing process and in the delivery of health care

ƒ

Education that helps employees to become better consumers of health care164

May 2008

37

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

Pay-for performance (“P4P”), a reimbursement arrangement in which a portion of provider
payments are tied to performance measures associated with quality of care, has been proposed as
one means of enabling value-based purchases.165, 166 By 2005, there were more than 150 P4P
programs and initiatives in the U.S., sponsored by various health care plans, employers, and
government purchasers.147, 167 Approximately 80% of P4P programs are sponsored by private
health plans, but some are sponsored by government payers, including Medicare and state
Medicaid programs.147 Primary care physicians are the main target of P4P initiatives while 60% of
programs involve specialists and 20% involve hospitals.
Expert testimony to Congress on the subject of value-based purchasing of physicians’ services
has routinely cited variation in laboratory testing and physicians’ use of laboratory testing as a
way to measure the efficiency and quality of care being provided.168-170 For example, laboratory
testing provides clinicians with objective means for improving performance, such as ordering
appropriate follow-up testing for patients with test results indicating out-of-range or otherwise
abnormal values.171 In turn, data that include laboratory testing can be used by consumers and
purchasers to assess the quality of health care provided by different clinicians or organizations.
Laboratory-related professional organizations, such as the College of American Pathologists
(CAP), are partners in the development of P4P measures. CAP led development of P4P
pathology-related measures for breast and colon cancer, which were approved by the American
Medical Association (AMA) in June 2007.172
Still, P4P represents a relatively new type of financing mechanism for the health care system, and
its evidence base is small. A 2006 systematic review of the published literature examining P4P
programs for the period 1980-2005 identified only 17 empirical studies of explicit financial
incentives for quality, and could conclude little about the impact of these arrangements.173 No
studies examined the optimal duration of financial incentives for quality or the persistence of their
effects after termination, and only one study addressed cost-effectiveness. The investigators
called for ongoing monitoring of the impact of current programs and further research to guide
implementation of financial incentives and to assess their cost-effectiveness.

VALUE TO PUBLIC HEALTH AND SURVEILLANCE
Public health surveillance refers to ongoing, systematic collection, analysis, and interpretation
of data related to health that is vital to the planning, implementation, and evaluation of public
health practice. It relies on facets of clinical and public health laboratory testing.174 Services
provided by clinical laboratories help to identify the nature of public health threats at both the
individual and population level, including infections acquired during care, development of
drug resistance, infectious disease outbreaks, and biological threats.

Health Care Associated Infections
Health care associated infections (also known as nosocomial infections) are those that develop in
hospitalized patients in the absence of evidence that the infections were present or incubating at
the time of admission.175 The greatest common risk faced by hospitalized patients is health care
associated infection, a major source of morbidity and mortality in the U.S. Up to 2 million
patients experience a health care associated infection every year, and approximately 88,000 people
die annually as a result of these infections.176 Monitoring health care associated infection rates

May 2008

38

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

and understanding their causes are necessary for hospitals to reduce the incidence of these
infections and their impact on health outcomes and costs.
Microbiology laboratories help hospitals to monitor and control health care associated infection
rates at individual and population levels. They are responsible for providing easy access to highquality, timely data and support for epidemiological analyses.175 Activities may include providing
training on basic microbiology to hospital infection control program staff, monitoring laboratory
results for unusual findings (e.g., clusters of pathogens indicating an outbreak, emergence of multidrug-resistant organisms), strain typing of results, and storing specimens for further study.
Hospital laboratories with infection control practitioners voluntarily report health care associated
infection rates to CDC. The National Nosocomial Infections Surveillance (NNIS) systemf was
established by CDC in the 1970s to monitor the incidence of health care associated infections in
the U.S. and to assist infection control professionals in managing endemic and epidemic health
care associated infection outbreaks.178 NNIS has contributed to significant reductions in
bloodstream infection rates in coronary, medical, pediatric, and surgical ICUs.179 More than 300
hospitals participate in the NNIS; microbiology laboratories at each of these hospitals provide the
NNIS with information related to health care associated infection rates.178 The system has been
updated and expanded into the National Healthcare Safety Network, a web-based surveillance
system that was launched by CDC’s Division of Healthcare Quality Promotion in 2005. The
National Healthcare Safety Network aims to improve patient and health care worker safety by
monitoring adverse events associated with devices, procedures, and medications, providing
comparative data for performance improvement, and ensuring access to prevention tools and
information about lessons learned and best practices.177

Multi-drug-resistant Organisms
Another common problem faced by health care facilities is multi-drug-resistant organisms
(MDROs). MDROs are microorganisms, predominantly bacteria, that are resistant to one or more
classes of antimicrobial agents. They are difficult to treat and can result in increased length of
hospital stay, higher cost, and greater chance of mortality in infected individuals.180-183 MDROs are
often the cause of health care associated infections. Health care facilities at particular risk for MDRO
outbreaks include those caring for older patients, acute care settings, and ICUs.184 Varying
temporally, geographically, and by health care setting, the prevalence of MDROs in the U.S. has
increased steadily in recent decades. While only 20-25% of Staphylococcus aureus was resistant to
methicillin and other antibiotics in the early 1990s, resistance rose to 59% by 2003.185, 186
Clinical laboratories monitor MDROs at the levels of patients, institutions, and populations. At
the level of individual patients, laboratories conduct antimicrobial susceptibility testing, which
involves isolating and testing pathogenic bacteria to determine the ability of antimicrobial agents
to inhibit their growth.50 The results of susceptibility testing help guide clinician decisions about
how to treat specific infections. Many of the roles played by microbiology laboratories with
regard to health care associated infection surveillance and education also apply to MDROs.175 In

f

The NNIS, with the National Surveillance System for Health Care Workers and the Dialysis Surveillance Network,
is undertaking a major redesign to become the National Healthcare Safety Network, which will cover new areas
of patient safety monitoring and evaluation.177

May 2008

39

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

2000, the CAP Microbiology Surveys Program assessed the frequency and accuracy of
susceptibility testing at 3,857 microbiology laboratories in the U.S. CAP found that laboratories
generally demonstrated acceptable test accuracy and reproducibility for the most commonly used
antimicrobial susceptibility testing systems or methods.187
Laboratories support national monitoring of MDROs. The National Antimicrobial Resistance
Monitoring System for Enteric Bacteria is part of CDC’s Emerging Infections Program’s
Epidemiology and Laboratory Capacity Program and the Foodborne Diseases Active Surveillance
Network.188 In 2004, the most recent year for which data are available, public health laboratories
from all 50 states sent isolates to CDC laboratories for antimicrobial susceptibility testing.189 CDC’s
Epidemiology and Laboratory Capacity for Infectious Diseases program, part of the National Center
for Infectious Diseases, has cooperative agreements with state and local public health agencies in all
50 states to identify, characterize, and respond to infectious diseases, including MDROs.190

Public Health Reporting of Adverse Events
Laboratories participate in the reporting of adverse events related to testing technologies and
abnormal test results that result in patient harm. FDA’s MedWatch program gathers data on all
marketed medical products submitted by clinicians, manufacturers, and consumers and includes
adverse event reporting for laboratory test failures that result in harm to a patient. The Joint
Commission’s (formerly known as the Joint Commission on Accreditation of Healthcare
Organizations) Sentinel Event database collects data on unexpected occurrences involving serious
physical or psychological injury or risk thereof; laboratories accredited by the Joint Commission
are required to submit reports to the database.191 The Joint Commission publishes Sentinel Event
Alerts for its accredited organizations that identify specific events and their underlying causes
and suggests ways to prevent such occurrences in the future.

Natural Disasters and Biological and Chemical Threats
Laboratories assist in meeting the challenges of natural disasters and biological and chemical
events. Several national programs aim to provide a mechanism for communication between
laboratories and a means to link public and private laboratories. Some of these initiatives include
the following:

ƒ

The National Laboratory System is an initiative founded by CDC to improve
cooperation and coordination among all public and private U.S. laboratories.
Development of this system is still underway.192 The goal of the National Laboratory
System is to promote public health laboratory leadership by improving the overall
quality of laboratory testing and communication between public health and private
clinical laboratories.

ƒ

The Public Health Information Network (PHIN) is a CDC initiative to promote national
advancement of fully interoperable information systems in organizations that participate
in public health and public health preparedness.193 The goal of PHIN is to ensure that
public health programs have near real-time access to data during acts of terrorism or
disease outbreaks by developing, promoting, and using industry standards for data and
technology.194 In 2005, PHIN described functional requirements and general workflow
for information systems responsible for managing laboratory testing.195 The
interoperability requirements include such basic functions as assignment of

May 2008

40

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

unambiguous identifiers to laboratory data, adherence to PHIN standards for message
exchange systems, and vocabulary standards. The Laboratory Response Network
Results Manager supports the functional requirements established by PHIN.193

ƒ

The Early Warning Infectious Disease Surveillance (EWIDS) was created in 2003 and
is funded by DHHS.196 EWIDS comprises state, federal, and international partners
working together to ensure rapid, effective laboratory confirmation of infectious
disease reports in the border regions of the U.S., Canada, and Mexico. Ongoing
EWIDS projects include the development of cross-border surveillance protocols, a
database directory of laboratories, and a Health Alert Network and cross-border secure
internet information exchange. In 2006, DHHS announced a $5 million contract to
assist the six Mexican states in creating EWIDS systems at the U.S.-Mexico border that
are coordinated and interoperable with currently existing EWIDS systems in the U.S.197

ƒ

The Laboratory Response Network was established by CDC in 1999 as a network of
laboratories capable of quickly responding to biological and chemical terrorism and
other threats to public health.198 It includes approximately 150 laboratories responsible
for biological response. National laboratories located at CDC and the U.S. Army
Medical Research Institute for Infectious Diseases in Maryland are the national
laboratories that perform confirmatory testing for disease agents.199 Reference
laboratories, such as those run by state and local public health departments, perform
confirmatory tests for biological agents, allowing local authorities to respond more
quickly to positive test results. Sentinel laboratories are private, commercial, and
hospital-based and test patient specimens as part of their daily testing regimen,
allowing them to identify unusual results and refer suspicious specimens to the
network’s reference laboratories. This network provides a chain-of-command for
reporting laboratory results and sharing laboratory data.

Another laboratory role in public health surveillance is response to biological or chemical
terrorism. In cases of an overt biological or chemical threat, law enforcement officials are usually
notified first, followed by notification of the Federal Bureau of Investigation, state emergency
management, and state or local public health officials.199 These agencies, in turn, are to contact
CDC for a joint assessment of the validity of the threat. In the event of a valid threat, state or local
public health laboratories that are part of the Laboratory Response Network are to test samples of
the suspicious substance. Laboratory staff can identify the unknown substance and perform
confirmatory testing to validate test results.
Laboratories are involved in mitigating adverse outcomes during natural disasters. Laboratories
diagnose and confirm the presence of communicable diseases and ensure that basic laboratory
tests are available to use in caring for injured individuals.200 Increasingly, POCT is being used to
enable faster and more accurate diagnosis, triage, and patient monitoring during disasters. POCT
was especially vital in the aftermath of Hurricane Katrina in August 2005.201 The hurricane
resulted in temporary closing of 12 hospitals (and their laboratories), local trauma centers, and an
independent laboratory. POCT devices already on site, including glucose meters, whole-blood
analyzers, and tests for infectious diseases, pregnancy, and prothrombin time, were used to
provide care to thousands of affected people. Public health laboratories in states not affected by
the hurricane assisted Louisiana and Mississippi public health laboratories, providing such
services as newborn screening and water safety tests.202

May 2008

41

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

QUANTIFYING VALUE USING COST-EFFECTIVENESS ANALYSIS
Analyses of the tradeoffs of the costs and benefits of health care are of increasing interest,
particularly in instances where interventions offering marginal improvements over standard care
are accompanied by large price increments.203 At present, coverage policies of Medicare and most
commercial payers is based entirely or largely on clinical evidence rather than on costeffectiveness or any other economic-related assessment.204 However, more commercial payers,
hospitals and other health care institutions, and policy makers are seeking information from
manufacturers and vendors of health care technologies, including laboratory tests, regarding
whether their new and existing technologies are cost-saving, and sometimes more cost-effective,
than alternatives. Interest in economic analyses that can inform these decisions appears to be
growing among health plans and public and private sector payers. Health plans are using this
type of information to develop drug formularies and tiered co-payment systems. At certain
points throughout its history, CMS has proposed using, though has not employed, costeffectiveness as an explicit criterion for determining coverage of new medical technologies.205
FDA performs cost analyses in certain instances, particularly to fulfill requirements for
assessment of the economic impact of regulations; but the agency does not perform such analyses
to inform market approval or clearance decisions for particular products.206 A survey conducted
in 2001 of medical directors of 228 managed care plans found that 90% of plans considered cost in
some regard when evaluating new medical intervention; however, only 40% reported using
formal economic analyses for these purposes.207
Cost-effectiveness analysis (CEA) is one main type of economic analysis that is used to evaluate
and compare the economic impact of health technologies and medical procedures. It quantifies
the incremental (marginal) cost per incremental unit of effectiveness achieved with a technology
versus the standard of care. Units of effectiveness are typically “natural” health units, such as
case of cancer detected or life-year saved. Costs accrue differently to various stakeholders;
depending on its purpose, a CEA can be conducted from the perspective of, e.g., the health care
provider, payer/health plan, or society-at-large.
Another unit used in CEA is the quality-adjusted life year (QALY). QALYs are units comprising
quality of life (assessed as patient utility for a given health state, ranging from 0.0 for death to 1.0 for
perfect health) and length of life, and, thus, are not confined to use for particular diseases or
conditions.208 (CEAs that assess tradeoffs between costs and some measure of patient utility for a
health state or outcome are also known as cost-utility analyses.) Though used more in other
industrialized countries, cost per QALY is used informally as a means to gauge value-for-money by
some commercial U.S. health plans, though not by Medicare.205, 209-211 Though no formal threshold is
used by U.S. payers, there is informal recognition that incremental cost-effectiveness ratios (ICERs)
of up to $50,000-$100,000 per QALY are of acceptable value. It is also recognized that the ratios for
many technologies in mainstream care exceed that magnitude.212 (In the U.K., technologies with
ICERs approaching £30,000 per QALY tend to draw greater scrutiny by the National Health Service,
although there is no formal cut-off level for inclusion as a health care benefit in that system.213) Even
though a laboratory test may result in a clinically significant improvement in health outcomes (e.g.,
via an informed treatment decision and course of care), doing so at a high cost may decrease payers’
willingness to cover it or to pay a premium price for it.

May 2008

42

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

CEA of Genetics and Pharmacogenomic Tests
Among laboratory testing services, the potential economic impacts of genetic and PGx testing are
gaining greater scrutiny. Even so, the number and quality of available CEAs remain limited. A
small numberg of systematic reviews of economic evaluations of genetic testing (not including
PGx) have been published in recent years.
Most of the economic evaluations of genetic testing found in the literature are CEAs, with
relatively few cost-utility analyses and cost-benefit analyses. One review counted 37 CEAs, 16
cost-utility analyses, 12 cost-benefit analyses, and 4 cost-minimization analyses published
between 1990 and 2004. Another review counted 47 CEA, 13 cost-benefit analyses, 7 cost-utility
analyses, and 22 “other” economic evaluations published between 1983 and 2005.217, 218 These
systematic reviews are limited because they excluded all disease-specific studies that did not
explicitly refer to “genetics.” For example, at least 14 economic evaluations of screening for
hereditary hemochromatosis (an inherited disorder that increases the amount of iron absorbed
from the gut) were excluded in these four reviews.
In one of the systematic reviews of economic analyses for genetic testing published during 1990
through 2004, outcomes assessed in a majority of the studies were life-years gained or, simply,
cases detected. QALYs were used in another 25% of the studies. Nearly 40% of the studies
addressed cancer (21%) or aneuploidies (abnormal number of chromosomes) (18%). Common
shortcomings among these analyses included lack of specifying the economic perspective, lack of
discussion of potential bias, and lack of disclosure of funding sources.217
As pertains to other interventions that involve the use of laboratory results to inform patient
management decisions, the cost-effectiveness of PGx is subject to multiple factors or conditions.
One paper suggested that a PGx strategy is likely to be cost-effective when:

ƒ

The polymorphism under consideration is prevalent in the population and has a high
degree of penetrance.

ƒ

Genetic testing is highly sensitive and specific, and less costly alternative tests that
could be used to individualize therapy are not readily available.

ƒ

The disease state involves outcomes with significant morbidity or mortality if left untreated.

ƒ

The treatment involves significant outcomes and/or costs that can be affected by
genotype-individualized therapy.219

As such, PGx strategies are not practical for all drug prescribing or dosing regimens, and
decisions to invest in developing genomic-based therapies should be determined on a case-bycase basis.

g

One review, not discussed here, only addressed molecular genetic testing in familial cancers.214 Two reviews were
restricted to predictive molecular genetic tests.215, 216 The remaining two reviews concerned biochemical,
cytogenetic, and molecular testing.217, 218

May 2008

43

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

CEA of Laboratory Tests: Four Examples
Compared to therapeutic interventions and other diagnostic interventions, the literature on CEA
of laboratory testing is small, although a stronger literature on how to apply CEA to laboratory
testing is emerging.220 The following are examples of CEAs of four types of clinical laboratory
tests that have been under economic scrutiny by payers, clinicians, and other stakeholders. These
examples illustrate how CEAs are conducted and reported across diverse population groups and
indications, as well as the range of results of such analyses, including some suggesting that certain
tests are good value for money and others that are not, at least based on incremental cost
effectiveness ratio (ICER). The four examples are: screening for Chlamydia trachomatis,
immunoassay fecal-occult blood testing for colorectal cancer, HER-2/neu testing of breast cancer,
and nucleic acid testing (NAT) for safety of donated blood. The example of immunoassay fecaloccult blood testing includes discussion of how this rare request by CMS for a CEA was made and
how it was used in a Medicare coverage determination.

Chlamydia Screening
The literature on economics of laboratory testing recognizes that cost-effectiveness of a given test
can vary by the frequency and ordering of tests as well as characteristics of the target population.
A CEA published in 2004 compared alternative strategies for screening for Chlamydia trachomatis,
which clinical guidelines have recommended be conducted annually in women younger than 25
years of age. Using a simulation model, this analysis compared four strategies targeted to three
specific age groups (15-19, 15-24, and 15-29 years) of sexually active women in the U.S.: 1) no
screening, 2) annual screening for all women, 3) annual screening followed by one repeated test
within 3-6 months after a positive test result, and 4) annual screening followed by selective
semiannual screening for women with a history of infection.221
This simulation showed that the most cost-effective strategy was annual screening in women 1529 years of age, followed by semiannual screening for those with a history of infection. This
strategy consistently had an ICER less than $25,000 per QALY compared with the next most
effective strategy. All of the strategies became more cost-effective when the indirect transmission
effects of a 10-year screening program on the probability of infection in uninfected women (that is,
per-susceptible rate of infection) were incorporated into the simulation. Results of the simulation
were sensitive to such factors as the annual incidence of chlamydia, probability of persistent
infection, screening test costs, and costs of treating long-term complications. Accounting for
feasible variations in these factors in a large number of simulated scenarios, the strategy of annual
screening in women 15-29 years of age followed by semiannual screening for those with a history
of infection maintained an ICER less than $50,000 per QALY in 99% of the simulations.
A systematic review published in 2006 sought to produce comparable estimates of relative health
impact and cost-effectiveness for services deemed effective by the USPSTF and DHHS Advisory
Committee on Immunization Practices. The review found that screening young women for
chlamydia has an ICER less than $15,000 per QALY.222 The investigators concluded that screening
young women for chlamydia was one of the most valuable clinical preventive services that can be
offered in medical practice.

May 2008

44

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

Immunoassay Fecal-occult Blood Testing
CEAs can be used to inform coverage decisions by public and private sector third-party payers.
Although this is rarely done for the Medicare program, CMS requested that AHRQ conduct a
comparative clinical and cost-effectiveness study regarding screening immunoassay fecal-occult
blood testing (iFOBT) for a coverage review in 2003.223 Medicare was already covering colorectal
cancer screening with a payment level of $4.50 for the standard guaiac-based fecal-occult blood
test (gFOBT).
AHRQ tasked this study to a team of investigators at Erasmus University, Memorial SloanKettering Institute Cancer Center, and the National Cancer Institute. The investigators used a
micro-simulation model to derive cost-effectiveness estimates. The analysis compared iFOBT
(priced at the manufacturer-recommended amount of $28) to two gFOBTs, Hemoccult II® and
Hemoccult Sensa®, at the Medicare reimbursement of $4.50.h The sensitivities and specificities of
the tests were varied according to their most likely values based on an extensive literature review.
The investigators found that:

ƒ

Assuming sensitivities of 40%, 70%, and 70% for Hemoccult II®, Hemoccult Sensa®,
and iFOBT respectively, iFOBT would detect more cancers than Hemoccult II® and a
similar number of cancers as Hemoccult Sensa.®

ƒ

All FOBTs were cost-effective. Hemoccult II® at $4.50 had a cost-effectiveness ratio of
$1,071 per life-year gained and iFOBT at $28 had a cost-effectiveness ratio of $4,500 per
life year saved, assuming 100% compliance. (Lower levels of compliance would
increase the cost per life-year gained.)

ƒ

At $28 for iFOBT and $4.50 for Hemoccult II, the ICER for iFOBT was $11,000 per
additional life-year saved, assuming 98% specificity for iFOBT, and $21,000 per
additional life-year saved, assuming 95% specificity for iFOBT.

ƒ

Compared to Hemoccult II® gFOBT at $4.50, iFOBT would have an equal costeffectiveness if priced at $13 with the more favorable assumption of 98% specificity.224

In the conclusion of its decision memorandum, CMS stated: “While the unit cost of iFOBT is
generally higher than gFOBT, both are considered cost-effective compared to other cancer
screening tests.” Noting that the major challenge of colorectal cancer screening was to increase
compliance from current low levels, CMS added that “It is likely that simple interventions to
increase compliance would be highly cost-effective as well.” The decision summary stated that
“CMS concludes that there is adequate evidence to determine that the iFOBT is an appropriate
and effective colorectal cancer screening fecal-occult blood test for Medicare beneficiaries aged 50
years and older.”

HER-2/neu Testing
The use of PGx to direct the use of therapies for targeted populations, especially where the
therapies are expensive and where the burden of disease is great for the affected population, calls
for careful weighing of health and economic tradeoffs. An instructive example of this

h

The analysis assumed that screening was confined to people aged 65 years and older, with a base case compliance
level of 100%.

May 2008

45

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

consideration involves testing HER-2/neu status in women with newly diagnosed breast cancer
for informing the use of trastuzumab (Herceptin) for treatment of those with HER-2/neu positive
breast cancer. HER-2/neu gene testing limits the risk that patients whose breast cancers do not
overexpress HER-2/neu will experience potentially serious side effects of treatment with
Herceptin and helps to avoid associated costs.
Two tests are used for HER-2/neu testing: immunohistochemical assays (IHC) and fluorescence
in situ hybridization (FISH). These tests have different mechanisms; IHC detects protein
overexpression and FISH detects gene amplification. Decisions about using these tests involve
tradeoffs between the additional costs associated with using FISH and the higher rate of falsepositive results associated with IHC.
A Canadian study published in 2007 compared the cost-effectiveness of various strategies of
testing HER-2/neu status in women with newly diagnosed breast cancer for informing the use of
trastuzumab for treatment of those with HER-2/neu positive breast cancer.225 The study involved
a systematic review and meta-analysis of data comparing the agreement of IHC and FISH testing
to determine HER-2/neu status. The investigators calculated the accuracy and incremental cost
per accurate diagnosis for alternative testing strategies compared with the base strategy of IHC
testing, followed by confirmation of 2+ scores (where the range of such scores is 0, 1+, 2+, 3+) by
FISH. They observed that the median percentage of patients in each category of IHC score was: 0:
36.1%; 1+: 35.5%; 2+: 12.0%; and 3+: 16.2%. The median percentage of results of FISH that were
positive in each IHC category was: 0: 1.6%; 1+: 4.9%; 2+: 29.8%; and 3+: 92.4%. The base strategy
was expected to determine correctly the HER-2/neu status of 96% of patients with breast cancer.
Compared to the base strategy, confirmation of HER-2/neu status by FISH in cases that received a
score of 3+ reduced the percentage of false positive results to 0% and increased the percentage of
accurately determined HER-2/neu results to 97.6%. This yielded a median ICER of CA$6,175 per
case of accurately determined HER-2/neu status compared to the base strategy. In comparison,
the strategy of performing FISH testing in all cases of breast cancer yielded a median ICER of
CA$8,401 per case of accurately determined HER-2/neu status. The investigators concluded that
the strategy with the lowest cost-effectiveness ratio involved screening all newly diagnosed cases
of breast cancer with IHC and confirming scores of 2+ or 3+ with FISH.
According to a CEA published in 2004 of HER-2/neu testing and trastuzumab for metastatic
breast cancer, IHC with confirmatory FISH testing resulted in an ICER of $125,000 per QALY and
initial FISH testing (without IHC) resulted in an ICER of $145,000 per QALY.226 These ratios are
higher than the $100,000 per QALY level cited as an informal acceptable threshold in the U.S. by
some health economists227 and thresholds cited in western Europe and certain other industrialized
countries. Even so, these ICERs for HER-2/neu testing are of similar or lesser magnitude than
those for various other cancer therapies and many other widely used health care interventions.
Continued improvements in testing, targeted treatments and lower costs that may arise with
competing interventions should improve the cost-effectiveness of HER-2/neu testing and related
tests for breast cancer.154
A study published in 2007 modeled the incremental cost-effectiveness of adding trastuzumab to
chemotherapy regimens in patients with HER-2/neu-positive breast cancer. The model assumed
that both IHC and FISH were used to determine HER-2 status. The study assumed that, on
average, five tests were performed for every patient identified for trastuzumab treatment; 30% of

May 2008

46

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

tests were assumed to be FISH in the base case. While this study did not analyze the costeffectiveness of the testing in particular, it projected that the incremental cost per QALY gained of
adding trastuzumab to chemotherapy, including the cost of testing to inform the therapeutic
decision, was $26,417. The cost-effectiveness of treatment with trastuzumab over a 20-year
horizon was projected to be $34,201 per QALY gained.228

Nucleic Acid Testing for Blood Safety
As described above, the accessibility of safe blood is vital for millions of people in the U.S. The
blood supply is rigorously screened and tested throughout the collection and transfusion process.
Scientific and technological advances have led to continued improvements in the safety of the
blood supply, so that transmission of infectious agents of greatest concern to health—HIV and
hepatitis B and C viruses—is rare. Recent advances have focused on decreasing the “window
period” of detecting viral antibodies and antigens, so as to diminish the chances of failing to
detect infected blood donations shortly after donors have contracted the pathogen. To the extent
that newer, more sensitive tests become available, any additional cost of detecting incrementally
more cases raises questions about the cost-effectiveness of these tests.i,206
NAT, which relies on polymerase chain reaction techniques, can detect the presence of viral genes
in blood. NAT can be conducted on “minipools” of 16-24 samples (minipool NAT) or on
individual donations of blood (individual donation NAT). Alternative testing strategies can yield
significantly different cost-effectiveness ratios. NAT has been the topic of several CEAs.
Based on detailed CEAs, researchers have concluded that use of NAT to screen all donated blood
for HIV and hepatitis C virus results in ICERs of more than $5 million per QALY saved. In addition
to the cost of testing many units of donated blood for the rare ones that cannot be detected by other
means, the short life expectancy of many people who receive donated blood products constrains the
health impact, and therefore the cost-effectiveness, of successful testing.203 A CEA published in 2003
examined the marginal cost-effectiveness of using NAT for HIV and hepatitis B and C viruses in
whole-blood donations using 2001 disease incidence data from the American Red Cross.231 This
study estimated that the addition of minipool NAT to HIV antigen testing would add a total of 62
QALYs per year in the U.S., while the addition of individual donation NAT would add a total of 90
QALYs per year. The cost per QALY gained for adding individual donation NAT in addition to
HIV antigen testing ranged from $8.4 million to $9.1 million, depending on whether hepatitis B
virus testing was included.j Eliminating HIV antigen testing and using only NAT would reduce
costs, but cost per QALY gained would remain above $4 million. The investigators concluded that
the cost-effectiveness of adding NAT to serologic testing is poor.
An analysis published in 2004 examined the cost-effectiveness of adding either individual
donation NAT or minipool NAT to pre-existing serologic testing protocols for HIV and hepatitis B
and C viruses with scenarios that included and excluded HIV antigen testing for blood donated in

As is so for many other avenues of health care, the increases in the safety of the blood supply achieved with additional
testing have represented diminishing returns over time.203 Whereas the ICER of HIV antibody testing, implemented
in 1985, was less than $5,000 per QALY saved,229, 230 the ICER for HIV p24 antigen testing, introduced in 1996, was
estimated to exceed $2 million per additional QALY saved.
j Adding minipool NAT to HIV antigen testing would result in a cost per QALY gained ranging from $5.8 million to
$7.6 million, depending on whether or not a test for hepatitis B virus was included.
i

May 2008

47

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

the U.S. Compared with serologic testing alone, minipool NAT was found to save a total of 102
additional QALYs and individual donation NAT was found to save an additional 115 QALYs.
Serologic testing (excluding HIV antigen testing) coupled with minipool NAT resulted in an ICER
of $1.5 million per QALY gained. Serologic testing (excluding HIV antigen testing) in conjunction
with individual donation NAT was found to be associated with an ICER of $7.3 million per QALY
gained. This study confirmed that the cost-effectiveness of adding NAT to current serological
testing far exceeds the traditionally recognized, though informal in the U.S., threshold of $50,000
to $100,000 per QALY gained. The authors concluded that the cost per QALY of adding NAT
screening is beyond the typical range for most health care interventions, but not for established
blood safety measures.232
Notwithstanding its unfavorable cost-effectiveness, NAT screening has been widely adopted. The
American Red Cross uses minipool NAT to test for HIV in all donated blood. The main justification
for implementing NAT screening was the concern of the public and policy makers arising from the
potential, however rare, for patients to contract a catastrophic illness from the blood supply.231, 232

CONCLUSIONS
Laboratory testing is integral to many clinical decisions, providing physicians, nurses, and other
health care providers with often pivotal information that aids in prevention, diagnosis, treatment,
and management of disease. Despite their impact, spending on laboratory services accounts for
only 2.3% of health care expenditures and 2% of Medicare expenditures.

ƒ

Laboratory medicine supports the practice of evidence-based medicine and
development of clinical practice guidelines, which assist practitioners and patients in
making decisions about health care in specific circumstances.

ƒ

Laboratory tests provide objective data about patient health that enable screening for
risk factors, accurate and early diagnosis, determination of disease severity and
likelihood of recovery, selection and monitoring of treatment, and evaluation of
potential adverse outcomes. Laboratory tests also are vital to patient self management
of chronic conditions, supporting their ability to monitor their health status daily,
adjust therapies, and evaluate progress with healthy lifestyle choices.

ƒ

Information provided by laboratory testing is a critical component of quality and
safety, including the prevention of adverse reactions. Laboratories protect the blood
supply from pathogens and accurately match patients and blood products. For
managing medication, testing provides information for maintaining optimum drug
levels, helps to detect and recover from medication errors, and enables use of genetic
information to guide personalized health care.

ƒ

Laboratory testing is incorporated into indicators used to assess quality of care,
particularly for diseases of high health and economic burden such as diabetes, heart
failure, and colon cancer. Laboratory data also can be used in new and emerging
approaches to value-based purchasing of health care.

ƒ

The evidence base for the cost-effectiveness of laboratory tests, and the broader therapeutic
regimens and other interventions of which they are a part, is growing, though still limited.
This evidence is helping to inform test selection and sequencing, technology acquisition

May 2008

48

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

decisions, formulary design (including for PGx-mediated therapies), and screening and
other population-based interventions. Though used less in the U.S. than in other
developed nations, it is being considered in selected coverage and payment policies of
some health plans and other third-party payers.

ƒ

Services provided by clinical laboratories also are critical to public health at the individual
and population levels by identifying nosocomial infections, antimicrobial resistance,
infectious disease outbreaks, exposure to toxic substances, and chemical and biological
threats. Laboratories also help to mitigate the effects of natural disasters by enabling rapid
turnaround of tests used during triage and emergency care of individual patients as well
as tests to confirm the presence of communicable diseases that threaten the population.

ƒ

Consistent with other sectors of health care, laboratory medicine is under greater
scrutiny for demonstrating its value for patients, providers, payers, and other
stakeholders. Value must be documented based on rigorous clinical, public health, and
economic evidence. Whether in the form of quality indicators, practice guidelines,
coverage policies, value-based purchasing or related payment policies, or simply
market share for laboratory services, the value of laboratory medicine is being
explicitly incorporated into health care decisions.

May 2008

49

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

REFERENCE LIST
1. The value of clinical laboratory services. Washington, DC: American Clinical Laboratory
Association, 2007. (Accessed May 15, 2007, at http://www.clinicallabs.org/issues/value/index.shtml.)
2. Terry M. Lab industry strategic outlook: market trends and analysis 2007. New York, NY:
Washington G-2 Reports, 2007.
3. Institute of Medicine. Crossing the quality chasm: a new health system for the 21st
century. Washington, DC: National Academy Press, 2001.
4. McGlynn EA, Asch SM, Adams J, et al. The quality of health care delivered to adults in the
United States. N Engl J Med 2003;348(26):2635-45.
5. Institute of Medicine. To err is human: building a safer health system. Washington, DC:
National Academy Press, 2000.
6. Leape LL, Berwick DM. Safe health care: are we up to it? BMJ 2000;320(7237):725-6.
7. Behal R. Identification of performance measures of importance for quality in laboratory
medicine. Washington, DC: National Quality Forum, 2007.
http://216.122.138.39/pdf/projects/lab-med/txlabpaper_behal_final%2005-21-07.pdf.
8. Through the patient's eyes: understanding and promoting patient-centered care. Gerteis
M, Edgman-Levitan S, Daley J, eds. San Francisco, CA: Jossey-Bass, 2002.
9. Sipkoff M. The new consensus favoring IOM's definition of quality. Manag Care
2004;13(6):18-7.
10. Steindel SJ, Howanitz PJ. Physician satisfaction and emergency department laboratory test
turnaround time. Arch Pathol Lab Med 2001;125(7):863-71.
11. Sackett DL, Rosenberg WM, Gray JA, Haynes RB, Richardson WS. Evidence based
medicine: what it is and what it isn't. BMJ 1996;312(7023):71-2.
12. Eddy DM. Evidence-based medicine: a unified approach. Health Aff (Millwood )
2005;24(1):9-17.
13. Timmermans S, Mauck A. The promises and pitfalls of evidence-based medicine. Health
Aff (Millwood ) 2005;24(1):18-28.
14. Sackett DL, Rosenberg WM. The need for evidence-based medicine. J R Soc Med
1995;88(11):620-4.
15. Seaborg E. Evidence-based approach to medicine improves patient care. Washington, DC:
Lab Tests Online, 2006. (Accessed June 13, 2007, at
http://www.labtestsonline.org/understanding/features/ebm-2.html.)
16. Diabetes control and complications trial (DCCT). Bethesda, MD: National Diabetes
Information Clearinghouse, National Institute of Diabetes and Digestive and Kidney
Diseases, National Institutes of Health, 2001. (Accessed June 13, 2007, at
http://diabetes.niddk.nih.gov/dm/pubs/control/#management.)

May 2008

50

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

17. Field M, Lohr KN. Attributes of good practice guidelines. In: Clinical practice guidelines:
directions for a new program. Field M, Lohr KN, ed. Washington, DC: National Academy
Press, 1990.
18. Farquhar CM, Kofa EW, Slutsky JR. Clinicians' attitudes to clinical practice guidelines: a
systematic review. Med J Aust 2002;177(9):502-6.
19. Shekelle PG, Woolf SH, Eccles M, Grimshaw J. Clinical guidelines: developing guidelines.
BMJ 1999;318(7183):593-6.
20. Woolf SH. Practice guidelines, a new reality in medicine. II. Methods of developing
guidelines. Arch Intern Med 1992;152(5):946-52.
21. Cutler DM. Your money or your life: strong medicine for America's health care system.
Oxford, England: Oxford University Press, 2004.
22. 2006 National Healthcare Quality Report. Report No. 07-0013. Rockville, MD: Agency for
Healthcare Research and Quality, 2006.
23. Value of clinical laboratory services in health care. Bethesda, MD: American Society for
Clinical Laboratory Science, 2005. (Accessed May 15, 2007, at
http://www.ascls.org/position/ValueCLS.asp.)
24. Screening. Bethesda, MD: National Cancer Institute, National Institutes of Health, 2007.
(Accessed June 19, 2007, at
http://www.cancer.gov/Templates/db_alpha.aspx?CdrID=46171.)
25. Screening tests for adults (ages 30-49). Washington, DC: Lab Tests Online, 2005. (Accessed
May 16, 2007, at http://www.labtestsonline.org/understanding/wellness/d_adult.html.)
26. Screening tests for young adults (ages 19-29). Washington, DC: Lab Tests Online, 2005.
(Accessed May 16, 2007, at
http://www.labtestsonline.org/understanding/wellness/c_youngadult1.html.)
27. Medicare preventive benefits begin January 2005 with a goal of healthier seniors.
Baltimore, MD: Centers for Medicare and Medicaid Services, 2004. (Accessed May 16,
2007, at http://www.hhs.gov/news/press/2004pres/20041109.html.)
28. Preventive services: a healthier U.S. starts here. Baltimore, MD: Centers for Medicare and
Medicaid Services, 2007. (Accessed May 16, 2007, at
http://www.medicare.gov/Health/Overview.asp.)
29. Institute of Medicine. Care without coverage: too little too late. Washington, DC: National
Academy Press, 2002.
30. Huynh PT, Schoen C, Osborn R, Holmgren AL. The U.S. health care divide: disparities in
primary care experiences by income. Findings from The Commonwealth Fund 2004
International Health Policy Survey. New York, NY: The Commonwealth Fund, 2006.
http://www.commonwealthfund.org/usr_doc/Huynh_UShltcaredivide_900.pdf?section=4039.
31. About genetic services. Washington, DC: GeneTests.org, 2004. (Accessed July 30, 2007, at
http://www.genetests.org/servlet/access?id=8888891&key=2Sg7gGJY9YmUh&fcn=y&f
w=i03Q&filename=/concepts/primer/primerwhatistest.html.)

May 2008

51

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

32. U.S. system of oversight of genetic testing: a response to the charge of the Secretary of
DHHS. Draft report of the Secretary's Advisory Committee on Genetics, Health, and
Society (available for public comment November 5-December 21, 2007). Bethesda, MD:
Secretary's Advisory Committee on Genetics, Health, and Society, 2007.
http://www4.od.nih.gov/oba/SACGHS/reports/SACGHS%20Draft%20Report%20on%
20the%20Oversight%20of%20Genetic%20Testing%2011-5-2007.pdf.
33. What is genetic testing? Berkeley, CA: Lawrence Berkeley National Laboratory, 2007.
(Accessed May 16, 2007, at http://www.lbl.gov/Education/ELSI/genetic-testing.html.)
34. GeneTests. Seattle, WA: GeneTests.org, 2008. (Accessed April 18, 2008, at
http://www.genetests.org/servlet/access?id=8888891&key=Z1o3AV3PRzDMu&fcn=y&f
w=apWV&filename=/.)
35. Border B. The universe of genetic testing. Washington, DC: Lab Tests Online, 2005.
(Accessed July 31, 2007, at
http://www.labtestsonline.org/understanding/features/genetics.html.)
36. Fogarty M. Genetic testing for people without symptoms of Alzheimer's. Palo Alto, CA:
GeneticHealth, 2001. (Accessed May 16, 2007, at
http://www.genetichealth.com/ALZ_Genetics_Testing_in_Healthy_People.shtml.)
37. Genomics and population health: United States 2003. Atlanta, GA: Centers for Disease
Control and Prevention, Office of Genomics and Disease Prevention, 2004.
http://www.cdc.gov/genomics/activities/file/print/2003report/2003Full.pdf.
38. Pap smear. Washington, DC: Lab Tests Online, 2005. (Accessed May 16, 2007, at
http://www.labtestsonline.org/understanding/analytes/pap/glance.html.)
39. SEER cancer statistics review, 1975-2004. Bethesda, MD: National Cancer Institute,
National Institutes of Health, 2006. http://seer.cancer.gov/csr/1975_2004/.
40. Cancer facts and figures 2007. Atlanta, GA: American Cancer Society, 2007.
http://www.cancer.org/downloads/STT/CAFF2007PWSecured.pdf.
41. Hewitt M, Devesa SS, Breen N. Cervical cancer screening among U.S. women: analyses of
the 2000 National Health Interview Survey. Prev Med 2004;39(2):270-8.
42. Colorectal cancer: early detection. Atlanta, GA: American Cancer Society, 2007. (Accessed
May 16, 2007, at
http://www.cancer.org/docroot/CRI/content/CRI_2_6X_Colorectal_Cancer_Early_Dete
ction_10.asp?sitearea=&level=.)
43. Williams N. How reliable is laboratory testing? Washington, DC: Lab Tests Online, 2005.
(Accessed May 17, 2007, at
http://www.labtestsonline.org/understanding/features/reliability.html.)
44. Staging: questions and answers. Bethesda, MD: National Cancer Institute, National
Institutes of Health, 2004. (Accessed May 17, 2007, at
http://www.cancer.gov/cancertopics/factsheet/Detection/staging.)
45. Diabetes. Bethesda, MD: Medline Plus, National Library of Medicine, National Institutes
of Health, 2007. (Accessed May 22, 2007, at
http://www.nlm.nih.gov/medlineplus/diabetes.html .)

May 2008

52

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

46. Smith DS. Pharyngitis. Bethesda, MD: Medline Plus, National Library of Medicine,
National Institutes of Health, 2007. (Accessed May 17, 2007, at
http://www.nlm.nih.gov/medlineplus/ency/article/000655.htm.)
47. FDA task force on antimicrobial resistance: key recommendations and report. Rockville,
MD: Food and Drug Administration, 2000.
http://www.fda.gov/oc/antimicrobial/taskforce2000.html.
48. Vanderweil SG, Pelletier AJ, Hamedani AG, Gonzales R, Metlay JP, Camargo CA, Jr.
Declining antibiotic prescriptions for upper respiratory infections, 1993-2004. Acad Emerg
Med 2007;14(4):366-9.
49. Martin JM, Green M, Barbadora KA, Wald ER. Erythromycin-resistant group A
streptococci in schoolchildren in Pittsburgh. N Engl J Med 2002;346(16):1200-6.
50. Susceptibility testing. Washington, DC: Lab Tests Online, 2005. (Accessed May 30, 2007, at
http://www.labtestsonline.org/understanding/analytes/susceptibility/glance.html.)
51. Mueller C, Scholer A, Laule-Kilian K, et al. Use of B-type natriuretic peptide in the
evaluation and management of acute dyspnea. N Engl J Med 2004;350(7):647-54.
52. Bhatia V, Nayyar P, Dhindsa S. Brain natriuretic peptide in diagnosis and treatment of
heart failure. J Postgrad Med 2003;49(2):182-5.
53. Martin JN, Jr., May WL, Magann EF, Terrone DA, Rinehart BK, Blake PG. Early risk
assessment of severe preeclampsia: admission battery of symptoms and laboratory tests to
predict likelihood of subsequent significant maternal morbidity. Am J Obstet Gynecol
1999;180(6 Pt 1):1407-14.
54. Thompson SR. Preeclampsia. Bethesda, MD: Medline Plus, National Library of Medicine,
National Institutes of Health, 2007. (Accessed May 17, 2007, at
http://www.nlm.nih.gov/medlineplus/ency/article/000898.htm.)
55. Sjauw KD, van dH, I, Nijsten MW, Nieuwland W, Zijlstra F. Value of routine admission
laboratory tests to predict thirty-day mortality in patients with acute myocardial
infarction. Am J Cardiol 2006;97(10):1435-40.
56. Factor V Leiden mutation and PT 20210 mutation. Washington, DC: Lab Tests Online,
2007. (Accessed November 21, 2007, at
http://www.labtestsonline.org/understanding/analytes/factor_v_and_pt20210/glance.html.)
57. FDA clears breast cancer specific molecular prognostic test. Rockville, MD: Food and Drug
Administration, 2007. (Accessed May 17, 2007, at
http://www.fda.gov/bbs/topics/NEWS/2007/NEW01555.html.)
58. Pollack A. Test to predict breast cancer relapse is approved. The New York Times.
February 7, 2007: Health.
59. About Oncotype DX. Redwood City, CA: Genomic Health, Incorporated, 2007. (Accessed
June 27, 2007, at http://www.genomichealth.com/oncotype/about/hcp.aspx.)
60. Harris L, Fritsche H, Mennel R, et al. American Society of Clinical Oncology 2007 update
of recommendations for the use of tumor markers in breast cancer. J Clin Oncol
2007;25(33):5287-312.

May 2008

53

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

61. NCCN Clinical Practice Guidelines in Oncology: breast cancer. V 2.2008. Fort Washington,
PA: National Comprehensive Cancer Network, 2008. (Accessed March 1, 2008, at
http://www.nccn.org/professionals/physician_gls/PDF/breast.pdf.)
62. Lab tests. Rockville, MD: Office of In Vitro Diagnostic Device Evaluation and Safety, Food
and Drug Administration, 2005. (Accessed May 21, 2007, at
http://www.fda.gov/cdrh/oivd/consumer-lab.html.)
63. Biel-Cunningham S. Understanding your labs. New York, NY: Body Health Resources
Corporation, 2004. (Accessed May 21, 2007, at
http://www.thebody.com/content/treat/art32195.html.)
64. Juan D. Thyroid stimulating hormone. Bethesda, MD: Medline Plus, National Library of
Medicine, National Institutes of Health, 2006. (Accessed May 21, 2007, at
http://www.nlm.nih.gov/medlineplus/ency/article/003684.htm.)
65. Soper JT, Mutch DG, Schink JC. Diagnosis and treatment of gestational trophoblastic
disease: ACOG Practice Bulletin No. 53. Gynecol Oncol 2004;93(3):575-85.
66. Frassica JJ. Frequency of laboratory test utilization in the intensive care unit and its
implications for large-scale data collection efforts. J Am Med Inform Assoc 2005;12(2):229-33.
67. Basic metabolic panel. Washington, DC: Lab Tests Online, 2005. (Accessed May 21, 2007,
at http://www.labtestsonline.org/understanding/analytes/bmp/bmp.html.)
68. Giuliano KK, Grant ME. Blood analysis at the point of care: issues in application for use in
critically ill patients. AACN Clin Issues 2002;13(2):204-20.
69. Kilgore ML, Steindel SJ, Smith JA. Evaluating stat testing options in an academic health
center: therapeutic turnaround time and staff satisfaction. Clin Chem 1998;44(8 Pt
1):1597-603.
70. Kost GJ, Ehrmeyer SS, Chernow B, et al. The laboratory-clinical interface: point-of-care
testing. Chest 1999;115(4):1140-54.
71. Storrow AB, Gibler WB. The role of cardiac markers in the emergency department. Clin
Chim Acta 1999;284(2):187-96.
72. Wells PS, Brill-Edwards P, Stevens P, et al. A novel and rapid whole-blood assay for Ddimer in patients with clinically suspected deep vein thrombosis. Circulation
1995;91(8):2184-7.
73. Jackson RG, Samra GS, Radcliffe J, Clark GH, Price CP. The early fall in levels of S-100 beta
in traumatic brain injury. Clin Chem Lab Med 2000;38(11):1165-7.
74. Scalise D. Poised for growth. Point-of-care testing. Hosp Health Netw 2006;80(9):77-83.
75. Gruszecki AC, Hortin G, Lam J, et al. Utilization, reliability, and clinical impact of pointof-care testing during critical care transport: six years of experience. Clin Chem 2003;49(6
Pt 1):1017-9.
76. Fenwick EA, Briggs AH, Hawke CI. Management of urinary tract infection in general
practice: a cost-effectiveness analysis. Br J Gen Pract 2000;50(457):635-9.
77. Jones R, Phillips I, Felix G, Tait C. An evaluation of near-patient testing for Helicobacter
pylori in general practice. Aliment Pharmacol Ther 1997;11(1):101-5.

May 2008

54

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

78. Dahler-Eriksen BS, Lauritzen T, Lassen JF, Lund ED, Brandslund I. Near-patient test for Creactive protein in general practice: assessment of clinical, organizational, and economic
outcomes. Clin Chem 1999;45(4):478-85.
79. Price CP. Point of care testing. BMJ 2001;322(7297):1285-8.
80. Deville WL, Yzermans JC, van Duijn NP, Bezemer PD, van der Windt DA, Bouter LM. The
urine dipstick test useful to rule out infections. A meta-analysis of the accuracy. BMC Urol
2004;4:4.
81. Holman H, Lorig K. Patients as partners in managing chronic disease. Partnership is a
prerequisite for effective and efficient health care. BMJ 2000;320(7234):526-7.
82. Healthy aging: preserving function and improving quality of life among older Americans
2007. Atlanta, GA: Coordinating Center for Health Promotion, Centers for Disease
Control and Prevention, 2007.
http://www.cdc.gov/nccdphp/publications/aag/pdf/healthy_aging.pdf.
83. Chronic disease overview. Atlanta, GA: Centers for Disease Control and Prevention, 2005.
(Accessed May 21, 2007, at http://www.cdc.gov/nccdphp/overview.htm.)
84. The benefits of cholesterol lowering. Bethesda, MD: National Cholesterol Education
Program, National Heart Lung and Blood Institute, National Institutes of Health, 2007.
(Accessed May 21, 2007, at http://www.nhlbi.nih.gov/chd/why4.htm.)
85. Chronic kidney disease. New York, NY: National Kidney Foundation, 2007. (Accessed
May 21, 2007, at http://www.kidney.org/kidneydisease/ckd/index.cfm.)
86. Position statement: advancing the integration of accredited disease management programs
in federal health care programs. Washington, DC: Disease Management Association of
America, 2003. http://www.dmaa.org/pdf/AdvancingIntegration032003.pdf.
87. DMAA definition of disease management. Washington, DC: Disease Management
Association of America, 2007. (Accessed May 21, 2007, at
http://www.dmaa.org/dm_definition.asp.)
88. Chronic disease care reports and initiatives: patient self-management. Oakland, CA:
California Healthcare Foundation, 2007. (Accessed May 22, 2007, at
http://www.chcf.org/topics/chronicdisease/index.cfm?subtopic=CL613.)
89. Benjamin EM. Self-monitoring of blood glucose: the basics. Clinical Diabetes 2002;20(1):45-7.
90. Strategies for reducing morbidity and mortality from diabetes through health-care system
interventions and diabetes self-management education in community settings. A report on
recommendations of the Task Force on Community Preventive Services. MMWR Recomm
Rep 2001;50(RR-16):1-15.
91. Klonoff DC. Continuous glucose monitoring: roadmap for 21st century diabetes therapy.
Diabetes Care 2005;28(5):1231-9.
92. Therapeutic drug monitoring. Washington, DC: Lab Tests Online, 2006. (Accessed May 22,
2007, at http://www.labtestsonline.org/understanding/analytes/thdm/glance.html.)
93. Stuart TL. Home monitoring and management of warfarin therapy: an anticoagulation
clinic perspective. J Thromb Thrombolysis 2000;10:55-7.

May 2008

55

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

94. Schumacher GE, Barr JT. Total testing process applied to therapeutic drug monitoring:
impact on patients' outcomes and economics. Clin Chem 1998;44(2):370-4.
95. Wu AH. On-site tests for therapeutic drugs. In: On-site drug testing. Jenkins AJ,
Goldberger BA, ed. Totowa, NJ: Humana Press, Incorporated, 2001.
96. Biosensor for therapeutic drug monitoring (TDM) in point-of-care testing (POCT). Basel,
Switzerland: Roche Diagnostics, 2003. (Accessed June 28, 2007, at
http://www.fuentek.com/images/TDM/TDM-brochure.pdf.)
97. Adverse drug reaction. Rockville, MD: Patient Safety Network, Agency for Healthcare
Research and Quality, 2007. (Accessed June 20, 2007, at
http://psnet.ahrq.gov/glossary.aspx.)
98. What is a medication error? Oakbrook Terrace, IL: National Coordinating Council for
Medication Error Reporting and Prevention, 2007. (Accessed June 27, 2007, at
http://www.nccmerp.org/aboutMedErrors.html.)
99. Institute of Medicine. Preventing medication errors. Washington, DC: National Academy
Press, 2006.
100. Bates DW, Cullen DJ, Laird N, et al. Incidence of adverse drug events and potential
adverse drug events. Implications for prevention. ADE Prevention Study Group. JAMA
1995;274(1):29-34.
101. Miller LK, Nelson MS, Spurlock BW. A compendium of suggested practices for
preventing and reducing medication errors. Sacramento, CA: The California Institute for
Health Systems Performance, 2001.
http://www.cihsp.org/pdf/Articles/CompendiumFinal.pdf.
102. Flynn EA, Barker KN, Pepper GA, Bates DW, Mikeal RL. Comparison of methods for
detecting medication errors in 36 hospitals and skilled-nursing facilities. Am J Health Syst
Pharm 2002;59(5):436-46.
103. Just the facts: a basic introduction to the science underlying NCBI resources; one size does
not fit all: the promise of pharmacogenomics. Bethesda, MD: National Center for
Biotechnology Information, National Library of Medicine, 2004. (Accessed June 20, 2007, at
http://www.ncbi.nlm.nih.gov/About/primer/pharm.html.)
104. Shastry BS. Pharmacogenetics and the concept of individualized medicine.
Pharmacogenomics J 2006;6(1):16-21.
105. Valdes R, Jr., Linder MW. Fine-tuning pharmacogenetics: paradigm for linking laboratory
results to clinical action. Clin Chem 2004;50(9):1498-9.
106. General information about breast cancer. Bethesda, MD: National Cancer Institute,
National Institutes of Health, 2007. (Accessed May 17, 2007, at
http://www.cancer.gov/cancertopics/pdq/treatment/breast/patient.)
107. Diaz NM. Laboratory testing for HER2/neu in breast carcinoma: an evolving strategy to
predict response to targeted therapy. Cancer Control 2001;8(5):415-8.
108. Telenti A, Aubert V, Spertini F. Individualising HIV treatment--pharmacogenetics and
immunogenetics. Lancet 2002;359(9308):722-3.

May 2008

56

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

109. Warfarin. Bethesda, MD: Medline Plus, National Library of Medicine, National Institutes
of Health, 2007. (Accessed June 19, 2007, at
http://www.nlm.nih.gov/medlineplus/druginfo/medmaster/a682277.html.)
110. Rieder MJ, Reiner AP, Gage BF, et al. Effect of VKORC1 haplotypes on transcriptional
regulation and warfarin dose. N Engl J Med 2005;352(22):2285-93.
111. Freeman BD, McLeod HL. Challenges of implementing pharmacogenetics in the critical
care environment. Nat Rev Drug Discov 2004;3(1):88-93.
112. Phillips KA, Veenstra DL, Oren E, Lee JK, Sadee W. Potential role of pharmacogenomics in
reducing adverse drug reactions: a systematic review. JAMA 2001;286(18):2270-9.
113. Matchar DB, Thakur ME, Grossman I, et al. Testing for cytochrome P450 polymorphisms
in adults with non-psychotic depression treated with selective serotonin reuptake
inhibitors (SSRIs). Evid Rep Technol Assess (Full Rep) 2007(146):1-77.
114. Piper MA. Special report: genotyping for cytochrome P450 polymorphisms to determine
drug-metabolizer status. Chicago, IL: Blue Cross Blue Shield Technology Evaluation
Center, 2004. http://www.bcbs.com/betterknowledge/tec/vols/19/19_09.pdf.
115. Pharmacogenetics. Burlington, NC: Laboratory Corporation of America, 2007. (Accessed
May 22, 2007, at
http://www.labcorp.com/services/hcp/pharmacogenetics/pharmacogenetics.html.)
116. Ezzeldin H, Diasio R. Dihydropyrimidine dehydrogenase deficiency, a pharmacogenetic
syndrome associated with potentially life-threatening toxicity following 5-fluorouracil
administration. Clin Colorectal Cancer 2004;4(3):181-9.
117. Weiss R. Clinical laboratories in the 21st century: reengineering and changing technology.
In: Clinical diagnostic technology: the total testing process. Volume 3: the postanalytical
phase. Ward-Cook K, Lehman CA, Schoeff LE, Williams RH, ed. Washington, DC:
American Association of Clinical Chemistry, 2003.
118. Valdes R, Jr., Linder MW, Jortani SA. What is next in pharmacogenomics? Translating it to
clinical practice. Pharmacogenomics 2003;4(4):499-505.
119. Schmitz G, Aslanidis C, Lackner KJ. Pharmacogenomics: implications for laboratory
medicine. Clin Chim Acta 2001;308(1-2):43-53.
120. Roden DM, Altman RB, Benowitz NL, et al. Pharmacogenomics: challenges and
opportunities. Ann Intern Med 2006;145(10):749-57.
121. Blood facts. Charlotte, NC: American Red Cross Southeast Division, 2007. (Accessed May
22, 2007, at http://www.redcrossblood.org/division/div_facts.html.)
122. Frequently asked questions. Bethesda, MD: AABB, 2007. (Accessed May 22, 2007, at
http://www.aabb.org/Content/About_Blood/FAQ/bloodfaq.htm.)
123. Custer B. Economic analyses of blood safety and transfusion medicine interventions: a
systematic review. Transfus Med Rev 2004;18(2):127-43.
124. FDA approves first nucleic acid test (NAT) system to screen whole blood donors for
infections with human immunodeficiency virus (HIV) and hepatitis C virus (HCV).

May 2008

57

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

Rockville, MD: Food and Drug Administration, 2002. (Accessed May 23, 2007, at
http://www.fda.gov/bbs/topics/ANSWERS/2002/ANS01140.html.)
125. Blood testing. Dedham, MA: American Red Cross Blood Services- New England Region,
2007. (Accessed May 23, 2007, at http://www.newenglandblood.org/library/testing.pdf.)
126. Transfusion-transmitted diseases: viruses. Bethesda, MD: AABB, 2007. (Accessed May 23,
2007, at
http://www.aabb.org/Content/About_Blood/Facts_About_Blood_and_Blood_Banking/
fabloodtrans.htm.)
127. Saxena S, Nelson JM, Osby M, Shah M, Kempf R, Shulman IA. Ensuring timely
completion of type and screen testing and the verification of ABO/Rh status for elective
surgical patients. Arch Pathol Lab Med 2007;131(4):576-81.
128. Complications of transfusion. Whitehouse Station, NJ: Merck and Company, Incorporated,
2006. (Accessed May 23, 2007, at
http://www.merck.com/mmpe/sec11/ch146/ch146e.html.)
129. Blood transfusion errors: preventing future occurrences. Oakbrook Terrace, IL: The Joint
Commission, 1999.
http://www.jointcommission.org/SentinelEvents/SentinelEventAlert/sea_10.htm.
130. Nichols JH, Bartholomew C, Brunton M, et al. Reducing medical errors through barcoding
at the point of care. Clin Leadersh Manag Rev 2004;18(6):328-34.
131. McCaig L, Nawar E. National hospital ambulatory medical care survey: 2004 emergency
department summary. Report No. 372. Atlanta, GA: Division of Health Care Statistics,
Centers for Disease Control and Prevention, 2006.
http://www.cdc.gov/nchs/data/ad/ad372.pdf.
132. Maio RF, Waller PF, Blow FC, Hill EM, Singer KM. Alcohol abuse/dependence in motor
vehicle crash victims presenting to the emergency department. Acad Emerg Med
1997;4(4):256-62.
133. Lai MW, Klein-Schwartz W, Rodgers GC, et al. 2005 annual Report of the American
Association of Poison Control Centers' national poisoning and exposure database. Clin
Toxicol (Phila) 2006;44(6-7):803-932.
134. Wu AH, McKay C, Broussard LA, et al. National Academy of Clinical Biochemistry
laboratory medicine practice guidelines: recommendations for the use of laboratory tests
to support poisoned patients who present to the emergency department. Clin Chem
2003;49(3):357-79.
135. Alexander DR. Toxicology screen. Bethesda, MD: Medline Plus, National Library of
Medicine, National Institutes of Health, 2006. (Accessed May 24, 2007, at
http://www.nlm.nih.gov/medlineplus/ency/article/003578.htm.)
136. Phillips JE, Bogema S, Fu P, et al. Signify ER Drug Screen Test evaluation: comparison to
Triage Drug of Abuse Panel plus tricyclic antidepressants. Clin Chim Acta 2003;328(1-2):31-8.
137. Tomaszewski C, Runge J, Gibbs M, Colucciello S, Price M. Evaluation of a rapid bedside
toxicology screen in patients suspected of drug toxicity. J Emerg Med 2005;28(4):389-94.

May 2008

58

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

138. Long H, Nelson LS, Hoffman RS. A rapid qualitative test for suspected ethylene glycol
poisoning. Acad Emerg Med 2004;11(5):530.
139. Adults with co-occurring serious mental illness and a substance use disorder: the National
Survey on Drug Use and Health Report. Rockville, MD: Office of Applied Studies,
Substance Abuse and Mental Health Services Administration, 2004.
http://www.oas.samhsa.gov/2k4/coOccurring/coOccurring.pdf.
140. Schiller MJ, Shumway M, Batki SL. Utility of routine drug screening in a psychiatric
emergency setting. Psychiatr Serv 2000;51(4):474-8.
141. Institute of Medicine. Medicare: a strategy for quality assurance. Washington, DC:
National Academy Press, 1990.
142. Using measures. Rockville, MD: National Quality Measures Clearing House, Agency for
Healthcare Research and Quality, 2007. (Accessed June 15, 2077, at
http://www.qualitymeasures.ahrq.gov/resources/measure_use.aspx#attributes.)
143. Donabedian A. Definition of quality and approaches to its assessment. Ann Arbor, MI:
Health Administration Press, 1980.
144. Schuster M, McGlynn E, Pham C, Spar M, Brook R. The quality of health care in the
United States: a review of articles since 1987. In: Crossing the quality chasm: a new health
system for the 21st century. Washington, DC: National Academy Press, 2001.
145. Noble MA. An overview of the essential elements of a PT/EQA program. Division of
Laboratory Systems, Centers for Disease Control and Prevention, Atlanta, GA: 2007. Slide.
146. Institute of Medicine. Performance measurement: accelerating improvement.
Washington, DC: National Academy Press, 2006.
147. Hilborne L. Measuring, monitoring and improving quality: the role of laboratory
indicators. RAND Global Health, Los Angeles, CA: 2006. Slide.
148. Comments on proposed NCVHS recommendations for measuring quality. Testimony of
Dr. Charles Cutler, National Medical Director, Aetna National Quality Management, to
the National Committee on Vital and Health Statistics. 2004.
http://www.ncvhs.hhs.gov/040625p1.htm.
149. Measuring health care quality: obstacles and opportunities. Washington, DC: Workgroup
on Quality, National Committee on Vital and Health Statistics, 2004.
http://www.ncvhs.hhs.gov/040531rp.pdf.
150. AQA approved quality measures: diabetes. Washington, DC: AQA, 2007.
http://www.aqaalliance.org/files/ApprovedPerformanceMeasures.xls.)
151. What is HEDIS? Washington, DC: National Committee for Quality Assurance, 2007.
(Accessed June 15, 2007, at http://web.ncqa.org/tabid/187/Default.aspx.)
152. About NQMC. Rockville, MD: National Quality Measures Clearing House, Agency for
Healthcare Research and Quality, 2007. (Accessed June 15, 2007, at
http://www.qualitymeasures.ahrq.gov/about/about.aspx.)
153. NCQA timeline. Washington, DC: National Committee for Quality Assurance, 2005.
(Accessed May 23, 2007, at http://www.ncqa.org/about/timeline.htm.)

May 2008

59

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

154. The Lewin Group. The value of diagnostics innovation, adoption and diffusion into health
care: final report. Washington, DC: Advanced Medical Technology Association, 2005.
155. The state of health care quality 2006. Washington, DC: National Committee on Quality
Assurance, 2006.
156. Strombler RE, Pollock A, Taylor JR, et al. Institute for Quality in Laboratory Medicine:
Recognizing Excellence in Practice: highlights from first landmark summit--an
opportunity to enhance medical care; April 28-30, 2005; Atlanta, Georgia. MedGenMed
2005;7(3):57.
157. Edson D, Massey LD. Proficiency testing performance in physician's office, clinic, and
small hospital laboratories, 1994-2004. Laboratory Medicine 2007;38(4):237.
158. Ehrmeyer SS, Laessig RH. Has compliance with CLIA requirements really improved
quality in US clinical laboratories? Clinica Chimica Acta 2004;346(1):37-43.
159. Lippi G, Guidi GC. Preanalytic indicators of laboratory performances and quality
improvement of laboratory testing. Clin Lab 2006;52(9-10):457-62.
160. Evidence-based laboratory medicine: quality/performance measure evaluation. Atlanta,
GA: Centers for Disease Control and Prevention, 2007. (Accessed December 12, 2007, at
http://www.cdc.gov/od/pgo/funding/CI07-710.htm.)
161. Standards for pathology and clinical laboratory services. Oakbrook Terrace, IL: The Joint
Commission on Accreditation of Healthcare Organizations, 1998.
162. Zarbo RJ, Jones BA, Friedberg RC, et al. Q-Tracks: a College of American Pathologists
program of continuous laboratory monitoring and longitudinal tracking. Arch Pathol Lab
Med 2002;126(9):1036-44.
163. Hilborne L. Quality Indicators Workgroup presentation. 2005 Institute for Quality in
Laboratory Medicine Conference: Recognizing Excellence in Practice. April 29, 2005.
Atlanta, GA: Centers for Disease Control and Prevention, 2005.
164. Meyer J, Rybowski L, Eichler R. Theory and reality of value-based purchasing: lessons
from the pioneers. Report No. 98-0004. Rockville, MD: Agency for Health Care Policy and
Research, 1997. http://www.ahrq.gov/qual/meyerrpt.htm.
165. Dudley RA. Pay-for-performance research: how to learn what clinicians and policy
makers need to know. JAMA 2005;294(14):1821-3.
166. Christianson JB, Knutson DJ, Mazze RS. Physician pay-for-performance. Implementation
and research issues. J Gen Intern Med 2006;21 Suppl 2:S9-S13.
167. Baker G, Carter B. Provider pay-for-performance incentive programs: 2004 national study
results. San Francisco, CA: Med-Vantage, Incorporated, 2005.
http://www.medvantageinc.com/Pdf/MV_2004_P4P_National_Study_ResultsExec_Summary.pdf.
168. H.R. 3617--109th Congress (2005): Medicare Value-Based Purchasing for Physicians'
Services Act of 2005. GovTrack.us, 2007. (Accessed November 12, 2007, at
http://www.govtrack.us/congress/bill.xpd?bill=h109-3617.)

May 2008

60

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

169. Hearing on physician quality and efficiency. Testimony of Herb Kuhn, Acting Deputy
Administrator, Centers for Medicare and Medicaid Services, before the House Ways and
Means Subcommittee on Health, 110th Congress, 1st session. 2007.
http://waysandmeans.house.gov/Media/pdf/110/5-10-07/KUHNTestimony.pdf.
170. Statement of Mark B. McClellan, MD, Ph.D., Administrator, Centers for Medicare and
Medicare Services on value-based purchasing for physicians under Medicare before the
House Ways and Means Subcommittee on Health, 109th Congress, 1st session. 2005.
http://www.hhs.gov/asl/testify/t050721.html.
171. Klein S. Issue of the month: Medicare P4P demo pushes physician care for chronically ill.
The Commonwealth Fund 2006;20.
172. AMA approves CAP pay for performance measures. Stateline: Federal and State News
Briefs 2007;23(12).
173. Petersen LA, Woodard LD, Urech T, Daw C, Sookanan S. Does pay-for-performance
improve the quality of health care? Ann Intern Med 2006;145(4):265-72.
174. Koo D. Overview of public health surveillance. Epidemiology Program Office, Centers for
Disease Control and Prevention, Atlanta, GA: 2006. Slide.
175. Emori TG, Gaynes RP. An overview of nosocomial infections, including the role of the
microbiology laboratory. Clin Microbiol Rev 1993;6(4):428-42.
176. Ranji SR, Shetty K, Posley KA, et al. Prevention of healthcare-associated infections. Report
No. 04(07)-0051-6. Rockville, MD: Agency for Healthcare Research and Quality, 2007.
http://www.ahrq.gov/downloads/pub/evidence/pdf/qualgap6/hainfgap.pdf.
177. Life after NNIS: National Healthcare Safety Network. National Center for Infectious
Diseases, Centers for Disease Control and Prevention, Atlanta, GA: 2002. Slide.
178. About NNIS. Atlanta, GA: Division of Healthcare Quality Promotion, Centers for Disease
Control and Prevention, 2005. (Accessed May 25, 2007, at
http://www.cdc.gov/ncidod/dhqp/nnis.html.)
179. Monitoring hospital-acquired infections to promote patient safety--United States, 19901999. MMWR Morb Mortal Wkly Rep 2000;49(8):149-53.
180. Antimicrobial resistance: issues and options. Harrison PF, Lederberg J, eds. Washington,
DC: National Academy Press, 1998.
181. Song X, Srinivasan A, Plaut D, Perl TM. Effect of nosocomial vancomycin-resistant
enterococcal bacteremia on mortality, length of stay, and costs. Infect Control Hosp
Epidemiol 2003;24(4):251-6.
182. Wilson SJ, Knipe CJ, Zieger MJ, et al. Direct costs of multidrug-resistant Acinetobacter
baumannii in the burn unit of a public teaching hospital. Am J Infect Control
2004;32(6):342-4.
183. Qavi A, Segal-Maurer S, Mariano N, et al. Increased mortality associated with a clonal
outbreak of ceftazidime-resistant Klebsiella pneumoniae: a case-control study. Infect
Control Hosp Epidemiol 2005;26(1):63-8.

May 2008

61

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

184. Management of multidrug-resistant organisms (MDRO) in healthcare settings:
CDC/HICPAC, October 2006, introduction and highlights. Office of Epidemiology and
Disease Control Programs, Department of Health and Mental Hygiene, State of Maryland:
2006. Slide.
185. Boyce JM, Jackson MM, Pugliese G, et al. Methicillin-resistant Staphylococcus aureus
(MRSA): a briefing for acute care hospitals and nursing facilities. The AHA Technical
Panel on Infections Within Hospitals. Infect Control Hosp Epidemiol 1994;15(2):105-15.
186. National Nosocomial Infections Surveillance (NNIS) system report, data summary from
January 1992 through June 2003, issued August 2003. Am J Infect Control 2003;31(8):481-98.
187. Jones RN. Method preferences and test accuracy of antimicrobial susceptibility testing:
updates from the College of American Pathologists Microbiology Surveys Program. Arch
Pathol Lab Med 2001;125(10):1285-9.
188. National Antimicrobial Resistance Monitoring System (NARMS): enteric bacteria. Atlanta,
GA: Centers for Disease Control and Prevention, 2007. (Accessed May 31, 2007, at
http://www.cdc.gov/narms/.)
189. National Antimicrobial Resistance Monitoring System for Enteric Bacteria (NARMS):
human isolates final report, 2004. Atlanta, GA: Centers for Disease Control and
Prevention, 2004.
190. Epi and Lab Capacity Program: home. Atlanta, GA: Centers for Disease Control and
Prevention, 2005. (Accessed June 1, 2007, at
http://www.cdc.gov/ncidod/osr/site/epi_lab/index.htm.)
191. Sentinel events. Oakbrook Terrace, IL: The Joint Commission, 2006. (Accessed June 12,
2007, at http://www.jointcommission.org/NR/rdonlyres/690008C7-EAB2-4275-BC7B68B37481D658/0/SE_Chap_Sept06.pdf.)
192. The National Laboratory System--public and private benefits. Centers for Disease Control
and Prevention, Atlanta, GA: 2007. Slide.
193. PHIN: overview. Atlanta, GA: Centers for Disease Control and Prevention, 2007.
(Accessed March 15, 2007, at http://www.cdc.gov/phin/about.html.)
194. PHIN: frequently asked questions. Atlanta, GA: Centers for Disease Control and
Prevention, 2007. (Accessed March 15, 2007, at http://www.cdc.gov/phin/faq.html.)
195. PHIN preparedness: connecting laboratory systems functional requirements and process
flows. Atlanta, GA: Centers for Disease Control and Prevention, 2005.
http://www.cdc.gov/phin/preparedness/CLS_RSv1.0.pdf.
196. Early Warning Infectious Disease Surveillance (EWIDS) program activities on the
Northern and Southern border states. Atlanta, GA: Centers for Disease Control and
Prevention, 2007. (Accessed March 15, 2007, at
http://www.bt.cdc.gov/surveillance/ewids/.)
197. HHS announces Early Warning Infectious Disease Surveillance project with Mexico.
Washington, DC: Office of Global Health Affairs, 2006. (Accessed March 15, 2007, at
http://www.globalhealth.gov/MexicoEWIDS.shtml.)

May 2008

62

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

198. Facts about the Laboratory Response Network. Atlanta, GA: Centers for Disease Control
and Prevention, 2006. (Accessed March 15, 2007, at
http://www.bt.cdc.gov/lrn/factsheet.asp.)
199. CDC laboratory preparedness for emergencies: CDC's laboratory response to suspicious
substances. Atlanta, GA: Centers for Disease Control and Prevention, 2004.
http://www.bt.cdc.gov/labissues/pdf/substanceresponse.pdf.
200. The role of laboratories and blood banks in disaster situations. Washington, DC: Pan
American Health Organization, Regional Office of the World Health Organization, 2002.
http://www.paho.org/English/AD/THS/EV/LAB_laboratories.pdf.
201. Kost GJ, Tran NK, Tuntideelert M, Kulrattanamaneeporn S, Peungposop N. Katrina, the
tsunami, and point-of-care testing: optimizing rapid response diagnosis in disasters. Am J
Clin Pathol 2006;126(4):513-20.
202. Townsend FF. The federal response to Hurricane Katrina: lessons learned. Washington,
DC: The White House, 2006. http://www.whitehouse.gov/reports/katrina-lessonslearned.pdf.
203. Grosse SD, Teutsch SM, Haddix AC. Lessons from cost-effectiveness research for United
States public health policy. Annu Rev Public Health 2007;28:365-91.
204. Garber AM. Cost-effectiveness and evidence evaluation as criteria for coverage policy.
Health Aff (Millwood) 2004;Suppl Web Exclusives:W4-96.
205. Medicare program; criteria and procedures for making medical services coverage decision
that relate to health care technology. Fed Regist 1989;54(30):4302-18.
206. Goodman C, Karnes E, Faulkner E, et al. Cost-effectiveness considerations in the approval
and adoption of new health technologies. Prepared for the Office of the Assistance
Secretary for Planning and Evaluation, Department of Health and Human Services
(Contract No. HHSP23300005T). Washington, DC: Office for Science and Data Policy,
Office of the Assistance Secretary for Planning and Evaluation, Department of Health and
Human Services, 2007. http://aspe.hhs.gov/sp/reports/2007/cecht/report.pdf.
207. Bergthold LA, Singer SS, Huang A, Gemperli M, Garber A, Osterhoff R. Using evidence
and cost in managed care decision-making. Health Affairs, 2002.
http://content.healthaffairs.org/cgi/data/hlthaff.w4.284v1/DC2/1.
208. Cost-effectiveness in health and medicine. Gold MR, Siegel JE, Russell LB, Weinstein MC,
eds. New York, NY: Oxford University Press, 1996.
209. Siegel JE. Cost-effectiveness analysis in US healthcare decision-making: where is it going?
Med Care 2005;43(7 Suppl):1-4.
210. Luce BR. What will it take to make cost-effectiveness analysis acceptable in the United
States? Med Care 2005;43(7 Suppl):44-8.
211. Neumann PJ, Rosen AB, Weinstein MC. Medicare and cost-effectiveness analysis. N Engl J
Med 2005;353(14):1516-22.
212. Veenstra DL, Higashi MK, Phillips KA. Assessing the cost-effectiveness of
pharmacogenomics. AAPS PharmSci 2000;2(3):1-11.

May 2008

63

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

213. Guideline to the methods of technology appraisal. London, England: National Institute
for Clinical Excellence, 2004. http://www.nice.org.uk/download.aspx?o=201973.
214. Griffith GL, Edwards RT, Gray J, et al. Estimating the survival benefits gained from
providing national cancer genetic services to women with a family history of breast
cancer. Br J Cancer 2004;90(10):1912-9.
215. Giacomini M, Miller F, O'Brien BJ. Economic considerations for health insurance coverage
of emerging genetic tests. Community Genetics 2003;6(2):61-73.
216. Rogowski W. Genetic screening by DNA technology: a systematic review of health
economic evidence. Int J Technol Assess Health Care 2006;22(3):327-37.
217. Carlson JJ, Henrikson NB, Veenstra DL, Ramsey SD. Economic analyses of human
genetics services: a systematic review. Genetics in Medicine 2005;7(8):519-23.
218. Jarrett J, Mugford M. Genetic health technology and economic evaluation: a critical
review. Applied Health Economics and Health Policy 2006;5(1):27-35.
219. Flowers CR, Veenstra D. The role of cost-effectiveness analysis in the era of
pharmacogenomics. Pharmacoeconomics 2004;22(8):481-93.
220. Hernandez JS. Cost-effectiveness of laboratory testing. Arch Pathol Lab Med
2003;127(4):440-5.
221. Hu D, Hook EW, III, Goldie SJ. Screening for Chlamydia trachomatis in women 15 to 29
years of age: a cost-effectiveness analysis. Ann Intern Med 2004;141(7):501-13.
222. Maciosek MV, Coffield AB, Edwards NM, Flottemesch TJ, Goodman MJ, Solberg LI.
Priorities among effective clinical preventive services: results of a systematic review and
analysis. Am J Prev Med 2006;31(1):52-61.
223. Decision memo for screening immunoassay fecal-occult blood test (CAG-00180N).
Baltimore, MD: Centers for Medicare and Medicaid Services, 2007. (Accessed June 10,
2007, at http://www.cms.hhs.gov/mcd/viewdecisionmemo.asp?id=87.)
224. van Ballegooijen M, Habbema JDF, Boer R, Zauber AG, Brown ML. Report to the Agency
for Healthcare Research and Quality: a comparison of the cost-effectiveness of fecal occult
blood tests with different test characteristics in the context of annual screening in the
Medicare population. Rockville, MD: Agency for Healthcare Research and Quality, 2003.
225. Dendukuri N, Khetani K, McIsaac M, Brophy J. Testing for HER2-positive breast cancer: a
systematic review and cost-effectiveness analysis. CMAJ 2007;176(10):1429-34.
226. Elkin EB, Weinstein MC, Winer EP, Kuntz KM, Schnitt SJ, Weeks JC. HER-2 testing and
trastuzumab therapy for metastatic breast cancer: a cost-effectiveness analysis. J Clin
Oncol 2004;22(5):854-63.
227. Ubel PA, Hirth RA, Chernew ME, Fendrick AM. What is the price of life and why doesn't
it increase at the rate of inflation? Arch Intern Med 2003;163(14):1637-41.
228. Garrison LP, Jr., Lubeck D, Lalla D, Paton V, Dueck A, Perez EA. Cost-effectiveness
analysis of trastuzumab in the adjuvant setting for treatment of HER2-positive breast
cancer. Cancer 2007;110(3):489-98.

May 2008

64

Laboratory Medicine: A National Status Report

Chapter I – The Value of Laboratory Medicine to Health Care

229. AuBuchon JP, Birkmeyer JD, Busch MP. Safety of the blood supply in the United States:
opportunities and controversies. Ann Intern Med 1997;127(10):904-9.
230. van HM, de Wolf JT, Staginnus U, Ruitenberg EJ, Postma MJ. Pharmaco-economics of
blood transfusion safety: review of the available evidence. Vox Sang 2002;83(2):146-55.
231. Jackson BR, Busch MP, Stramer SL, AuBuchon JP. The cost-effectiveness of NAT for HIV,
HCV, and HBV in whole-blood donations. Transfusion 2003;43(6):721-9.
232. Marshall DA, Kleinman SH, Wong JB, et al. Cost-effectiveness of nucleic acid test
screening of volunteer blood donations for hepatitis B, hepatitis C and human
immunodeficiency virus in the United States. Vox Sang 2004;86(1):28-40.

May 2008

65

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

CHAPTER II
MARKET PROFILE OF THE LABORATORY MEDICINE SECTOR
This chapter provides an overview of the size and structure of the U.S. laboratory sector,
including revenue, spending, test volume, and other key elements. Market information is
delineated by laboratory setting and test type. Settings include hospitals, physicians’ offices,
other clinical settings (e.g., skilled nursing facilities), and independently operated sites in the
community. Examining market by test type highlights trends in the traditional broad disciplines
of clinical pathology and anatomic pathology as well as the emerging interdisciplinary areas
associated with molecular pathology. Depending on the type of test and the state of origin,
consumers may perform self testing at home, work, or another site, or order tests directly through
a clinical laboratory or the Internet.
Publicly available information about the economic status and quality of the laboratory medicine
sector is limited. For purposes of this chapter, data were compiled from multiple public and
private sector sources. Among these sources were market research reports, such as Lab Industry
Strategic Outlook: Market Trends and Analysis 2007a, the CMS OSCAR database, which contains
information about laboratory certification, accreditation, and proficiency testing; other
government reports; published literature; and personal communications with industry experts
and government officials.

U.S. MARKET SIZE
The revenue, spending, and test volume of the U.S. clinical laboratory testing market has grown
steadily over the past decade. Market data indicate that:

a



Revenues have increased by more than 40% since 1998 (not adjusted for inflation). In
2006, laboratory industry revenues (i.e., gross earnings) were valued at approximately
$48.5 billion, with predicted growth of 6.5% to $51.7 billion in 2007.1



Spending growth on clinical laboratory services averaged 7% annually over the 2003 to
2006 period, just slightly less than the growth rate in total national health care
spending at 8%.1 However, laboratory expenditures as a percentage of total health care
spending have remained relatively stable at 2 to 3% from 1998 to 2007 (see Figure 2.1).

The Lab Industry Strategic Outlook: Market Trends and Analysis, published by Washington G-2 Reports, compiled data
from its own surveys; proprietary surveys of hospitals and independent laboratories; data from CMS; financial
reports filed with the Securities and Exchange Commission; population and business data from the U.S. Census
Bureau; and interviews with laboratory industry executives, hospital laboratory managers, consultants, and
government executives. One of the surveys conducted for the 2007 report was sent to 12,000 laboratories in their
database, of which 141 responded. Approximately 55% of respondents were hospital/health system laboratories,
17% were independent or commercial laboratories, 2% were pathology groups, 7% were POLs, and 19% were
categorized as “other” (i.e., a combination of one or more type of laboratory). However, because of the very low
response rate, the results of this survey may be limited in terms of generalizability to the larger laboratory sector.

May 2008 v2

67

Laboratory Medicine: A National Status Report



Chapter II - Market Profile of the Laboratory Medicine Sector

Total volume of laboratory testing was projected to be approximately 6.8 billion tests in
February 2008, according to data self reported by laboratories and compiled in the CMS
OSCAR database.2 However, higher figures have been reported by another organization.b

Expenditures in billions of dollars

Figure 2.1: Total Health Care and Laboratory Expenditures, United States
1998 - 2007

$2,000
$1,500
$1,000
$500
$0
1998

1999

2000

2001

2002

Laboratory testing expenditures

2003

2004

2005

2006

2007

Health care expenditures

Sources: Terry M. Lab industry strategic outlook: market trends and analysis 2007. New York, NY: Washington G-2 Reports, 2007.
National health expenditures by type of service and source of funds: CY 1960-2005. Baltimore, MD: Centers for Medicare and
Medicaid Services, 2007. (Accessed July 23, 2007, at
http://www.cms.hhs.gov/NationalHealthExpendData/02_NationalHealthAccountsHistorical.asp#TopOfPage.)
National health expenditures projections 2006-2016, forecast summary and selected tables. Baltimore, MD: Centers for
Medicare and Medicaid Services, 2006. (Accessed July 23, 2007, at
http://www.cms.hhs.gov/NationalHealthExpendData/03_NationalHealthAccountsProjected.asp#TopOfPage.)

Many factors contribute to market growth in the laboratory sector, including:

b



Aging of the population and corresponding increase in the prevalence of chronic
diseases, which are highly dependent on use of laboratory testing and services for
diagnosis, treatment, and ongoing management. More than 90 million Americans live
with chronic illnesses, and about half of adults aged 65 and older live with at least two
chronic medical conditions.3, 4



Growth of the U.S. population from 266 million in 1997 to 301 million in 2007, a 13%
increase that has contributed to higher rates of spending on laboratory tests.1, 5, 6

The true number of laboratory tests conducted annually in the U.S. is not known. A 1996 report published by the
National Inventory of Clinical Laboratory Testing Services estimated the annual number of tests to be 7.25 billion.
Using the OSCAR database, CDC estimated annual volume at 6.8 billion tests in February 2008 (as cited above).2
Yet, there are some limitations to the data. First, laboratories with a certificate of waiver usually do not provide
updates of test volume after the initial application. Second, OSCAR data is self-reported by laboratories and is
not confirmed by CMS or any other organization. Lastly, CLIA may count tests differently than other laboratories
or other organizations (e.g., for chemistry profiles, CLIA counts each individual analyte separately).

May 2008 v2

68

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

Approximately 60% of this population growth is attributed to the net difference
between birth and death rates; 40% is attributed to immigration.7



Research in molecular diagnostics and development of new tests in the areas of genomics
and proteomics to support more targeted clinical care.8, 9 The U.S. government
continues to devote great resources to expand research beyond the Human Genome
Project. Such efforts are complemented by a vibrant, innovative private sector.



Continued miniaturization of testing equipment, increasing use of point-of-care devices,
and development of high-throughput automated systems. Great advances in
miniaturization of assay technologies enable more efficient, high-volume testing.10, 11



Increased incorporation of laboratory testing as an indicator of quality of care and provider
performance. Evidence continues to accumulate on the association of biomarkers with
health care outcomes of interest to consumers, purchasers, and payers.12, 13



Increased media attention to health-related issues has increased consumer awareness of
and requests for certain laboratory tests.1, 14 For example, a 2003 report by CDC found
that providers in metropolitan areas where a genetic test for breast and ovarian cancer
susceptibility was marketed directly to consumers ordered more tests.15

MARKET BY LABORATORY TYPE AND SETTING
Laboratory types vary according to their settings, including hospitals, POLs, independent
laboratories, and public health laboratories, among others. Laboratories must register with
CMS, obtain the appropriate certificate, and comply with CLIA requirements. Under CLIA,
laboratory tests are categorized as either waived or non-waived. Laboratories performing
waivedc testing must obtain a certificate of waiver (CW), while those conducting non-waived
PPM must obtain a certificate for PPM procedures. Laboratories conducting other non-waived
testing must obtain a certificate of compliance (COC) following inspection or a certificate of
accreditation (COA) from a CMS-approved private sector accreditation organization.
The number of CLIA-certified laboratories in the U.S. has increased steadily since
implementation of the regulations. The number of laboratories grew 28% from 154,740 in 1993
to 198,232 in 2006.16 According to CMS, there were 203,939 CLIA-certified laboratories
providing testing services in the U.S. as of December 2007.17 Figure 2.2 depicts the breakdown
of the most common laboratories by type.

c

Waived tests are those whose methods are judged to be sufficiently simple and accurate that the likelihood of
erroneous results is negligible, that they pose no reasonable risk of harm to the patient if performed incorrectly,
and that have been cleared by FDA for home use.

May 2008 v2

69

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

Figure 2.2: Most Common Types of Laboratories, 2006

End Stage Renal Disease
Dialysis Facility
2.1%

Other
21.5%

Independent
2.7%

Physician Office
53.6%

Community Clinic
3.3%
Hospital
4.4%
Home Health Agency
5.1%
Skilled Nursing Facility/
Nursing Facility
7.4%

Source: CLIA update—December 2006, laboratories by type of facility.
Baltimore, MD: Centers for Medicare and Medicaid Services, 2006.

Although the raw number of hospital laboratories displayed in Figure 2.2 is relatively small,
they held the largest market share by both test volume and revenue in 2006, as presented in
Figure 2.3.1 Independent laboratories had the second-highest market share, followed by POLs.
Figure 2.3: U.S. Laboratory Industry Market Share by Test Volume, 2006
Other
5%

POL
8%

Independent
32%

Hospital
55%

Source: CMS-CLIA laboratory application forms. Terry M.
Lab industry strategic outlook: market trends and analysis
2007. New York, NY: Washington G-2 Reports, 2007.

May 2008 v2

70

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

Hospital-based Laboratories
Hospital laboratories are the chief provider of laboratory services for their inpatient population
as well as outpatient population receiving care from physicians who are affiliated with the
hospital.18 Outpatient laboratory testing is conducted on patients who are receiving care from
providers affiliated with the hospital but who have not been admitted to the hospital. Also,
hospital laboratories often conduct outreach testing, serving as the reference laboratory for
others in the community with limited testing capabilities.1
In 2006, there were 8,680 hospital-based clinical laboratories in the U.S., which together conducted
about 3.3 billion laboratory tests.16, 19 The number of hospital laboratories increased by 6%
between 2000 and 2004, but decreased by 0.8% between 2004 and 2006 due to competition from
independent laboratories and consolidation through mergers and acquisitions.1 Generally, the
number of hospital-based laboratories exceeds the number of hospitals (5,756 hospitals in 2006).20,
21 In addition to a main laboratory, hospitals may operate laboratories in their emergency
department, intensive care unit, pulmonary service, surgical service, and satellite sites.22-24
Revenue. In 2007, revenues for hospital laboratories are projected to reach $28.4 billion, an
increase of more than 6% over 2006.1
Volume. In 2005, hospital test volume grew by a median of 5% and an average of 6% over 2004,
including inpatient and outpatient testing.1 Hospital laboratories with the lowest test volumes
(fewer than 250,000 tests per year) had the highest growth rates (see Table 2.1).
Table 2.1: Hospital Laboratory Median and Average Test Volume Growth, 2005
Annual Volume

Median Growth

Average Growth

<250,000

6.4%

7.5%

250,000 – 499,999

4.3%

5.6%

500,000 - 999,999

5.0%

5.9%

>1 million

5.0%

6.7%

Overall

5.0%

6.4%

Source: Reprinted with permission from Washington G-2 reports. Third annual outreach
survey. Terry M. Lab industry strategic outlook: market trends and analysis 2007. New
York, NY: Washington G-2 Reports, 2007.

Budget. A 2005 survey of laboratories found that about 36% of hospital laboratories operated
with a budget of less than $5 million per year, while the budget of another 37% was between $5
million and $20 million.1 Over one-third of these budgets is spent on staff salaries, more than
20% on supplies, nearly 9% on employee benefits, and approximately 7% on blood-related
expenses. The remaining 30% is devoted to rent, repairs, internal test transfers, and other
expenses. About half of the laboratories responding to the survey cited staff salaries as the
fastest-growing component of their budgets.1

May 2008 v2

71

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

Staff. Approximately 60% of laboratory technologists/scientistsd and 43% of technicians
worked in laboratories based in general medical and surgical hospitals in 2006.25, 26 The
percentages of these professionals employed in general medical and surgical hospitals has
remained steady since 2002.

Hospital Outreach Testing
Hospital laboratories that perform outreach testing function as reference laboratories for other
hospitals, community clinics, POLs, and other facilities.27 Specimens tested through outreach
services are rarely collected by the outreach laboratory itself.28
Hospitals have marketed outreach testing services to generate additional revenue and compete
with independent laboratories. While 74% of hospital laboratories actively marketed their
outreach services in 2003, 91% marketed these services in 2005.1 Outreach testing volumes
increased from 16% of hospital laboratory testing volume (2003) to 29% (2005) as a result of
these marketing activities. Profitability has risen as well—nearly 85% of hospital laboratory
outreach programs surveyed in 2005 reported a profit. Figure 2.4 provides a comparison of
hospital inpatient, outpatient, and outreach testing.
Figure 2.4: Components of Hospital Laboratory Testing, 2003 and 2005
60.0%

51.0%
45.2%

50.0%
40.0%

33.0%

29.2%

25.2%

30.0%

16.0%

20.0%
10.0%
0.0%
Inpatient Testing
(patients admitted to hospital)

Outpatient Testing
(non-admitted patients)

2003

Outreach Testing
(specimens from patients not
affiliated with the hospital)

2005

Source: Reprinted with permission from Washington G-2 reports. First national hospital laboratory survey and third annual
outreach survey. Terry M. Lab industry strategic outlook: market trends and analysis 2007. New York, NY: Washington G-2
Reports, 2007.

d

Clinical laboratory technologists/scientists conduct laboratory tests on tissues, blood, and other bodily fluids and
perform a full range of complex chemical, biological, hematological, and immunologic tests. Laboratory technicians
work under the supervision of a technologist/scientist and conduct less complex tests in all areas of the laboratory.

May 2008 v2

72

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

To further increase competitiveness, many outreach laboratories have invested in Internet
connectivity, improved billing and pricing of tests, and focused on faster test TATs. The
efficiency of their billing systems is close to that of their competitors.1 Efficiency gains, as
determined by the number of days in accounts receivable,e are depicted in Figure 2.5. In 2006,
more than 45% of hospital outreach laboratories reported that they were holding their market
share and 38% reported that they were gaining market share.
Figure 2.5: Median Days in Accounts Receivable, 2003 versus 2005
120

100
Number of Days

100
80
60

48

46

53

60

52

40
20
0
Quest Diagnostics

LabCorp
2003

Hospital Outreach
2005

Source: Park City Solutions (now Chi Solutions, Inc.). Fourth comprehensive national laboratory outreach survey 2005. Ann
Arbor, MI: Chi Solutions, Inc., 2005.
Reprinted with permission from Washington G-2 reports. First national hospital laboratory survey and third annual
outreach survey. Terry M. Lab industry strategic outlook: market trends and analysis 2007. New York, NY: Washington G-2
Reports, 2007.

Physician Office Laboratories
POLs comprise the largest portion of clinical laboratories in the sector. As of December 2006,
there were 106,190 POLs, about 54% of the total number of CLIA-certified laboratories in the
U.S.16 From 2000 to 2006, the number of POLs increased 12% and the total number of all
laboratories increased by approximately 17%.16, 30
Testing in POLs tends to be limited to a small number of specimens and is often conducted by
medical assistants.31 On-site testing in POLs allows for immediate availability of results to
clinicians.21, 32 More than 80% of POLs are certified to perform only waived and/or PPM tests.1
However, some POLs that serve large group medical practices are certified to perform certain

e

Days in accounts receivable can be calculated by determining the number of days between date of service to date of
payment and averaging across all claims or by calculating the ratio of accounts receivable to average daily
charges and multiplying by 365 days.29 Days in accounts receivable measures a company’s billing efficiency; a
higher figure is inversely related to lower billing efficiency.1

May 2008 v2

73

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

moderate and high complexity tests that are typically provided by hospital and independent
laboratories.
Data provided by the American Academy of Family Physicians estimate that nearly 50% of testing
conducted in family physician POLs is waived, 13% of testing is PPM, 22% is moderate
complexity (e.g., tests to measure theophylline levels), and 4% is high complexity (e.g., tests to
measure antibiotic susceptibility).33 Tests commonly performed in family physician POLs are
highlighted in Table 2.2.f
Table 2.2: Ten Most Commonly Offered Laboratory Tests at Family
Physician’s Offices and Percent of Offices Performing Test On-Site, 2005
Name of Laboratory Test

Percent of Physician’s Offices
Performing Test On-Site

Dipstick/tablet urinalysis

97.5%

Fecal occult blood

92.2%

Urine pregnancy test

87.2%

Rapid strep (direct antigen)

86.5%

Vaginal smear/wet mount

76.6%

Glucose, using a waived instrument

68.3%

Urine microscopic exam

61.1%

Infectious mononucleosis screen

39.2%

Prothrombin time

39.0%

Glucose, visual whole blood glucose dipstick
(performed via fingerprick)

35.3%

Sources: American Academy of Family Physicians. Performance of laboratory services in family
physician practice settings. Practice Profile II Survey, October 2006. Leawood, KS: American Academy
of Family Physicians, 2006.
Reprinted with permission from Washington G-2 Reports. Terry M. Lab industry strategic outlook:
Market trends and analysis 2007. New York, NY: Washington G-2 Reports, 2007.

Revenue. In 2006, POLs generated $2.5 billion in revenues, comprising 5% of total revenues for
the industry. Revenues from POLs increased between 2000 and 2003 by approximately 22%;
however, slowed growth in the number of POLs is reflected in the small but consistent revenue
declines since 2003 (see Figure 2.6). Decreasing revenue also is attributed to shifts in the types of,
and payments for, tests provided by POLs. Manufacturers have significantly increased the
availability and marketing of waived tests to POLs, promoting ease of use, point-of-care access to
test results, and convenience for the patient. The shift toward less expensive testing also has
been fueled by declining reimbursement rates. Medicare reimbursement for the top 20 tests
provided by POLs in 2006 averaged almost $1.00 less than that for the top 20 POL tests in 2002.g,1

Dipstick urinalysis, fecal occult blood, and streptococcal antigen detection also were among the top five most commonly
offered tests according to two surveys, one conducted in 2003 by CMS of CW sites and one conducted of pediatricbased POLs in 1996 by the Illinois chapter of the American Academy of Family Physicians.34, 35
g This comparison was based on the difference between the average reimbursement for the top 20 tests in 2002 and
2006; the sets of top 20 tests differed in the two years.
f

May 2008 v2

74

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

Revenue in billions of dollars

Figure 2.6: Physician Office Laboratory Revenue, 2000 - 2006
$3.5
$3.0
$2.5

$2.3

$2.4

2000

2001

$2.6

$2.8

$2.7

$2.6

$2.5

2004

2005

2006

$2.0
$1.5
$1.0
$0.5
$0.0
2002

2003

Source: Reprinted with permission from Washington G-2 Reports. Terry M. Lab industry strategic outlook: market trends and
analysis 2007. New York, NY: Washington G-2 Reports, 2007.

Volume. POLs conduct an estimated 525 million laboratory tests annually; approximately 29.5%
of these tests are waived.19 POLs accounted for approximately 8% of the total number of
laboratory tests performed in the U.S. in 2005, a decrease of 3% from 2003.1
Staff. From 2002 to 2005, the proportion of the clinical laboratory workforce employed in POLs
declined, including a drop from 10% to 8% for technologists/scientists and 18% to 17% for
technicians (i.e., as a percentage of all technologists/scientists and technicians in the U.S.).25, 26, 36, 37
This decrease may be due to slower revenue growth among POLs (and subsequently, fewer funds
available for salaries) between 2003 and 2006. The decrease in the number of skilled laboratory
personnel is linked to the increase in waived testing during this time.35 The decline in the number
of technologists/scientists and technicians employed in POLs accompanies the increasing number
of POLs holding only a CW, under which there are no personnel requirements.

Independent Laboratories
In 2006, there were 5,414 CLIA-certified, privately-owned independent laboratories in the U.S.
operated by non-profit and for-profit corporations.16 Privately-owned independent laboratories
conduct approximately 1.5 billion tests annually. Approximately 74% of these laboratories are
certified to perform non-waived testing.19
The independent laboratory sector accounts for 32% of clinical laboratory testing and 35% of the
revenue produced by the industry.1 From 2000 to 2006, revenue produced by independent
laboratories grew from $10.6 billion to $15.5 billion, an increase of approximately 46% (see Figure
2.7). Growth in independent laboratory revenue is expected to continue at a rate of nearly 6% to
$16.5 billion in 2007. Table 2.3 provides market data of the seven publicly traded companies with
the highest revenue in the first half of 2005.

May 2008 v2

75

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

Figure 2.7: Independent Laboratory Revenue, 2000 – 2007

Revenue in billions of dollars

$18.0
$16.0
$14.0
$12.0

$10.6

$11.7

$12.4

$13.0

$13.7

$14.6

$15.5

$16.5

$10.0
$8.0
$6.0
$4.0
$2.0
$0.0
2000

2001

2002

2003

2004

2005

2006

2007

Source: Reprinted with permission from Washington G-2 Reports. Terry M. Lab industry strategic outlook: market trends and
analysis 2007. New York, NY: Washington G-2 Reports, 2007.

Table 2.3: Publicly Traded Laboratory Companies with Highest Revenue
First Half of 2005
Company

Revenue First Half 2005

Quest Diagnostics

$2.70 billion

LabCorp

$1.65 billion

AmeriPath

$278 million

LabOne

$252 million

Genzyme Genetics

$107 million

Bio-Reference*

$76.9 million

Specialty Laboratory

$75.7 million

*Bio-Reference’s revenue is for six months ended April 30
Source: Reprinted with permission from Washington G-2 Reports. Laboratory
Industry Report from company reports. Terry M. Lab industry strategic outlook:
market trends and analysis 2007. New York, NY: Washington G-2 Reports, 2007.

Market Competition. Small- and mid-sized independent laboratories, as well as hospital outreach
laboratories, face significant competition from the large laboratory corporations, which have
sizable financial resources and economies of scale. Eleven mid-sized independent laboratories
with annual revenues between $10 and $500 million had combined total revenues of $1.04 billion
in 2005, a 19% increase from 2004. According to survey data, three small, independent
laboratories with annual revenue of less than $10 million estimated an increase in revenue of 15%
between 2006 and 2007.1

May 2008 v2

76

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

Several factors have contributed to the competitive advantages of the large corporations:
national managed care contracts; efficient, centralized billing management; lower supply costs;
extensive high complexity testing capabilities; and the ability to invest in Web-based systems.
The two largest laboratories have contracts with the three largest managed care organizations
(i.e., UnitedHealthcare, Cigna, and Aetna).1 These contracts generated 42 to 50% of their total
revenues in 2006.
Given their test volume, large laboratories are able to negotiate more favorable contracts with
reagent and supply vendors, sometimes at costs 30 to 50% less than those paid by hospitals and
smaller independent laboratories. Financial resources have allowed large laboratories to build
capacity for high complexity testing, upgrade billing management systems, and invest in Webbased systems to improve efficiency in ordering, billing, and result reporting. However, the sheer
size of their operations can result in disadvantages in physician communication, specimen pickup
scheduling, and test TATs, areas in which smaller laboratories can excel. Smaller laboratories are
often located closer to their clients, facilitating greater laboratory-client interaction. They also may
offer more flexibility in scheduling specimen pickup times than large laboratories that are tied to
specific times for daily specimen pick ups.1

Public Health Laboratories
Public health laboratories are government laboratories dedicated to safeguarding the public’s
health.38 While their responsibilities and roles intersect at many points, public health laboratories
generally do not compete with private clinical laboratories because their focus is on population
health and security rather than individual patient testing. Responsibilities of public health
laboratories include:



Specialized disease testing to detect and monitor newly emerging infectious diseases,
such as West Nile virus, severe acute respiratory syndrome, and new influenza strains



Newborn screening to detect potentially life-threatening metabolic and genetic
disorders, such as phenylketonuria, sickle cell disease, and cystic fibrosis



Molecular analyses to differentiate one strain of a disease organism from another (e.g.,
E. coli O157:H7, which causes foodborne illness)



Confirmatory testing to verify the identity of microbes and other suspect agents



Testing to detect the presence of sexually-transmitted diseases



Testing to detect the presence of bacteria, parasites, pesticides, and other potentially
harmful agents in the environment, especially drinking water



Routine surveillance of public health threats and emergencies by analyzing samples,
providing information to support effective response, and working with other health
authorities to protect citizens38-40

State Laboratories. Public health laboratories include state and local facilities. There are a total of
56 state public health laboratories in the U.S., including one in every state and U.S. territory.h,41
State public health laboratories, usually part of the state health agency, are responsible for
h

U.S. territories include American Samoa, Guam, the Marshall Islands, Puerto Rico, the Mariana Islands, and the U.S.
Virgin Islands.

May 2008 v2

77

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

participating in national public health surveillance activities and monitoring disease trends.38
Often, state public health laboratories provide training to workers in private sector and local
public health laboratories, and in some states provide regulatory oversight of clinical and/or local
public health laboratories.
Local Laboratories. Local public health laboratories function at the city or county level. The
Association of Public Health Laboratories estimates that there are between 600 and 800 local
public health laboratories in the U.S. (The precise number and capabilities of these laboratories
varies according to how the laboratory is defined.)41 Most states have several local public health
laboratories that range from large urban laboratories employing hundreds of scientists to small
rural laboratories staffed by one or two scientists.39
In general, local public health laboratories have greater familiarity with local health problems and
stronger ties to their communities than state-level laboratories.42 A 2003 survey of local public
health agencies found that more than two-thirds rely on in-house public health laboratories for at
least some of their testing needs. Agencies located in larger counties or cities were more likely to
use their own laboratory than those located in smaller or rural locations. When testing is required
beyond the local agency, the specimen is usually sent to the state public health laboratory. Other
referral sites include independent laboratories, hospital laboratories, other state public health
laboratories, and university laboratories.
A 2003 survey indicated that 39% of local public health laboratories were certified to perform
waived testing. Of the 61% that were certified to perform non-waived testing, 39% were certified
to perform moderate complexity tests and 22% were certified to perform high complexity tests,
e.g., tests for the fungal disease candidiasis and the enteric pathogen shigella.42 Nearly 70% of
testing fell into the waived category, compared to 20% for moderate complexity, and 11% for high
complexity. Public health laboratories serving larger populations tended to offer highercomplexity testing.
Local public health laboratories derive revenue from a variety of sources, as shown in Figure 2.8,
about half of which is provided by local and/or state governments.
State and local public health laboratories are likely to face significant challenges over the next
several years. Emphasis on public health preparedness and its ability to respond quickly and
effectively to biological or chemical terrorist threats has led to increased scrutiny of public health
laboratory infrastructure.43 Public health laboratories are being called upon to foster and lead
preparedness and response planning efforts for emerging infectious diseases, such as pandemic
influenza.44 Biomonitoring, or the measurement of chemical levels in the human body (e.g., in
blood, urine, or saliva) to assess human exposure to pollution is an area where state public health
laboratories are continuously called upon to play a large and leading role.45 Collaboration
between public and private laboratories and continued funding of current and future initiatives
will remain vital components critical to the success of public health laboratories.

May 2008 v2

78

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

Figure 2.8: Sources of Revenue for Local Public Health Laboratories, 2003
Other
4%

Patient
Payments
13%

Federal
Funds
10%
State
Funds
19%

Medicare/
Medicaid
19%

Local Funds
33%
Private
Insurance
2%
Source: Assessing America's local public health
laboratory capacity. Silver Spring, MD: Association of
Public Health Laboratories, 2004.

Skilled Nursing/Nursing Facility Laboratories
Skilled nursing is health or rehabilitation care that can only be provided safely and correctly by a
registered nurse or licensed practical nurse.46 According to CMS, there were 14,838 skilled
nursing facilities in the U.S. in December 2003.47 As of December 2006, a total of 14,760 skilled
nursing or nursing facilities had their own clinical laboratories, comprising 7.36% of all
laboratories.19 Skilled nursing or nursing facility laboratories conduct approximately 60.4 million
tests annually. More than 99% of skilled nursing or nursing facility laboratories are certified to
perform waived or PPM testing.

Home Health Agency Laboratories
Home health care usually refers to medical care that is provided to elderly and other patients with
the goal of enabling them to regain their independence and retain the highest degree of selfsufficiency without being confined in a hospital.48 Home health care can include skilled nursing
care, physical and occupational therapy, speech-language therapy, mental health services,
palliative care, and medical social services.49 Organizations whose main function is to provide
home health care services and supplies must meet federal and state requirements for licensure
and certification.50 As of March 2007, there were approximately 11,130 Medicare-certified home
health agencies in the U.S.51
Many home health agencies operate their own clinical laboratories to serve the needs of their
clients. In 2006, there were 10,134 home health agency-based laboratories, accounting for more
than 5% of all laboratories in the U.S.19 The annual volume of testing conducted by home health
laboratories is approximately 18.4 million. About 98% of testing conducted in home health
agencies is waived and more than 99% of home health agency laboratories are certified to conduct
waived or PPM testing only.

May 2008 v2

79

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

Other Types and Settings of Laboratory Services
Clinical laboratories are present in additional settings, including community clinics, ambulatory
surgical centers, student health facilities, and pharmacies which, together, account for
approximately 26% of all laboratories (see Table 2.4).16
In 2006, their estimated combined revenue was $2.9 billion. Growth among these laboratories is
slowing primarily because of lower overall revenues at the contract level and costs associated
with CLIA regulatory requirements.1, 35, 52
Table 2.4: Other types of laboratories by prevalence in the U.S., 2006

Type of Laboratory

Number

Percent of
Total
Laboratories

Total
Annual Test
Volume
(million)

Waived
Testing as
Percentage
of Total
Annual Test
Volume

Community Clinic

6,588

3.32%

75.9

28.7%

End Stage Renal Disease Dialysis Facility

4,099

2.07%

12.9

83.0%

Ambulatory Surgical Centers

4,023

2.03%

25.7

13.5%

Pharmacy

3,815

1.92%

73.3

99.9%

Ambulance

2,831

1.43%

3.0

73.5%

Ancillary Test Site in Health Care Facility

2,722

1.37%

67.7

36.9%

Other Practitioner

2,554

1.29%

29.2

11.9%

School/Student Health Facility

1,873

0.95%

5.5

42.3%

Hospice

1,872

0.94%

3.5

48.3%

Industrial

1,677

0.85%

2.3

67.2%

Rural Health Clinic

1,226

0.62%

3.9

47.6%

Mobile Laboratory

1,117

0.56%

5.0

58.6%

Interm. Care Facility, Mentally Retarded

985

0.50%

1.7

52.5%

Health Maintenance Organization

677

0.34%

100.7

4.3%

Health Fair

516

0.26%

13.0

7.0%

Federally Qualified Health Center

444

0.22%

1.4

52.7%

Blood Banks

375

0.19%

83.9

12.2%

Comprehensive Outpatient Rehab. Facility

272

0.14%

1.7

67.5%

Insurance

42

0.02%

5.4

0.8%

Tissue Bank/Repositories

36

0.02%

0.895

1.3%

15,300

7.72%

267

25.3%

Not Otherwise Specified

Source: CLIA update―December 2006, laboratories by type of facility. Baltimore, MD: Centers for Medicare and Medicaid Services, 2006.

May 2008 v2

80

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

MARKET FOR LABORATORY TESTS
There is no definitive estimate of how many clinical laboratory tests are available. However, a
large independent clinical laboratory currently lists over 4,000 tests on its testing menu.53
According to the American Society for Clinical Laboratory Science (ASCLS), of the 1,162 tests
reimbursed by Medicare, about 500 are performed regularly.54
Laboratory tests are categorized by the following key areas:





Anatomic pathology


Cytology



Surgical pathology



Oncology



Neuropathology



Immunohistochemistry

Clinical pathology


Molecular pathology
-

Cytogenetics

-

Genetics



Histocompatibility



Microbiology



Immunology



Chemistry (including toxicology and drugs of abuse testing)



Hematology



Immunohematology



Radiobioassay14

Most of the tests are performed at the request of a clinician, although patients and other
consumers may order tests directly from the laboratory in some states. Discussion of the market
for consumer direct access testing (DAT)i and over-the-counter (OTC) tests is provided
subsequently in this chapter.
The market for clinical pathology is estimated to be $31.9 billion in 2006, comprising nearly twothirds of the industry (Figure 2.9).1 Anatomic pathology and cytology are estimated to have
markets valued at $9 billion and $2 billion, respectively. The market for molecular and other
esoteric testsj is about $4.1 billion, and that for drugs of abuse testing is $1.5 billion.

DAT is known by a variety of other names, including consumer ordered tests, patient-directed tests, consumer
driven tests, patient authorized tests, and consumer self-orders.55, 56
j The term “esoteric” refers to new molecular pathology tests and certain other relatively low volume tests. Most
esoteric tests use molecular-based laboratory techniques.
i

May 2008 v2

81

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

Figure 2.9: Major Testing Segments in the U.S. Laboratory Industry, 2006

Molecular Pathology/
Esoteric Testing
$4.1 billion
8%

Drugs of Abuse
$1.5 billion
3%

Anatomic
Pathology
$9.0 billion
19%

Cytology
$2.0 billion
4%

Clinical Pathology
$31.9 billion
66%

Source: Reprinted with permission from Washington G-2 reports.
Terry M. Lab industry strategic outlook: market trends and analysis
2007. New York, NY: Washington G-2 Reports, 2007.

Figure 2.10 highlights the two-year average growth rate in each category. The market for all testing
areas is expanding, except drugs of abuse testing. Growth in clinical pathology and
molecular/esoteric testing is expected to average 11% in 2007. Contributing to the growth in
molecular pathology testing are higher reimbursement rates, greater demand from both consumers
and providers, and increases in the number of new predictive laboratory tests available.
Growth in anatomic pathology is expected to average 5% in 2007.1 This growth is due mainly to
changes in the burden of disease in the population, stakeholder interest in expanding methods
of early detection to decrease health care costs, and technological innovation that has simplified
testing techniques in areas such as flow cytometry and immunohistochemistry, allowing for
more ubiquitous anatomic pathology testing in hospital and independent laboratories. The
largest increase in test volumes is expected to occur in FISH and polymerase chain reaction
(PCR)-based testing.57

May 2008 v2

82

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

Figure 2.10: U.S. Laboratory Segment Annual Growth Rates, 2004 - 2006
14.60%

16%
14%
12%

9.80%

10%
8%

6.10%

6%

4.00%

4%

2.40%

2%
0%
Clinical Pathology

Molecular
Anatomic pathology
Pathology/Esoteric
Testing

Drugs of abuse

Cytology

Source: Reprinted with permission from Washington G-2 reports. Terry M. Lab industry strategic outlook: market trends and
analysis 2007. New York, NY: Washington G-2 Reports, 2007.

Market for Anatomic Pathology Testing
Anatomic pathology consists of the subspecialties of cytology, immunohistochemistry,
neuropathology, dermatopathology, oral pathology, forensic pathology, autopsy pathology, and
histology. Cytology refers to the diagnosis of cells from all systems and areas of the body.58
Histology involves the study of tissues and cells under a microscope.59 Immunohistochemistry
involves use of antibodies to detect specific proteins that are expressed in tumors.k,60
Neuropathology is the branch of medicine dealing with diseases of nervous system tissue,
specifically the brain and spinal cord.61 Dermatopathology is the study of diseases of the skin,
including infectious, immunologic, degenerative, and neoplastic diseases.62 Oral pathology refers
to the study of diseases affecting the oral and maxillofacial regions.63 Forensic pathology concerns
the examination of living or dead persons in order to provide an opinion on the cause,
mechanism, and manner of disease, injury, or death.64 Autopsy pathology refers specifically to
the external and internal examination of the body after death.65
Together, anatomic pathology and cytology account for about 23% of the laboratory industry by
testing volume. Aside from cytology, market data for the other anatomic pathology testing areas
are not available.
Three major privately-owned independent laboratories collected an estimated $2 billion in
anatomic pathology revenues in 2006.1 Despite consolidation efforts among privately-owned
independent laboratories, the anatomic pathology market remains relatively fragmented. More
than 70% of the anatomic pathology market is occupied by thousands of pathology groups,
including small and mid-sized private practices, independent laboratories, and hospitals.66
k

Immunohistochemistry is considered anatomic pathology because it is conducted on biopsies and surgical
specimens as opposed to bodily fluids (e.g., blood, urine).

May 2008 v2

83

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

According to CMS data, about 8,015 laboratories perform anatomic pathology tests, generating an
annual volume of nearly 1.45 million tests.67
Table 2.5 summarizes responses from 190 anatomic pathology laboratories to a survey regarding
average annual testing volumes conducted in 2007. Respondents self-identified as belonging to
one of four groups, three of which were hospital-related.57 While average testing volumes in
independent laboratories exceeds average testing volumes in some hospital-based laboratory
arrangements for certain types of testing (e.g., surgical pathology accessions,
immunohistochemistry), these results indicate that the majority of anatomic pathology testing is
conducted in hospital laboratories. For example, 82% of liquid-based Pap smears and 86% of
other molecular diagnostic tests are performed in hospital-related laboratories.
Table 2.5: Self-Reported Anatomic Pathology Testing Volumes by Type of Practice, 2007
Hospital-Related Sites

Total
Volume
by
Testing
Type

% of Work
Conducted
in HospitalRelated
Labs

Hospitala

Pathology
Practice
Groupb

Surgical Pathology
Accessions (All)

18,322

30,057

29,723

48,577

126,679

78.4%

- Biopsies

12,914

17,368

20,451

23,937

74,670

83.3%

5,305

3,290

4,010

10,334

22,939

80.3%

469

215

86

1,331

2,101

72.4%

Cytopathology

15,380

18,592

19,208

34,181

87,361

80.0%

Liquid-based Pap Smears
(All)

16,023

27,339

17,774

32,786

93,922

82.1%

- Thin-layer Automated
Imaging of Pap Smears

9,540

13,530

10,308

22,476

55,854

79.3%

Other Molecular
Diagnostic Tests

501

10

3,541

879

4,931

86.3%

Polymerase-Chain
Reaction-Based Tests

926

2,162

1

3,106

6,195

73.7%

9,652

13,169

15,226

21,108

59,155

64.3%

61,273

91,544

85,559

141,968

380,344

80.1%

- Immunohistochemistry
Fluorescent In Situ
Hybridization

All Other Non-Surgical
Pathology Accessions
(e.g., electronic
microscopy accessions)
Total Volume by
Practice Type*

Otherc

Independent
Labd

a

Refers to pathologists who receive a salary from the hospital and conduct testing of specimens collected from hospital
inpatients and outpatients

b

Refers to pathologists who have formed a private business group and who have contracted with a hospital to perform testing in
that hospital’s laboratory. These pathologists conduct inpatient, outpatient, and outreach testing. They bill insurance
companies directly for the work they perform (professional component only); they maintain their own billing and revenue
systems
c

Refers to pathologists who work in multihospital systems, integrated delivery systems, academic medical centers, or university
hospitals
d

Refers to pathologists who work in an independent laboratory where the laboratory, testing equipment, and office space is not
owned by a hospital system. These pathologists usually conduct testing on specimens collected in outpatient settings, but also

May 2008 v2

84

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

can function as reference laboratories for hospitals and other facilities. They bill insurance companies for both the technical and
professional components of testing.
*Biopsies, immunohistochemistry, and thin-layer automated imaging of Pap smears were not included in calculation of total
volume by type of practice because they are sub-categories of surgical pathology and liquid-based Pap smears, respectively.
Volumes for these tests are included here to show breakdowns of specific tests.

This category may include some molecular diagnostic testing (e.g., FISH, PCR-based testing) because some respondents may
have included all molecular diagnostic testing in the “non-surgical pathology accessions” category rather than delineating these
tests by specific category.

Source: Modified and reprinted with permission from Washington G-2 reports. Business strategies for anatomic pathology,
including state of the market report, 2007. New York, NY: Washington G-2 Reports, 2007.

Anatomic pathologists/technologists in all settings are becoming more specialized.68 Some
laboratories and practice groups hire staff members with expertise in a particular organ system
or testing method. Other groups hire individuals with doctoral degrees in specific areas, such
as molecular biology, to research and develop new testing procedures.69 To expand their
business, these groups also are beginning to market specific tests or services directly to
physicians and patients.70

Cytology
Cytologic testing remains the gold standard in detection of many types of diseases, including
common forms of cancer (e.g., uterine and cervical cancers, leukemia, lymphomas).71 In some
instances, traditional cytologic tests are being replaced by newer technology. For example, an
inexpensive point-of-care test to detect bladder cancer recently developed may be substituted for
traditional laboratory-based cytologic testing.72
The value of the cytology market is approximately $2 billion.1 From 2003 to 2006, cytological testing
grew by 60%.66 Although the cytology market has been undergoing some consolidation, it remains
fragmented, with the majority of the market held by independent laboratories and hospitals.
A major contributor to the growth of cytology has been the nearly complete transition from
traditional Pap smear tests to liquid-based thin-layer slide preparation testing methods for
cervical cancer screening.73 Whereas traditional Pap smear testing involves using a fixative to
spray specimen cells onto a glass slide prior to sending them to a laboratory for analysis, the thinlayer testing method allows the specimen to be mixed into a vial of liquid preservative, which is
then sent to the laboratory and made into slide samples, providing for cleaner and more uniform
analysis.74 In 2006, 92% of laboratories used thin-layer Pap test method to conduct cervical cancer
screenings; however, by 2007, this number was projected to increase to 99%.1, 75
Thin-layer Pap smear tests are reimbursed approximately $10 more than conventional tests, and
increased use of thin-layer tests has contributed significantly to the growth in cytology-related
revenue over the past several years.l From 1998 to 2005, revenue generated by gynecologic
cytology testing has more than doubled, with revenues in 2005 exceeding $1.38 billion.1 More
than 80% of the gynecologic cytology manufacturing market is dominated by two privately
owned companies.

l

Medicare reimbursed thin-layer tests at the same rate as traditional Pap smear tests until April 2001; Medicare’s
increase in reimbursement for thin-layer tests is believed to have accelerated acceptance of this form of testing.1

May 2008 v2

85

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

Market for Molecular Pathology
Although molecular laboratory tests fall under the umbrella of clinical pathology, molecular
testing techniques are increasingly applied in anatomic pathology. Thus, many experts are now
discussing the blurring of the traditional lines between clinical and anatomic pathology. This
discipline includes the categories of histocompatibility (e.g., assays to determine whether recipient
and donor share antigens to ensure that a donated graft is accepted and remains functional);
cytogenetics (e.g., analysis of amniotic fluid to detect fetal genetic abnormalities);
hematopathology (e.g., tests to determine recipient genotype prior to bone marrow
transplantation); infectious disease (e.g., HIV genotyping and determination of status of hepatitis
B virus infection); inherited disease (e.g., cystic fibrosis carrier screening); and pharmacogenomics
(e.g., genotyping to guide warfarin dosing).76-79
In 2007, the U.S. molecular diagnostic testing market was valued at approximately $4.1 billion,
representing the fastest-growing and most-profitable area of the clinical laboratory industry.80
Market researchers anticipate continued growth in the molecular diagnostic market of
approximately 19% per year over the next three years. The exact number of genetic tests available
is not known, but an estimated 1,430 diseases are currently detectable using genetic testing (287
diseases are tested only in research settings).81
Key areas of growth include infectious disease testing, pharmacogenomics, genetic testing, and
oncology testing. Figure 2.11 displays predicted revenues for each of these areas within the
context of the worldwide molecular diagnostic market in 2016. By then, the U.S. is expected to
generate nearly $46.2 billion in revenues, half of the world market. Infectious disease testing is
the largest area of the molecular diagnostic market, due to the high incidence of infectious disease
and the relative ease with which genetic information required for identifying pathogens is
obtained from bacterial and viral species.82 Pharmacogenomic, genetic, and chromosome testing
represent the next largest areas of growth. For example, cytogenetic laboratory tests involve
preparation of cells and isolation of chromosomes in order to identify chromosomal abnormalities
(e.g., tests to detect the presence of fragile X syndrome, the most common inherited cause of
mental impairment and the most common known genetic cause of autism). About 1.6 million
cytogenetic tests are performed annually in 373 laboratories.67
Molecular diagnostic testing is changing the practice of laboratory medicine. For example,
laboratory testing is expected to have an important role in the growth of personalized and
preventive medicine.83 Molecular testing techniques are increasingly being applied to oncology and
cardiology and tests to determine genetic expression and gene profiles, allowing clinicians to detect
disease at much earlier stages. Although many molecular tests are currently still in the research
stage, they are expected to become part of routine clinical practice over the next several years.
Laboratory experts predict that all neoplastic tissues will eventually be analyzed biochemically
and morphologically.84 Expression profiling, by which expression patterns of thousands of
individual genes in a given cell or tissue sample can be discerned, may become standard practice
alongside traditional histopathology.85 If the historical line separating anatomic and clinical
pathology continues to dissolve, pathology residency programs will likely need to adapt to
emphasize training in both the morphologic and molecular basis of disease.84

May 2008 v2

86

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

$70.0

$60.8

$60.0
$50.0

dollars)

Predicted revenue (in billions of U.S.

Figure 2.11: Predicted Average Revenue in 2016 for Segments of the International
Molecular Diagnostic Market

$40.0
$30.0
$20.0

$12.4

$9.8

$10.0

$5.3

$0.0
Infectious Disease

Pharmacogenomics

Oncology

Genetic Testing

Source: Molecular diagnostics: major world markets. New York, NY: Kalorama Information, 2007.

Techniques from molecular biology, identity testing, transplantation, and anatomic pathology are
fueling many of the advances in molecular diagnostics.86 For example, rapid molecular testing is
now available using nucleic acid extraction techniques, allowing for more immediate initiation of
therapy. Information derived from the sequencing of the human genome is enabling creation of
molecular diagnostic tools that help to reveal the presence and roles of additional genes and gene
products in complex diseases. Advances in innovative automated systems to support molecular
diagnostic testing technologies also are improving laboratory practice by decreasing TATs,
improving precision of quantitative results, and reducing costs (relative to manual testing).87
Higher reimbursement rates for molecular tests are contributing to the accelerated growth in
this area. According to a recent market analysis, the average charge for a non-molecular test is
approximately $30.39, compared to $176.77 per molecular test.73 Automation of molecular and
genetic testing will likely affect the budgets and operating costs of laboratories performing these
tests.

Market for Clinical Pathology Testing
Clinical pathology comprises the largest segment within laboratory testing; the market is
currently valued at $31.9 billion, or approximately 66% of the industry.1
Waived Tests. According to CMS, waived testing accounts for approximately 11% of annual
laboratory testing volume.67 In 2006, annual volume of waived testing approached 700 million
tests and generated annual revenue of $1 billion. Approximately 75% of waived testing is
conducted in laboratories certified to perform only waived or PPM testing, although about 17% is
conducted by laboratories with a COA and about 8% by those with a COC (see Table 2.6).19

May 2008 v2

87

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

Table 2.6: Number of Waived and Non-Waived Tests Performed According to Laboratory
Data Self-Reported to CMS, 2006

Number of
Waived Tests
Performed
Number of
Non-Waived
Tests Performed

Laboratories
Certified to
Perform only
Waived or PPM
Testing

Laboratories with
Certificate of
Accreditation

Laboratories with
Certificate of
Compliance

Total Number of
Tests Performed

523,797,097

121,240,212

54,890,056

699,927,365

Not applicable

4,614,509,645

921,013,600

5,535,523,245

Source: OSCAR/CLIA data base: Test volume information based on laboratory self-reporting on CMS-116. Zeller C, ed.
Baltimore, MD: Centers for Medicare and Medicaid Services, March 2006.

According to OSCAR data obtained by Washington G-2 Reports, the highest-volume waived tests
performed by all laboratories include those to evaluate prothrombin, urinalysis, ovulation,
glucose, fecal occult blood test, and the lipid panel (Figure 2.12).1 From 2000 to 2006, tests for 36
analytes (for which waived tests were not previously available) were granted CLIA waived
status.88 With new technology and simplification of testing techniques, the number of tests
receiving waived status will continue to increase.

Number of waived tests in millions

Figure 2.12: Five Highest Volume Waived Tests
30.0

25.4

25.0

23.5

20.0

13

15.0

12

11.5

Glucose

Fecal Occult
Blood Test

10.0
5.0
0.0
Prothrombin

Urinalysis

Ovulation

Sources: Medicare part B physician/supplier national data CY 2005: Top 100 lab procedures. Baltimore, MD: Centers for
Medicare and Medicaid Services, 2005. (Accessed November 10, 2007, at
http://www.cms.hhs.gov/MedicareFeeforSvcPartsAB/Downloads/LabServ05.pdf?agree=yes&next=Accept.)
Terry M. Lab industry strategic outlook: Market trends and analysis 2007. New York, NY: Washington G-2 Reports, 2007.

Moderate and High Complexity Tests. Although only 20% of laboratories are certified to perform
moderate and high complexity tests, approximately 89% of laboratory testing volume is moderate
or high complexity (non-waived) testing.19 In 2006, the volume of non-waived testing exceeded

May 2008 v2

88

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

5.5 billion (Table 2.6). The majority of non-waived testing is conducted in laboratories that have a
certificate of accreditation.
As per CLIA regulations, CMS delineates non-waived clinical pathology laboratory tests into
selected specialty areas. For clinical pathology, these areas include: chemistry, hematology,
diagnostic immunology, microbiology, immunohematology, histocompatibility, and
radiobioassay.89 While the information provided here is based on data collected by CMS, CLIA
specialties are not all inclusive of the menu of available clinical pathology laboratory tests. For
example, CLIA does not include a separate specialty for genetic testing or toxicology. An
overview of each CLIA specialty, including purpose, examples of specific tests, annual volume,
and number of laboratories performing that type of test is displayed in Table 2.7.
Data reported by laboratories to CMS indicate that 62% of all non-waived tests performed are
chemistry tests, which measure compounds and chemical reactions in the body and include
routine chemistry, endocrinology, urinalysis, and toxicology tests.67, 89 Commonly performed
routine chemistry tests include measurements of total cholesterol, calcium, triglycerides, glucose,
electrolytes, blood urea nitrogen, creatine, and prostate specific antigen (PSA). Examples of
endocrinology tests include measurements of testosterone, thyroid-stimulating hormone, and
progesterone; toxicology tests include measurements of acetaminophen, blood alcohol, digoxin,
and lithium.
Accounting for 24% of all laboratory testing, hematology studies the blood, blood-producing
organs, and cells of the body.67, 90 Examples of hematological laboratory tests include
measurements of red and white blood cells, differential blood counts, prothrombin time, and
activated clotting time.89
Diagnostic immunology, which involves measurement of the body’s response to infection and
inflammation, accounts for approximately 4.3% of testing.67 Frequently ordered immunological
tests include mononucleosis assays, antibody assays for viruses such as herpes and hepatitis,
mycoplasma pneumoniae assays, and tests to detect rheumatoid arthritis.78, 89
Microbiological testing also comprises 4% of non-waived testing and refers to the detection,
identification, and measurement of disease-causing microorganisms (e.g., viruses, bacteria, fungi,
algae, parasites).67, 90 For example, antigen assays detect streptococcal infection and culture
assays detect and identify bacteria that cause urinary tract infection.89
Immunohematology, or blood banking, involves the preparation of blood and blood components
for transfusion and the selection of appropriate and compatible blood components for
transfusion.91 Approximately 2.4% of moderate and high complexity testing falls into the
category of immunohematology.67 Common immunohematology tests are those that determine
blood and Rh type and that screen and identify antibodies.89
Smaller percentages of non-waived testing falls into the areas of radiobioassay (0.2%),
histocompatibility (0.7%), and cytogenetics (0.03%).67, 92 Radiobioassay tests determine the type,
quantity, concentration, and location of radioactive material in the body (e.g., test of red cell
volume, Schilling’s test for B12 absorption).89, 93 Histocompatibility tests assess the extent to which
an organ donor and organ recipient share antigens and an organ can be successfully transplanted

May 2008 v2

89

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

(e.g., human leukocyte antigen typing to determine the proteins on white blood cells that make
each person’s tissue unique).78
Table 2.7: Annual Testing Volume, and Number of Laboratories Performing Clinical
Pathology Laboratory Testing According to CMS Laboratory Test Categorization
Examples of Commonly Ordered
Laboratory Tests

Clinical Area

Chemistry

Hematology

Diagnostic
Immunology

Microbiology

Immunohematology

Radiobioassay±
Histocompatibility





























Approximate
Annual Test
Volume

Number of
Laboratories
Performing Tests

3.5 billion

25,755

1.4 billion

24,663

238 million

12,804

225 million

20,056

131 million

6,956

10 million

611

4.0 million

260

Cholesterol, total
Uric acid
Glucose (blood sugar)
Ferritin
Folate
Blood alcohol
Acetaminophen
Red blood count
White blood count
Platelet count
Hemoglobin
Hematocrit
Mononucleosis assays
Rheumatoid arthritis
Febrile agglutinins
Human Immunodeficiency Virus
Hepatitis or herpes antibody assays
Streptococcal testing
Bacterial cultures
Gram stains
ABO blood type
Rh(D) type
Compatibility testing (cross-matching)
Antibody screening
Red cell volume
Schilling’s test
Human leukocyte antigen typing
(disease associated antigens)

±

Radiobioassay testing may not always be considered part of the clinical laboratory and instead may be classified as a part of
nuclear medicine.
Sources: Histocompatibility and immunogenetics terminology. Mt. Laurel, NJ: The American Society for Histocompatibility and
Immunogenetics, 2007. (Accessed April 27, 2007, at http://www.ashi-hla.org/).

Consumer laboratory test information page. Bethesda, MD: American Society for Clinical Laboratory Science, 2007. (Accessed on
April 25, 2007, at http://www.ascls.org/labtesting/index.asp).
Kotrla, K. Immunohematology. Austin, TX: Austin Community College, 2004. (Accessed April 27, 2007, at
http://www.austincc.edu/kotrla/immunohe.htm).

May 2008 v2

90

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

Laboratory departments. East Lansing, MI: Michigan Association of Laboratory Science Educators, 2007. (Accessed on April 27,
2007, at http://www.malse.org/labdepts.htm#).
Definition of terms. Raleigh, NC: North Carolina Department of Environment and Natural Resources, 2007. (Accessed on April
27, 2007, at http://www.ncradiation.net/RMS/rmsglossary.htm#B).
Clinical Laboratory Improvement Amendments of 1988 (CLIA) application for certification. Baltimore, MD: Centers for Medicare
and Medicaid Services, 2007. (Accessed April 27, 2007, at http://www.cms.hhs.gov/cmsforms/downloads/cms116.pdf).
OSCAR/CLIA data base: Test volume information based on laboratory self-reporting on CMS-116. Zeller C, ed. Baltimore, MD:
Centers for Medicare and Medicaid Services, April 2007.

Market for Drugs of Abuse Testing
Drugs of abuse testing is used to determine the presence or absence of illegal drugs in an
individual’s body. Such testing is periodically conducted for designated workers in the public
and private sectors.m In the federal government, specific federal employees are required to
undergo random drugs of abuse testing, including federal employees determined to be “testingdesignated personnel” under the Federal Drug-Free Workplace Program, Department of Defense
employees, Department of Transportation employees involved in aviation, railroads, mass transit,
pipelines, and other transportation industries, and Nuclear Regulatory Commission employees
and contractors.94 In total, this includes about 16 million people. Under the Drug-Free Workplace
Act of 1988, contractors and grantees of federal agencies must agree to provide drug-free
workplaces prior to receiving a contract or grant; however, these programs do not require random
drugs of abuse testing.95 In the private sector, companies can voluntarily elect to implement drugfree workplace programs such as drug abuse prevention education.n
The drugs of abuse testing market was valued at approximately $1.5 billion in 2006. About 33
million tests are conducted for employers annually. About 20% of these tests (approximately 7.5
million) are conducted for federal employees who are required to undergo drugs of abuse testing.
A key factor contributing to market growth for drugs of abuse testing was the passage of the
Drug-Free Workplace Act in 1988. The new rules required that all federal contractors and
grantees ensure a drug-free workplace, prompting steep increases in drugs of abuse testing from
21% of employers in 1987 to 81% in 1996.
In order to conduct tests for federal agencies, laboratories must be certified by the government
through the National Laboratory Certification Program of the Substance Abuse and Mental
Health Services Administration (SAMHSA).o, 96 While private companies can elect to use
laboratories that are not certified by SAMHSA, many use these laboratories, considered to be the
“gold standard” for drugs of abuse testing. Of the 45 laboratories currently certified to conduct
federal workplace drugs of abuse testing, 11 are owned or operated by the two largest laboratory
corporations.97 In 2005, their combined revenue for drug testing was $264 million, almost 28% of
the market but a small percentage of their total revenue.1 Other smaller companies may derive
the majority of their revenue from drugs of abuse screening. Table 2.8 provides the estimated
revenue for top 8 laboratories engaged in drugs of abuse testing in 2005.

Drugs of abuse testing also are an important part of forensic testing, although this topic is outside the scope of this
report.
n Some states offer financial benefits to private companies that enact drug-free workplace programs. Some insurance
companies provide lower rates to companies that have such programs in place.
o SAMHSA oversees the Federal Drug-Free Workplace Program.
m

May 2008 v2

91

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

Table 2.8: Estimated 2005 Revenue at Top Eight Employee Drug Testing Laboratories
Company
Quest Diagnostics

Estimated Revenue 2005
$165 million*

LabCorp

$99 million*

First Advantage Enterprise Screening

$72 million

Medtox

$32 million

Psychmedics

$21 million

Advanced Toxicology Network

$19 million

Kroll Laboratory Specialists

$18 million

Clinical Reference Lab

$13 million
Total, top 8 companies

$439 million

*Based on percentage of total revenue attributed to drug testing
Source: Reprinted with permission from Washington G-2 reports. Terry M. Lab industry strategic outlook: market trends
and analysis 2007. New York, NY: Washington G-2 Reports, 2007.

Aside from federal employers, the number of employers requiring pre-employment drugs of
abuse screening has decreased by 11% from 1999 to 2005.1 Drugs of abuse testing of all employees
on a regular basis has declined steadily from 1999 to 2004. Many employers have reported that
drugs of abuse testing is not cost-effective, particularly when the actual number of positive test
results is quite low. In 1999, the most current year for which data of this nature is available, the
American Civil Liberties Union reported that, of the $11.7 million (1990 dollars) spent by the
federal government to screen 29,000 employees, only 153 tests were positive.98
To decrease expenses, drugs of abuse testing for non-federal employees is increasingly being
conducted in POLs, clinics, or on site in employer-based health departments or clinics using
waived test kits.p Kits sold over-the-counter through retailers (e.g., CVS, Wal-Mart) typically cost
$3 to $5, compared to laboratory-performed tests, which cost $25 to $50.1 Although OTC testing
of drugs of abuse may be less accurate and reliable than laboratory-performed tests, this shift in
testing location has resulted in the closing of many drug testing laboratories. From 1998 to 2007,
the number of SAMHSA-certified laboratories decreased from 71 to 45, a 36% drop.1, 97 While
many states do not have special requirements to govern employers who perform their own drugs
of abuse testing, some states do require that all positive test results be confirmed in a SAMHSAcertified laboratory.99

Market for Consumer-Directed Tests
An increase in the amount of information available to the public via the Internet and news media
attention to personal health issues have enhanced consumer interest in understanding, directing,
and managing their health care.84,100 The increasing number of Americans who are uninsured or
underinsured may be contributing as well, to the extent that they seek personal health information
outside of the health care system. There is now a substantial market for health care services and
products that support consumers in their self-care associated with prevention and disease
management. Two chief avenues for consumers to engage in self-directed laboratory testing are
DAT at an established laboratory and OTC tests.

p

Between 2000 and 2006, FDA approved 35 waived tests, 5 specifically for drug testing.

May 2008 v2

92

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

Direct Access Testing. Consumers have demonstrated strong interest in the ability to order
laboratory tests and obtain results independently of a health care provider.101 Typically, DAT
services are purchased out-of-pocket by the consumer without physician consultation.55 A major
concern regarding most DAT is that, in the absence of clinician prescribing and test interpretation,
consumers are largely responsible for interpreting DAT laboratory results and may only follow
up with their clinician if they deem it necessary.
Since CLIA or other federal regulations do not address DAT, its governance falls under state
law.55 Some states prohibit hospital laboratories from performing DAT, while others prohibit
hospitals only from conducting DAT for inpatients. In 2007, 13 states prohibited DAT completely
and 12 states permitted DAT with specific limitations.102 The remaining 25 states and the District
of Columbia allow DAT either because statutes explicitly provided for it or there is no law against
it. In 2004, 10-15% of hospital and commercial clinical laboratories offered some form of DAT.103
Consumer access to DAT varies based on the type of test and geographic location. Internet
websites, telephone services, freestanding stores, hospital and commercial laboratory facilities,
and pharmacies are currently the primary methods of access. While many DAT laboratories offer
only simple tests, other laboratories offer more complex tests. Examples of tests commonly
ordered via DAT services include those that measure complete blood counts, cholesterol levels,
throat and urine infection, diabetes, HIV antibody tests, and blood type.56
Another factor calling attention to DAT is direct-to-consumer advertising of laboratory tests,
specifically genetic tests. For example, a provider of clinical BRCA1/2 tests, for detecting genetic
mutations associated with predisposition for inherited breast and ovarian cancer, directly marketed
these tests to consumers over a 6-month period, marking the first time a genetic test was marketed
to the public.15 Providers in the pilot cities reported ordering more tests and reported an increase in
the number of patients who asked about testing, asked for genetic counseling referrals to consider
testing, and requested testing. The chapter on regulation of laboratory medicine in this report
provides a more detailed discussion of direct-to-consumer advertising of genetic testing.
Over-the-Counter Tests. Consumers can purchase laboratory kits for certain types of tests and
perform the tests themselves at home.104 These tests are purchased OTC at drugstores and
supermarkets as well as on the Internet. OTC laboratory kits available for home use include (but
are not limited to) ovulation detection tests, pregnancy tests, and certain tests that measure and
monitor cholesterol, glucose, fecal occult blood, and urinary tract infections. The average price of
individual tests varies, ranging from $20 for ovulation detection and cholesterol tests to $30-$100
for instruments that monitor glucose levels. Currently, FDA has approved more than 800
laboratory tests that can be sold OTC; more than 160 tests were approved in 2007 alone.105
Laboratory tests performed at home by the consumer can offer certain advantages. They provide
a means by which individuals can monitor a disease that has already been detected by a
physician.104 Secondly, home testing also allows individuals to detect certain conditions in the
privacy of their home. However, laboratory tests performed at home also pose the risk of
inaccurate test results or incorrect interpretation of results, and failure to receive medical advice
and attention if needed.
Research examining the accuracy of home-use tests versus tests performed in a central laboratory
show mixed results. For example, a study of 111 patients with type 1 and 2 adult diabetes found
May 2008 v2

93

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

that, compared to blood glucose values obtained from a calibrated hand-held glucose monitor,
53% of blood glucose measurements using home testing kits were within 10% of the control value,
84% were within 20% of the control value, and 16% varied by 20% or more from the control
value.106 A 2006 review of available evidence on self-monitoring using home-use glucose meters
found that, while these devices are far more accurate than the earlier approach of matching colors,
they are not as accurate as those derived from laboratory testing. Even so, if used properly, they
give an acceptably accurate reflection of immediate plasma glucose levels. The authors noted
that, in the relatively near term, self-monitoring of blood glucose could be replaced gradually by
continuous glucose monitoring.107

RESEARCH-SUPPORT TESTING
Research-support testing is often used in clinical research trials. Although research laboratories
devoted to clinical trial support are exempt from CLIA requirements, they must comply with
FDA regulations and inspections governing the studies.
In recent years, the pharmaceutical industry has outsourced the management and design of clinical
trials to contract research organizations (CROs).108 The CRO industry has grown with the expansion
of clinical trials. Enrollment of individuals in clinical trials also increased from 7 million in 1992 to
20 million in 2001. In 2006, the market for Phase I-IV studiesq by CROs was valued at
approximately $8.5 billion; 8 firms hold 63% of the market.110 The largest providers of phase I-IV
and central laboratory services in the CRO industry are Quintiles, Pharmaceutical Product
Development, Inc., and Covance. Together, these three account for 37% of the CRO market. CROs
can be involved in many different aspects of clinical trials, including preclinical safety analysis,
study design, clinical trial management, laboratory services, statistical analysis, and regulatory
services.108 There are approximately 1,500 CROs in existence today.111 Roughly 20 CROs conduct
business on a global basis, while the remaining ones conduct clinical trials in smaller regions.
While there are no reliable figures, approximately half of CROs in business today operate their own
laboratories.111 Many of these laboratories tend to be small, focusing solely on either basic or
esoteric tests. The number of CROs offering laboratory services has increased as drug and medical
device developers have demanded that CROs play a larger role in all phases of research.112
Covance reports that it has increased its volume of laboratory testing by more than 40% during
the last two years. As part of this expansion, the company implemented a global automated
specimen collection kit production line in which barcoded testing kits are assembled and sent to
clinicians to simplify specimen collection and identification from research subjects. This method
of production also enables monitoring each step involved in kit production, thereby achieving
greater process control.111, 113
Anecdotal evidence from industry experts suggests a synergistic relationship between larger and
smaller CROs in regard to laboratory services and business markets. For instance, larger CROs
might use the framework and relationships established by smaller, ”niche” CROs to gain entry to
q

Clinical trials of most pharmaceuticals and biologicals and some medical devices are conducted in phases.109 For most
experimental products, Phase I and II trials involve determination of treatment safety, dosage, and side effects; Phase
III trials involve confirmation of efficacy and side effects and allow comparison to commonly used treatments.
Phase IV trials are post-marketing studies to obtain additional information about risks, benefits, and optimal use.

May 2008 v2

94

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

the clinical research market in a foreign country. Smaller CROs, particularly those that do not
house their own clinical laboratory services, may use the laboratories of larger CROs to facilitate
their own clinical research.111

CONCLUSIONS
The revenue, spending, and test volume of the U.S. clinical laboratory testing market has grown
steadily over the past decade. Market expansion is attributed to changes in population
demographics and burden of disease; scientific, medical, and technological advancements
driving innovative research and development; and increased consumer awareness of and
demand for high quality, safe health care.



According to CMS, nearly 6.8 billion laboratory tests are performed annually in the
U.S., with projected revenues of $52 billion in 2007.


Clinical pathology testing comprises 66% of all laboratory tests and $32 billion
in revenue



Anatomic pathology and cytology account for 23% and $11 billion in revenue



Molecular and esoteric testing account for 8% and $4 billion in revenue



Drugs of abuse testing accounts for 3% and $1.5 billion in revenue



Today, over 4,000 thousand laboratory tests are currently available for clinical use. Of
the 1,162 tests that are reimbursed by Medicare, about 500 are performed regularly.
Medicare covers genetic tests only in a very limited capacity; most are not eligible for
coverage unless they are indicated for symptomatic patients or are used to determine
how a patient will respond to particular therapies.



The number of genetic tests available is growing, although many of these tests are only
used in research settings. Specifically, an estimated 1,430 diseases are currently detectable
using genetic testing. Of these, 287 diseases are tested only in research settings.



The number of CLIA-certified laboratories grew by 28% from 1993 to 2006, and
exceeded 200,000 in 2007. POLs represent the largest number of clinical laboratories in
this sector (106,190 or 54%); approximately 80% are certified to perform only waived
and/or provider-performed microscopy tests including urinalysis, fecal occult blood,
urine pregnancy, rapid streptococcal, and glucose tests.



Hospital-based laboratories account for the largest proportion of total testing volume
(55%) and generate the highest proportion of total testing revenue (54%), projected at
$28.4 billion for 2007. From 1999-2006, the average annual growth rates of both test
volume and revenue were approximately 6 to 7%. In 2006, privately-owned
laboratories generated revenues of $15.5 billion (32% of total revenue), performing
mostly routine, high-volume tests and molecular/esoteric tests.



Consumer-directed testing in the form of DAT and OTC tests is a key area for current
and future market growth. In 2004, 10-15% of hospital and commercial clinical
laboratories offered some form of DAT.

May 2008 v2

95

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

Gaps, Needs, and Challenges:


Publicly available information about the economic status and quality of the laboratory
medicine sector is limited. The main sources currently available are CMS’ OSCAR
database, ad hoc surveys, and commercial market reports used for investment
purposes. As a group, these leave certain gaps in covering the laboratory market,
including precise figures of market revenues, spending, test volume, and laboratory
testing trends. As a result, estimates about these facets and other descriptors are likely
to be incomplete and imprecise. This lack of data inhibits the ability of researchers,
regulators, payers, providers, and other stakeholders to adequately describe, anticipate
the direction of, and influence this key health care sector. Better data sources would
support improved assessment of the value of laboratory medicine, workforce
development, growth and directions of test use, financing, quality improvement, and
organizational and strategic planning.



POLs are undergoing a major shift toward performing primarily waived testing. This
shift is attributable to the growing number to FDA-approved waived tests available,
industry’s targeted marketing of waived tests to POLs, higher costs associated with
workforce training, and other expenses required to comply with CLIA regulations for
moderate and high complexity testing. The effect on quality of patient care of the shift to
waived testing is unclear and may require further study.



As consumer-driven testing increases, laboratories should be prepared to assume a
greater advisory role to individuals seeking DAT and using OTC tests. Consumer
demand for DAT is already creating incentives for laboratories to provide additional
services. For instance, some laboratories offer consumers the opportunity to consult
with a medical doctor via e-mail prior to ordering tests and to receive test results via email. In addition, for all tests, laboratories can develop consumer-friendly, easily
understandable laboratory reports, written interpretations, and related support to
promote more informed self care by consumers, patient safety, and quality of care.

May 2008 v2

96

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

Reference List
1. Terry M. Lab industry strategic outlook: market trends and analysis 2007. New York,
NY: Washington G-2 Reports, 2007.
2. OSCAR/CLIA data base February 2008. Handsfield J, ed. Atlanta, GA: Centers for
Disease Control and Prevention, 2008.
3. Healthy aging: preserving function and improving quality of life among older
Americans 2007. Atlanta, GA: Coordinating Center for Health Promotion, Centers for
Disease Control and Prevention, 2007.
http://www.cdc.gov/nccdphp/publications/aag/pdf/healthy_aging.pdf.
4. Chronic disease overview. Atlanta, GA: Centers for Disease Control and Prevention,
2005. (Accessed May 21, 2007, at http://www.cdc.gov/nccdphp/overview.htm.)
5. The world factbook: United States. Washington, DC: Central Intelligence Agency, 2007.
(Accessed May 3, 2007, at
https://www.cia.gov/cia/publications/factbook/geos/us.html.)
6. 1997 population profile of the United States. Washington, DC: Department of
Commerce, 1998. http://www.census.gov/prod/3/98pubs/p23-194.pdf.
7. Camarota SA. 100 million more: projecting the impact of immigration on the U.S.
population, 2007 to 2060. Washington, DC: Center for Immigration Studies, 2007.
http://www.cis.org/articles/2007/back707.pdf.
8. Tsongalis GJ. Genetic testing: current and future trends. MLO Med Lab Obs
2006;38(10):42, 44.
9. Hortin GL, Jortani SA, Ritchie JC, Jr., Valdes R, Jr., Chan DW. Proteomics: a new
diagnostic frontier. Clin Chem 2006;52(7):1218-22.
10. Narayanan S. Miniaturization in futuristic clinical laboratory medicine. Laboratory
Medicine 2005;36(12):748-52.
11. Ahn CH, Choi J, Beaucage G, et al. Disposable smart lab on a chip for point-of-care
clinical diagnostics. Proceedings of the Institute of Electrical and Electronics Engineers
2004;92(1):154-73.
12. Comments on proposed NCVHS recommendations for measuring quality. Testimony of
Dr. Charles Cutler, National Medical Director, Aetna National Quality Management, to
the National Committee on Vital and Health Statistics. 2004.
http://www.ncvhs.hhs.gov/040625p1.htm.

May 2008 v2

97

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

13. Measuring health care quality: obstacles and opportunities. Washington, DC:
Workgroup on Quality, National Committee on Vital and Health Statistics, 2004.
http://www.ncvhs.hhs.gov/040531rp.pdf.
14. Knowledge Source. Clinical laboratory testing market overview--2006. Trumbull, CT:
Knowledge Source, Inc., 2006.
15. Genetic testing for breast and ovarian cancer susceptibility: evaluating direct-toconsumer marketing--Atlanta, Denver, Raleigh-Durham, and Seattle, 2003. MMWR
Morb Mortal Wkly Rep 2004;53(27):603-6.
16. CLIA update--December 2006, laboratories by type of facility. Baltimore, MD: Centers
for Medicare and Medicaid Services, 2006. (Accessed January 3, 2007, at
http://www.cms.hhs.gov/CLIA/17_CLIA_Statistical_Tables_Graphs.asp#TopOfPage.)
17. CLIA update--December 2007. Baltimore, MD: Centers for Medicare and Medicaid
Services, 2007. (Accessed February 13, 2008, at
http://www.cms.hhs.gov/CLIA/downloads/statupda.pdf.)
18. Where lab tests are performed: in a hospital. Washington, DC: Lab Tests Online, 2004.
(Accessed November 4, 2006, at www.labtestsonline.org/lab/labtypes-4.html.)
19. OSCAR/CLIA data base March 2006. Baltimore, MD: Centers for Medicare and
Medicaid Services, 2006.
20. Fast facts on US hospitals. Washington, DC: American Hospital Association, 2006.
(Accessed February 27, 2007, at http://www.aha.org/aha/resource-center/Statisticsand-Studies/fast-facts.html#registered.)
21. Institute of Medicine. Medicare laboratory payment policy: now and in the future.
Washington, DC: National Academy Press, 2000.
22. Steindel SJ, Howanitz PJ. Physician satisfaction and emergency department laboratory
test turnaround time. Arch Pathol Lab Med 2001;125(7):863-71.
23. Singer A, Viccellio P, Niegelber E, Thode H, Henry M. Introduction of a STAT
laboratory reduces ED length of stay. Acad Emerg Med 2007;14(5 (Suppl 1)):79.
24. Johnson BD, Beck KC, Zeballos RJ, Weisman IM. Advances in pulmonary laboratory
testing. Chest 1999;116(5):1377-87.
25. Occupational employment and wages, May 2006, medical and clinical laboratory
technologists. Washington, DC: Bureau of Labor Statistics, Department of Labor, 2007.
(Accessed November 27, 2007, at http://www.bls.gov/oes/current/oes292011.htm.)
26. Occupational employment and wages, May 2006, medical and clinical laboratory
technicians. Washington, DC: Bureau of Labor Statistics, Department of Labor, 2007.
(Accessed November 27, 2007, at http://www.bls.gov/oes/current/oes292012.htm.)

May 2008 v2

98

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

27. Terry M. Washington G-2 Reports. Personal communication. July 23, 2007.
28. Terry M. Washington G-2 Reports. Personal communication. April 23, 2007.
29. Medical billing performance metrics. Billing Precision, 2007. (Accessed April 19, 2007, at
http://www.billingprecision.com/Medical-Billing-Performance-Metrics-Article.htm.)
30. Overview of CLIA statistics for the last decade. Baltimore, MD: Centers for Medicare
and Medicaid Services, 2005. (Accessed November 14, 2006, at
http://www.cms.hhs.gov/CLIA/downloads/decadestats.pdf.)
31. Where lab tests are performed: at the point of care. Washington, DC: Lab Tests Online,
2004. (Accessed December 3, 2006, at http://www.labtestsonline.org/lab/labquality3.html.)
32. Clinical lab quality: CMS and survey organization oversight should be strengthened.
Report No. GAO-06-416. Washington, DC: Government Accountability Office, 2006.
www.gao.gov/cgi-bin/getrpt?GAO-06-416.
33. CLIA office laboratory classification of family physicians by census division and urban
or rural practice location, October 2006. Practice Profile Survey II. Leawood, KS:
American Academy of Family Physicians, 2006.
34. Binns HJ, LeBailly S, Gardner HG. The physicians' office laboratory: 1988 and 1996
survey of Illinois pediatricians. Pediatric Practice Research Group. Arch Pediatr Adolesc
Med 1998;152(6):585-92.
35. Howerton D, Anderson N, Bosse D, Granade S, Westbrook G. Good laboratory practices
for waived testing sites: survey findings from testing sites holding a certificate of waiver
under the clinical laboratory improvement amendments of 1988 and recommendations
for promoting quality testing. MMWR Recommendations and Reports 2005;54(RR-13):125.
36. Occupational employment and wages, 2002, medical and clinical laboratory technicians.
Washington, DC: Bureau of Labor Statistics, Department of Labor, 2003. (Accessed
November 10, 2006, at http://www.bls.gov/oes/2002/oes292012.htm.)
37. Occupational employment and wages, 2002, medical and clinical laboratory
technologists. Washington, DC: Bureau of Labor Statistics, Department of Labor, 2003.
(Accessed November 10, 2006, at http://www.bls.gov/oes/2002/oes292011.htm.)
38. Public health laboratories--analysis, answers, action. Silver Spring, MD: Association of
Public Health Laboratories, 2004. http://www.aphl.org/about_aphl/pages/action.aspx.
39. Defending the public's health. Silver Spring, MD: Association of Public Health
Laboratories, 2007. http://www.aphl.org/about_aphl/Documents/defending_phl.pdf.

May 2008 v2

99

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

40. Dicker LW, Mosure DJ, Steece R, Stone KM. Testing for sexually transmitted diseases in
U.S. Public health laboratories in 2004. Sex Transm Dis 2007;34(1):41-6.
41. DeVoll J. Association of Public Health Laboratories. Personal communication. April 23,
2007.
42. Assessing America's local public health laboratory capacity. Silver Spring, MD:
Association of Public Health Laboratories, 2004.
http://www.aphl.org/about_aphl/products_and_publications/Documents/laboratory
_capacity_12-04.pdf.
43. Public health laboratories: unprepared and overwhelmed. Washington, DC: Trust for
America's Health, 2003. http://healthyamericans.org/reports/files/LabReport.pdf.
44. Shult PA. Beyond the checklist: addressing shortfalls in national pandemic influenza
preparedness. Statement by Dr. Peter A. Shult on behalf of the Association of Public
Health Laboratories, director of the Communicable Diseases Division of the Wisconsin
State Laboratory of Hygiene, before the Committee on Homeland Security, United States
House of Representatives. Silver Spring, MD: Association of Public Health Laboratories,
2007. http://www.aphl.org/policy/Documents/Pan_flu_testimony.pdf.
45. Biomonitoring: measuring chemicals in people. Silver Spring, MD: Association of Public
Health Laboratories, 2004.
http://www.aphl.org/Documents/Global_docs/biomonitoring.pdf.
46. Medicare glossary. Baltimore, MD: Centers for Medicare and Medicaid Services, 2007.
(Accessed April 26, 2007, at
http://www.medicare.gov/Glossary/search.asp?SelectAlphabet=S&Language=English
#Content.)
47. Skilled nursing facilities: 2004. Washington, DC: Census Bureau, 2004. (Accessed May
15, 2007, at http://www.census.gov/compendia/statab/tables/07s0160.xls.)
48. Choosing a home health care agency: what is home health care? Englewood, CO:
Colorado Foundation for Medical Care, 2007. (Accessed February 26, 2007, at
http://www.cfmc.org/consumers/consumers_hh.htm.)
49. Older adults: home health agencies. Pittsburgh, PA: University of Pittsburgh Institute on
Aging, 2007. (Accessed February 26, 2007, at
http://www.aging.upmc.com/seniors/home-care/home-care-agencies.asp.)
50. Home health care. Whitehouse Station, NJ: The Merck Manual of Geriatrics, 2007.
(Accessed February 26, 2007, at
http://www.merck.com/mrkshared/mmg/sec1/ch11/ch11b.jsp.)
51. Home health compare. Baltimore, MD: Centers for Medicare and Medicaid Services,
2007. (Accessed March 22, 2007, at
http://www.medicare.gov/HHCompare/Home.asp?version=default&browser=IE%7C

May 2008 v2

100

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

6%7CWinXP&language=English&pagelist=Home&CookiesEnabledStatus=True&dest=
NAV|Home|Search|SearchCriteria&Type=State#astep1a.)
52. Borgert N. POLs under the gun. Clinical Lab Products 2007.
53. LabCorp managed care. Burlington, NC: Laboratory Corporation of America, 2007.
(Accessed December 12, 2007, at http://www.labcorp.com/managed_care/.)
54. Consumer laboratory testing information page. Bethesda, MD: American Society for
Clinical Laboratory Science, 2002. (Accessed February 16, 2007, at
http://www.ascls.org/labtesting/index.asp.)
55. Consumer access to laboratory testing and information classification: position paper.
Bethesda, MD: American Society for Clinical Laboratory Science, 2004.
http://www.ascls.org/position/DAT.asp.
56. O'Connell J. Where lab tests are performed: through a direct access laboratory.
Washington, DC: Lab Tests Online, 2004. (Accessed November 14, 2006, at
http://www.labtestsonline.org/lab/labtypes-6.html.)
57. Business strategies for anatomic pathology, including state of the market report 2007.
New York, NY: Washington G2 Reports, 2007.
58. Cytology. National Cancer Institute, National Institutes of Health, 2007. (Accessed
December 12, 2007, at
http://www.cancer.gov/Templates/db_alpha.aspx?CdrID=335081.)
59. Histology. National Cancer Institute, National Institutes of Health, 2007. (Accessed
December 12, 2007, at
http://www.cancer.gov/Templates/db_alpha.aspx?CdrID=44171.)
60. Countryman G. Immunohistochemistry Laboratory, Oregon Health and Science
University. Personal communication. November 26, 2007.
61. Dorland's illustrated medical dictionary. 3rd ed. Philadelphia, PA: Saunders, 2003.
62. About physician specialties: pathology. Evanston, IL: American Board of Medical
Specialties, 2007. (Accessed March 6, 2007, at
http://www.abms.org/Who_We_Help/Consumers/About_Physician_Specialties/path
ology.aspx.)
63. What is oral pathology? Maxillofacial Center for Education and Research, 2007.
(Accessed December 12, 2007, at http://www.oralpath.com/About.htm#OralPath.)
64. Forensic pathology. Northfield, IL: College of American Pathologists, 2007. (Accessed
November 8, 2007, at
http://www.cap.org/apps/cap.portal?_nfpb=true&cntvwrPtlt_actionOverride=%2Fpor
tlets%2FcontentViewer%2Fshow&_windowLabel=cntvwrPtlt&cntvwrPtlt%7BactionFor

May 2008 v2

101

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

m.contentReference%7D=disciplines%2Fforensic_medicine%2Fforensicpath_index.html
&_state=maximized&_pageLabel=cntvwr.)
65. Autopsy. Northfield, IL: College of American Pathologists, 2007. (Accessed November 8,
2007, at
http://www.cap.org/apps/cap.portal?_nfpb=true&cntvwrPtlt_actionOverride=%2Fpor
tlets%2FcontentViewer%2Fshow&_windowLabel=cntvwrPtlt&cntvwrPtlt%7BactionFor
m.contentReference%7D=autopsy%2Fautopsy_index.html&_state=maximized&_pageLa
bel=cntvwr.)
66. Anatomic pathology and cytology: continued growth driven by Pap testing. New York,
NY: Washington G-2 Reports, 2007.
67. OSCAR/CLIA data base April 2007. Zeller C, ed. Baltimore, MD: Centers for Medicare
and Medicaid Services, 2007.
68. McGonnagle R. Like it or not, AP's future will be digitally driven. CAP Today 2007.
69. Southwick K. Staying ahead of the AP pack--Three key steps. CAP Today 2004.
70. Paxton A. Rich niche: AP outreach. CAP Today 2005.
71. Bates J. Innovations in cancer: novel therapeutics, new diagnostics and future R&D.
London, England: Business Insights Limited, 2006.
72. Marchant J. Innovations in diagnostics: next generation molecular and point-of-care
diagnostics driving personalized healthcare. London, England: Business Insights
Limited, 2005.
73. Laboratory market leaders report 2008. Terry M, ed. New York, NY: Washington G-2
Reports, 2007.
74. Cramer B. New Pap smear provides more accurate test results. Nashville, TN:
Vanderbilt Medical Center, 2000.
http://www.mc.vanderbilt.edu/reporter/index.html?ID=1053.
75. Jhala D, Eltoum I. Barriers to adoption of recent technology in cervical screening.
Cytojournal 2007;4:16.
76. Reference resources and publications. Northfield, IL: College of American Pathologists,
2007. (Accessed May 2, 2007, at
http://www.cap.org/apps/cap.portal?_nfpb=true&_pageLabel=reference.)
77. Molecular diagnostics test requirement table. Richmond, VA: Department of Pathology,
Virginia Commonwealth University, 2007. (Accessed November 7, 2007, at
http://www.pathology.vcu.edu/clinical/moldx/table.html.)

May 2008 v2

102

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

78. Histocompatibility and immunogenetics terminology. Mt. Laurel, NJ: American Society
for Histocompatibility and Immunogenetics, 2007. (Accessed April 27, 2007, at
http://www.ashi-hla.org/.)
79. Explanation of cytogenetic tests. Madison, WI: University of Wisconsin Cytogenetic
Services, 2007. (Accessed November 20, 2007, at
http://www.slh.wisc.edu/wps/wcm/connect/extranet/cytogenetics/tests/explanatio
n.php.)
80. Business strategies for molecular diagnostics in the lab, including state of the market
report 2007. Terry M, Murg S, ed. New York, NY: Washington G-2 Reports, 2007.
81. GeneTests. Seattle, WA: GeneTests.org, 2008. (Accessed April 18, 2008, at
http://www.genetests.org/servlet/access?id=8888891&key=Z1o3AV3PRzDMu&fcn=y
&fw=apWV&filename=/.)
82. Park R. Explosive growth anticipated for molecular diagnostics. Medical Device Link
2007.
83. Tufel G. Industry trends: midyear report. Clinical Lab Products 2007.
84. Friedman BA. Challenges to pathology informatics in the era of the electronic medical
record. Advancing Practice, Instruction, and Innovation through Informatics (APIII
2006). August 16, 2006. Pittsburgh, PA: APIII, 2006.
85. Ladanyi M, Chan WC, Triche TJ, Gerald WL. Expression profiling of human tumors: the
end of surgical pathology? Journal of Molecular Diagnostics 2001;3(3):92-7.
86. Farkas DH, Bernard DW. SWOT analysis for molecular diagnostics: strengths,
weaknesses, opportunities, and threats. Biotechnology Healthcare 2004:46-51.
87. Mifflin TE. Developments in molecular diagnostic automation. Symposium on
Laboratory Automation. November 8, 2003. Washington, DC: American Association for
Clinical Chemistry, 2003.
88. CLIA database. Rockville, MD: Center for Devices and Radiological Health, Food and
Drug Administration, 2007. (Accessed November 8, 2007, at
http://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfCLIA/search.cfm.)
89. Clinical Laboratory Improvement Amendments of 1988 (CLIA) application for
certification. Baltimore, MD: Centers for Medicare and Medicaid Services, 2007.
(Accessed April 27, 2007, at
http://www.cms.hhs.gov/cmsforms/downloads/cms116.pdf.)
90. Laboratory departments. East Lansing, MI: Michigan Association of Laboratory Science
Educators, 2007. (Accessed April 27, 2007, at http://www.malse.org/labdepts.htm.)

May 2008 v2

103

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

91. Kotrla T. Immunohematology. Austin, TX: Austin Community College Medical
Laboratory Technology, 2007. (Accessed April 27, 2007, at
http://www.austincc.edu/kotrla/immunohe.htm.)
92. Definition of terms. Raleigh, NC: North Carolina Radiation Protection, 2007. (Accessed
April 27, 2007, at http://www.ncradiation.net/RMS/rmsglossary.htm#B.)
93. Schilling test. Bethesda, MD: Medline Plus, National Library of Medicine, National
Institutes of Health, 2007. (Accessed April 27, 2007, at
http://www.nlm.nih.gov/medlineplus/ency/article/003572.htm.)
94. Federal programs. Rockville, MD: Division of Workplace Programs, Substance Abuse
and Mental Health Services Administration, 2007. (Accessed November 8, 2007, at
http://workplace.samhsa.gov/FedPgms/Fed_DFWP.aspx.)
95. Drug-Free Workplace Act of 1988: determining coverage. Washington, DC: Drug-Free
Workplace Advisor, Department of Labor, 2007. (Accessed November 8, 2007, at
http://www.dol.gov/elaws/asp/drugfree/howto.htm.)
96. About the Division of Workplace Programs. Rockville, MD: Substance Abuse and
Mental Health Services Administration, 2005.
http://www.workplace.samhsa.gov/DivisionWP/DWPMissionStatement062105.pdf.
97. Current list of laboratories which meet minimum standards to engage in urine drug
testing for federal agencies. Fed Regist 2007;72(63):15890-1.
98. Drug testing: a bad investment. American Civil Liberties Union, 1999.
http://www.workrights.org/issue_drugtest/dt_drugtesting.pdf.
99. On-site drug testing regulations 2004. Canton, MA: Avitar On Site Diagnostics, 2007.
(Accessed November 8, 2007, at
http://www.avitarinc.com/Resources/Drug_Testing_State_Regulations.cfm.)
100. Institute of Medicine. Culture and society. In: Health literacy: a prescription to end
confusion. Washington, DC: National Academy Press, 2004.
101. Whalen R. Direct access testing (DAT): Clinical Laboratory Improvement Amendments
(CLIA) requirements. Clinical Laboratory Improvement Advisory Committee meeting.
March 12, 2003. Atlanta, GA: Centers for Disease Control and Prevention, 2003.
102. Survey of direct-to-consumer testing statutes and regulations. Washington, DC: Genetics
and Public Policy Center, 2007. (Accessed February 12, 2008, at
http://www.dnapolicy.org/resources/DTCStateLawChart.pdf.)
103. Merlin T. Direct access testing: overview and summary. Clinical Laboratory
Improvement Advisory Committee meeting. September 18, 2003. Atlanta, GA: Centers
for Disease Control and Prevention, 2003.

May 2008 v2

104

Laboratory Medicine: A National Status Report

Chapter II - Market Profile of the Laboratory Medicine Sector

104. Home health care testing information page. Bethesda, MD: American Society for Clinical
Laboratory Science, 2007. (Accessed February 26, 2007, at
http://www.ascls.org/labtesting/hometest.asp.)
105. IVD over the counter database. Rockville, MD: Office of In Vitro Diagnostic Device
Evaluation and Safety, Food and Drug Administration, 2007. (Accessed November 8,
2007, at http://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfIVD/Search.cfm.)
106. Alto WA, Meyer D, Schneid J, Bryson P, Kindig J. Assuring the accuracy of home
glucose monitoring. J Am Board Fam Pract 2002;15(1):1-6.
107. Saudek CD, Derr RL, Kalyani RR. Assessing glycemia in diabetes using self-monitoring
blood glucose and hemoglobin A1c. JAMA 2006;295(14):1688-97.
108. Jayashree M. Clinical research outsourcing: overview, current scenario, and future
outlook. Austin, TX: International Biopharmaceutical Association Newsletter, 2005.
(Accessed April 20, 2007, at
http://www.ibpassociation.org/IBPA_articles/Outsoursing.htm.)
109. An introduction to clinical trials. Bethesda, MD: National Institutes of Health, 2006.
(Accessed April 11, 2007, at http://clinicaltrials.gov/ct/info/whatis#whatis.)
110. Ratliff J. Beating the competition. Research Triangle Park, NC: Quintiles Transnational,
2007.
111. San Filippo J. Becton, Dickinson and Company. Personal communication. April 30, 2007.
112. Upman P, Langenderfer T. Testing: the expanding role of contract research
organizations. Medical Device and Diagnostic Industry 2005:76.
113. Global automated production of visit-specific patient kits. Princeton, NJ: Covance, 2007.
(Accessed April 30, 2007, at
http://www.covance.com/central_lab/svc_automated.php.)

May 2008 v2

105

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

CHAPTER III
LABORATORY MEDICINE WORKFORCE
The laboratory medicine workforce plays a vital role in the health care system, managing and
applying evidence-based, scientific testing techniques to support patient care and protect against
public health threats.1 However, there is growing concern regarding a significant shortage in the
number of laboratory professionals entering the workforce. The shortage could become
pronounced soon with the forthcoming retirement of many laboratorians. At the same time, the
demand for laboratory services continues to increase given such factors as the aging of the
population, growing prevalence of chronic diseases, and availability of new testing methods.
Innovative technologies also are changing the practice of laboratory medicine and, in turn, the
educational requirements and staff qualifications needed to provide quality testing services.
This chapter provides an overview of the professionals in the laboratory medicine workforce,
including their day-to-day responsibilities, demographic characteristics, vacancy rates, and
wages. The chapter also addresses the status of educational programs for each professional
group, including specialty education programs, changes in curriculum, and data on program
enrollees and graduates. Lastly, this chapter discusses licensing and certification at the federal
and state level.

TYPES OF PROFESSIONALS
Clinical laboratories typically are staffed with a medical team comprising pathologists, doctorallevel laboratory scientists, laboratory technologists and technicians, and phlebotomists. In addition
to pathologists, personnel in anatomic pathology laboratories include histotechnologists/
histotechnicians, cytotechnologists, and pathologists’ assistants. POLs are typically staffed by
medical assistants and/or laboratory technologists and technicians. Many of these professionals
acquire additional training as a specialist within a subdiscipline of clinical laboratory testing.2 While
there can be overlap in some of their tasks, their contributions and responsibilities differ in certain
important ways.

Pathologists and Doctoral-Level Laboratory Scientists
Pathologists and doctoral scientists frequently operate at the more senior levels in clinical
laboratories. Often they serve as directors of clinical laboratories, responsible for oversight of
staff, testing processes, quality control procedures, quality of laboratory care for patients, and
other managerial functions. Pathologists are licensed medical doctors who have graduated
from an allopathic or osteopathic school of medicine. They examine samples, interpret results
of laboratory tests, ensure the accuracy of laboratory tests, and consult with clinicians.3 There is
growing recognition of the vital roles of pathologists and doctoral-level laboratory scientists in
the delivery of patient care by consulting with clinicians on ordering laboratory tests and
interpreting the results.4 In most cases, pathologists and doctoral scientists can interpret and
sign off on laboratory test results.a,5
a

Some health care payers will not pay for laboratory test interpretation that is not conducted by a pathologist; in these
cases, pathologists must interpret and sign off on laboratory tests. Some hospitals also require pathologist approval.5

May 2008

105

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

Pathologists in all branches of laboratory medicine (i.e., anatomic pathology, clinical pathology,
and interdisciplinary subjects) tend to specialize in and practice specific clinical disciplines, as
outlined in Box 3.1. For pathologists and doctoral-level laboratory scientists, pathology-related
subspecialties in which certification is available include: immunohematology and transfusion
medicine, chemical pathology, cytopathology, dermatopathology, forensic pathology,
hematology, histocompatibility, immunology, microbiology, molecular diagnostics, molecular
genetic pathology, neuropathology, oral pathology, and pediatric pathology.6
Box 3.1: Certifications Available for Anatomic, Clinical,
and Molecular Pathology Specialties
Clinical Pathology
Chemical Pathology (P, DLLS, T/S)
Hematology (P, DLLS, T/S)
Immunology (DLLS)
Microbiology (P, DLLS, T/S)
Histocompatibility* (P, DLLS)
Immunohematology and Transfusion Medicine
(P, T/S)

Anatomic Pathology
Cytopathology (P, T/S)
Neuropathology (P)
Dermatopathology (P)
Oral Pathology (P)
Forensic Pathology (P)
Histology (T/S, T)
Pediatric Pathology± (P)

- Hemapheresis** (T/S)
- Apheresis** (T)
Phlebotomy (T)
Virology (T/S)

Molecular Pathology
Molecular Genetic Pathology (P, T/S)
Cytogenetics (T/S)
Molecular Diagnostics (DLLS)

P= Certification available for pathologists
DLLS= Certification available for doctoral-level laboratory scientists
T/S= Certification available for technologists/scientists
T= Certification available for technicians
±Pediatric pathology can consist of one or both of anatomic and clinical pathology.
* Pathologists can become certified as histocompatibility laboratory directors through the American Board of
Histocompatibility and Immunogenetics.
**Specialists in hemapheresis have knowledge of all aspects of donor and therapeutic procedures. They have managerial
responsibilities and make clinical and therapeutic decisions. Apheresis technicians work in donor and therapeutic settings
and perform basic and intermediate apheresis procedures; they do not have managerial responsibilities and are not
generally involved in clinical and therapeutic decision-making.7
Sources: About physician specialties: pathology. Evanston, IL: American Board of Medical Specialties, 2007.
Clinical Laboratory Improvement Amendments of 1988 (CLIA) application for certification. Baltimore, MD: Centers for
Medicare and Medicaid Services, 2007.
Candidate handbook. Lenexa, KS: American Board of Histocompatibility and Immunogenetics, 2008. (Accessed April 28,
2008, at http://www.ashi-hla.org/abhi/ABHI-handbook.pdf.)
Reference resources and publications. Northfield, IL: College of American Pathologists, 2007. (Accessed May 2, 2007, at
http://www.cap.org/apps/cap.portal?_nfpb=true&_pageLabel=reference.)

May 2008

106

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

Doctoral-level laboratory scientists have earned advanced degrees in such laboratory-related
fields as clinical chemistry, immunology, genetics, or microbiology; some have also completed
postgraduate fellowships. They are often responsible for establishing new laboratory tests,
interpreting test results, and ensuring accurate and appropriate test outcomes.8 Doctoral-level
laboratory scientists can serve as laboratory directors, with responsibility for supervising the
laboratory’s technical and scientific functions.9, 10 With greater automation of microbiological
laboratory testing, doctoral-level laboratory scientists are shifting to emerging areas of laboratory
testing involving genetics and pharmacogenomics.

Technologists/Scientists and Technicians
These laboratorians represent the two largest groups of laboratory professionals and can be
involved in many aspects of laboratory specimen collection, preparation, and testing, depending
on the nature of the test and their qualifications.
Each group is referred to by interchangeable titles by professional organizations, educational
programs, and other clinical laboratory professionals:

ƒ

Medical technologists/clinical laboratory scientists (MT/CLSs)

ƒ

Medical laboratory technicians/clinical laboratory technicians (MLT/CLTs)

The largest certifier of laboratorians, the American Society for Clinical Pathology (ASCP), uses the
titles of MT and MLT, whereas another certifier, the National Credentialing Agency for
Laboratory Personnel (NCA), uses CLS/CLT. In 2005, ASCP and NCA began discussing the
formation of one credentialing agency for laboratory personnel and unifying professional titles.11,
12 Discussions are ongoing.
In order to minimize use of abbreviations and simplify terminology for this report, MT/CLSs will
be referred to as technologists/scientists and MLT/CLTs as technicians. Unless otherwise noted, use
of these personnel titles includes individuals that have received general certification and those
that have received subspecialty certification.

Technologists/Scientists
Technologists/scientists have earned a bachelor’s degree from an accredited college or university
and have completed an accredited education program specific to medical technology or have met
the education requirements and have obtained the laboratory experience defined by a certifying
agency.13, 14 Their training allows them to conduct laboratory tests on tissues, blood, and other
bodily fluids and perform a range of complex chemical, biological, hematological, and
immunologic tests.15 In addition, they may evaluate and confirm the accuracy of test results,
report laboratory findings to pathologists and other physicians, and develop and alter test
procedures.15, 16 Technologists/scientists often advance to supervisory and
managerial/administrative positions in which they provide day-to-day management of
laboratory operations.
Technologists/scientists can pursue additional education to become certified in the specific areas
of laboratory medicine noted in the section on certification, below. They may receive specialty
certification in blood banking, chemistry, cytogenetics, cytology, hematology,

May 2008

107

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

immunohematology, hemapheresis, histology, laboratory safety, microbiology, molecular
pathology, and virology.13, 14 About 60% of technologists/scientists work in general medical and
surgical hospitals, followed by 14% that work in independent laboratories, and 8% in physician
offices, among other settings (see Figure 3.1).17

Technicians
Technicians perform less complex tasks under the supervision of a technologist/scientist. They
may prepare samples, operate automated analyzers, and perform manual laboratory tests in the
areas of blood banking, chemistry, hematology, immunology, and microbiology.15 Technicians
usually have an associate’s degree from an accredited college or university, with training from an
accredited laboratory technician education program or the equivalent.13, 14 Technicians can pursue
additional education to become certified as a technologist/scientist.13, 14
About 43% of technicians are employed in general medical and surgical hospital settings, 16% in
physician offices, and 14% in independent laboratories (Figure 3.1).18
Figure 3.1: Distribution of Settings of Work for Technologists/Scientists and Technicians
Technologists/Scientists
Government
3%
Colleges, Universities &
Professional Schools
4%

Other
11%

Offices of
Physicians
8%
Medical &
Diagnostic
Laboratories
14%

Technicians
Other Ambulatory
Health Care Services
4%

General Medical &
Surgical Hospitals
60%

Other
15%

Colleges, Universities &
Professional Schools
8%
Medical &
Diagnostic
Laboratories
14%

General Medical &
Surgical Hospitals
43%

Offices of
Physicians
16%

Sources: Occupational employment and wages, May 2006; Medical and clinical laboratory technologists. Washington, DC: Bureau
of Labor Statistics, 2007. http://www.bls.gov/oes/current/oes292011.htm
Occupational employment and wages, May 2006; Medical and clinical laboratory technicians. Washington, DC: Bureau of Labor
Statistics, 2007. http://www.bls.gov/oes/current/oes292012.htm

Pathologists’ Assistants
Through special training, individuals may become pathologists’ assistants, who are often the first
staff to look at an anatomic laboratory sample, provide the pathologist with a description of the
sample, and assist in determining whether further analysis is required.19 Pathologists’ assistants
perform a variety of tasks and are primarily responsible for gross examination of surgical
pathology specimens and conducting autopsies.20 Prior to January 1, 2008, pathways to becoming
a certified pathologist’s assistant included earning a bachelor’s degree and completing an
accredited pathologists’ assistant program or completing three years full-time experience as a
pathologists’ assistant under the supervision of a pathologist.19 This on-the-job training route has
been discontinued; all certified pathologists’ assistants must now complete a formal training
program in order to qualify for certification.

May 2008

108

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

Histotechnologists/Histotechnicians
Histotechnologists (HTLs) and histotechnicians (HTs) work in anatomic pathology laboratories to
prepare tissue for microscopic examination by a pathologist.21 HTLs have advanced training in
understanding how and why specimens are collected and processed. They are responsible for
solving technical and instrumental problems that may arise in the laboratory, understanding the
reasons for unusual test results, and evaluating new laboratory techniques and procedures. In
order to qualify for certification as an HTL, individuals must have a bachelor’s degree and must
complete an accredited HTL program or an associate degree and one year full time experience in
a histopathology laboratory.13, 22

Cytotechnologists
Cytotechnologists are responsible for the microscopic examination of cell samples to detect signs
of cancer and other diseases.23 Cytotechnologists specifically analyze cell changes, both in the
nucleus and cytoplasm, and compare these changes to normal cells from the same site. When
findings for certain specimens are normal, cytotechnologists can issue the final laboratory report;
when abnormal cells are detected, cytotechnologists work with pathologists to determine the final
diagnosis. Individuals must have a bachelor’s degree and complete an accredited cytotechnology
educational program in order to become certified.24

Medical Assistants
Approximately 62% of medical assistants are employed in POLs.25 In this setting, medical
assistants can be responsible for drawing blood and preparing laboratory specimens and
performing basic laboratory tests.26 While some medical assistants receive on-the-job training,
this is becoming less frequent, and medical assistants increasingly complete formal educational
programs that include clinical and academic training in areas such as medical terminology,
laboratory techniques, clinical and diagnostic procedures, and patient relations.25 Medical
assistant educational programs must receive accreditation from designated agencies; graduates of
these programs can apply for certification by organizations such as the American Association of
Medical Assistants.

WORKFORCE DEMOGRAPHICS
Pathologists and Doctoral-Level Laboratory Scientists
In 2005, there were an estimated 19,339 clinical and anatomic pathologists in the U.S.27 Of these,
2,533 were residents and fellows in either discipline. According to the Intersociety Committee on
Pathology Information, 80% of pathologists work in community practice, 15% work in academic
practice and medical school administration, 3% work in industry, and 1% hold government,
public health, and regulatory positions.28 Data from 2006 show that slightly more than half of
clinical and anatomic pathology residents and fellows were female and 34% were graduates of
non-U.S. medical schools.6 While the percentage of female residents remained the same relative
to 2005, approximately 37% of residents were graduates of non-U.S. medical schools in 2005.
About 32% of full-time physician faculty members in clinical and anatomical pathology were
female in 2006, compared to 31% in 2005.

May 2008

109

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

According to the AMA, the pathologist workforce tends to be largely white (46% in 2005),27 with
11% Asian, 3% Hispanic, 1.3% African American, 0.02% American Indian or Alaskan Native, and
2% identified as other; race/ethnicity was not identified by 36% of respondents. Figure 3.2
compares race/ethnicity data for pathologists, technologists/scientists, and technicians. These
percentages are fairly representative of the ethnic and racial composition of the physician
workforce as a whole. The geographic concentration of clinical and anatomic pathologists is fairly
evenly dispersed throughout the U.S. In 2005, approximately 24% of the workforce was
concentrated in the northeastern U.S., 22% were in the north central U.S., 34% were located in the
southern U.S., and 19% were in the western U.S.29
Demographic data for doctoral-level professionals in the laboratory medicine workforce are not
available, though some rough inferences can be made from the profile of students receiving
doctoral degrees in the life and physical sciences. In 2003, 52% of students awarded a doctoral
degree in the life sciences were male, as were 72% of students receiving doctoral degrees in the
physical sciences.30, 31 More than 80% of life and physical science doctoral degree recipients
identified themselves as white, non-Hispanic. However, these percentages are shifting. The
percentage of women awarded degrees in the life and physical sciences has been increasing over
the past several years.
Figure 3.2: Comparison of Racial and Ethnic composition of Laboratorians, 2005
Pathologists

Unknown
36.4%

Technologists/Scientists & Technicians

White
46.3%

African
American
11.4%
Hispanic
7.4%
Asian
12.1%
White
69.1%

Other
2.3%
American Indian
or Alaskan Native
0.02%
African
American
1.3%

Asian
10.5%
Hispanic
3.1%

Sources: Physician characteristics and distribution in the U.S., 2007 edition. Chicago, IL: American Medical Association, 2007.
Employed civilians by occupation, sex, race, and Hispanic origin: 2005. Washington, DC: U.S. Census Bureau, 2005.
http://www.census.gov/compendia/statab/tables/07s0602.xls

Technologists/Scientists and Technicians
The technologist/scientist and technician workforce also has a female majority and is more
representative of the racial/ethnic makeup of the U.S. population.1 In 2006, there were an estimated
160,760 technologists/scientists and 144,710 technicians employed in the U.S.17, 18 Of these
professionals, 74% were female.32 The racial/ethnicity distribution of the combined laboratorian

May 2008

110

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

groups was 69% white, 12% Asian, 11% African American, and 8% Hispanic (Figure 3.2). In 2002,
the last year in which such data is available, the median age of these laboratorians was 41.1
The geographical distribution of technologists/scientists and technicians varies across the U.S.
The Bureau of Labor Statistics defines workforce concentration as the number of people employed
in a specific profession divided by the total number of people employed in all sectors in a defined
geographic area. Concentrations also vary by urban, suburban, and rural status, and may not
reflect the state concentrations listed above.
Based on data from multiple sources, the geographic distribution of these laboratorians is
approximately as follows:

ƒ

The five states with the highest workforce concentration of technologists/scientists are
Massachusetts, South Dakota, Louisiana, Nebraska, and Pennsylvania.17 About 58% of
technologists/scientists are employed in an urban environment, 24% work in suburban
areas, and 18% are employed in a rural location.33

ƒ

The five states with the highest concentrations of technicians are Massachusetts,
Pennsylvania, Tennessee, Kansas, and Indiana.18

VACANCY RATES
There is significant concern across the laboratory medicine sector about the growing shortage of
clinical laboratory workers, which is expected to worsen over the next decade, particularly for
technologist/scientist and technician positions.1 Several factors are cited as contributing to this
shortage: the pending retirement of many members of the workforce, competing career
opportunities, and difficulty recruiting and retaining staff.1, 34 The effects of technological
advancement on the workforce appear to be mixed. While greater automation can decrease the
need for personnel in the laboratory and enable non-laboratory personnel to perform tests,
operating more advanced laboratory equipment can require more highly-trained
technologists/scientists and technicians. These effects may shift the skill sets required for
laboratory personnel at all levels.15

Pathologists and Doctoral-Level Laboratory Scientists
Currently available data do not indicate a shortage of pathologists.35 However, virtually every
academic department in the U.S. is reported to have a vacancy in pathology.28 According to the
ASCP’s 2006 Resident Council Fellowship and Job Market Survey, 85% of respondents in their final
year of residency or fellowship who had applied for a job had at least one offer of employment, an
increase of 10% from the 2005 survey.36 The remaining 15% are reported to have taken fellowship
positions. Of those applying to pathology fellowship positions, 88% received at least one offer, a
decrease from the 97% of applicants who were offered at least one fellowship in 2005.b
Data on vacancies in doctoral-level laboratory scientists is not available, but anecdotal evidence
from the American Board of Clinical Chemistry (ABCC) suggests that the number of individuals
certified by the board each year has been decreasing over the past several years.37
b

The decrease in the percentage of fellowship applicants who were awarded at least one fellowship is attributed to
the increasing number of pathology residents applying for fellowships.36

May 2008

111

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

Other Laboratory Personnel
Vacancies for technologists/scientists and technicians, especially at the phlebotomist and
cytotechnologist levels, are more extensive than for pathologists and doctoral-level laboratory
scientists. The ASCP Board of Registry has been collecting information on vacancy rates on a
biannual basis since 1988.1 Their 2005 survey of wages and vacancies (Table 3.1) showed that
vacancy rates for both were highest prior to 2002 before dropping in 2003.34 From 2003 to 2005,
vacancy rates increased slightly for technologists/scientists at all levels (i.e., staff, supervisory, and
managerial), and increased for technicians at the supervisory level (including phlebotomists).
ASCP identified several trends in vacancy rates by laboratory setting:

c

ƒ

Reference laboratories had higher-than-average vacancy rates for certified
technologists/scientists and technicians.

ƒ

Hospital-based laboratories reported slightly higher-than-average vacancy rates for
phlebotomy staff and supervisors.

ƒ

Vacancy rates for certified HTLs and HTs were higher for laboratories housed in
facilities with 500 beds or more. These laboratories also reported higher-than-average
vacancy rates for certified technologists/scientists staff and supervisors.

ƒ

Laboratories located in the northeastern U.S. reported high vacancy rates for certified
HTLs.

ƒ

Vacancies for certified technologists/scientists staff were higher than average in the far
western and northeastern U.S., while vacancies for certified phlebotomy staff were
highest in the far western U.S.c,34

California recently began requiring certification of phlebotomists and imposing more stringent regulations
regarding phlebotomy certification.38 The higher-than-average vacancy rate among phlebotomy staff in
the far western U.S. may be attributable to California’s phlebotomy certification requirement.

May 2008

112

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

Table 3.1: Vacancy Rates for Various Clinical Laboratory Personnel Positions in the U.S.
Selected Years 2000-2005
Position

2000

2002

2003

2005

7%
6%
4%

4%
3%
2%

6%
4%
4%

Medical Technologist/Clinical Laboratory Scientist
Staff
Supervisory
Manager

11%
13%
13%

Medical Laboratory Technician/Clinical Laboratory Technician
Staff
Supervisory

14%
n/a

9%
7%

6%
2%

6%
3%

18%
n/a

9%
8%

7%
3%

7%
5%

16%
22%
20%

9%
11%
6%

6%
4%
5%

4%
7%
4%

21%
10%

8%
6%

4%
2%

3%
2%

Phlebotomist
Staff
Supervisory
Histologist
Histotechnician
Histotechnologist
Supervisory
Cytologist
Staff
Supervisory

Source: Steward CA, Thompson NN. ASCP 2005 wage and vacancy survey of medical laboratories. Laboratory
Medicine 2006;37(8):465-469.
Note: Vacancy rates varied by geographic region.

WAGES
Pathologists
Pathologists earn salaries comparable to other physicians. Starting salaries for most pathologists
exceed $125,000 per year and vary by geographic region and place of employment.3 The ASCP’s
2006 job market survey asked pathology residents and fellows about their job offers.36 Of those
respondents who had been offered a job, including both residents and fellows in their final year of
training, 12% were offered less than $100,000 per year, 39% were offered a salary that ranged from
$100,000 to $150,000, 44% were offered a salary between $150,000 and $200,000, and 5% were
offered a salary greater than $250,000. After their first and second year of training, another study
of allied physicians found that pathologists earned a base salary of $169,000; those with more than
three years of practice earned $321,000.d,39 In 2007, the Medical Laboratory Observer annual
survey of 1,873 laboratory personnel found that the median annual salary of pathologists was
$190,000.40 Table 3.2 provides summary datae compiled by the Modern Healthcare Physician
d
e

This survey did not report whether figures represented median or average incomes.
The following surveys were included in the data: American Medical Group Association: data from more than 32,000
member medical group physicians in 123 specialties. Hay Group: surveyed 98 organizations. Hospital and
Healthcare Compensation Service: 17,650 physicians in 44 specialties. Medical Group Management Association:
40,295 physicians in more than 105 specialties. Sullivan, Cotter and Associates: more than 175 organizations.
Warren Surveys: more than 7,000 physicians in 32 specialties. Merritt, Hawkins and Associates: 2,687 physicians.

May 2008

113

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

Compensation Review of pathologist salary ranges from seven different survey instruments as
reported in 2005.
Table 3.2: Comparison of Pathologist Salary Ranges to Other Physician Groups, 2005
Physician Specialty
Pathology

Salary Range
$193,477 - 350,286

Anesthesiology

$274,886 - 338,722

Cardiology

$287,907 - 387,800

Internal Medicine

$163,250 - 180,800

Obstetrics/Gynecology

$222,838 - 275,800

Oncology

$241,628 - 320,200

Pediatrics

$144,000 - 184,900

Radiology

$209,365 - 411,131

Source: 2005 Modern Healthcare Physician Compensation Review. Irving, TX: Merritt, Hawkins & Associates, 2005.

Doctoral-Level Laboratory Scientists
Although salary information specifically for doctoral-level laboratory scientists is unavailable,
surveys of laboratory directors and laboratory professionals with postgraduate degrees have been
conducted. A 2004 survey of 2,128 laboratory professionals conducted by Medical Laboratory
Observer found the median salary for laboratory directors was $86,238, approximately 3% greater
than the median salary reported in the 2003 survey.41, 42 Median salary for laboratory
professionals with a postgraduate degree was $76,454.
In addition, several organizations track salary information for professionals who have achieved
doctoral-level chemistry and toxicology degrees. A 2006 salary survey conducted by the
American Chemical Society, completed by 8,580 respondents, found that the median base salary
for professionals with doctoral-level chemistry degrees was $95,000.43 Average annual salaries
increased by 3.4% between 1996 and 2006, adjusting for inflation. A 2004 survey of individuals
with doctoral-level degrees in toxicology conducted by the American College of Toxicology and
the Society of Toxicology found that salary varied by years of experience following terminal
degree and by sex.44 Mean annual salary for individuals employed in contract laboratories with
three to five years experience was $81,000 for men and $90,000 for women; mean annual salary for
individuals employed by the federal government with three to five years experience was $78,000
for men and $75,000 for women.

Technologists/Scientists and Technicians
Significant differences in salary for technologists/scientists are based on education, geographic
location, job function, and work experience.40 Nationally, annual salaries in 2005 ranged from
$24,419 for staff phlebotomists to $66,539 for technologist managers.34 Laboratory employees with
a postgraduate college degree made about $22,000 more than employees with a high school
diploma. Table 3.3 provides specific wage information delineated according to position.
ASCP found that the median average hourly salaries for personnel working in hospital-based and
reference laboratories were more likely to be higher than personnel working in POLs.34 Salaries

May 2008

114

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

also were higher in larger laboratories whose test volume exceeded one million tests per year.
Other data indicate that most laboratory technologists/scientists working with management and
technical consulting services earned higher wages. Yet, laboratory managers and administrators
and information systems managers earned more money on average than medical technologists,
section managers and section manager supervisors.40
Geographic assessment of wages indicate that California, Alaska, and Nevada were the toppaying states for technologists/scientists, whereas Rhode Island, Alaska, and Hawaii were the
top-paying for laboratory technicians.17, 18 Conversely, professionals working in the mountain
and southeast areas of the U.S. were paid less on average than those in other regions.
Table 3.3: Median Average Wages for all
Clinical Laboratory Positions and Levels in the U.S., 2005
Position

Median Average Annual Salary*

Technologist/Scientist
Staff
Supervisory
Manager

$44,762
$53,997
$66,539

Technician
Staff
Supervisory

$35,838
$41,642

Phlebotomist
Staff
Supervisory

$24,419
$34,923

Histologist
Histotechnologist
Histotechnician
Histotechnican- Supervisory

$44,990
$38,418
$54,018

Cytologist
Staff
Supervisory

$54,434
$63,523

*Median average annual salary calculated from median average hourly wage using
assumption that employees are working 2,088 hours per year, or 40 hours per
week for 52 weeks.
Source: Adapted from Steward CA, Thompson NN. ASCP 2005 wage and vacancy
survey of medical laboratories. Laboratory Medicine 2006;37(8):465-469.

May 2008

115

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

EDUCATION, TRAINING, AND RECRUITMENT
Medical Schools
The educational requirements for pathologists, whether for an Doctor of Medicine (MD) degree or
Doctor of Osteopathy (DO) degree, match those of all medical doctors, which include premedical school requirements, four years of medical school, and three-to-six years of post-graduate
training.3, 45 For purposes of comparison, allopathic medicine (traditional medicine) is
characterized as using remedies to counter the effects produced by the disease and emphasizes
diagnosis and treatment of specific diseases within the body.46, 47 Traditional allopathic medical
school curricula consist of two years of basic science education and two years of clinical rotations.
Osteopathic medicine is characterized by its emphasis on the connection between structure and
function and the body’s inherent ability to heal itself.48 Similar to MDs, doctors of osteopathic
medicine can choose any medical specialty, prescribe drugs, perform surgery, and practice
medicine anywhere in the U.S.49 Schools for osteopathic medicine also require two years of basic
science education followed by two years of clinical clerkships and three-to-six years of postgraduate training.50
Current data indicate that:

ƒ

There are 148 medical schools in the U.S., of which 125 teach allopathic medicine and
23 teach osteopathic medicine.51, 52 In 2006, an estimated 69,167 students were enrolled
in allopathic medical schools, a 5% increase since 2002.53 Male-female ratios were
relatively even (49% female).

ƒ

Enrollment in osteopathic schools of medicine, which totaled 13,406, grew at a
substantially higher rate of 17% during this same period.54, 55 Approximately 50% of all
osteopathic medical school students were female.

The need for more physicians in the U.S. and predictions of physician shortages are contributing to
a push for increased enrollment in medical schools. The Council on Graduate Medical Education,
an advisory group to the Health Resources and Services Administration, has recommended that
allopathic and osteopathic medical schools increase their enrollment by 15% from their 2002 levels
over the next 10 years, both by expanding the size of first-year classes and by building new medical
schools.56 Emphasis on recruitment and enrollment are proving successful.

ƒ

In 2007, a total of 42,315 people applied to allopathic medical schools, an increase of 8%
since 2006.57 Earlier data (2005) found a similar trend with osteopathic medical
schools, which received 8,258 applications, a 14% increase over the previous year.58

ƒ

Actual enrollments in medical school are projected to increase. First-year enrollment in
allopathic medical schools is expected to increase by 17% to nearly 19,300 students by
2012.59 Osteopathic medical school enrollment may double between 2002 and 2015.60

Residency and Fellowship Programs
Following medical school, allopathic and osteopathic graduates can apply to pathology
residency programs through the National Residency Match Program (NRMP), which is a
private, not-for-profit corporation that provides a uniform date of appointment to all entering

May 2008

116

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

medical residents.61 The length of time required to complete residency and post-graduate
training in pathology ranges from three to six years, depending on field of pathology in which a
physician is specializing.45 Residency programs that combine clinical and anatomic pathology
require four years of training, while those that focus on either clinical or anatomic pathology
require three years. Training in a specific pathology subspecialty requires an additional year,
with the exception of neuropathology which requires an additional two years. Residency
programs for pathology have not been developed by osteopathic-related organizations. As a
result, osteopathic graduates must enter NRMP allopathic residency programs in order to
practice pathology. However, the Osteopathic Postdoctoral Training Institute-Westf is
developing pathology residencies in California and Oregon.62, 63

Residency Programs
As of 2007, there were 150 NRMP-accredited anatomic and clinical pathology residency
programs.6 In 2006, there were an estimated 2,603 active pathology residents and fellows.36 There
are notable differences in trends between the 2001-2005 and 2005-2007 periods, as follows.

ƒ

The total number of pathology residency positions offered through the NRMP
increased from 383 to 526 positions from 2001 to 2005, but decreased by 13 positions
between 2005 and 2007.64

ƒ

The number of allopathic medical school seniors entering pathology residency
programs increased by an average of 19% annually from 2002 to 2005, but decreased by
an average of 5% from 2005 to 2007.65 Still, only about 2% of all medical students enter
pathology residencies.64

ƒ

Allopathic seniors filled 50% of pathology residency positions in 2002, and 55% in
2007.64 Graduates of international and osteopathic medical schools and prior graduates
of allopathic medical schools filled approximately 34% of pathology residency
positions in 2002 and 33% in 2007.64, 65

ƒ

Approximately 9% of pathology residency positions remained vacant in 2007,
compared to 16% in 2002.

ƒ

Fewer than 1% of osteopathic graduates have chosen a career in pathology during the
past 10 years.54

Fellowship Programs
Following completion of a pathology residency, many physicians pursue specialty training through
fellowship programs. According to the ASCP’s 2006 job market survey, which was completed by
742 residents in their final year of training, 702 applied for fellowships.36 More than half of
respondents attributed their pursuit of fellowship training to their long-term career interests; 29%
indicated that the completion of a fellowship was necessary to secure eventual employment in their
desired position. Subspecialty training programs and enrollment for residents and fellows for
pathology programs leading to subspecialty certification are provided in Table 3.4.

f

Osteopathic Postdoctoral Training Institute is an educational consortium with 8 member institutions that supports
osteopathic graduate medical education programs in the western U.S.

May 2008

117

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

Table 3.4: Number of Accredited Pathology Specialty Programs and Total Number of
Active Residents/Fellows in Program in the U.S., 2006g
Number of
Accredited Programs

Total Number of Active
Residents/Fellows in Program

Blood Banking/Transfusion Medicine

46

36

Chemical Pathology

3h

2

Cytopathology

85

109

Dermatopathology

47

51

Forensic Pathology

38

28

Hematology

77

88

Medical Microbiology

12

9

Molecular Genetic Pathology

18

12

Pathology Specialty

Neuropathology

35

38

Pediatric Pathology

25

18

Source: FREIDA online specialty training statistics information. Chicago, IL: American Medical Association, 2005. Accessed
February 28, 2007. http://www.ama-assn.org/vapp/freida/spcstsc/0,1238,300,00.html.

Doctoral-Level Laboratory Science Programs
Professionals can enter the laboratory workforce as graduates of doctoral programs in laboratoryrelated fields such as clinical chemistry, toxicology, and microbiology. Requirements for entry
into doctoral programs vary according to the institution, but most U.S. science graduate programs
require applicants to have completed a bachelor’s and/or master’s degree and required entrance
examinations.67, 68
Doctoral study typically involves three stages of academic work.69 The first stage consists of
preliminary course, seminar, and laboratory studies. The second stage includes a set of advanced
seminars and consortia during which students select a dissertation subject and design their
research. Independent research as well as the writing, presentation, and defense of the thesis
encompass the final stage of doctoral education. A doctorate of philosophy or doctorate of
sciencei in a laboratory-related subject area is required for board certification.70, 71
Most laboratory doctoral scientists also complete postdoctoral fellowships that last one or two years
and prepare them to serve as a director and/or laboratory consultant in a variety of laboratory
settings, including microbiology, immunology, and public health laboratories.72 Postdoctoral
fellowship programs are accredited by a variety of professional societies. For example, there are
currently 12 postdoctoral training programs located in medical centers across the U.S. approved by
the American College of Microbiology’s Committee on Postgraduate Educational Programs.73 The
Commission on Accreditation in Clinical Chemistry currently accredits postdoctoral fellowship
training programs at 18 institutions across the U.S. and Canada.74
There is no subspecialty certification program for surgical pathology; therefore, it was not included in this table.
The Accreditation Council for Graduate Medical Education only recently began accrediting chemical pathology
programs and, thus, the number of programs is currently low but expected to increase in coming years.66
i In some cases, individuals who have earned a doctoral-level degree in osteopathic medicine, veterinary medicine,
public health, or dental surgery can be accepted for board-level certification as a doctoral-level laboratory scientist if
they have met the requirements for postdoctoral training and laboratory-related experience.70
g

h

May 2008

118

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

A total of 15,560 doctoral degrees were awarded in biology, biomedicine, health and clinical
science, physical science, and science technologies during the 2004-2005 academic year, an
increase of approximately 16% relative to 2003-2004.75 About 61% were awarded by public
universities. Degrees awarded in these areas represent approximately 28% of all doctoral degrees.
This percentage is representative of the overall increase in doctoral degrees awarded over the past
25 years, as life sciences doctorates have increased 57% and physical sciences doctorates have
increased 30%.30, 31 However, these professionals are not necessarily eligible to perform the duties
of a doctoral-level laboratory scientist.
ASCLS and other organizations are developing educational programs that lead directly to a
doctorate in clinical laboratory science (DCLS).76 The DCLS would provide an alternative to
traditional master’s and research-based doctoral degrees. The National Accrediting Agency for
Clinical Laboratory Sciences (NAACLS) approved standards for accreditation of DCLS programs
in September 2006 and began reviewing applications for programs interested in offering the
DCLS degree in April 2007.77

Technologist/Scientist and Technician Education Programs
Educational requirements for technologists/scientists are more stringent than those for
technicians. Becoming a technologist requires a bachelor’s degree in medical technology, clinical
laboratory science, or one of the life sciences.1, 15 Technicians, on the other hand, typically have an
associate’s degree from a junior or community college or training from a hospital, a technical or
vocational school, or from the U.S. Armed Forces.15 It also is possible to become a technician
without obtaining an associate’s degree by completing required coursework in biology and
chemistry at an accredited college or university.13 Along with formal training, both
technologists/scientists and technicians must complete additional clinical education in a medical
technology or clinical laboratory science program accredited by NAACLS.j,16 Depending on the
state in which they practice and the setting in which they work, further training beyond these
requirements may be necessary.
As depicted in Figure 3.3, the majority of these educational opportunities are provided in
hospital-based programs, universities, and community colleges.78 There are clear differences in
educational sites between technologist/scientist programs and technicians.

j

Depending on the route followed, not all certified technologists/scientists and technicians complete NAACLS-accredited
programs. For example, some technicians have an associate’s degree or 60 semester hours of academic credit, have
completed required coursework, and have completed a 50-week U.S. military medical laboratory training course.

May 2008

119

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

Figure 3.3: Educational Program Settings for Technologists/Scientists and Technicians,
2006
Technologists/Scientists
Community
College
Independent 0.6%
Other
Laboratory
0.6%
1.1%

Technicians
Hospital/Medical
Center/Blood
Military
Bank
Facility
Other 1.2%
0.6%
4.3%

University/
College
9.9%
University/
College
44.3%

Hospital/Medical
Center/Blood
Bank
53.4%
Community
College
84.0%

Source: Bugbee AC. ASCP Board of Registry’s 2006 annual survey of medical laboratory science programs. Lab Medicine,
2007;38(8):463-471

Programs
All technologist/scientist and technician education programs must be accredited by the NAACLS
or the Commission on Accreditation of Allied Health Education Programs.1 NAACLS accredits
more than 469 programs for technologists/scientists and technicians, HTLs, HTs, pathologists’
assistants, diagnostic molecular scientists, cytogenetic technologists, and phlebotomists.15, 79 The
Commission on Accreditation of Allied Health Education Programs accredits 47 programs in
cytotechnology and 15 programs for blood banking specialists.80, 81 Figure 3.4 portrays trends in
the numbers of NAACLS-accredited education programs for specific laboratory professional
groups between 1970 and 2007. Among these trends:

ƒ

The number of education programs for technologists/scientists and technicians have
declined since 1975, and enrollment in these programs has declined over 50% since
1980. (See discussion on student population.)1, 82

ƒ

The most dramatic decline has been in technologists/scientists programs,
approximately 71% of which closed between 1970 and 2007.83 Four programs closed
between 2006 and 2007 alone.

ƒ

From 1985 to 2007, the number of technician education programs declined by
approximately 27.0%. However, four new programs were created during 2006-2007.

ƒ

Declines in the number of education programs fall into specific disciplines. The
number of HTL and HT programs diminished from 49 in 1983 to 30 in 2006, a decline
of 38.8%, while the number of phlebotomy training programs increased six-fold, from 9
to 58 from 1987 to 2003.

May 2008

120

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

Number of Accredited Programs

Figure 3.4: Trend in Number of NAACLS-Accredited Education Programs in
Clinical Laboratory Sciences, 1970-2007
900
800
700
600
500
400
300
200
100
0
1970 1977 1983 1987 1989 1991 1993 1995 1997 1999 2001 2003 2005 2007

Year
Technologists/Scientists

Technicians

Histotechnologists/Histotechnicians

Source: NAACLS table of program and graduate numbers. Tice D, ed. Chicago, IL: National Accrediting Agency for the Clinical
Laboratory Sciences, 2007.

A variety of factors have affected the laboratory medicine educational programs. The Medicare
Prospective Payment System changed the hospital payment structure such that clinical laboratories
(including outreach testing), once a source of revenue, became cost centers.82 Increases in treatment
via the outpatient setting further decreased hospital revenue by diminishing the number of
inpatients requiring laboratory tests. Other factors often cited as reasons for the decline in
laboratorian education programs include their operating expenses, the number of different
instructors and faculty members required, and the lack of outside funding. The decreasing number
of students entering laboratorian education programs is attributed to additional factors.
Historically, women have dominated the technologist/scientist and technician workforce;
however, as wider employment opportunities have arisen, they have been entering other positions
in science and medicine. Salary and career advancement opportunities associated with laboratory
medicine often are less desirable than those in other health-related industries. Work schedules for
laboratorians, which can require long hours and overnight shifts, may be less attractive than for
some other health professions and other competing employment opportunities.1, 84

Curriculum
Due to the changing nature of laboratory medicine (e.g., increases in genetic testing, increased
use of technology systems and automation), more than half of all medical laboratory science
program directors have reported changes to the educational curriculum in 2002, the last year
that this data was reported by ASCP. Among programs reporting curricular changes,
approximately 24% adjusted molecular science content, 18% changed management skills
content, and 15% altered online content during the 2002-2003 academic year (Figure 3.5).85
Fewer than 5% of programs reported deleting any curricular content. The long-term benefit of
these changes is unclear at this time.

May 2008

121

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

Proposals for widespread, systemic changes include raising awareness of clinical laboratory
professional’s role in health care, lowering the operational costs of training programs (e.g.,
incorporating distance learning, founding education programs at independent laboratories),
and educating laboratory professionals in new technology-related areas such as POCT and
informatics.1, 82

Percentage of Programs Responding

Figure 3.5: Percentage of Technologist/Scientist and Technician Programs
Reporting New Content to Curricula, 2002
30%

24.0%

25%
20%

18.0%
15.0%

17.0%

15%
10%

6.0%

5%

4.0%

4.8%

0%
Management

Molecular

Online

Other

Research

Shared

Deletions

Type of Program Content Added
Source: Ward-Cook K, Chapman S, Lindler V. Executive summary: 2002 annual survey of accredited/approved medical
laboratory science programs. Chicago, IL: American Society for Clinical Pathology, 2002.
http://www.ascp.org/Certification/ForProgramDirectors/research/documents/Svy02.pdf

Student Population
Enrollment
Data on the number of students entering technologist/scientist, technician, and HT/HTL
programs are available only through 2002.k In this year, a total of 4,782 students entered
NAACLS-accredited programs for technologist/ scientist, technician, and HT/HTL.1, 83 In that
year, 3,974 graduated.
Of students entering laboratory medicine education programs, 40% entered technologist/scientist
programs, 54% entered technician programs, and over 5% entered HT/HTL programs.
Recruitment efforts targeting minorities and males have resulted in recent increases in enrollees
for blood banking and histotechnology. Given the importance of active recruitment, about half of
programs have staff and one-third have special budgets dedicated to recruiting new students.

k

Although NAACLS collects enrollment data, there are discrepancies in defining point of enrollment that affect
counts (i.e., according to when the enrollee begins their educations, enters the professional sequence, or enters the
clinical sequence). For this reason, this enrollment data are used only internally by NAACLS.83

May 2008

122

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

A 2006 survey that examined medical laboratory science program directors’ perceptions of the
quality of applicants found that about 40% perceived no change in quality, 21% perceived an
increase in quality, and 13% perceived a decrease in quality. The remaining 27% gave more than
one answer to this question. Figure 3.6 provides a comparison of program directors’ perceptions
in 2002 and 2006.78, 85
Figure 3.6: Change in Quality of Applicants as Perceived by
Laboratory Science Program Directors, 2002 and 2006

60%
50%
40%

52%
40%

38%

30%

21%

20%

10%

13%

10%
0%
Increase

Decrease
2002

No Change
2006

Note: Percentages for 2006 do not add to 100 because some respondents gave more than one answer.
Sources: Ward-Cook K, Chapman S, Lindler V. Executive summary: 2002 annual survey of accredited/approved medical
laboratory science programs. Chicago, IL: American Society for Clinical Pathology, 2002.
http://www.ascp.org/Certification/ForProgramDirectors/research/documents/Svy02.pdf
Bugbee AC. ASCP Board of Registry’s 2006 annual survey of medical laboratory science programs. Lab Medicine 2007;38(8):463-471.

Graduates
There have been two periods of steep decline in the number of graduates from laboratory
education programs. The first was a decrease of 42% that occurred from 1977 to 1990; the second
was a decrease of 45% that occurred from 1994 to 2002.83 Since 2002, the number of graduates has
increased steadily by about 7% per year. Figure 3.7 shows the student populations graduating
from NAACLS-accredited technologist/scientist, technician, and HT/HTL programs for the years
1970 through 2006.
Concerted efforts to promote laboratory programs have succeeded in increasing some graduate
rates. Since 2002, the numbers of technologist/scientist and technician graduates have increased
30% and 33%, respectively.83 The number of HT/HTL graduates increased by 17% from 2001 to
2003, then declined by 11% from 2004 to 2006. As noted above, these changes have accompanied
the decrease in number of programs.

May 2008

123

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

Figure 3.7: Graduates of NAACLS-approved Technologists/Scientists, Technicians, and
Histotechnologists/Histotechnicians Programs by Program, 1970-2006
7,000
6,000
5,000
4,000
3,000
2,000
1,000
0
2006

2005

2004

2003

2002

2001

2000

1999

1998

1997

1996

Technicians

1995

1994

1993

1992

1991

1990

1989

1988

1987

1985

1983

1980

1977

1975

1970

Technologists/Scientists

Histotechnologists/Histotechnicians

Source: NAACLS table of program and graduate numbers. Tice D, ed. Chicago, IL: National Accrediting Agency for the Clinical
Laboratory Sciences, 2007.

LICENSING AND CERTIFICATION
State licensure and certification requirements for laboratory directors are embodied in the CLIA
regulations and are based on the type of testing performed in the laboratory. Laboratories
performing only waived tests do not have requirements for laboratory directors or other
personnel.86

Licensure
Licensure, sometimes referred to as “right of practice,” is the most restrictive form of professional
and occupational regulation.1 CLIA requires state licensure for individuals serving as directors of
PPM laboratories and clinical consultants and for individuals with MD or DO degrees serving as
directors of non-waived laboratories.9, 87, 88 Under CLIA, laboratory personnel performing PPM
testing must also be licensed to practice in their state.89 CLIA does not require licensure of
doctoral-level laboratory scientists.

Pathologists
As with all physicians, pathologists must receive a medical license to practice. Licensure for
physicians is granted by state boards of medicine and is required to guarantee to the public that a
physician has successfully completed medical education and passed an examination or other form
of certification demonstrating competency and appropriate qualifications to practice medicine.90
All states, the District of Columbia, and the U.S. territories, a total of 54 jurisdictions, require

May 2008

124

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

physicians to be licensed before they can legally practice medicine.91,l A license is necessary for
each state in which a physician wishes to practice. According to the Federation of State Medical
Boards and the Bureau of Labor Statistics, physicians must provide proof of graduation from an
accredited medical school, complete at least one year of residency training, and pass a licensing
examination.90, 91 Physicians who were educated outside of the U.S. or Canada are required to
complete a residency in the U.S. prior to obtaining a license to practice medicine in the U.S., even
if they are licensed to practice in another country.90
There are no licensure requirements for doctoral-level laboratory scientists.

Technologists/Scientists and Technicians
In recent years, several state-based initiatives have been undertaken to institute licensure
requirements for technologists/scientists and technicians. Table 3.5 summarizes current status of
licensure activities.
Table 3.5: Status of Licensure Requirements for
Technologists/Scientists and Technicians by State, 2007
Licensure of Laboratory
Personnel Required
California
Florida
Georgia
Hawaii
Louisiana
Montana
New York
Nevada
North Dakota
Rhode Island
Tennessee
West Virginia
Puerto Rico

Licensure Legislation
Introduced/Will Be Introduced in
Next Legislative Session
Illinois
Massachusetts
Michigan
Minnesota
Missouri
Ohio
Texas

Considering a Licensure
Initiative
Indiana
Iowa
Nebraska
Virginia
Wisconsin

Source: Garrott P. American Society for Clinical Laboratory Science. Personal communication. December 10, 2007.

In licensure states, laboratorians can practice only in the areas for which they are licensed.
Employers can hire only licensed individuals to complete tasks within the defined scope of the
licensure. However, the provisions governing licensure can vary by state. Most states require
licensed personnel to pay an annual or bi-annual licensing fee, participate in continuing
education, and meet minimum educational and professional competency requirements.93 Some
states require fingerprinting, documentation of certification from an accredited certifying agency,
and documentation of a defined number of hours of continuing education.

l

State medical boards can prepare their own licensing examinations or they can administer one prepared by and
purchased from a specialized agency, such as the U.S. Medical Licensing Examination or the Comprehensive
Osteopathic Medical Licensing Examination.92

May 2008

125

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

Certification
Under CLIA, board certification is required for doctoral-level laboratory scientists, who are not
previously grandfathered, to serve as laboratory directors or clinical consultants for non-waived
testing laboratories.9, 87 Some routes by which a pathologist can serve as a laboratory director
also require board certification.9, 87 Certification is a voluntary process for other clinical
laboratory personnel (i.e., pathologists, technologists/scientists, and technicians) by which
nongovernmental agencies grant recognition to professionals whose levels of competence meet
specific standards.9, 15, 87, 94, 95

Pathologists
Physicians wishing to practice a medical specialty in the U.S. can seek certification from 24
nationally recognized medical specialty boards.96 Certification of physicians practicing in a
specialty is recognized by physicians, health care institutions, insurers, and patients as proof of a
physician’s knowledge and skills. Although physician certification is not required by law, some
health care plans provide additional benefits and recognition to physicians who are board
certified and who maintain their certification.97
The American Board of Pathology (ABP), under the American Board of Medical Specialties,
provides primary certification in clinical pathology, anatomic pathology, or both.m ABP also
offers certification in 10 pathology subspecialties.99 Examinations comprise written and practical
components, both of which candidates must pass in the same sitting.99 In order to apply for ABP
certification, applicants must have graduated from a U.S. or Canadian medical school accredited
by the Liaison Committee on Medical Education or from an osteopathic medical school accredited
by the Bureau of Professional Education of the American Osteopathic Association (AOA).
Applicants for primary and subspecialty certification also must provide proof of valid licensure to
practice medicine in a state or jurisdiction of the U.S. or Canada.
The American Osteopathic Board of Pathology (AOBP), part of the AOA, is responsible for
evaluation and recommendation of osteopathic physician candidates for certification in anatomic
pathology, laboratory medicine, and forensic pathology.100 In order to receive AOBP certification,
candidates must be a graduate of an AOA-accredited college of osteopathic medicine, licensed in
the state or territory of practice, a member of the AOA or the Canadian Osteopathic Association
for two years prior to the date of certification, complete one year of AOA-approved internship
plus four years of additional training in clinical or anatomic pathology, and provide evidence of
conformity to the AOA Code of Ethics.101
Effective January 1, 2006, all primary and subspecialty certificates issued by the ABP expire after
10 years.102 Pathologists must fulfill requirements in key areas to be recertified.n Pathologists
who received ABP certification prior to this date can participate in the maintenance of certification
The ABP administers all certification examinations in computer-based format at the APB Examination Center in
Tampa, FL.98 Primary certification examinations are offered twice a year; pathology subspecialty certification
examinations in pathology, medical microbiology, molecular genetic pathology, neuropathology, and pediatric
pathology are given every two years.
n ABP recertification requirements include: proof of unrestricted license to practice medicine; participation in
continuing medical education; completion of a recertification exam; and provision of peer attestations of
interpersonal and communication skills, professionalism, ethics, and effectiveness in systems-based practice.
m

May 2008

126

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

program on a voluntary basis. Similarly, on January 1, 1995, AOBP’s recertificationo requirements
for osteopathic pathologists became valid for 10 years. However, for those certified prior to 1995,
recertification is voluntary.

Doctoral-Level Laboratory Scientists
CLIA requires board certification for all doctoral-level laboratory scientists who are serving as a
laboratory director and clinical consultant. Four organizations are currently approved by DHHS
to certify doctoral-level laboratory scientists:

ƒ

American Board of Bioanalysis

ƒ

American Board of Clinical Chemistry

ƒ

American Board of Medical Laboratory Immunology

ƒ

American Board of Medical Microbiology

Candidates must demonstrate appropriate education, postdoctoral training, and laboratory
experience.70, 71, 103, 104 The length of training or experience required varies by board and by the
educational route chosen by the candidate (e.g., completion of a postdoctoral fellowship), but
most boards require a minimum of three years of postdoctoral experience prior to certification. In
some cases, individuals who participate in postdoctoral fellowships are eligible to apply for early
admission to the examination. For example, applicants who have completed at least one year of a
postdoctoral clinical chemistry training program accredited by the Commission of Accreditation
in Clinical Chemistry are eligible to take the ABCC certification examination prior to completing
their fellowship.p,103
Candidates who have met all certification requirements are eligible to take a written certification
exam. Candidates who pass the examination are recognized as diplomates. All four boards
require diplomates to participate in continuing education programs in order to maintain
certification. Table 3.6 displays the number of diplomates certified by each board in 2007 and the
total number of active diplomates certified by each board.
The ABCC reports that, over the past 15 years it, has certified an average of 11 diplomates in
clinical chemistry, 2 diplomates in toxicology, and 2 diplomates in molecular diagnostics.37
According to this and other organizations, the number of diplomates has been decreasing over the
past several years. As in other areas of laboratory medicine, the decrease is attributed to increases
in use of advanced technology systems in the laboratory and market consolidation. Anticipated
retirement of a significant number of doctoral-level laboratory scientists over the next several
years is expected to increase the demand for new diplomates.

For AOBP recertification, osteopathic pathologists must: hold certification for at least 8 years; present written
evidence of continuous compliance with the initial requirements of the AOA for specialty certification; and
submit documents that provide evidence of having attended, presented, or participated in at least 75 hours of
education programs, seminars, lectures, or other academic sessions relating to pathology or a division of
pathology during the preceding three-year period.
p After obtaining certification, these applicants must complete their fellowship and meet all requirements before
becoming a director of a high complexity laboratory or a clinical laboratory consultant.
o

May 2008

127

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

Table 3.6: Number of Diplomates Certified in 2007 and Total Number of
Active Diplomates by Doctoral-Level Laboratory Scientist Certification Boards
Number of Diplomates
Certified in 2007

Total Number of
Active Diplomates

American Board of Bioanalysis

Not Available

Not Available

American Board of Clinical Chemistry

16

346

American Board of Medical Laboratory Immunology

3

72

American Board of Medical Microbiology

25

310

Sources: ABCC register of active diplomates. Washington, DC: American Board of Clinical Chemistry, 2007.
ASM ABMLI diplomates database. Washington, DC: American Board of Medical Laboratory Immunology, 2007.
ASM ABMM diplomates database. Washington, DC: American Board of Medical Microbiology, 2007.

Technologists/Scientists and Technicians
Voluntary certification is the professional standard for technologists/scientists and technicians.
Some employers require that employees maintain their certification for the duration of their
employment. Organizations offering certification for these laboratorians include the:

ƒ

Board of Registry of the American Society for Clinical Pathology

ƒ

National Credentialing Agency for Laboratory Personnel

ƒ

Board of Registry of the American Association of Bioanalysts

ƒ

American Medical Technologists

ƒ

National Registry of Clinical Chemistry

ƒ

National Registry of Microbiologists1

ƒ

State certification programs, such as that in California105

The ASCP and the NCA provide the majorityq of certifications for technologists/scientists and
technicians.1 In 2006, ASCP certified 2,050 new technologists/scientists and 1,479 new
technicians. From April 2006 through March 2007, NCA certified 312 new
technologists/scientists and 121 new technicians.106, 107
Technologists/scientists can earn ASCP specialty certification in blood banking, chemistry,
cytology, hematology, histology, microbiology, hemapheresis, laboratory safety, virology, and
molecular pathology.13 In some instances, categorical certification in one of these areas can be
obtained without first becoming a certified (general) medical technologist/clinical laboratory
scientist. For technicians, ASCP certification is available in apheresis, phlebotomy and donor
phlebotomy, and histology.
In 2004, the ASCP initiated a certification maintenance program that extends three years and
initially applied only to newly certified individuals in the entry level categories.108 However, the
requirement was extended to all individuals in 2006. ASCP certification maintenance varies
q

As of December 2006, the ASCP had certified a total of 223,958 technologists/scientists and 70,674 technicians.106
NCA does not track the total number of professionals certified.107 In general, among first-time takers of
certification examinations, ASCP certifies approximately 2,000 technologists/scientists and 1,300 technicians each
year, while NCA certifies approximately 655 technologists/scientists and 200 technicians annually.1

May 2008

128

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

according to certification type but generally involves participation in formal or employer-based
continuing education courses relating to various aspects of the laboratory. The NCA began
requiring certification renewal in 1980 via continuing education or re-examination.109
Recertification by either method extends NCA certification by three years.

CONCLUSIONS
Clinical laboratories conducting non-waived testing typically are staffed with a medical team
comprising pathologists, doctoral-level laboratory scientists, technologists/scientists, and
technicians. While some of the tasks they perform on a daily basis may overlap, the contributions
and responsibilities of each differ in several ways.

ƒ

Pathologists (medical doctors) and doctoral-level laboratory scientists operate at the
highest levels within clinical laboratories, often serving as laboratory directors
responsible for staff oversight, testing processes, quality control procedures, quality of
patient care, and other functions. In 2005, there were an estimated 19,339 pathologists
in the U.S.; 80% work in community practice. Minorities are under-represented in the
discipline of pathology with 10% identified as Asian, 3% Hispanic, and 1% African
American. Slightly over half of pathology residents are female.

ƒ

Approximately 160,760 medical technologists/scientists and 144,710 technicians were
employed in the U.S. in 2006. While this workforce also is female dominated (74%), it is
more representative of the diverse ethnic make up of the population with 12% Asian,
11% African American, and 7% Hispanic. Geographically, 58% of
technologists/scientists work in an urban setting, 24% in suburban, and 18% in rural.

ƒ

Overall, the number of technologist/scientist and technician education programs has
declined by over 50% since 1970, with the most dramatic decline in
technologist/scientist programs—71% closed between 1970 and 2007. In contrast, the
number of phlebotomy programs increased six-fold from 1987 to 2003. Factors
contributing to the changes include decreases in hospital revenues resulting from
changes to the Medicare payment system as well as the expense of operating clinical
laboratory science programs.

ƒ

Current enrollment in specialized technologist/scientist and technician educational
programs is lowest in blood banking and histotechnology. However, recent recruiting
efforts programs appears to be effective, specifically those targeted at recruiting
minorities and male students, raising awareness of laboratory careers among students,
and dedicating program staff and budget specifically to recruitment.

ƒ

At present, 12 states and one territory require licensure of technologists/scientists and
technicians, and 12 other states are involved in some phase of licensure activity.
However, the specific requirements for licensure vary by state.

Gaps, Needs, and Challenges:
ƒ

May 2008

There is significant concern regarding the growing shortage of technologists/scientists
and technicians, which is expected to worsen over the next decade. The shortages are
attributed to the aging of the workforce, competing career opportunities, and difficulty
recruiting and retaining staff. Although vacancy rates at the staff level were highest in
129

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

2000 (11 to 22%), they remained steady from 2002 to 2005 at an annual rate of 4 to 7%.
Vacancies vary according to staff position, laboratory type and size, and geographic
location.

ƒ

May 2008

Technological advancement of laboratory testing, emerging pharmacogenomic and
proteomic testing, and greater laboratory automation could change the qualifications
required of next generation laboratory professionals. The laboratory sector needs to
clearly redefine staffing qualifications and workforce level requirements to meet these
forthcoming advancements.

130

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

REFERENCE LIST
1. Chapman SA, Franks PE, Lindler V, Ward-Cook K. The clinical laboratory workforce: the
changing picture of supply, demand, education and practice. Rockville, MD: Health
Resources and Services Administration, 2005.
2. Careers in the medical laboratory. Chicago, IL: American Society for Clinical Pathology,
2007. (Accessed February 28, 2007, at http://www.ascp.org/CareerLinks/default.aspx.)
3. The pathologist. Chicago, IL: American Society for Clinical Pathology, 2007. (Accessed
February 27, 2007, at http://www.ascp.org/careerLinks/pdf/thePathologist.pdf.)
4. Friedman BA. Challenges to pathology informatics in the era of the electronic medical
record. Advancing Practice, Instruction, and Innovation through Informatics (APIII 2006).
August 16, 2006. Pittsburgh, PA: APIII, 2006.
5. Murray R. Lutheran General Hospital (Park Ridge, IL). Personal communication.
November 29, 2007.
6. FREIDA online specialty training search. Chicago, IL: American Medical Association,
2007. (Accessed February 28, 2007, at http://www.ama-assn.org/vapp/freida/srch/.)
7. Holladay EB. American Society for Clinical Pathology. Personal communication.
March 19, 2008.
8. Murray R. Lutheran General Hospital (Park Ridge, IL). Personal communication.
April 28, 2007.
9. Laboratory requirements. Code of Federal Regulations, Title 42, Section 493.1405. Subpart M-personnel for nonwaived testing; laboratories performing moderate complexity testing;
laboratory director qualifications.
10. Who's who in the lab: a look at laboratory professionals. Washington, DC: Lab Tests
Online, 2005. (Accessed April 11, 2007, at
http://www.labtestsonline.org/lab/who-2.html.)
11. ASCP and NCA take another step toward merging certification bodies. Clinical Lab
Products 2006.
12. DiIulio R. The science of credentialing scientists. Clinical Lab Products 2006.
13. ASCP Board of Registry procedures for examination and registry. Chicago, IL: American
Society for Clinical Pathology, 2007. (Accessed February 2, 2007, at
http://www.ascp.org/Certification/pdf/booklet.pdf.)
14. NCA candidate handbook. Lenexa, KS: National Credentialing Agency for Laboratory
Personnel, 2007. (Accessed February 2, 2007, at http://www.ncainfo.org/pdfs/NCAHandbookFeb2007.pdf.)
15. Clinical laboratory technologists and technicians. Report No. O*NET 29-2011.00,292012.00. Washington, DC: Bureau of Labor Statistics, Department of Labor, 2006.
http://www.bls.gov/oco/pdf/ocos096.pdf.

May 2008

131

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

16. The medical technologist and medical laboratory technician. Chicago, IL: American
Society for Clinical Pathology, 2007. (Accessed December 20, 2006, at
http://www.ascp.org/CareerLinks/LabCareers/default.aspx#.)
17. Occupational employment and wages, May 2006, medical and clinical laboratory
technologists. Washington, DC: Bureau of Labor Statistics, Department of Labor, 2007.
(Accessed November 27, 2007, at http://www.bls.gov/oes/current/oes292011.htm.)
18. Occupational employment and wages, May 2006, medical and clinical laboratory
technicians. Washington, DC: Bureau of Labor Statistics, Department of Labor, 2007.
(Accessed November 27, 2007, at http://www.bls.gov/oes/current/oes292012.htm.)
19. The pathologists' assistant. Chicago, IL: American Society for Clinical Pathology, 2007.
(Accessed April 12, 2007, at
http://www.ascp.org/careerLinks/pdf/thePathologistsAssistant.pdf .)
20. What is a pathologists' assistant? Roseville, MN: American Association of Pathologists'
Assistants, 2008. (Accessed March 25, 2008, at
http://www.pathologistsassistants.org/Default.aspx?m=1012&s=142.)
21. American Society for Clinical Pathology. The histotechnologist and histotechnician.
Chicago, IL: ASCP, 2007. (Accessed December 20, 2006, at
http://www.ascp.org/CareerLinks/LabCareers/default.aspx#.)
22. Technician certification. Chicago, IL: American Society for Clinical Pathology, 2008.
(Accessed March 25, 2008, at
http://www.ascp.org/FunctionalNavigation/certification/GetCertified/TechnicianCertif
ication.aspx.)
23. The cytotechnologist. Chicago, IL: American Society for Clinical Pathology, 2007.
(Accessed December 20, 2006, at
http://www.ascp.org/CareerLinks/LabCareers/default.aspx#.)
24. Technologist certification. Chicago, IL: American Society for Clinical Pathology, 2008.
(Accessed March 25, 2008, at
http://www.ascp.org/FunctionalNavigation/certification/GetCertified/TechnologistCer
tification.aspx.)
25. Medical assistants. Report No. O*NET 31-9092.00. Washington, DC: Bureau of Labor
Statistics, Department of Labor, 2007. http://www.bls.gov/oco/ocos164.htm.
26. Is medical assisting for you? Chicago, IL: American Association of Medical Assistants,
2008. (Accessed March 18, 2008, at http://www.aamantl.org/medassisting/right4you.aspx.)
27. Physicians by race/ethnicity. Chicago, IL: American Medical Association, 2005.
28. Career opportunities in pathology. Bethesda, MD: Association of Pathology Chairs, 2007.
(Accessed February 28, 2007, at http://www.apcprods.org/Library/recruit.ppt.)
29. Physicians by census region, self-designated specialty, and activity. Chicago, IL: American
Medical Association, 2005.
30. Statistical profile of persons receiving doctor's degrees in the life sciences: selected years,
1979-80 through 2002-03. Washington, DC: National Center for Education Statistics,

May 2008

132

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

Department of Education, 2005.
http://nces.ed.gov/programs/digest/d05/tables/dt05_299.asp.
31. Statistical profile of persons receiving doctor's degrees in the physical sciences: selected
years, 1979-80 through 2002-03. Washington, DC: National Center for Education Statistics,
Department of Education, 2005.
http://nces.ed.gov/programs/digest/d05/tables/dt05_300.asp.
32. Employed civilians by occupation, sex, race, and Hispanic origin, 2005. Washington, DC:
Census Bureau, 2007. (Accessed December 17, 2006, at
http://www.census.gov/compendia/statab/tables/07s0602.xls.)
33. Grover A. Clinical laboratory workers. Clinical Laboratory Improvement Advisory
Committee meeting. September 12, 2002. Rockville, MD: National Center for Health
Workforce Information and Analysis, Bureau of Health Professions, Health Services and
Research Administration, 2002.
34. Steward CA, Thompson NN. ASCP 2005 wage and vacancy survey of medical
laboratories. Laboratory Medicine 2006;37(8):465-9.
35. The Lewin Group. Physician supply and demand: projections to 2020. Rockville, MD:
Bureau of Health Professions, Health Resources and Services Administration, 2006.
ftp://ftp.hrsa.gov/bhpr/workforce/PhysicianForecastingPaperfinal.pdf.
36. Rittershaus AC. American Society for Clinical Pathology 2006 Resident Council fellowship
and job market survey. Chicago, IL: American Society for Clinical Pathology, 2007.
http://www.ascp.org/careerLinks/pdf/2006JobMarketSurvey.pdf.
37. Jones P. American Association for Clinical Chemistry. Personal communication.
November 29, 2007.
38. Frequently asked questions: phlebotomy certification. Richmond, CA: Division of
Laboratory Science, Laboratory Field Services Branch, 2007. (Accessed May 2, 2007, at
http://www.dhs.ca.gov/ps/ls/lfsb/pdf/FREQUENTLYASKEDQUESTIONS071106.doc.)
39. Physician salaries and salary surveys. Rehoboth Beach, DE: Allied Physicians, 2006.
(Accessed May 2, 2007, at http://allied-physicians.com/salary-surveys/physicians/#top.)
40. Long A. Lady luck smiles on the salary jackpot. MLO Med Lab Obs 2007;39(3):34-6.
41. Haigh A. Survival of the fittest. MLO Med Lab Obs 2005;37(3):28, 30.
42. Dunham D. Every calling is great when greatly pursued. MLO Med Lab Obs
2004;36(3):24-6.
43. Heylin M. Employment and salary survey. Chemistry and Engineering News 2006:42-51.
44. Gad SC. Sixth triennial toxicology salary survey. Reston, VA: Society of Toxicology, 2004.
http://www.toxicology.org/AI/APT/sixthtrisurvey.asp.
45. Careers in medicine: pathology. Washington, DC: Association of American Medical
Colleges, 2007. (Accessed February 28, 2007, at
www.aamc.org/students/cim/pub_pathology.htm.)
46. Definition of allopathic medicine. Atlanta, GA: MedicineNet.com, 2007. (Accessed
March 2, 2007, at http://www.medterms.com/script/main/art.asp?articlekey=33612.)

May 2008

133

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

47. Glossary. Tucson, AZ: Tucson Osteopathic Medical Foundation, 2007. (Accessed May 2,
2007, at http://www.tomf.org/osteopathic_medicine/glossary.html.)
48. Glossary of osteopathic terminology. Chevy Chase, MD: American Association of Colleges
of Osteopathic Medicine, 2006. Accessed on March 5, 2007.
http://www.aacom.org/om/Glossary.pdf.
49. Osteopathic medicine. Chevy Chase, MD: American Association of Colleges of
Osteopathic Medicine, 2007. (Accessed March 2, 2007, at
http://www.aacom.org/om.html.)
50. Osteopathic medical college information book: 2007 entering class. 2nd edition. Chevy
Chase, MD: American Association of Colleges of Osteopathic Medicine, 2007.
http://www.aacom.org/om/Glossary.pdf.
51. Fact sheet 2006. Chicago, IL: American Osteopathic Association, 2006. (Accessed May 2,
2007, at http://www.osteopathic.org/pdf/ost_factsheet.pdf.)
52. Medical schools. Washington, DC: American Association of Medical Colleges, 2007.
(Accessed May 2, 2007, at http://www.aamc.org/medicalschools.htm.)
53. Total enrollment by sex and school, 2002-2006. Washington, DC: Association of American
Medical Colleges, 2006. (Accessed November 9, 2006, at
http://www.aamc.org/data/facts/2006/factsenrl.htm.)
54. Van Akkeren L. Association of American Colleges of Osteopathic Medicine. Personal
communication. March 5, 2007.
55. Singer AM. 2004 annual report on osteopathic medical education. Chevy Chase, MD:
American Association of Colleges of Osteopathic Medicine, 2005.
http://www.aacom.org/data/annualreport/AROME2004.pdf.
56. Physician workforce policy guidelines for the United States, 2000-2020. Report No. 16.
Rockville, MD: Council on Graduate Medical Education, 2005.
http://www.cogme.gov/16.pdf.
57. Applicants, first-time applicants, acceptees, and matriculants to U.S. medical schools by
sex, 1996-2007. Washington, DC: Association of American Medical Colleges, 2006.
(Accessed November 28, 2007, at
http://www.aamc.org/data/facts/2007/2007summary2.htm.)
58. Annual statistical report on osteopathic medical education. Chevy Chase, MD: American
Association of Colleges of Osteopathic Medicine, 2007.
http://www.aacom.org/data/annualreport/ASROME2006.pdf.
59. U.S. medical school enrollment projected to increase by 17 percent. Washington, DC:
Association of American Medical Colleges, 2007. (Accessed March 5, 2007, at
http://www.aamc.org/newsroom/pressrel/2007/070212.htm.)
60. Salsberg E. Increasing medical school enrollment: rational and implications for medical
schools and GME. AMA Section on Medical Schools. November 11, 2006. Chicago, IL:
American Medical Association, 2007.
61. About the NRMP. Washington, DC: National Residency Matching Program, 2006.
(Accessed February 6, 2007, at http://www.nrmp.org/about_nrmp/index.html.)

May 2008

134

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

62. Obradovic J. American Osteopathic Association. Personal communication. March 7, 2007.
63. Introduction. Pomona, CA: OPTI-West Educational Consortium, 2007. (Accessed March 8,
2007, at http://www.westernu.edu/OPTI-West/intro.htm.)
64. Advance data tables for 2007 main residency match. Washington, DC: National Residency
Match Program, 2007. http://www.nrmp.org/advancedata2007.pdf.
65. Positions offered and filled by U.S. seniors and all applicants 2002-2006. Washington, DC:
National Residency Match Program, 2006. (Accessed March 6, 2007, at
http://www.nrmp.org/res_match/tables/table5_06.pdf.)
66. Etzel S. American Medical Association. Personal communication. May 3, 2007.
67. About the GRE. New York, NY: The Princeton Review, 2007. (Accessed April 16, 2007, at
http://www.princetonreview.com/grad/testprep/testprep.asp?TPRPAGE=10&TYPE=G
RE-ABOUT.)
68. So you want to attend grad school in the United States: a guide for international
applicants. New York, NY: The Princeton Review, 2007. (Accessed April 16, 2007, at
http://www.princetonreview.com/grad/apply/articles/process/gradUS.asp.)
69. Research doctorate programs. Washington, DC: Network for Education Information,
Department of Education, 2007. (Accessed April 16, 2007, at
http://www.ed.gov/about/offices/list/ous/international/usnei/us/edlite-researchdoctorate.html.)
70. Eligibility requirements and examination information. Washington, DC: American Board
of Medical Laboratory Immunology, 2007. (Accessed November 30, 2007, at
http://www.asm.org/Academy/index.asp?bid=3955.)
71. Eligibility requirements and examination information--eligibility requirements revised
June 2007. Washington, DC: American Board of Medical Microbiology, 2007. (Accessed
November 30, 2007, at http://www.asm.org/Academy/index.asp?bid=4121.)
72. Postdoctoral training programs in medical microbiology and immunology accredited by
the Committee on Postgraduate Education Programs. Washington, DC: American Society
for Microbiology, 2007. (Accessed November 28, 2007, at
http://www.asm.org/Academy/index.asp?bid=2108.)
73. CPEP-approved postgraduate training programs. Washington, DC: American Society for
Microbiology, 2007. (Accessed November 28, 2007, at
http://www.asm.org/Academy/index.asp?bid=2262.)
74. Training programs. Washington, DC: Commission on Accreditation in Clinical Chemistry,
2007. (Accessed November 28, 2007, at http://apps.aacc.org/comacc/training.stm.)
75. Degrees conferred by degree-granting institutions, by control of institution, level of
degree, and discipline division, 2004-05. Washington, DC: National Center for Education
Statistics, Department of Education, 2005.
http://nces.ed.gov/programs/digest/d06/tables/dt06_260.asp.
76. Advanced practice: doctorate in clinical laboratory science. Bethesda, MD: American
Society for Clinical Laboratory Science, 2005.
http://www.ascls.org/position/AdvPractice.asp.

May 2008

135

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

77. Gale DD. NAACLS approves standards for the clinical doctorate. NAACLS News
2006;94:1-2.
78. Bugbee AC. ASCP Board of Registry's 2006 annual survey of medical laboratory science
programs. Laboratory Medicine 2007;38(8):463-71.
79. Guide to accreditation for clinical laboratory scientist/medical technologist programs.
Chicago, IL: National Accrediting Agency for Clinical Laboratory Sciences, 2007.
http://www.naacls.org/PDFviewer.asp?mainUrl=/docs/guide_cls-mt.pdf.
80. What is CAAHEP? Clearwater, FL: Commission on Accreditation of Allied Health
Education Programs, 2007. (Accessed January 5, 2007, at http://www.caahep.org/.)
81. CAAHEP annual report: July 1, 2006-June 30, 2007. Clearwater, FL: Commission on
Accreditation of Allied Health Education Programs, 2006.
http://www.caahep.org/documents/Publications/AnnualReport.pdf.
82. Castillo JB. The decline of clinical laboratory science programs in colleges and universities.
J Allied Health 2000;29(1):30-5.
83. NAACLS table of program and graduate numbers. Chicago, IL: National Accrediting
Agency for the Clinical Laboratory Sciences, 2007.
84. LaBeau KM, Simon M, Granade S, Steindel SJ. The Pacific Northwest Laboratory Medicine
Sentinel Monitoring Network final report of the findings of questionnaire 13: laboratory
testing personnel shortages. Atlanta, GA: Centers for Disease Control and Prevention,
2000. http://wwwn.cdc.gov/mlp/pdf/LMSMN/PNW/report13.pdf.
85. Ward-Cook K, Chapman S, Lindler V. Executive summary: 2002 annual survey of
accredited/approved medical laboratory science programs. Chicago, IL: American
Society for Clinical Pathology, 2002.
http://www.ascp.org/Certification/ForProgramDirectors/research/documents/Svy02.pdf.
86. CLIA personnel requirements. Leawood, KS: American Academy of Family Physicians,
2007. (Accessed April 13, 2007, at
http://www.aafp.org/online/en/home/practicemgt/pt/clia/requirements.html.)
87. Laboratory requirements. Code of Federal Regulations, Title 42, Section 493.1443. Subpart M-personnel for nonwaived testing; laboratories performing high complexity testing;
laboratory director qualifications.
88. Laboratory requirements. Code of Federal Regulations, Title 42, Section 493.1357. Subpart M-personnel for nonwaived testing; laboratories performing PPM procedures; laboratory
director qualifications.
89. Laboratory requirements. Code of Federal Regulations, Title 42, Section 493.1363. Subpart M-personnel for nonwaived testing; laboratories performing PPM procedures; PPM testing
personnel qualifications.
90. Physician education, licensure, and certification. Chicago, IL: American Medical
Association, 2007. (Accessed March 6, 2007, at http://www.amaassn.org/aps/physcred.html#educ.)

May 2008

136

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

91. Physicians and surgeons. Report No. O*NET 29-1061.00, 29-1062.00, 29-1063.00, 291064.00, 29-1065.00, 29-1066.00, 29-1067.00, 29-1069.99. Washington, DC: Bureau of Labor
Statistics, Department of Labor, 2005. http://www.bls.gov/oco/pdf/ocos074.pdf.
92. Physician licensing overview. Chicago, IL: American Osteopathic Association, 2007.
(Accessed March 6, 2007, at
http://www.osteopathic.org/index.cfm?PageID=ado_license.)
93. Personnel licensure. Bethesda, MD: American Society for Clinical Laboratory Science,
2007. (Accessed January 16, 2006, at
http://www.ascls.org/jobs/grads/personnel_licensure.asp.)
94. Laboratory requirements. Code of Federal Regulations, Title 42, Section 493.1425. Subpart M-personnel for nonwaived testing; laboratories performing moderate complexity testing;
testing personnel qualifications.
95. Laboratory requirements. Code of Federal Regulations, Title 42, Section 493.1489. Subpart M-personnel for nonwaived testing; laboratories performing high complexity testing; testing
personnel qualifications.
96. What board certification means. Evanston, IL: American Board of Medical Specialists,
2007. (Accessed May 2, 2007, at
http://www.abms.org/About_Board_Certification/means.aspx.)
97. UnitedHealthcare's premium designation program adds new recognition component to
promote quality and professional development by the nation's largest medical specialty
board. Minneapolis, MN: UnitedHealth Group, 2006. (Accessed November 29, 2007, at
http://www.unitedhealthgroup.com/news/rel2006/0627ABIM.htm.)
98. Examination dates. Tampa, FL: The American Board of Pathology, 2007. (Accessed March
6, 2007, at http://www.abpath.org/ExamDates.htm.)
99. Requirements for primary and subspecialty certification. Tampa, FL: The American Board
of Pathology, 2006. (Accessed March 6, 2007, at
http://www.abpath.org/ReqForCert.htm.)
100. American Osteopathic Board of Pathology. Chicago, IL: American Osteopathic Board of
Pathology, 2007. (Accessed March 6, 2007, at http://www.aobpath.org/.)
101. Application for board eligible classification in pathology. Chicago, IL: American
Osteopathic Board of Pathology, 2007. (Accessed March 6, 2007, at
http://www.aobpath.org/AOBPaApplication%20for%20Board%20Eligible%20Classification.
pdf.)
102. Maintenance of certification. Tampa, FL: The American Board of Pathology, 2006.
(Accessed March 6, 2007, at http://www.abpath.org/MOCIndex.htm.)
103. Why should you be certified by the American Board of Clinical Chemistry? Washington,
DC: American Board of Clinical Chemistry, 2007. (Accessed November 29, 2007, at
http://apps.aacc.org/abcc/why.stm.)
104. Certification standards for bioanalyst clinical laboratory director (BCLD). St. Louis, MO:
American Board of Bioanalysts, 2007. (Accessed November 30, 2007, at
http://www.aab.org/bcld.htm.)

May 2008

137

Laboratory Medicine: A National Status Report

Chapter III – Laboratory Medicine Workforce

105. Laboratory personnel website. Richmond, CA: Division of Laboratory Science, Laboratory
Field Services Branch, 2007. (Accessed March 11, 2007, at
http://www.applications.dhs.ca.gov/lpw/.)
106. Examination statistics for January-December 2006. Chicago, IL: American Society for
Clinical Pathology, 2007. (Accessed May 3, 2007, at
http://www.ascp.org/Certification/ForProgramDirectors/statistics/examstat2006.pdf.)
107. Annual report/examination summary April 2006-April 2007. NCA Clipboard 2006;11(1):3.
108. Certification Maintenance Program (CMP). Chicago, IL: American Society for Clinical
Pathology, 2007. (Accessed January 5, 1006, at
http://www.ascp.org/certification/CMP/.)
109. Frequently asked questions: recertification questions. Lenexa, KS: National Credentialing
Agency for Laboratory Personnel, 2007. (Accessed January 16, 2007, at http://www.ncainfo.org/faqs_recertification.asp.)

May 2008

138

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

CHAPTER IV
QUALITY AND THE TOTAL TESTING PROCESS
Improving quality has been a core goal of the laboratory medicine sector for decades, beginning
with proficiency testing (PT) in the 1930s.a Quality-related initiatives have been an important part
of laboratory operations ever since.
A milestone in quality improvement occurred in 1986, when CDC hosted the first in a series of
Institutes on Critical Issues in Health Laboratory Practice devoted to improving laboratory quality.1
Representatives assembled from diverse sectors within the health laboratory community including
laboratorians, providers, public health practitioners, industry representatives, regulators, and
payers. Participants defined the roles and responsibilities to be assumed in the processes associated
with laboratory testing. At this critical meeting, participants were introduced to the concept of the
total testing process (TTP), a systems-based framework for examining all possible interactions and
activities that can affect the quality of laboratory tests.1 The aim of introducing the TTP was to
design and implement interventions, restrictions, and limits that could reduce or eliminate errors
that adversely affect testing and patient health outcomes.
Today, the TTP remains the conceptual framework for understanding the dynamics of laboratory
medicine as well as quality measures to improve care. This chapter provides an overview of the
TTP and examines the main types of error and other challenges to quality that occur in each phase.

DEFINITION OF THE TOTAL TESTING PROCESS
The initial definition of the TTP espoused at the 1986 CDC Institute still serves as the primary
point of reference for addressing quality in laboratory medicine. Since its inception, the
definition has been refined and expanded to encompass all components that complete the test
cycle, from the point of the clinical question to the point of clinical action, known as the “brainto-brain” model.2 In this regard, the TTP is defined by the activities in three distinct phases that
align with clinical workflow outside and inside the laboratory:
(1) Preanalytic: clinician test selection, test ordering, patient preparation, specimen
collection, patient and specimen identification, and specimen transport
(2) Analytic: specimen processing and preparation, testing of the specimen, results review
and verification, and quality control (QC) checks
(3) Postanalytic: TAT, critical value reporting, report formatting, general results reporting,
clinician interpretation and follow-up, laboratory interpretive consultation services,
specimen storage and, if applicable, daily laboratory shutdown3, 4
In most instances, preanalytic activities occur outside the laboratory. The specimen is collected by
the clinician at the site of care (e.g., hospital, physician’s office, patient’s home). However, there
are instances in which the specimen is collected at the laboratory. Some experts in laboratory
medicine include specimen preparation in preanalytic activities. In the “brain-to-brain” model,
a

Further discussion on the history and development of quality-related initiatives in the laboratory sector is provided in the
Quality Systems and Performance Measurement Chapter and Federal Regulatory Oversight Chapter of this report.

May 2008

139

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

these activities are identified as analytic-related and, therefore, will be included in the discussion
of the analytic phase for the purposes of this report. Similarly, postanalytic activities that involve
laboratory communication with clinicians take place in both practice areas. Figure 4.1 presents a
streamlined diagram of the key components of each phase of the TTP.
Figure 4.1: Phases of the Total Testing Process

Source: Adapted from Boone J. Presentation at the Institute on critical issues in health laboratory practice: managing for better
health, September 23-26, 2007. Atlanta, GA: Centers for Disease Control and Prevention.

May 2008

140

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

QUALITY AND ERRORS
Most quality initiatives in laboratory medicine have focused, historically, on the analytic phase of
testing; however, root cause analyses and other medical error studies confirm that more errors
occur in the preanalytic and postanalytic phases of testing. The distribution of these errors by
phase varies among settings and institutions. An extensive review of reported errors in
laboratory medicine published from 1992 to 2001 found great heterogeneity in study designs and
reporting of errors. The distribution of errors was 32-75% in the preanalytic phase, 13-32% in the
analytic phase, and 9-31% in the postanalytic (administration) phase.5 This review included
studies of error rates associated with clinical chemistry, the whole laboratory, primary care, stat
laboratory, and molecular genetic testing. One of these studies (whole laboratory) estimated that
8% of errors had the potential for serious harm.6 Error distribution in transfusion medicine is
reported to be somewhat higher in the postanalytic phase. In a large study of errors detected in
blood banks, the distribution was 41% in the preanalytical phase, 4% in the analytic phase, and
55% in the postanalytical phase.7
Poor communicationb between laboratory professionals and clinicians is generally cited as the
chief issue affecting quality of laboratory services during the preanalytic and postanalytic
phases.9-12 Throughout the health care system, communication failures are a leading cause of
shortfalls in quality, particularly of preventable errors that harm patients.13-15 Widely overlooked
in training of health care providers is that few clinicians or laboratory professionals receive formal
training in effective communications.
Preanalytic communication involves discussion between the clinician and the laboratory to select
an appropriate test or set of tests and the communication of appropriate patient information on
requisition slips. These communications may involve an extensive set of medical professionals
including physicians, nurses, pathologists, medical technologists, laboratory technicians, and
clerical staff. They may communicate about test orders, patient identification information, and
specimen adequacy. Postanalytic communication entails laboratory professionals’
communications with the clinician about critical values and interpretation of laboratory findings.
Breakdowns in pre- and postanalytic communication lead to errors, patient safety events, and
inefficient and ineffective use of health care resources.16, 17
The next sections review each component of the TTP and provide examples of the types of quality
issues and errors that are documented in the literature. In addition, the general discussion of the
TTP is followed by a more specific discussion of quality issues and errors associated with POCT.
Much of the information is derived from research conducted by CAP through its Q-Probes® and
Q-Tracks® studies.

b

Communication is defined as the effective transmission of knowledge or information from one individual to
another. It requires clear and concise formulation of data, transformation of the data into useful information, and
an agreed method of communication understood by the sender and receiver.8

May 2008

141

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

PREANALYTIC PHASE
Clinician Knowledge of Diagnostic Tests
Many laboratory test selection errors arise because clinicians lack adequate knowledge for
decision-making when ordering complex testing regimens. Physician knowledge of laboratory
tests and ability to order appropriately is complicated by two factors: (1) rapid proliferation of
new tests and tests for additional analytes; (2) lack of formal education in laboratory testing.
Recent advances in biochemical, molecular, and genetic sciences have led to a plethora of new
laboratory diagnostics for clinical use. Currently, there are more than 4,000 different laboratory
tests on the market, including new genetic tests that can be used for an estimated 1,430 diseases.
Although orders for routine genetic tests and newer assays of increased breadth and complexity
are increasing, medical students are exposed to only 29 hours on average of didactic coursework
in medical genetics.18-20
Primary care and specialist physicians need to be familiar with broadening sets of existing and
emerging screening and diagnostic tests. According to a 1999 survey, 25% of primary care
physicians indicated that their medical knowledge base is insufficient for the scope of the care
they are expected to provide.19 Some 38% of specialists believe that primary care physicians
cannot maintain adequate expertise to deal with the overload of new clinical information. Even
when physicians lack knowledge for diagnostic or management decisions that poses threats to
quality or medical malpractice, they only obtain definitive answers to address their uncertainties
about 30-50% of the time.21 Day-to-day clinician demands leave little time to acquire this intricate
knowledge.19, 20 Continued medical and scientific advancement will compound challenges
associated with ordering the optimal sequence of tests, correctly interpreting results, and
incorporating genetic information into clinical practice.9

Appropriateness of Clinician Test Selection
Definitions of appropriate care vary, e.g., as follows:

ƒ

According to IOM, the best care is synonymous with appropriate care and results from
the conscientious, explicit, and judicious use of current best evidence and knowledge
of patient values by well-trained, experienced clinicians.14

ƒ

RAND has defined appropriate care as that in which the expected benefits exceed the
expected risks by a sufficient margin such that the service is worth doing.

ƒ

CAP defines appropriateness as the extent to which a particular procedure, treatment, test
or service is effective, clearly indicated, non-excessive, adequate in quantity and provided
in the inpatient, outpatient, home or other setting best suited to the patient’s needs.22

Principles of appropriateness in laboratory medicine are embodied in selecting the right test at the
right time for the right patient. Test appropriateness is inherent to an understanding of the
specific clinical condition and the value of a particular test to the respective patient. The ability to
make these determinations varies among clinicians. Standard measures of appropriateness do not
prevail currently, though their development is viewed as important. Instead, clinical guideline
performance indicators of care quality and measures of test use (including underuse and overuse)
have been the basis for drawing conclusions about appropriateness. For example, the sentinel

May 2008

142

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

2003 study by McGlynn et al. on the quality of U.S. health care evaluated clinician performance on
439 indicators of quality, 131 of which involved laboratory testing or radiography.23 Only 61.7%
of patients received the recommended laboratory or radiology tests for preventive, acute, and
chronic care associated with priority health conditions. The study findings called attention to
serious shortfalls in use of key tests that support care quality.
Studies of appropriateness can be undertaken for different applications of laboratory testing,
including screening for disease in asymptomatic individuals, establishing a diagnosis in persons
presenting with signs and symptoms of disease, and monitoring the course of disease or its
treatment. Screening may include panels of laboratory tests performed as part of periodic health
evaluations, or tests performed at the time of hospital admission, pre-operative evaluation, or
prenatal evaluation. Examples of screening tests include fecal occult blood testing for colorectal
cancer, cholesterol testing to detect risk of coronary artery disease, and Pap smears for cervical
cancer.c,27 Underuse is a common problem for several tests that reduce mortality by early detection
of disease or prompting interventions to control risk. The 2006 National Healthcare Quality Report
from AHRQ reported that 51.7% of adults over age 50 had colorectal cancer screening in the
previous two years (i.e., sigmoidoscopy, colonoscopy, proctoscopy, or fecal occult blood testing),
indicating that half of adults in this population are not receiving needed cancer screening.28
Other studies have demonstrated overuse of certain screening tests. For example, while Pap smears
are recommended for most women, they are not routinely recommended for women older than 65
or in women following hysterectomy (unless they have previous cervical neoplasia). In 1996, the
USPSTF recommended that routine Pap smears are unnecessary in women who have undergone a
hysterectomy for benign disease and no longer have a cervix. Yet, based on findings from largescale U.S. survey data for 1992 and 2002, among women who had ever had a hysterectomy, 68.5% in
1992 and 69.1% in 2002 had a Pap smear within the previous three years of the survey. After
adjusting for Pap smears performed prior to a hysterectomy, and hysterectomies that spared the
cervix or were performed for cervical neoplasia, the investigators estimated that 10 million women,
or half of all women who had undergone hysterectomy, were being screened unnecessarily.29 Still,
some clinicians and patients often prefer a cautious approach, especially if neoplasia was present.
Other bodies of research show that some screening tests, such as routine admission or preoperative
panels of tests, are of limited or no utility.30, 31
Monitoring involves testing to track the course of disease, make necessary adjustments in therapy,
or detecting complications of care. Examples include testing for HbA1c in diabetic patients and
lipid levels in patients with diabetes or known coronary artery disease. According to the 2006
National Healthcare Quality Report, performance rates of monitoring tests for diabetic patients
were relatively good. Of adult diabetic patients, about 90% had HbA1c measured in the past year,
but only 45.5% had their HbA1c level under optimal control and only 48.1% had their total
cholesterol under control.28 While these figures represent an improvement over previous years,
improved use of laboratory testing likely would facilitate better control. Of course, measurement
alone does not ensure desired outcomes. In a study of more than 1,700 diabetic patients at 30 U.S.
academic medical centers during 2000-2002, high annual rates of testing for HbA1c, blood

c

Use of other screening tests, such as glucose testing for diabetes, prostate specific antigen testing for prostate cancer,
and thyroid stimulating hormone, testing for thyroid disease, is supported by some groups; however, a strong
evidence-base for their preventive value is required by the USPSTF before recommendation.24-26

May 2008

143

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

pressure, and cholesterol did not translate to effective metabolic control. Only 40.4% of patients
received needed therapeutic adjustments for high HbA1c values.32
Estimates of diagnostic testing appropriateness are derived mostly from estimates of test use.
However, drawing broad conclusions about rates of inappropriate use across tests, settings, and
timeframes can be flawed. In a review of 44 eligible reports in the laboratory utilization literature
from 1966 to 1997, investigators found that reported rates of inappropriate use ranged from 4.5%
to 95%, suggesting significant inconsistencies in the validity of implicit or explicit criteria for
appropriateness and reliability of their application.16 Even for laboratory use among physicians
treating patients with the same diagnosis, there is considerable variability.33 Further research to
develop and validate criteria for appropriateness evaluations is needed.
More recent studies using clearly defined algorithms found that, when guidelines were applied,
20-25% of frequently ordered tests, such as autoantibody tests, infectious disease serologic tests,
and hepatitis serology tests were inappropriately requested.34-36 Redundant test requests also are
common and contributed to inappropriate use. In a multicenter CAP study, 1.5% of thyroid
stimulating hormone (TSH) requests appeared to be redundant. When applied to high volume
testing, even this apparently low rate can result in substantial unnecessary costs. In 2005, an
estimated 12.3 million TSH assays were performed at a cost of $288 million.37 Applying the CAP
data, the annual cost of a 1.5% rate of redundant testing is $4.3 million. A randomized trial of a
computer-based intervention to reduce redundant test use found that 1.2% of tests were
redundant, but use of computer alerts led to test cancellation of 69% of these redundant tests.
Even so, the net impact of the computer alerts was small because less than half of the redundant
tests were ordered via computer, only half of the computer orders were screened for redundancy,
and almost one-third of the reminders were overridden.38
Physicians often order certain laboratory tests habitually or because they are grouped together for
convenience. Multiple factors lead to inappropriate use of tests, including test panels that contain
unnecessary tests, delays in performing tests, and difficulties caused by ordering forms or
menus.33 Efforts to decrease utilization and improve appropriateness have focused on changing
physician behavior by altering requisition forms, changing policy, instituting computer rules and
reminders, and changing reimbursement. Those that target multiple components of behavior
modification appear to be most effective.39 For example,

ƒ

In a randomized trial conducted in hospitals in France, redesign of laboratory ordering
requisitions resulted in higher rates of compliance (83%) to ordering guidelines for
thyroid function tests compared to conventional ordering (63%), reducing
inappropriate test requests.40

ƒ

In a time-series study of interventions to improve rates of appropriate testing in nonhospital clinical laboratories in Ontario from 1991 to 1997, the combination of physician
guidelines, laboratory requisition form modification, and changes in reimbursement
policy for commonly used tests resulted in large shifts in the ordering of certain tests.
Specifically, orders decreased for total thyroxine tests by 96%, iron-related tests by
80%, urea-related tests by 58%, erythrocyte sedimentation rate tests by 57%, and TSH
tests by 12%, along with increases in certain tests encouraged for substitution.41

ƒ

A computer alert system with basic metabolic panel rules to be applied after hospital
admission in a large U.S. academic medical center decreased panel orders by almost

May 2008

144

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

60% without changing patient length of stay, mortality, readmission rates, or transfer
rates to ICUs.42
Because laboratory test results help clinicians determine diagnoses, therapies, and need for
follow-up tests, inappropriate test use can compromise case management and increase cost per
patient and rates of adverse health outcomes.16

Test Ordering
When a clinician decides to order a laboratory test, a requisition slip (order form) is completed in
writing or electronically and submitted with the specimen to the laboratory. Information on the
order forms can directly affect processing and analysis of the specimens.
Inaccurate or incomplete requisitions are another source of error and can affect the quality of
laboratory testing.43 A CAP Q-Probes study of 577 institutions reported in 1995 that examined how
accurately physicians’ test orders for inpatients were transmitted to the laboratory found that 97.1%
of documented physician orders were completed by the laboratory. However, 1.9% (median) of test
orders were not completed and, of the 17,085 patient records examined, 10% were not completed at
all. Reasons given as the most likely cause for not completing ordered tests included:

ƒ

Failure to enter orders correctly into hospital computer (41.8%)

ƒ

Test requisition improperly filled out (12.8%)

ƒ

Physician handwriting was unclear (4.1%)

ƒ

Failure to enter orders correctly in the laboratory computer (1.4%)

ƒ

Other (23.0%) and not applicable (16.9%)

The study also found that the specific laboratory test was not listed on the requisition or in the
patient’s medical record for 2.5% of the 224,431 laboratory tests performed; this occurred most
often in hospitals where verbal orders are more frequent.44
Similar findings were demonstrated in anatomic pathology. A 1996 CAP study of surgical
pathology specimens reported that missing or incorrect information on order forms accounted for
77% of all deficiencies. Specifically, the most common of these deficiencies was no clinical history
or diagnosis (40%). Smaller hospitals and laboratories had more cases with inadequate
documentation than larger hospitals. The additional clinical information, when obtained,
confirmed the diagnostic impression in 59.4% or was not relevant to pathologic diagnosis in 25.1%
of cases. However, the diagnosis was changed substantially in 4.2% of cases and required a report
revision in 1.9% of cases.45
Computerized physician order entry (CPOE) systems have been promoted as a means of
improving the quality of care, reducing errors, and increasing efficiency.15, 46-48 Because CPOE
systems are complex and costly to implement, efforts thus far have focused primarily on the use
of CPOE for medication ordering in hospital settings.49 However, a recently published literature
review identified 19 studiesd of the impact of CPOE on pathology services from 1990 to 2004.50
d

Fifteen studies compared CPOE with and without specific decision support mechanisms and four studies compared
setting with and without CPOE systems.

May 2008

145

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

Use of CPOE (as compared to conventional ordering) resulted in statistically significant decreases
in the volume of test ordering for blood count, chemistry, serum, and stat tests; when CPOE was
linked to additional decision support features, volume decreases ranged from 9.5% (per patient
per day) to 45.6% (per hours per patient day). Costs associated with laboratory ordering also
decreased by up to 28% for certain tests.
Direct access testing, in which individuals can directly request that certain tests be performed on
their own blood or urine specimens, is permitted in 26 of 50 states and the District of Columbia.51
An additional 12 states permit limited DAT. About 10-15% of laboratories offer direct access
testing.52 Although these tests are increasingly marketed and used, few data exist on their
utilization, appropriateness, accuracy, or impact on consumer decisions or health outcomes.53

Patient Preparation
During the next steps of the preanalytic phase, several factors can affect the quality of
laboratory results. Whether at the physician’s office, the laboratory, or the patient’s bedside, the
most important factors include patient preparation and identification and specimen collection
and labeling.
Inadequate patient preparation is a common source of misleading laboratory test results.54
Preparation factors that may affect laboratory test results are food intake, time of day, exercise,
stress, posture, time in menstrual cycle, medications, smoking, and illness unrelated to the condition
for which the test was ordered.55 For example, accurate glucose determination requires a fasting
specimen and tests for drug levels need to be performed when drugs are at steady state.
Calculating medication dosages based on inaccurate drug levels can have adverse consequences. In
a CAP Q-Probes study reported in 1993 of 666 institutions and more than 18,000 toxic levels, 22-31%
of digoxin levels in the toxic range were collected before the steady state of the drug was reached.
The investigators found that small institutions, outpatients, stat specimens, and laboratory policies
not requiring the time of the last dose before measurement were associated with higher percentages
of specimens drawn before the recommended time had elapsed.56 Laboratories do not have direct
control over these factors, especially when non-laboratory personnel collect samples; however, the
laboratory can develop and disseminate information about appropriate patient preparation.

Patient Identification
Patient identification problems are one of the most common causes of erroneous laboratory
results.57 Quality assurance (QA) and professional organizations (e.g., The Joint Commission)
have cited accurate patient identification as a key indicator in patient safety and quality
improvement initiatives. Progress with the use of patient wristbands with appropriate
identifying information in the hospital setting have been made, though errors can still occur. For
example, a study of electronic barcode systems demonstrated their success in reducing and
eliminating identification errors for all patients requiring blood transfusion from 1999 to 2002. A
portable, hand-held, scan-and-print device was used to verify and document patients’ identify at
two critical points of transfusion: blood sampling for compatibility and blood administration. In
the first three years of hospital-wide use of the device, no incidents occurred of blood transfusion
to wrong patients or wrong labeling of blood samples with 41,000 blood sampling procedures and
administration of 27,000 units of blood.58

May 2008

146

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

A two-year CAP study included wristband errors as a quality indicator.59 Continuous monitoring
led to significant decreases in wristband errors of all types from 7.4% (mean) at the start of the
study to 3.1% (mean) at its conclusion, although at the 10th percentile, there were wristband
errors in 11.4% or more of patients. As a percentage of all errors, the most common errors were:

ƒ

Missing wristbands (71.6%)

ƒ

Missing patient identification information (9%)

ƒ

Illegible wristband (7.7%)

ƒ

Erroneous identification information (6.8%)

ƒ

Conflicting information (3.7%)

ƒ

Wrong wristband (1%)

Hospital practices associated with fewer patient identification errors include written protocols
governing patient identification, placing new wristbands immediately when needed, and having
dedicated phlebotomists collect blood specimens. Laboratories with policies that required
phlebotomists to refuse to draw blood from patients with wristband errors reported the fewest
specimen identification errors.
Barcode systems have been developed for use at the point of phlebotomy, but little data have
been published on their use. In one study, using a beta version of a commercial system reduced
error by 77%.60 As with computerized order systems, however, use of barcodes may increase
certain error types. An evaluation of the Department of Veterans Affairs barcode medication
administration system identified five common error patterns, including degraded coordination
between physicians and nurses and decreased ability to adapt to changes from routine.61

Specimen Labeling/Identification
Patient identification problems can easily translate to specimen labeling problems.
Typically, labels are generated and applied to containers prior to specimen collection. Additional
labeling and numbering of the specimens may occur as specimens are processed for analysis.
Different institutions use different rules for counting specimens. Some assign a single accession
number to each specimen, while others assign the same number to all specimens from a single
phlebotomy or outpatient encounter, or assign separate numbers for aliquots destined for
different laboratory divisions.62 In surgical pathology, a single case number is usually assigned to
multiple specimens; then, each specimen is assigned a separate specimen identifier in addition to
the case number.
Specimen identification errors can result in serious patient injury such as wrong-patient cancer
resections and fatal transfusion reactions.63, 64 While error rates for individual institutions may be
relatively low, those error rates at the national level may yield a significant number of events. For
instance, one CAP study reported in 2002 detected specimen-related errors at 0.3% of total errors,
of which 5.8% were associated with inadequate labeling.65 In a 2006 study of these errors in
laboratories associated with 120 institutions (teaching, private, and local hospitals, independent
laboratories, federal government facilities), the authors reported adverse events resulting from 1
of every 18 specimen identification errors (5.6%), which, if extrapolated to the nation’s 6,000

May 2008

147

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

hospital laboratories, would result in 160,000 events.66 Such extrapolations are difficult to validate
because of the lack of standardized measurementse for reporting data in CAP studies and because
the laboratories in the studies are not a representative national sample.
Labeling errors have been studied extensively in transfusion medicine, where correct specimen
identification is critical to avoiding fatal transfusion reactions. In a study of transfusion errors in
New York State, undetected phlebotomyf errors were the cause of 13% of ABO-incompatibleg
transfusions, while failure to properly identify the patient at the time of transfusion caused
another 38% of incompatible transfusions.67 An international study of 82 facilities in 10 countries
determined that mislabeled specimens (defined as missing some or all identifying information
needed for acceptance) occurred in 1 of 165 specimens, while misidentified specimens (in which
the blood appeared to be from a patient other than who was identified on the label) occurred in an
estimated 1 of 1,986 specimens.68
Mislabeling/misidentification of specimens is a common error in anatomic pathology. Errors
usually occur when patient or specimen information on containers is missing or inaccurate. For
example, specimen-related errors can include misidentification of the origin of the tissue
specimen (e.g., stomach vs. colon), anatomic location (e.g., ascending vs. sigmoid colon), and
laterality of the tissue (e.g., right vs. left breast).69 A 1996 study detected identification and
accessioning errors in 6% of more than 1 million surgical pathology specimens. Specimen
identification errors accounted for 9.6% of these deficiencies, 77% of which were due to missing or
incorrect information. Among the specific deficiencies were:

ƒ

Illegible patient name or universal patient identifier (UPI) on either container or requisition

ƒ

No label on container

ƒ

No patient identification on container

ƒ

Patient name/UPI on container does not match that on requisition

ƒ

Patient name/UPI on container or requisition does not match master list

ƒ

Wrong patient name/UPI on both container and requisition

ƒ

No tissue source indicated on container or requisition

ƒ

No date of procedure

ƒ

No name of submitting physician

ƒ

Incorrect information other than patient name/UPI on container or requisition
(e.g., sex, age)

Laboratories performing 20,000 or more accessions per year had the highest rate of deficiencies at
6.0% compared to those performing fewer than 20,000, which ranged from 3.2 to 4.5%. Also,
institutions that labeled containers with the patient’s name and UPI had fewer deficiencies than
those that uses either one alone.45
CAP data collection typically is not standardized with clear definition of a numerator and denominator. Thus, data
from each institution may measure indicators differently and may use different detection and reporting criteria.
f Phlebotomy refers to venous blood drawing.
g ABO-compatibility refers to the laboratory testing to determine blood type A, B, or O compatibility between the
donor and recipient.
e

May 2008

148

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

Use of relatively new information and communication technology has led to substantial
improvements in patient and specimen identification.70 Computerized specimen-handling systems
include portable data loggers for laboratory personnel, and automatically upload patient and
specimen information into the laboratory information system (LIS). Barcode identification systems
are being used successfully in more institutions to create standardized labels for laboratory
specimens.71, 72 Errors still can occur when barcoded labels are transferred to the specimen
container. The risk of this error is higher in busy emergency rooms or ICUs where multiple labels
for different patients may be printed in advance of the specimen collection. Other approaches
under evaluation include radiofrequency identification chips and patient imbedded chips.73

Specimen Collection
Clinicians (e.g., nurses) or laboratorians (e.g., phlebotomists) may collect patient specimens.
Blood is the most common specimen submitted to laboratories. In CAP Q-Probe studies of
phlebotomy,h reported rates of success (i.e., specimens judged by the laboratory to be suitable for
analysis) have been 99.6% among ambulatory patients65 and 93.2% among inpatients.75 Lower
rates have been reported for inpatients because phlebotomists had more difficulty in obtaining
specimens (1.6%) or patients were not available (2.3%). Other common types of clinical pathology
specimens collected for analysis include urine, cerebrospinal fluid, serous fluids, feces, gastric
fluid, and synovial fluid. Anatomic pathology specimens are those obtained by aspiration,
washing, smear or scraping for cytologic examination or tissue taken during biopsy or surgery.
As a group, specimen collection problems are some of the more common causes of preanalytic
variation and may be associated with failure to collect the correct type of specimen, correct
volume of specimen, or collection of an unusablei or misleading specimen. The specimen
adequacy often cannot be determined until the analytic phase, when the specimen is assessed for
acceptance or rejection. There are limited data on error frequency associated with collection of the
wrong type of specimen. A 1997 study of the stat testing section of a university-based laboratory
in Italy reported that 2.1% of all TTP errors were attributed to incorrect specimen collection.76
Inadequate volume for testing is a common cause of specimen rejection. This problem affects not
only blood specimens, but other tests such as Pap smears. In a multicenter study involving 768
laboratories, 0.5% (median) of Pap smears had inadequate specimen volume and another 5.8%
(median) had adequate volume but limited the ability to evaluate the specimen for cervical cancer
due to other specimen-related factors.77 Two studies evaluated specimen rejection for complete
blood count (CBC) and chemistry specimens. Of 703 hospitals, 0.45% of CBC specimens were
rejected; 10.1% of rejections were due to insufficient specimen quantity.78 The subsequent 1997
study of chemistry specimens reported that 0.35% were rejected prior to testing; of these specimens,
11.4% were rejected due to inadequate volume.79 In the stat testing study mentioned above,
specimen volume and adequacy for analysis were affected when phlebotomy was performed from
a patient’s infusion; this inappropriately diluted the specimen volume in 20.6% of cases.76

Most medical and laboratory professional are trained to collect blood specimens but a specialist in this area is the
phlebotomist.74 Blood specimens can be collected from various sources. Venipuncture and phlebotomy refer to
collection of blood from veins, while skin puncture refers to collection from capillaries. Arterial blood also is
collected for blood gas measurements and pH.
i An unusable specimen is generally due to trauma during its transport that results in hemolysis or clotting.
h

May 2008

149

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

To ensure that specimen volume is sufficient for testing, laboratories often recommend specimen
volumes that exceed those needed for testing. Several studies noted collection of up to 45 times
the amount of blood actually needed for testing, raising concerns about production of “iatrogenic
anemia” in high risk hospitalized patients (e.g., ICU patients, neonates, elderly) who undergo
repeated blood collection.80, 81 The decisive factor contributing to blood loss was rigid blood
drawing schemes in which repetitive drawings were performed regardless of the patient’s clinical
condition.82 Reporting of individual cumulative blood loss on the daily laboratory report may
influence the requesting clinician’s behavior. Earlier studies examined the use of a pediatric
collection container for adult patients, which led to a 50% reduction in the mean daily blood loss;
however, this change had no effect on the ordering clinician’s requests for laboratory tests that
were not medically necessary.83, 82, 84
Unusable (e.g., due to hemolysis, clotted blood specimens) or mislabeled specimens are the most
frequent reasons for specimen rejection. Proper specimen collection and labeling is essential for
hematology and coagulation testing, where attention to specimen quality is necessary for an
optimal laboratory result.17 Several studies reported that hemolysisj of chemistry specimens
caused 54-60% of specimen rejections and occurred five times more frequently than the secondmost cited reason of insufficient volume; clotted specimens caused 65% of CBC specimen
rejections.5, 78, 79 Among rejected chemistry and CBC tests, a higher percentage were collected in
pediatric microcollection tubes.
Contamination of specimens collected for bacterial culture is another common source of error,
estimated at 2.5% of positive blood cultures and 18% of urine cultures in reports published in
2007.85, 86 Because it can be difficult to distinguish a false positive from a truly positive culture,
patients with contaminated cultures are often given unneeded care, increasing hospital costs by
thousands of dollars per patient.87 Similar contamination rates have been reported in inpatient
and outpatient specimens.85 Continuous monitoring, use of dedicated phlebotomists, feedback to
those drawing specimens, and use of faster acting decontamination techniques all reduce the
frequency of contaminated cultures.88-91 There are many other sources of error in microbiology
testing, including wrong type of specimen collected for type of infection, insufficient specimen
collected, contamination of specimen collection containers and solutions, and poor timing of
specimen collection and transport.92-94 When microbiology specimens are collected during a
surgical procedure, contamination of surgical blades also may cause errors.95
Health care and laboratory settings also can affect specimen collection. In a recent study
comparing rates of and reasons for specimen rejection across different settings as well as
associations with patient demographic factors, of the 0.74% of specimens that were rejected, 47%
were from the inpatient setting, 27% from the emergency department, and 25% from outpatient
setting. After adjusting for the total number of specimens per site, the emergency department
rejection rates were twice as high as rates for inpatient services and five times as high as rates for
the outpatient setting. Also, the rejection rate of specimens from African Americans was twice
that of the rates for Caucasians and 30% higher than the rate for other races in the hospital setting
but indicated no difference in the outpatient setting.96 The investigators hypothesized that the
severity and seriousness of the diseases and comorbidities of patients admitted to emergency
department or inpatient services are contributing to the higher proportion of specimens rejected.
j

Breakage of red blood cells membranes causing the release of hemoglobin and other internal components into the
surrounding fluid.

May 2008

150

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

In general, African American patients are more likely to choose the emergency department as the
mode of entry to the system, independent of health insurance status, therefore increasing the
likelihood of errors.97

Specimen Delivery
Specimen quality can be compromised during delivery by excessive delay, adverse temperature,
or manual or mechanical trauma to the specimens.55 Different means have been employed to
transport specimens to and within the laboratory, such as human couriers, pneumatic-tube
systems, and other technologies, that have advantages and disadvantages. For example, using
human couriers is a “batch process” dependent on pickup at specific times.98 While reliable, it
entails significant training and management costs. Pneumatic-tube systems (i.e., tube transport
systems that move contents via vacuum and positive pressure) provide cost-effective, rapid
transport for specimen delivery in hospitals.99 Evidence on potential for specimen damage from
acceleration and deceleration forces is mixed. Although some evidence suggests that mechanical
factors—tube length and speed, and number of times transported—may result in hemolysis of
blood specimens,100 other evidence shows no clinically significant effect of these systems on
hematology and coagulation results.101 Other technologies such as track vehicles, mobile robots,
and conveyor belts are now widely used to transport specimens from one point within the
laboratory to another. Although safe to specimens, transport via these technologies tends to be
slower. Implementing all of these technologies entail capital expenditures for installation and
costs for transition from old to new systems, training, and maintenance.
Each specimen type has standards for timely delivery and conditions for transport in order to
maintain its integrity. Specimens for stat orders such as those collected in an emergency room
need to be delivered to the laboratory immediately. Most microbiologic organisms die quickly
after removal from the body and should be transported quickly. Specimens for some tests (e.g.,
cryoglobulins, cryofibrinogen) need to be kept at body temperature, while others need to be
protected from light (e.g., bilirubin, vitamin A).74, 102 Transporting and processing delays can
render a specimen invalid for analysis. For example, a 2000 study of one-hour delays in the
processing of blood samples found significant effects on the concentration of biomarkers.
Decreases in the concentrations occurred for red and white blood cells, high-density lipoprotein
cholesterol, glucose, and creatine. Increases in concentration were observed for total cholesterol,
total testosterone, free testosterone, alkaline phosphatase, total bilirubin, and thiobarbituric acidreactive substances.103

CLINICAL PATHOLOGY TRANSITIONAL PREANALYTIC
Specimen Processing and Preparation
Once received, the laboratory processes the specimen for analysis. Quality in processing requires
that the laboratory verify specimen labeling (e.g., patient identification, time of collection, initials
of sample collector) and information provided on the accompanying requisition (e.g., patient
identification, tests ordered, relevant clinical information).74 Pertinent information must be
accurately transcribed and logged manually or entered into the LIS. Specimens may arrive at the
laboratory with a barcoded label generated at the point of specimen collection. Next, the
specimen is evaluated according to guidelines on its acceptability for analysis. Accepted

May 2008

151

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

specimens are distributed to specific laboratory sections for analysis or additional preparation
(e.g., centrifugation). If a specimen is rejected, another must be obtained, increasing the cost of
care. Specimens that cannot be tested immediately and those tested only during certain
days/shifts are stored appropriately (e.g., refrigeration or freezing).
Quality problems in specimen processing (aside from patient misidentification described above)
include transcription errors and forwarding the wrong specimen type. Transcription presents
opportunities for multiple types of error. A study published in 1999 reported that 4.8% of
outpatient requisition slips had at least one laboratory order entry error type, including
discrepancies in the test ordered, physician’s name, and test priority status.43 Order entry errors
tended to be higher in facilities with a greater percentage of occupied hospital beds, those that
made extensive use of verbal orders (in person and by telephone), and federal facilities in both
urban and rural settings. In contrast, an Australian study of pathology laboratories reported in
1996 found transcription error rates of up to 39% among lowest performers and up to 15% among
best performers.104 Error types in order of prevalence included those related to patient and
physician identification, patient sex and age, tests requested, and patient ward location or
address. Transcription errors can be decreased through routine rechecking of orders entered
against requisitions and substitution of verbal orders with written and facsimile orders.43, 105, 106
The evaluation of specimen suitability is a critical factor in test result accuracy, precision, and
usefulness. Preanalytic problems (e.g., specimen collection) may not be discovered until the
examination for acceptance. Accepting unsuitable specimens can lead to erroneous information
that compromises patient care.107 Guidelines for evaluating specimens are integral to QC
procedures, and rates of and reasons for specimen rejection are evaluated as an indicator of
quality. The high rates of acceptance of inadequate specimens resulting in incorrect Pap smear
interpretation with subsequent adverse effects on patients in the late 1980s precipitated increased
regulation of the laboratory sector. It also helped to prompt development of new technology to
aid in detection of abnormalities. Proceeding with microbiology or other testing of specimens of
insufficient volume may result in false-negative blood cultures, adversely affecting antibiotic
management. As mentioned, leading reasons for rejecting specimens often are associated with
problems in the preanalytic phase, including hemolysis, improper method of collection, empty
specimen container, mislabeling of container, clotted specimen, delay in transport, and significant
platelet clumps.108
When accepted, specimens undergo additional preparation for analysis. Preparation of clinical
pathology specimens involves several activities: centrifugation,k aliquoting,l pipetting, diluting,
and sorting specimens with appropriate labels (often barcoded) into batches for introduction into
automated analyzers or other methods of analysis.109 Molecular testing methods have the
potential to replace many conventional microbiology laboratory assays.110, 111 Preparation of
microbiology specimens for these new molecular-based testing methods may involve nucleic acid
purification and amplification with automated systems instead of manual procedures. Studies of
resource allocation demonstrate that specimen preparation consumes a large portion of the
laboratory budget (19%) and staff time (37%), and exposes laboratory workers to the risks of
Centrifugation is the use of a mechanical device that uses centrifugal or rotational forces to separate substances of
different densities, such as solids from liquids or liquids from other liquids.
l Aliquoting is the separation of an equal fractional part from the whole, especially specimens of substances that have
the same volume or weight.
k

May 2008

152

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

handling infectious specimens.112 In addition, specimens requiring separation by centrifugation
historically have been bottlenecks in laboratory processes because of the time required to
manually load and unload the instruments.113 The introduction of automated preanalytical
processing systems has reduced labor, hazard, and errors associated with specimen processing.
In larger laboratories, preanalytic processing systems often are linked directly to analytic systems,
forming total laboratory automation systems whereas in other, usually medium and smaller
laboratories, they operate as modular or stand-alone systems. Because these systems can include
instruments for chemistry, immunoassay, hematology, coagulation, drug screening, and other
tests, they allow for dramatic gains in consolidation of work and personnel and the integrity of
specimen handling.114 Preanalytic processing units typically perform the basic tasks of reading
barcodes, centrifuging specimens when required, and decapping and sorting tubes, as well as
more complicated tasks such as checking the quality of a specimen with instruments designed to
detect substances that interfere with testing, such as hemoglobin, bilirubin, and lipoprotein
levels.17 Some units have capabilities for postanalytic storage and retrieval. Most importantly,
these systems eliminate much of the rote manual work of laboratory staff that causes errors due to
fatigue or distraction.115 In a recent evaluation of an automated preanalytical blood specimen
processing unit at two U.S. academic health centers, sorting and routing errors decreased from
7,950 to 477 per month and biohazard exposures decreased from 2,658 to 6 per month.113

CLINICAL PATHOLOGY ANALYTIC PHASE
Specimen Analysis
Along with preanalytic activities, the quality of test results depends on factors linked to the
analytical systems or processes used for testing itself. In clinical pathology, many tests are
conducted using automated laboratory instruments that analyze the specimen and generate results.
Technologies include, but are not limited to, chemistry analyzers; immunoassay instruments;
hematology, coagulation, and urinalysis equipment; electrochemistry,m electrophoresis,
chromatography, mass spectrometry, and flow cytometry instruments; and molecular diagnostic
technology.117 Although some analyzers require more operator involvement than others, in general,
high levels of automation have enabled laboratorians to concentrate more on QA and results
interpretation.118 In addition, most laboratory QC and assurance programs have targeted accuracy
and precision during specimen analysis. As a result, the risks and rates of testing errors have been
significantly reduced to the extent that analytic systems now have the lowest error rates when
compared to any step in the preanalytic and postanalytic phases.119 This trend is expected to
continue given recent advances in technology development supporting connections between
analytic systems and preanalytic processing units to create total laboratory automation.
The overarching goal of analytic system quality is to ensure run-to-run accuracy and precision
(i.e., reproducibility). Accuracy is the degree of conformity of a measured or calculated
quantity to its actual, nominal, absolute, or some other reference value. Precision refers to the
closeness of agreement between independent test results obtained under prescribed conditions;
it is a measure of reproducibility or random error. Achieving accuracy and precision requires
m

Electrochemistry involves the measurement of current or voltage generated due to the activity of specific ions.
Several different types of instruments are used for these measurements.116

May 2008

153

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

careful selection of reagents, rigorous use of controls, and strict adherence to established
protocols. While automated analytic systems have successfully minimized many errors, factors
such as interference and calibration can alter the conditions of measurement, create variance
and/or bias, and diminish accuracy and precision of testing systems, results, and interpretation.
A 2001 report of a CAP survey of PT specimens for therapeutic drug monitoring that evaluated
25 drugs among 5,000 laboratories found that 57.8% (range 35.3-73.7%) of variance was due to
long-term, within-laboratory variance, 25.0% (8.8-50.6%) was due to short-term withinlaboratory variance, and 17.3% (5.0-35.4%) was due to between-laboratory variance; total
laboratory variance was 82.7% (64.6-95.0%).120
Bias, or systematic error, refers to the extent to which a measurement, sampling, or analytic method
systematically underestimates or overestimates the true value. Analytic bias can directly affect
patient classification and clinical decision-making by shifting the distribution of test values.121 For
example, a major study examined the relationship between matrixn effects and the accuracy of
laboratory measurements for 11 analytes, based on CAP Comprehensive Chemistry Survey data for
1994, and using definitive methods at the National Institute of Standards and Technology and
CDC. Among the findings were that matrix biases affected results in 69% test comparisons and
that, because of matrix biases, the reference value was the correct target value only 32% of the time.
Other error was introduced by random matrix effects and calibration biases.122 Minimizing bias is
accomplished by examining contributors to variance in test values including sensitivity and
specificity, interference, instrument calibration error, reagent lot differences, personnel performance
error, and other factors.123-125
A common cause of bias is interference, which is caused by specimen components other than the
analyte that influence the concentration measurement.125 Interference may be caused by a single,
identifiable substance or property of the material. Major endogenous sources of interference
include hemoglobin, lipemia, bilirubin, and proteins and extraneous antibodies, while exogenous
sources include drugs, materials given to patients for diagnostic purposes, and additives from
collection containers (e.g., anticoagulants, preservatives).125 Interference can lead to falsely elevated
or lowered measurements. A 2002 study of 5,310 patients in the UK for whom common
immunoassay tests for TSH and/or gonadotropins were requested found that, of the instances of
interference in immunoassays for TSH, lutenizing hormone, and follicle-stimulating hormone, the
interference was large enough in 82% to cause inaccurate results and have potentially adverse
effects on patient care and health care costs.126 The magnitude of interference can vary significantly.
Whether or not the bias due to interference is clinically significant depends on the use of the test
results and the allowable error rate. In general, laboratorians are guided by the allowable error rates
given in the criteria for acceptable performance in PT specified in the CLIA regulations.
Other causes of bias that can result in spurious test results include instrument calibration error,
reagent lot differences, inaccurate mathematical correction for specimen dilution, and
misinterpretation of instrument codes.123, 124 Calibration is a measurable signal related to a
substance concentration or other reported result. It requires testing the specimen against one or
more materials (calibrators) that behave similarly to the specimen and for which the true result is
known (i.e., a comparator). The response should be linear over the reportable range (i.e., the
response should be proportional to the analyte concentration). Calibration drift is a systematic
n

A matrix is the biological medium such as blood, urine, or breath in which a substance is being detected or that is
being used for a reference standard.

May 2008

154

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

change in measurement that occurs over a time period of unadjusted, continuous operation of a
test instrument. In 2004, the National Institute of Standards and Technology released a report on
the impact of calibration error in clinical decision making and health care costs.127 This report
studied calcium results in about 89,000 Mayo Clinic patients during 1998-1999 and found that
calibration error in measurements of serum calcium levels led to analytic bias in 15% of tests
results. Not only were some test results passing decision thresholds specified in practice
guidelines, but estimated costs of the errors on a national scale were substantial, ranging from $60
million to $199 million per year. Similarly, in a study reported in 2000 of inaccurate test results
based on data from the New York State Department of Health PT program to characterize the
quality of toxicology testing, calibration drift was cited as the most frequent cause of analytic error
(48%), followed by method bias (14%), indeterminate source (11%), reportable range (9%), and
component failure (8%). This study also noted that approximately half of laboratories used an
allowable error for QC of analytic systems that exceeded the threshold error specified by
manufacturers for stable instrument performance.124 Nonconformity is documented to be highly
correlated with process complexity.128 There has been a growing trend to have all laboratory
measurements traceable to a primary method of analysis reference material, but this may not
always be possible when the material being analyzed is unstable.
Lot-to-lot variations in the manufacturing of calibrator and reagent concentrations or volume can
lead to bias affecting analytical performance.127 The lack of uniformity and standardization
among manufacturers makes the implementation of laboratory-based guidelines difficult because
each such guideline must have method-dependent decision limits.123, 129 The heterogeneity of test
values also makes it difficult to integrate data or test results into a patient’s medical record, or to
make use of the test results outside the institution or setting in which they are produced. This
issue is widely recognized in the laboratory medicine sector and efforts are underway to
standardize values. Certain tests, such as C-reactive protein, require higher sensitivity in order to
correlate test values to clinical diseases (e.g., cardiovascular disease). Efforts to define
performance criteria for such high-sensitivity tests may lead to improved standardization,
performance in quality assessment schemes, and enhanced risk prediction.130

Report Review, Interpretation, and Verification
Subsequent to specimen analysis, the next step in testing is report interpretation and verification.
In most large clinical laboratories, test results are produced from and stored in the LIS. In these
laboratories, test data may be entered manually into the LIS or automatically transferred to the
LIS from automated systems. However, smaller laboratories, POLs, and providers using POCT
devices often document values without use of an LIS. In these instances, the results reports may
be generated from a printer linked to analyzers or a POCT device, or documented directly in the
patient’s medical record.
Review of laboratory reports requires checking the results for any instrument error codes,
markedly abnormal (especially critical) results, or specimen integrity issues; comparing multiple
results on the same specimen if applicable; and determining appropriate commentary for
inclusion in the final report. Most laboratories have a few levels of review depending on the type
of test and values in the report.131 At the first level, the technician or technologist analyzing the
specimen reviews the test results. Then, the supervisor (technologist) approves results for certain
tests, and may report results to the clinician unless additional review by more senior professional

May 2008

155

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

staff is required. At the next level, a professional staff member (e.g., pathologist, doctoral-level
scientist, microbiologist, biochemist, bioanalyst) reviews and approves the test results for release.
Senior level review and approval usually is performed when test results indicate abnormal
values. Manual review of results is necessary in such instances as verification of anatomic
pathology results and critical test results. However, automated verification systems now exist for
tests such as chemistry and hematology assays.
Most laboratories continue to use manual processes for report validation. Manual test validation is
a time-consuming, tedious process with large interindividual variation that slows laboratory TAT.107
Common errors associated with manual processes are data entry/transcription errors that account
for about 4.6% of all hospital laboratory errors.43, 76
Automated verification (or autoverification), shows promise in validating results without review
when the results meet predefined criteria.132, 133 These mechanisms rely on knowledge-based
techniques such as internal consistency checks, delta checks,o and checks for specific errors in
addition to the standard reference range and pathological limit checks of the LIS.133 Also, report
processing by these systems is fast and highly efficient. For example, an immunoassay
autoverification system, under the direction of a supervisor, can validate close to 500 results in
about 30 minutes.135 Autovalidating can allow senior level laboratorians to focus on difficult cases
and interact with clinicians. Some autoverification systems lack the ability to scan control data
before the report is generated.136 Beyond these advantages, it remains to be demonstrated that
autoverification systems improve patient safety and outcomes.107

Reference Intervals
Reference intervals, also known as reference or normal values,p are pre-determined values against
which laboratory test results are compared in order to allow clinicians to make physiological
assessments, medical diagnoses, and management decisions.138 The intervals are established by
testing a group of individuals selected on the basis of well-defined criteria.139
The most common type of reference interval, health-associated intervals, is derived from a
reference sample of people who are in good health. For example, 95% of the healthy adult
population tested by many laboratories has a serum potassium level that is between 3.5 and 5.1
mEq/L; this range is used to define the serum potassium reference interval. Disease-associated
intervals, sometimes referred to as decision-based intervals, are specific medical decision limits
that allow clinicians to classify patients as having a disease, as being healthy, or to otherwise
manage patients.140 For example, hyperlipidemia guidelines typically recommend diet and
exercise to lower cholesterol in otherwise healthy adults once their serum cholesterol level
exceeds a certain threshold, e.g., 200 mg/dL.138 Disease-associated intervals are often defined
during clinical trials and incorporated into the medical literature and adopted by laboratories.
The creation of reliable reference intervals is an important task for both clinical laboratories and
for manufacturers of diagnostic tests. While individual laboratories independently may obtain
A delta check is a comparison of consecutive values for a given test in a patient's laboratory file used to detect abrupt
changes, usually generated as a part of computer-based QC programs. If a delta exceeds its threshold, the value for
“today” fails the check and is suspected of being erroneous.134
p The term reference interval is preferred over normal values because it can be defined for a given reference population.137
o

May 2008

156

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

similar test results, these results may be interpreted differently if different reference intervals are
being employed, leading to differences in what is considered healthy or pathological.109
However, reference intervals historically have been poorly defined and not determined using a
uniform process, leading to considerable variation in clinical laboratories.139 Inter- and intralaboratory differences in preanalytic and analytic factors also have been implicated as sources of
variation in the development and application of reference intervals.140
Some work has been done to standardize the reference interval process. CLIA requires that
laboratories introducing an unmodified, FDA-cleared or approved non-waived test system verify
that the reference intervals supplied by the manufacturer are appropriate for the laboratory’s
patient population before reporting patient test results.141 CLIA also requires that laboratories
modifying FDA-approved tests or developing their own tests establish reference intervals for their
assays prior to reporting patient test results. In all cases, reference intervals must be included in
laboratory reports or made available to individuals who order tests.142 In 2000, the Clinical and
Laboratory Standards Institute (CLSI) published a voluntary standard for clinical laboratories and
test manufacturers that provides information on defining and determining reference intervals with
the intention of achieving a level of reliability and accuracy across reference intervals.139 Assay
harmonization also has been proposed to eliminate the requirement that each laboratory establish
its own reference intervals.140 At present, however, there are few enforced regulations to ensure the
validity of the reference intervals defined by individual laboratories. A 2007 CAP Q-Probes study of
163 laboratories found that approximately half of all laboratories adopted reference intervals from
manufacturers without testing them on-site using healthy individuals.138
For many laboratory tests, no single reference interval applies to everyone because the test may be
affected by factors such as age and sex.137 For instance, the concentration of alkaline phosphatase,
a cellular enzyme responsible for creation of bone, rises in proportion to the production of new
bone cells. While children and adolescents should have high alkaline phosphatase levels, high
levels in adults can be indicative of disease. Similarly, hemoglobin and hematocrit both decline
naturally as part of the aging process. In many cases, reference intervals that are appropriate for
pediatric populations and elderly populations have not been widely developed. Thus, test values
that are considered healthy compared to aggregate reference intervals may be abnormal if agespecific intervals are used.140 The 2007 CAP Q-Probes study described above also found that few
laboratories test healthy children to establish pediatric reference intervals; most of the laboratories
that do test healthy children use in-laboratory testing only to validate reference intervals supplied
by manufacturers rather than to establish their own reference intervals.138

ANATOMIC PATHOLOGY TRANSITIONAL PREANALYIC
Specimen Processing/Accessioning and Preparation
Anatomic pathology tissue processing/accessioning,q preparation, and examination in the
analytic phase are generally more labor-intensive than for clinical pathology, which, in many
large laboratories, relies more on automated modules for analyzing specimens. The exception is
microbiology, which also relies on labor-intensive microscopic examination. Some of the
q

Accessioning refers to the process of recording and assigning a surgical pathology identifier number to a case and
specimen(s) that meet the laboratory’s acceptance criteria.

May 2008

157

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

processing tasks of anatomic pathology are analogous to those in clinical pathology. When
specimens arrive, information from requisition slips and orders must be accurately transcribed
and entered into the LIS or retrieved from a computer-based system designed for anatomic
pathology; a sheet of labels is created, the specimen and labels are forwarded to the appropriate
pathology section, and the specimen undergoes gross examination for acceptance or rejection.
The similarities between processing in anatomic and clinical pathology end there.
Preparation of an anatomic pathology tissue specimen is a complex, multistep process in which
the tissue is repeatedly cut, transferred, and relabeled. After initial preparatory cutting, the tissue
is embedded in wax to create blocks for sectioning from the gross (whole) specimen and produce
slides for examination under a microscope. Most tissue cutting is performed manually by trained
professional histotechnologists or pathologists’ assistants with the use of a microtome blade or
scalpel, though automated cutting systems are in early development. Though infrequent, injuries
to the laboratorian can occur during early stage specimen cutting. Usually, such injuries can be
avoided by complying with safeguards such as use of protective gloves, use of handles with
cutting blades, and avoidance of inherently dangerous maneuvers.143
There are many points along the anatomic pathology workflow where errors and other threats to
quality can arise.62 The most prevalent problems during the preparatory steps involve patient or
specimen identification errors and mix-ups, cognitive errors in the gross room, tissue cutting- or
staining-related issues, and specimen defects. As noted above, patient and specimen
identification errors that affect the analytic phase can begin in the preanalytic phase as a result of
incorrect information on the patient or specimen requisition slip, specimen placement in an
incorrectly identified container, specimen mix-ups and incorrect test ordering at the time of log in,
or transcription errors.45 In a broad study of identification errors and mix-ups in all three phases
of the testing process for surgical pathology, investigators found numerous sources of
identification error, many of which pertain to specimen accessioning or preparation of tissue
blocks and slides, including:

ƒ

Specimen accessioned to wrong patient in the laboratory

ƒ

Case misidentified in the gross room and dictated with wrong accession number or
wrong specimen number

ƒ

Specimen placed in or embedded into incorrect cassette

ƒ

Specimen sections mixed up in water bath

ƒ

Specimen placed on slide labeled for another specimen

ƒ

Incorrect permanent label affixed to slide

ƒ

Incorrect history/paperwork associated with slide62

Additional identification-related errors that take place during and after microscopic examination
are listed in the next section.
Cognitive errors can occur during preparatory gross room tasks or microscopic examination.
Little detailed information is available about these types of error.144 An example of a cognitive
error of omission, defined as the failure to perform pertinent ancillary studies at the tissue, block,
or slide level, may be the failure to take fresh tissue for flow cytometry or the failure to perform a

May 2008

158

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

culture. Other cognitive errors in the gross room may be inaccurate examinations with poor
descriptions (e.g., lack of appropriate measurements), lack of or incomplete lesional sampling,
and lack of sampling of pertinent areas necessary for proper lesional characterization or staging.
Cognitive errors associated with gross room tasks can be common though difficult to detect.
However, there is virtually no evidence on rates of cognitive errors in the laboratory.
Incorrect conclusions can be drawn from poor technique associated with tissue cutting (i.e.,
undercutting or overcutting specimens) and tissue staining (e.g., misidentification of the
appropriate stain, incorrect selection of reagents and staining protocol). Cross contamination by
“floaters” (i.e., foreign tissue fragments carried over from other sections or specimens) is a better
documented quality problem. In a 1996 CAP study of 275 laboratories, extraneous tissue
fragments were found in 0.6% of 321,750 slides examined prospectively and 2.9% of 57,083 slides
examined retrospectively. For the prospective study, 59.4% of the contaminants were found on
slides, 28.4% on paraffin blocks, and 12.2% were undocumented. In comparison, for the
retrospective study, 72.9% of contaminants were associated with slides, 15.9% with paraffin
blocks, and 11.2% were undocumented. The majority of extraneous tissue came from different
cases (63.2% in the prospective study and 48.5% in the retrospective study). Study findings
confirmed that more than 92%of instances of extraneous tissue originated in the laboratory.145

ANATOMIC PATHOLOGY ANALYTIC PHASE
Microscopic Specimen Examination
In anatomic pathology, microscopic examination is the primary method for making clinical
diagnoses, although innovative, automated technologies are increasingly being developed and
used in cytology and molecular pathology.146 Digital microscopes with advanced capabilities for
taking, storing, replicating, and cataloging digital images are used widely among laboratories.147
Certain microbiology tests also rely on microscopic examination.
Histotechnologists and cytotechnologists play an important role in the microscopic examination
of tissues. They formulate and document interpretive conclusions for presentation of cells,
biochemical components, tissue vascularity and, if applicable, presentation of disease, which may
include definition of grade and stage of disease, tumor type, measurements, and characteristics.148
These laboratorians also play an important role in the examination of infectious agents present in
tissue specimens. Once the result is approved by the technologist, the pathologist reviews the
findings. A secondary case review may be undertaken or required as part of a QC check or if the
diagnostic findings are unclear, as discussed further, below.
Because of pathology’s dependence on visual association, technologists and pathologists must
possess the cognitive and interpretive skills needed to evaluate accurately tissue specimens and
establish diagnoses beyond a reasonable doubt. The skill set requires visual pattern recognition of
cells and structures, recognition of whether the arrangement is normal or abnormal, association of
the features to diseases that mimic the pattern, development of multiple hypotheses or differential
diagnoses, and investigation of the clinical or histological possibilities to rule-in or rule-out
diagnoses.149 In surgical pathology, cognitive and interpretive skills are applied to histologic
grading and staging, considered the most important prognostic indicators. Although several
grading systems exist, the National Cancer Institute’s three-grade system based on histologic

May 2008

159

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

type, tumor necrosis, and mitotic activity serves as the general standard for determinations.
Similarly, the American Joint Committee on Cancer pathologic staging system is the standard to
guide evaluations of tumor size and depth. For gynecologic cytology, standards for examination
have been codified in CLIA.
QC, performance evaluation, and test reproducibility to minimize diagnostic discrepancies and
errors have been more easily defined and applied in clinical pathology than in anatomic
pathology.150 The reliance on subjective judgment and somewhat variable diagnostic thresholds
between individuals limits reproducibility and may introduce bias in areas of cytology and
surgical pathology. For example, one study conducted in 1995 in the area of neuropathology
found discrepancies between general pathologists and neuropathologists. The pathologists were
in disagreement in 42.8% of cases. Of those cases, 20.6% were considered serious (e.g., tumor
diagnosis changed to non-tumor, diagnosis changed within benign or malignant), 44.9% were less
serious but substantial (e.g., glioma type or grade changed), and the remaining 34.6% were minor
(e.g., tentative or doubtful diagnosis confirmed). These differences can have detrimental effects
on patient management and costs.151
When errors occur in anatomic pathology analytic processes, they can be classified as either
cognitive or identification/clerical. Cognitive errors at the microscope include slips and lapses
while analyzing slides, poor cognitive formulations, knowledge problems, communication
problems (e.g., poorly worded or unintelligible reports), and difficulties in using classification
models that have poorly defined criteria. Correct patient and specimen identification and record
keeping throughout the processes associated with microscopic observations and documentation
are crucial to quality and accuracy.17 Examples of identification and clerical errors include:
pathologist examines wrong slide, pathologist dictates/writes incorrect case number,
transcriptionist types diagnosis for incorrect case or specimen; downtime/temporary medical
number updated incorrectly, and report prints with wrong patient identifier due to LIS error.62
A literature review published in 2004 reported wide variability in anatomic pathology errors,
ranging from less than 1% to 43% (with mean error rates of 1%-5%) among institutions
participating in the studies reviewed.152 Error rates tended to be higher in cytology compared to
surgical pathology, with false-negative diagnosis as the greatest source of error. Most cognitive
and identification/clerical errors are discovered before sign-out, although a small percentage are
discovered thereafter. Discovery in either regard may be precipitated by receipt of additional
information or material for the current case or a different recent case, results review by the
pathologist or other laboratorian, or pathologist-initiated external consultation with an expert.69
Other methods of detection include intradepartmental review or a double-read of the results
before sign out, and preparation for or presentation at a conference with clinicians (e.g., board
review) after sign-out. The patient’s clinician also may question the pathologists’ diagnosis and
request re-evaluation of the specimen and/or findings, at which time an error may be discovered.

Results Review
In general, laboratory test results are transcribed into the LIS or another computer application,
reviewed and approved by the pathologist, and delivered to the clinician. In certain instances
(e.g., a difficult case), the pathologist may seek an external consultation with another pathologist
for a second opinion or specific expertise prior to finalizing the report. When an experienced

May 2008

160

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

pathologist finds that immediate recognition of the pathologic process does not occur, and that
applying familiar rules and criteria do not lead to a clear diagnosis, the pathologist may face an
unfamiliar scenario and can be prone to the same types of errors as the novice.17, 153 As such,
seeking consultation is based on self-knowledge of one’s limitations and the severity of
consequences of error.154 A study published in 2002 reported findings on the rates and
characteristics associated with expert consultation. Consultations were sought for 0.5% of cases,
of which 52% were sent to nationally known experts and 32% were went to local experts. Among
the consultations, 54.6% confirmed the original diagnosis. The referring pathologist’s assessment
of possible diagnoses was correct in only 21.5% of cases, 15.9% of consults confirmed the original
diagnosis but added significant information, 6.5% were discordant with the original diagnosis,
1.4% were attributed to unidentified other factors, and 0.7% were ambiguous and not helpful.155
After report sign-out, discrepancies and errors can be detected through secondary case review,
e.g., via conference review (such as an oncology board) or institutional review. The specific
methods employed to determine discrepancies are correlation of findings and assessment of
amended reports. In general, rates of discrepancy and errors after cases are signed out are
relatively low for cytology and surgical pathology. One study involving 74 hospitals reported
variability in rates and causes of laboratory discrepancies in anatomic pathology. While 10% of
hospitals reported no discrepancies, another 10% reported errors in at least 5% of diagnoses. The
mean frequency of laboratory discrepancies was 6.7%. About 47.8% of discrepancies resulted in a
change within the same category of interpretation (e.g., one tumor type was changed to another),
20.9% resulted in a change across categories of interpretation (e.g., benign diagnosis was changed
to malignant), 18.5% were from typographical errors, 9.1% were from a change in patient
information, and 3.7% were from a change in margin status. According to the investigators, 5.3%
of discrepancies had a moderate or marked effect on patient care.156
The most common method of detecting errors, before and after sign-out, is through the
correlation of findings between different tests on the same tissue specimen. Correlations of
frozen-permanent sections and of cytologic-histologic findings are well-documented in the
literature. For gynecologic cytology, correlation of cytologic and histologic findings is mandated
by CLIA. Discrepancy rates using correlation are generally low. For example, in a hospital study
published in 2002, investigators reported discrepancies in nongynecologic cases of 2.26% for
cytology and 0.44% for histology, and in gynecologic cases of 0.87% for cytology and 7.37% for
histology.157 These rates were consistent with error frequency rates of other studies. Several CAP
studies that examined diagnostic errors on frozen sections also published discrepancy rates in the
range of 1.4-1.7%.158, 159
Error rates and types have been assessed by calculating the rate of reports that must be
amended. In a large CAP Q-Probes study conducted in 1996 of surgical pathology in 359
laboratories, the aggregate rate of reports amended to change clinically significant information
was 1.9 per 1000 cases.160

May 2008

161

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

POSTANALYTIC PHASE
The main components of the postanalytic phase focus on results reporting and interactions
between laboratorians and clinicians. The core challenges for pathologists and other laboratory
professionals in their communication with physicians concern timeliness of reporting, notification
of significant abnormal test results, and presentation of relevant information through reports and
interpretive comments. Customer satisfaction surveys have found that all of these factors receive
high percentages of below average and poor ratings.12 These challenges are often cited as the
chief concerns in the postanalytic phase.

Test Turnaround Time
The timeliness with which test results are delivered is one of the most prominent parameters of
laboratory medicine and a common indicator of performance.161, 162 Common among these are test
TAT and time for notification of critical results. Automation of various steps in the analytic
phase, increased use of electronic results reporting, and development of automatic electronic
alerting systems for critical values have helped to decrease TATs.
TAT is typically assessed by determining the difference between recorded starting times (test
ordering) and ending points (test reporting time).163, 164 Some laboratories also analyze intervals
that comprise TAT, such as test order to specimen collection, collection to laboratory specimen
receipt time, and receipt time to reporting time, in order to determine the specific points at which
delays occur.164 A few laboratories also are expanding the scope of measurement by evaluating
”therapeutic TAT,” the time from initiation of the test order to the implementation of clinical
decisions (e.g., change in treatment).165
Regardless of method, TAT is viewed as a quality measure that reflects the performance of the
testing process as a whole. Prompt and predictable reporting of test results can increase efficiency
of patient care and improve clinician and patient satisfaction, even when it does not affect health
outcomes.163 Actions taken as a result of regularly monitoring TAT and other factors may
improve certain aspects of performance.162, 164 However, researchers also recognize that shorter
TAT measurements do not necessarily indicate superior service.162 That is, absolute
measurements of TAT do not reflect whether the laboratory services meet the expectations of the
clinicians using those services. Also, improving TAT can be challenging, not only because of the
contributing factors outside the control of the laboratory, but because laboratories frequently try
to improve TATs for a specific test, location, or specimen type by immediately identifying and
assaying those specimens in question, thereby extending the TATs of other tests.166
Most TAT studies have focused on inpatient and emergency care settings, though a few researchers
have ventured to outpatient settings. TAT varies depending on the location of the laboratory (e.g.,
satellite laboratories often have shorter turnaround times than central laboratories) and the analyte
(if other variables stay the same). Typical TAT for routine testing in the emergency setting is 12-48
minutes or less, particularly if POCT is available, a few hours for general testing in the inpatient
setting, and a few hours to the next morning or 24 hours for testing in the outpatient setting. One
study of stat laboratories for the emergency department and ICUs evaluated outlierr test TATs,
r

Outlier events are test TATs that exceed targeted or tolerable reporting times. These events are used to assess the
degree to which laboratory services meet the needs of clinicians.162

May 2008

162

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

accounting for about 10-15% of total events. The majority of problems directly affecting TAT for
selected chemistry tests are associated with preanalytic-related test ordering and specimen
collection (57%) and analytic-related personnel and technical problems (28%).164 Staff shortages in
both phases were the major cause of delays. The percentage of delays was 4.7% higher in ICUs than
emergency departments. Shortened TATs have been associated with rural locations, delivery of
specimens as collected, pneumatic tube delivery systems, and continuous versus batch testing.167
Innovative POCT devices also have had a significant impact of the reduction of TAT. For example,
a study of stat TAT in an academic health center reported TATs that were 1-2 minutes shorter for
bedside testing compared with a satellite laboratory and 9-12 minutes shorter in the satellite
laboratory compared to the central laboratory.165
Data from studies of TAT for outpatient testing in hospital laboratories has produced different
results. In 1997, a Q-Probes study of TAT for three common assays reported that 50% of
laboratories were able to verify 90% of results within 2.7 hours for the CBC, 3.5 hours for the
biochemical profile, and 21.6 hours for TSH.168 For half of participants, outpatient testing
accounted for about 46% of the typical hospital laboratory workload. A follow-up study reported
in 2002 found increases in TATs to 3.9 hours for the CBC, 4.9 hours for the biochemical profile,
and 38.1 hours for the TSH.169 TATs increased for specimens received later in the day for all
analytes and when specimen transport was not under the control of the laboratory.
TAT, laboratory practices, and specimen characteristics (e.g., type of specimen and findings)
associated with anatomic pathology (e.g., cytology, surgical pathology) have been examined as well.
Result availability and timeliness are considered indicators of quality for anatomic pathology.
Turnaround times varied substantially according to specimen type. A study of gynecologic cytology
found that 50% of laboratories had a mean TAT of 6 days or less, though the average laboratory
could complete 90% of their cases within 8 days; 10% of laboratories needed 13-19 days.s,170 TATs for
nongynegologic cytology (e.g., fine needle aspirations) are much shorter; 50% of laboratories had a
mean TAT of 1.6 days or less (3 days for 90% of cases); 10% needed 3.2-6 days.171 These cytology
studies found that longer TATs were associated with the need to contact the physician’s office for
additional information, and use of students, residents, or fellows in the evaluation. In addition to
these factors, gynecologic cytology TATs were influenced by use of reference laboratories for all or
part of the evaluation and provision of service on the weekend, whereas TATs for other cytologic
specimens were influenced by issuance of atypical/suspicious findings for malignancy diagnosis;
having to pull prior case material for review; having to perform cell blocks, special stains, or other
activities; and not having transcriptionists working weekends.
Many laboratories also monitor TAT for surgical pathology with the goal of having the majority
of cases signed out within 1-2 days.17 A 1995 Q-Probes study of biopsies and complex specimens
documented mean TATs of 1.5 days for complex cases, 1.3 days for routine cases, and 2.6 days for
cases requiring special handling.172 Factors associated with increased TAT included: institutional
bed size greater than 450, responsibility for gross section dissections assigned to residents only,
slides not available to pathologist before 12:00PM, resident involvement in sign-out, interposing a
day between availability of slides and final sign-out for resident education purposes, and a
greater number of pathologists on staff. These findings were confirmed in a subsequent study

s

They concluded that, if one week is acceptable TAT and actual screening time is 5-10 minutes per slide, there is
room for improvement.

May 2008

163

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

published in 1998. Pathologists had signed off on 85.9% of biopsy diagnoses by the second
working day, and surgeons received the hard-copy reports by the fourth working day.

Notification of critical values/test results
A critical value or test result represents a pathophysiologic state at such variance with normal as to
be potentially life-threatening unless something is done promptly and for which some corrective
action can be taken.173 Timely, if not immediate, notification of critical values or results is crucial to
patient safety. Regulatory bodies now require laboratories to have policies for notification of such
values, although performance benchmarks and definitions of specific critical values can vary by
accrediting organization, laboratory, or health care setting.174 In many instances, providers will
order laboratory tests stat even though the patient is not in a life-threatening pathophysiologic state.
The variance may be in the high and low limits of the analyte values.175 In 2002, a CAP study of
623 institutions confirmed wide variation in critical values for routine chemistry and hematology
tests, with no value being the same in 80% or more of laboratories.176 For certain common
analytes (e.g., glucose, potassium, hematocrit, white blood cell count), almost all laboratories had
high and low critical values. For other analytes, laboratories had either a high or low critical
value. About 72% of laboratories did not have a policy on repeat critical values for the same
patient. Of particular note is that more than 45% of critical values were unexpected and 65%
resulted in therapeutic change.
The Massachusetts Coalition for the Prevention of Medical Errors and the Massachusetts Hospital
Association are leading a state-wide initiative to standardize critical values and reporting
mechanisms for laboratory, cardiology, radiology, and other diagnostic tests across all health care
institutions.177 The initiative aims to address “errors in the process of communication of test
results that are both frequent and have the potential for serious harm.” A set of safe practice
recommendations has been developed to promote successful communication of test results. In
addition, the group established a standardized set of abnormal test values widely agreed to be
critical to patient health.
The limits chosen for any clinical result directly affect the institution’s workload, as notifying
clinicians of critical values is labor intensive. A CAP Q-Probes study of 623 institutions, published
in 2002, found that telephone contact is the primary method of reporting critical values by more
than 90% of laboratories, with calls taking an average of 6 minutes for inpatients and 14 minutes
for outpatients. The calls were made by the laboratorian performing the test, most often a
technologist. For certain types of tests, other methods of notification include facsimile, computer,
and voice mail.176
Misreading values is not the only problem of communication. In hospitals, accrediting
organizations have required communication only with the responsible physician or caregiver
(usually a nurse) since 2000. To minimize miscommunication of values during notification calls,
The Joint Commission requires the receiving clinician to ”read-back” the critical value along with
patient identifiers.178 They also recently added monitoring of TAT for critical value reporting as a
measure of quality and patient safety.
Previously, communication followed CLIA provisions, which permit reporting with the entity
or individual ordering or responsible for using the results, including unit secretaries. This is
May 2008

164

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

still the case in the outpatient setting. Nurses receive 65% of reports in hospitals and
unit/clerical staff receive the largest percentage (40%) of reports in physicians office practices.176
Communication with clerical staff instead of clinicians can result in another type of error—
undocumented critical values in the patient’s medical record.174 Lastly, some critical values
result from preanalytic problems, such as with specimen collection or analytic interference,
rather than medically significant values.179
While critical results in clinical pathology are time-sensitive, anatomic critical values are typically
viewed as information-sensitive in that the report content is highly important but usually does
not require immediate action.180 Occasional diagnoses in surgical pathology and cytology require
immediate notification of the clinician to rapidly initiate treatment.181 Guidelines for critical
results reporting in anatomic pathology have not been established. Consequently, there is a wide
range of opinion among pathologists about the need for an immediate telephone call and the
degree of urgency. Furthermore, most anatomic pathology results are not quantified with values
and limits. A 2004 study examined the potential for developing parameters to address critical
results in surgical pathology and identified 11 possible cases including, but not limited to, renal
biopsy specimens, vasculitis, and bacteria in heart valves or bone marrow.182 Differences in
opinion prevail; only 58% of these reports documented calls to the clinician. There was a greater
difference of opinion when a new metastasis was identified in a patient and when an organism
was identified in an immunocompetent patient.

Report Formatting
Laboratorians are trained to convert valuable data to useful clinical information in the form of
laboratory results reports—the major communication link between laboratorians and clinicians.
The reports are communicated to the clinician most often with the assumption that the basic
pathologic process described is understood correctly. While the clinician’s level of experience and
training has direct impact on their understanding, this is not always the case, particularly with
growing use of genetic tests and microbiology tests. Yet, little attention has been focused on
educating and training clinicians to more fully understand pathology reports.13 Few published
studies have evaluated comprehension of laboratory reports and, of those available, most focus
on anatomic pathology.
The content, format, and physical presentation of the information significantly affect the
interpretation and use of laboratory data by clinicians.76, 183 Mistakes in the content and
completeness of laboratory reports as well as misunderstanding by the treating physician as to the
significance of the information in the report, among other factors, can delay treatment of a serious
disease and alter outcomes.144 Problems can be magnified when a critical result is unanticipated
by the clinician.
Specific report content issues can include any of the following: uninterpretable information,
incorrect data or reference intervals, inaccurate patient identification, or incorrect physician or
patient location for reporting results. A 1992 CAP study of mistakes in clinical pathology
laboratory reports found that the frequency of content errors was lowest in blood bank reports,
intermediate in chemistry and microbiology reports, and highest in hematology reports.184
Clinicians’ limited willingness or ability to interpret laboratory reports may result in their
focusing inordinate attention on certain values while disregarding others of potential importance
May 2008

165

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

or failing to discern broader clinical implications. For example, in a survey of attending and
house staff physicians in a major hospital system reported in 2002, only 4 of 11 analytes routinely
reported in the CBC battery were selected as frequently or always useful by more than by 90% of
responding physicians: hemoglobin, hematocrit, platelet count, and white blood cell count.
Primary care physicians also chose mean cell volume as a useful parameter in the evaluation of
anemia. The investigators suggested that modifications to report formats were needed to
facilitate physician perceptions of the importance of hematology laboratory results.185
Laboratories also must be cognizant of how new medical findings affect results interpretation and
reporting. The incorporation of new evidence into clinical guidelines can expand indications for
treatment, requiring laboratories to update report content and interpretations. For example, in 2001,
the U.S. National Cholesterol Education Program issued new guidelines for the prevention and
management of high cholesterol in adults, based on accumulated evidence concerning the
contribution of lipoproteins (and other risk factors) to the development of coronary heart disease.186
The guidelines necessitated changes in laboratory analyses, reporting, and interpretations including:
modification of cut-points, standardization of measurements, fasting profile at initial screening, and
testing for emerging risk factors and secondary dyslipidemias.187 Values must be presented clearly
in reports with appropriate interpretive comments reflecting current guidelines.
Several organizations have developed voluntary minimum standards outlining the data elements
that should comprise reports. For example, CMS endorsed the Bethesda System for reporting
cervical cytology results, CDC issued standards for reporting HIV results, CLSI has suggested
standards for microbiology test results and others, CAP established several standards for specific
types of clinical and anatomic pathology reports, and the Association of Directors of Anatomic
and Surgical Pathology has focused on its respective areas of expertise.
The data elements for reports are selected with the primary aim of promoting accuracy and
completeness. The different standards are widely accepted; however, variability in report
accuracy and completeness remains among laboratories. For example, a CAP Q-Probes study of
bladder biopsies and curettings in 268 institutions noted that the presence or absence of
muscularis propria should be routinely included in reports of all biopsies. Yet, for invasive
carcinomas, definitive assessments were reported in only 53.3% of cases, and in only 30% of
noninvasive carcinomas.188 Another CAP study of breast biopsies, published in 1997, found that
23% of reports of malignant cases were missing information on tumor size, 17% on tumor grade,
and 8% on margin status, and 24% did not have information on the extent of intraductal
carcinoma.189 These studies and others also found that the single practice that was most effective
in ensuring completeness of reporting was use of a checklist. The checklist guides laboratorians
in documenting test results to ensure the inclusion of specific data in the final report.189, 190
The physical presentation of test results can influence the clinician’s understanding of them.8 Poorly
designed reports can result in misunderstandings and/or undervaluation of important
information.191 More specifically, the spacing, highlighting, font size, and formatting of content can
affect comprehension of computer-generated reports for clinical and anatomic pathology.
Grammar and word selection also are important for anatomic pathology reports where, unlike
clinical pathology reports, results are documented via selection of predefined phrases and drafting
of free text commentary. In general important, unusual, or critical results in clinical and anatomic
pathology reports can be emphasized with capitalization, a special mark in the margin, or

May 2008

166

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

underlining.192 Color highlighting in electronic reports or printouts can highlight a result for the
viewer’s attention. Unfortunately, most computer-generated reports still crowd data into columns
on each page, are single-test focused, and employ limited use of graphics for certain tests.123, 193
Because specific details of interest to the user are more difficult to abstract from sentences and
paragraphs, clinical and anatomic pathology reports now organize content in synoptic formats.194
Key information provided in tabular form is generally a reproduction of the checklist data. Variable
amounts of free text can be added in the form of special gross features and comments on other
important findings and implications. In general, the data elements are relatively consistent from
one computer program to another, but the presentation may differ according to vendor.
Laboratorian selection of data elements also may result in content differences. Synoptic formatting
generally has improved report legibility relative to straight textual summaries, although more work
is needed to ensure timely comprehension of test results and accompanying information.
A few researchers have examined the impact of redesigned report formats to more graphicallyoriented models.183 One recent study of surgical pathology reports assessed the effect of report
redesign on physician comprehension and found a 30% discordance rate between pathologists’
intended meanings and interpretation by surgeons.183 The authors stated that stylistic
improvements have the potential to interfere with comprehension and increase the number of
misunderstandings, and that further research is needed. Generally, researchers believe that the
forthcoming proliferation of proteomic and genomic tests will challenge clinical informatics and
prompt changes to existing report formats.193 To meet this challenge, the next generation of
laboratory reports must make better use of graphical displays to facilitate the rapid assimilation and
comprehension of important data. Developers have been using these concepts to improve the
design, function, and comprehensibility of electronic health records (EHRs) over the last decade.
Further advancements should incorporate user-friendly graphical displays of laboratory data.

Physician Interpretation and Follow-up
In the TTP, a laboratory test is not complete until its result has been interpreted by the clinician
and incorporated into patient care, as appropriate. There is considerable evidence indicating that,
for a variety of reasons, physicians often do not take, or do not document, appropriate actions in
response to abnormal laboratory test results. In a CAP Q-Probes study of elevated calcium levels
in 525 institutions, published in 2000, 3.5% of reports of abnormal levels were not entered into
patient medical records. Of reports that were entered, 23% did not contain physician progress
notes responding to elevated calcium results; this figure rose to 93% for patients with no known
history of hypercalcemia. Also, follow-up tests were not ordered for 13.8% of the elevated values.
However, in a follow-up survey of physicians for whose patients there was neither physician
documentation of an abnormal test nor ordering of the designated follow-up test, the majority of
responding physicians indicated that they did order the test and that the results led to further
action that was not documented in their notes.195 A retrospective study of 2000-2002 data in 30
U.S. academic medical centers reported that only 40% of diabetic patients with high HbA1c and
only 5.6% with high low-density lipoprotein cholesterol had adjustments in treatment based on
these results.32 A similar study of 1999-2000 data of elderly non-insulin-requiring diabetics in
Ontario showed that fewer than half of the patients of endocrinology specialists or primary care
providers had adjustments in treatment based on elevated HbA1c levels.196

May 2008

167

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

Interpretive Consultation Services
The actual and potential benefits of laboratorians providing information and consultation to
physicians on laboratory test selection and interpretation of patient-specific results is generally
recognized.10, 197, 198 While most laboratories provide some form of interpretive comments in
anatomic pathology reports, this is not always the case with clinical pathology reports. Medicare
reimburses consultations only for the 20 clinical pathology tests that frequently require
interpretation by a pathologist.199 Most private insurers and managed care organizations follow
this Medicare policy.
The primary barrier to interpretive commenting is the shortage of true experts with high
subspecialty expertise in clinical pathology. Other barriers contributing to this problem include:
generally low reimbursement for professional activities within clinical laboratories; more complex
criteria for reimbursement of selected clinical pathology tests; and a shift toward the practice of
anatomic pathology where reimbursement for interpretive comments is provided for all anatomic
pathology tests.197, 199 As a consequence, most current pathologists and scientists do not provide
clinically valuable interpretations in areas such as coagulation, autoimmunity, and other complex
areas of laboratory medicine, although some of these professionals do so despite lack of
incentives. A 2004 report of an evaluation of interpretive commenting in clinical chemistry found
that many of the comments were automatically generated by a computer, whereas others were
individually generated depending on the results and availability of clinical information. The
majority of comments were acceptable; however, some comments were inappropriate,
misleading, or, in few instances, dangerous to patient care.198 Inappropriate comments were the
result of inaccurate assumptions by staff when the clinical information available was insufficient
or when the expertise in clinical chemistry subspecialty areas (e.g., toxicology, endocrinology, and
tumor markers) was inadequate.
Despite these few challenges to interpretive commenting, the service generally is well received by
physicians.10, 12 Several organizations have been studying methods of improving the quality of
communication between clinicians and laboratorians during ordering and results interpretation by
developing programs that expand the use of interpretive commenting in combination with other
tools. For example, a major academic medical center instituted a broad laboratory medicine
interpretive service and undertook a series of studies to assess improvements. The service provides
a physician expert-written, evidence-based, patient-specific interpretation that accompanies the
results of complex laboratory testing panels.9, 197 A study of its coagulation service found that 98%
of physicians perceived the interpretations as useful or informative, 59% perceived reduced time to
diagnosis, 72% perceived that the interpretation reduced the number of laboratory tests required to
make a diagnosis, and 72% believed that it helped them to avoid misdiagnoses.10

POINT-OF-CARE TESTING
POCT, also known as bedside, near patient, decentralized, or alternative site testing, refers to
clinical laboratory testing that is conducted close to the site of patient care outside of the
traditional, core, or central laboratory.200, 201 The main objective of POCT is to produce a result
quickly, facilitating decisions about appropriate treatment and care to improve clinical or
economic outcome.202 POCT can be performed in various settings, including sites of primary care
(e.g., physician’s office and community clinic, community pharmacy, health center, workplace

May 2008

168

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

clinic) and secondary and tertiary care (e.g., emergency room, ICU, outpatient clinic, ambulatory
diagnostic and treatment center).t, 203
Similar to traditional laboratory testing, POCT can be delineated by preanalytic, analytic, and
postanalytic components.204, 205 Some benefits of POCT include decreases in the amount of
specimen required and minimization of delays that are associated with specimen transport,
processing, and preparation203
Ensuring high quality in POCT poses several challenges. Many POCT devices are granted CLIAwaived status and are operated by personnel whose primary training may not be in the clinical
laboratory sciences and who may, therefore, be unfamiliar with testing practices.202, 206 Unlike
laboratorians in the central laboratory, POCT operators often have minimal time to reflect on the
TTP and may not have access to ancillary information such as QC data. Non-adherence to test
protocols and procedures and operators’ use of reagents that are not controlled are two other
sources of error in POCT.204 Improved QC performance in POCT blood glucose monitoring has
been directly associated with supervision of QC programs by laboratory personnel rather than
nursing personnel.207
Laboratory experts have also identified possible sources of POCT error amplification, i.e.,
conditions that increase the frequency or likelihood of POCT error, including those that become
preventable adverse events.204 For example, suboptimal use of real-time POCT results, such as
initiating the incorrect therapeutic action or failing to immediately recognize the significance of a
test value, can amplify errors in POCT. While the immediate availability of POCT results can
significantly enhance the quality of care, additional research is needed to determine the best
methods for integrating POCT into day-to-day clinical processes (i.e., care pathways).208

PREANALYTIC PHASE
Selection of Testing Method
In the hospital setting, professional laboratory consultation and inclusion of the laboratory in the
selection of test methods and ongoing management of POCT are important components of
successful POCT programs.209, 210 Consultations with the core laboratory can assist clinicians by
providing information about the advantages and disadvantages of POCT versus core laboratory
testing in specific patient settings.205 Laboratory participation in the development of practice
guidelines for laboratory order sets can decrease practice variability and ensure that POCT
operators are following the most cost-effective pathway to the best patient outcome. Laboratory
staff also can create reference manuals for each nursing unit (or physician office) that contain
written procedures, policies, training checklists, and other information.205
Testing in physician offices differs considerably from that conducted in the hospital setting,
particularly in such matters as testing menus and knowledge of quality-related procedures.
Physician office personnel usually have been trained only in performing a few tests of limited
complexity.211 If questions arise pertaining to POCT, these personnel typically contact the
diagnostics manufacturer rather than consult with a hospital or independent laboratory.
t

While certain POCT can be conducted at home by individuals, this chapter focuses on clinical uses of POCT; home
laboratory testing and direct access testing are discussed elsewhere in the report.

May 2008

169

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

Test Ordering
Excessive or incorrect test ordering can contribute to medical errors at the point of care.204, 208, 212
Excessive ordering can confuse and overwhelm test interpreters. Increased use of CPOE, clinical
decision support systems (CDSSs), and EHRs may prevent incorrect, excessive, and redundant
POCT ordering by standardizing test ordering and reminding clinicians about previously ordered
laboratory tests.38, 213, 214 CPOE and CDSSs are discussed in greater detail in the Laboratory
Information Systems chapter of this report.
Another source of error related to test ordering is mistimed or uncoupled testing, which occurs
when the delivery of laboratory test results is not synchronized with the therapeutic intervention
during dynamic treatment.204 For example, if incorrectly timed or interpreted, receiving results of
POCT pH and bicarbonate tests during infusions of bicarbonate for patients undergoing
cardiopulmonary resuscitation may prompt test interpreters to react to test results stemming from
a transient or recent pathophysiologic state, rather than to the patient’s current state.

Patient/Specimen Identification
POCT error patterns related to patient and specimen identification tend to differ from those that
occur during traditional laboratory-based testing. Patient misidentification is more likely with
POCT in emergencies when there is a greater potential for a staff member dispatched to perform
POCT on a patient to mistakenly test or record results for a different patient.204 POCT operator
entry of an incorrect patient identification number can lead to such problems as posting of results
to the incorrect patient medical record, inappropriate medical treatment, and unavailability of
POCT results for comparison with previous and subsequent test results.204, 205 Incorrect patient
identification also can result in failure to post test results in the LIS and improper billing.
Patient identification errors in POCT can be reduced. Computerization of POCT devices
increasingly allows for electronic capture of information such as the date, time, operator and
patient identification, device serial number, reagent and control lots, and control ranges, thereby
automating this information capture.215 Some POCT devices have mechanisms that lock out
operators who repeatedly make identification errors, facilitating corrective counseling of these
operators prior to further use.205 For example, one health system that implemented a “three-strike
rule” found that glucose meter testing errors decreased significantly, although there was no
impact on the rate of blood gas errors.216 Other POCT devices can lock out testing if patient
identification is invalid or unavailable.217 Another method for safeguarding hospitalized patients
prior to POCT includes cross-checking patient information against the bed location.
Electronic barcodes have been installed in hospitals recently to automate data entry, and many
POCT devices have built-in barcode scanners.218 In order to be successful, all of the components
that must be identified to the system must be barcoded, including the patient, operator, testing
strips, and QC materials. Following implementation of an automated barcoding system, one health
system found significant reduction in error rates in the glucose and blood gas devices.216 Another
example of barcoding is a POCT system that uses a special syringe with a unique barcode identifier
that scans the barcode, the patient’s wrist band, and the phlebotomist’s identification badge.208 In
this system, the patient’s identification is maintained throughout all phases of testing and is linked
to the test results and other relevant information. However, limitations in barcoding (e.g., difficult
to read through blood stains and moisture, limited number of encodable characters, need for proper
May 2008

170

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

physical alignment of barcode and reader) have prompted interest in other systems for automatic
identification, including radiofrequency identification.218
A POCT error that is characteristic of outpatient settings results from batching of specimens.204 This
error most often occurs with urine tests and rapid tests for GAS antigen that are left on the counter.
Labeling of containers and swabs immediately upon collection reduces batching-related errors.

Specimen Collection
Specimen collection errors include inappropriate or inconsistent specimen type, volume, or
application to the testing surface or reaction chamber on the POCT device.204 Operator variability
and error rates in POCT specimen collection are influenced by several factors, including the extent
and effectiveness of operator training and the frequency with which POCT operators perform
specific tests and progress along the learning curve for each test.204 Several approaches can be
employed to reduce the likelihood of specimen collection error. For example, minimizing the
number of different types of POCT devices and the number of staff performing testing and
selecting a single manufacturer and model of device for hospital-wide use allow for using one
testing protocol and training program, resulting in less confusion for POCT operators who work
at several sites.205 Operators also need to be trained and revalidated. Several studies have
reported that non-laboratory professionals can obtain measurements that are just as accurate as
those obtained by laboratory professionals if properly trained in QA and device maintenance
prior to using the POCT device.219, 220
Automation in POCT devices can prompt the operator to enter specific information, ensuring that
every test follows the same sequence.205 Some POCT devices have functions that warn the
operator about common preanalytic errors and lock out untrained operators.

ANALYTIC PHASE
The relative importance of precision and accuracy in POCT depends on the site of testing. For
home-use POCT, precision is likely to be more important; while a POCT device may be biased,
the device is functional as long as the patient knows how to track and interpret results over time
and determine treatment based on the results generated by that device.221 In those circumstances,
absolute accuracy is not as important as the precision and daily consistency of results. Whether
they are evaluated in an emergency room, operating room, ICU, general medical unit, or
outpatient setting, the results of POCT must correlate closely to those generated by the central
laboratory via results verification, including delta checks, and interpretation.
An important POCT issue concerns the tradeoffs between rapid results and analytical test
performance.203 Several studies have assessed the analytic validity of POCT devices, primarily
comparing POCT results to those obtained in the central laboratory. Findings vary by the setting of
device use. For example, two recent studies confirmed the precision and accuracy of POCT glucose
meters using blood samples from patients attending outpatient clinics for routine checkups and
who were not suffering from underlying disease that required immediate hospitalization.222, 223
A study investigated the performance of POCT glucose meters in hospitalized patients with
serious underlying disorders in addition to their diabetes found that, at high and low glucose

May 2008

171

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

levels, there was significant disagreement between glucose meter readings and laboratory
analyzer readings. The authors concluded that clinicians should be cognizant of POCT meter
readings at the hypo- and hyperglycemic levels and corroborate these results with those obtained
by central laboratory analyzers whenever possible.224 Another study measured the accuracy and
clinical impact of three common POCT methods for glucose measurements in critically ill patients
receiving insulin infusions: glucose meter analysis of capillary blood (finger stick); glucose meter
analysis of arterial blood; and blood gas/chemistry analysis of arterial blood.225 All patients were
enrolled for a maximum of three days and had at most nine sets of measurements. Glucose meter
analysis of arterial and capillary blood tended to provide higher glucose values than blood
gas/chemistry analysis of arterial blood. The study reported that the magnitude of these
differences led to frequent clinical disagreements regarding insulin dose titration in insulin
infusion protocols for aggressive glucose control.

Specimen Analysis
Several factors can interfere with the operator’s ability to detect factors that can degrade POCT
test quality. Most POCT assays accept small sample volumes, in which evidence of hemolysis
(i.e., the breakdown of red blood cells) or clots can be difficult to detect. Some POCT devices now
contain a mechanism to detect the presence of clots or bubbles in the specimen.205 Common errors
also result from patient-related interference (e.g., non-specific agglutinins in precipitation slide
tests), specimen-related non-target influences (e.g., drugs that cause false results in chemistry
POCT devices), and specimen-reagent combination-related matrix effects.204 The design of most
POCT devices hides specimen reaction sites from view, making it difficult or impossible for the
operator to assess the progress of the reaction or to distinguish patient, specimen, and matrix
sources of error.
Another opportunity for error in the analytic phase of POCT arises from failure to calibrate a
POCT device, deviating from the calibration protocol, and misrecording calibration data.204
Similar to all laboratory instruments, calibration requirements for POCT devices differ depending
on such factors as how frequently the device is used and whether it is disposable after single use.
Among other requirements for non-waived POCT devices, the CAP 2006 checklist requires that
calibrators for POCT devices are properly labeled, calibration results are documented, criteria are
established for calibration verification, and test systems are recalibrated when calibration
verification fails to meet the established criteria.226 The CAP checklist for waived POCT requires
calibrators to be properly labeled and that POCT programs follow manufacturer instructions for
calibration and calibration verification.

Quality Control
Because central laboratory and POCT methods and techniques differ, traditional QC applied to
POCT can be costly and ineffective for identifying problems that can compromise the quality of
POCT results.227 It is difficult to apply conventional QC procedures to POCT for several reasons,
including: the number of different instruments, the diverse training and experience of POCT
operators, and the inability to view and assess the analytic reaction chamber of the devices.228
Some laboratory experts contend that QC is irrelevant and unnecessary in POCT because the
instruments are relatively error-free and the prevalence of preanalytical errors renders QC of the
analytical process useless.229 However, most experts believe that POCT must have a QA and QC

May 2008

172

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

program equivalent to those used in the central laboratory. The latter position is supported by the
findings of the CDC Pacific Northwest Monitoring Network study, which reported that 42% of
POCT waived tests were regarded as providing a definitive diagnosis without further
confirmation and 9% were used to monitor patients and whose results had the potential to
directly affect therapeutic interventions.230
The design of POCT devices, particularly those for single-use testing, has forced manufacturers to
assume responsibility for building QC into POCT products, a process often referred to as
“autonomation.”208, 229 Increasingly, if the instrument-controlled analytic process fails to meet the
manufacturer’s quality criteria, the patient’s data are not released, thereby allowing the
manufacturer to guarantee a statistically defined level of quality associated with each test result.208
Internal QC systems may encompass electronic checks to assess the performance of the
instrument’s electronic circuits, temperature, sample flow, electronic stability, and sensor
response.231 Built-in basic positive and negative controls (i.e., “test worked/test failed”) are
capable of assessing the viability of the analytic testing process, and more sophisticated,
quantitative measurements can provide numerical responses to the analyte concentrations in the
control. More advanced systems may include a series of integral liquid controls and calibrators
contained in closed reagent packs that have been validated by the manufacturer.208 Also, POCT
devices may automatically record QC data, producing charts and statistics and applying QC
algorithms to the data.231
In addition to these built-in QC functions, other forms of POCT QC are recommended. Specifically,
POCT operators should directly observe instrument or method function by performing and
recording results of QC testing each day.204 POCT supervisors should observe operators
performing and recording QC and quiz operators on various scenarios of QC failure. For both
operators and supervisors, checklists help to ensure that procedures are being followed.
Instrument or worksheet records can be compared to patient or maintenance records to identify
any inconsistencies in the transfer of information.
Laboratory experts agree that a QC check must be performed following the initiation of a new
batch of reagents and when the system is recalibrated.u,203 Many experts also suggest that QC be
performed when each sample is run and when each new operator uses the system. Methods to
determine the frequency of QC testing often are based on factors such as the overall analytic
performance and reproducibility of the system and the number and competence of the operators.
Typically, the frequency of QC depends on the type of POCT device. For bench top analyzers, QC
may be run at least once per shift (i.e., three times per day), whereas for critical care analyzers, QC
can be preprogrammed to occur at specific intervals.
Failure to perform QC has been cited as a common problem among facilities using POCT.233 CAP
Q-Probes studies demonstrated that POCT operators commonly neglect to perform QC and/or
report patient test results, even when QC procedures do not follow specified control guidelines.234
Several studies have examined optimal QC techniques for POCT used in a variety of medical
settings. A large university-based health system studied nearly 600 technicians and nurses who
had verified competency to perform POCT using two different devices to measure blood gas,
chemistry, and hematocrit levels. In this study, reported in 1999, investigators compared POCT
results and core laboratory results extracted from the LIS and from POCT data management
u

CLIA requires that operators of waived tests follow manufacturers’ QC directions.232

May 2008

173

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

stations. Discrepancies were noted and changes were implemented to improve POCT reliability
where problems were identified, including in-service training and provision of additional POCT
equipment. The investigators concluded that POCT processes could be monitored and corrected
using continuous quality improvement techniques.227
A similar study involved a formalized continuous quality improvement program in which all
testing sites in a large medical center were reviewed on a monthly basis for quality indicators
such as QC and maintenance performance, PT, patient identification, and alert value
confirmations.235 Major aspects of quality improvement requiring attention included
instrument maintenance documentation, QC documentation for manual tests, and
documentation of actions taken with the correction of failed QC tests. Significant improvement
was made in QC documentation of urine dipstick testing, including that the number of POCT
sites not documenting more than 5% of QC results was reduced from 30% to 15%.
Another facility’s QA program involved a database designed to incorporate five main
components:

ƒ

Documentation of initial device performance and reagent/control lots

ƒ

Documentation of operator competence and compliance with daily QC regulatory
requirementsv

ƒ

Storage of proficiency and patient correlation results

ƒ

Monitoring of performance and policy compliance

ƒ

Determination and documentation of the effect of POCT on patient outcome using
links between the POCT database, EHR, and LIS221

Drawing from a national survey and consensus process reported in 2001, a multidisciplinary
group of experts in critical care POCT programs and other hospital disciplines determined that
QC in POCT must involve a mechanism for blocking patient testing if required QC procedures
are not performed.217 These experts also recommended that requirements for QC and QC timing
be matched to clinical priorities, patient test results be suppressed if QC results are unacceptable,
exceptions to QC procedures be recognized for emergency situations, and that decisions be made
about when to implement routine, urgent, or critical interruptions to QC processes.

Result Generation and Verification
Because POCT data are generated in series, operators often are not afforded a real-time review of
trends and deviations in the sequence of test results that could enable identifying possible
errors.204 Whereas delta checks of routine and stat central laboratory testing tells whether a
patient’s laboratory test result differs significantly from previous results reported with the same
assay, POCT operators do not usually have automatic access to previous results or automated
statistical analysis, both of which are needed to perform delta checks.
Potential errors in POCT report generation also can occur when patient test results are outside the
device’s validated range.204 Organizations performing POCT must first ensure that the
v

By using QC conducted during routine POCT device use, this institution could avoid having to visually inspect
operators regularly and the additional testing and cost involved in routine inspections.

May 2008

174

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

manufacturer’s suggested reference interval range applies to their respective patient populations.236
Lack of QC, operator failure to recognize failures and problems in QC, and the absence or failure of
performance-control monitors also can lead to acceptance of invalid results.204 Linking the POCT
device to the hospital or laboratory information system allows for age-specific reference ranges,
physician alerts, and other warnings to be included with the test results.208
Verification of POCT results can be conducted using computer workstations; in this situation,
information flows from the POCT device to the data manager and awaits LIS verification.237
Expert decision-support functions in the LIS can autoverify the test results using predetermined
expert system rules that are consistent with pre-existing rules for results reporting.238, 239 While
few studies have examined autoverification of results received from POCT devices, the benefits of
autoverification have been assessed in central laboratory testing. Autoverification of results can
speed TAT, improve laboratory workflow, and ease the effect of shortages in laboratory
personnel; however, the autoverification process is not error-proof.240, 241 In order to prevent
autoverification of potentially invalid results, the LIS must be able to capture all error flags
generated by an instrument. While it raises other concerns, it is likely that autoverification of test
results received via POCT offers similar benefits.
Few studies have examined error during autoverification of results received via POCT devices. In
a study reported in 2005, a core laboratory located in a large health system analyzed data to
improve the rate of amended test results (i.e., correction of reported values after results
verification). Investigators reported that 34% of defects identified resulted from autoverification
errors made by the LIS.242 However, in a study reported in 2005, a large clinical chemistry and
urinalysis laboratory found that the number of inappropriately verified test results decreased
following implementation of results autoverification. In instances where results were
inappropriately autoverified or prevented from being autoverified, modification of exclusionary
setup rules eliminated the problem.241

POSTANALYTIC PHASE
Results Interpretation
Incorrect interpretation of POCT results is a source of error that can severely affect the quality of
patient care. The potential for results misinterpretation emphasizes the importance of training in
all phases of the testing process, including POCT results interpretation.205 It is vital for POCT
operators to be aware of the strengths, weaknesses, and limitations of POCT devices, particularly
where false positive results may arise from cross-reactivity with foods, medications, and/or
metabolites present in the specimen being tested.243, 244 The NACB’s laboratory medicine practice
guidelines recommend that organizations agree on and implement procedures that allow only
those individuals recognized as being competent to interpret POCT results to do so.245
Relatively few peer-reviewed scientific studies have examined the incidence of POCT errors that are
related to results misinterpretation. One study, reported in 2001, used simulated patient case
examples with results of blood glucose and urine dipsticks to assess the interpretive abilities of 250
nurses who routinely conduct point-of-care glucose testing. When the results simulated a
hypoglycemic patient, 84.1% of nurses correctly interpreted the results; 95.7% of nurses correctly
interpreted results simulating a patient with diabetes mellitus. However, only 5.4% of nurses

May 2008

175

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

correctly interpreted the POCT results of a simulation of a case in which the puncture site had
become contaminated, falsely raising the capillary blood glucose measurement that, if gone
unnoticed, could have led to inappropriate dosage of insulin and other potential consequences.205, 246
While this study reported no difference in test interpretation ability according to nurse seniority,
others have reported differences in interpretation of POCT results when experienced laboratory
personnel read the results versus when non-laboratory personnel perform the interpretation.247
As noted above, linkages between the POCT device and hospital and laboratory information
systems also have the potential to diminish errors in POCT results interpretation. Particularly in
critical care settings, a POCT operator who has access to the patient’s EHR and real-time
knowledge of the patient’s treatment regimen and other critical physiologic measurements (e.g.,
pH, PO2, and hematocrit levels) is more likely to avoid the adverse effects of drugs and
confounding variables on POCT device results.248, 249

Critical Value Reporting
The importance of timely and accurate critical value reporting for quality of care and medical
error prevention is receiving national recognition.250, 251 Reporting of critical values received via
POCT presents several opportunities for error. Among these, criticality of the results may not be
immediately recognized, criticality may not be noticed by the effective or designated clinical
decision maker; and critical results may not be documented sufficiently for subsequent
retrieval.204 These errors also can occur during reporting of non-critical POCT results.
In the national survey and consensus process noted above, a group of experts in POCT programs
in critical care and other hospital disciplines generated recommendations regarding critical, panic,
and alert value reporting and documentation in POCT situations.217 Specifically, a list of relevant
critical limits should be built into POCT devices and critical results should be stored in an easily
accessible manner and annotated. The individual obtaining or the clinician receiving the critical
results should be recorded, and verification of critical test results should be requested.
Improved techniques for critical values reporting have arisen from practical experience. As
described in a 2007 report, one emergency department at a tertiary care referral center with an
annual volume of more than 50,000 patients found that, after implementing arterial blood gas
testing at the point of care, critical values were not appropriately reported to physicians in 10 of the
first 664 samples run (1.5%).252 After adding a physician “pick list” as a mandatory field, this
problem was eliminated for future samples. Emphasis on establishing and improving linkages
between the POCT device and the laboratory computer system has played a major role in ensuring
that critical values derived from POCT are reported and marked appropriately. A detailed
discussion of connectivity can be found in the chapter on Laboratory Information Systems.

Report Formatting
Several types of error can occur during report formatting. Reports that lack units of measurement
or use inappropriate units of measurement can lead to harmful misinterpretation of results,
particularly when POCT is being used in critical care situations.204 Some POCT devices are not
connected to printers, and results are displayed on the device itself; in these cases, results can be
misperceived due to their appearance on a small screen.253 For printed results, POCT device
outputs that are unclear also are associated with errors.204
May 2008

176

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

Report Management
The error potential in the final postanalytic step, report management, has not been as widely
researched as results reporting. Errors that can occur during report management may result from
failure to check initially generated results against subsequently recorded results, leaving users
unaware of discrepancies between a POCT device’s test result and the patient record; failure to
record how clinical users acted upon POCT device results; and delayed recording of results,
thereby delaying clinical users’ awareness of potentially critical information.204

CONCLUSIONS
The TTP defines the preanalytic, analytic, and postanalytic phases of the laboratory testing cycle,
providing a systems-based framework for examining all possible interactions and activities that
can affect the quality of laboratory tests. This framework serves as the basis for designing and
implementing interventions, restrictions, or limits that can reduce or remove the likelihood of
errors that adversely affect testing and patient outcomes.

ƒ

Quality activities in laboratory medicine have historically focused on the analytic phase
of testing; however, available evidence demonstrates that a higher percentage of errors
occur in the pre- and postanalytic phases of testing. The distribution of errors varies
widely among institutions and settings. A review of several key studies found error
rates of 32-75% in the preanalytic phase, 13-32% in the analytic phase, and 9-31% in the
postanalytic phase.

ƒ

Poor communication between laboratorians and clinicians during test selection/ordering
and interpretation of laboratory findings is an important issue affecting the quality of
laboratory services. Although one out of four primary care physicians perceives that the
scope of care expected of them is beyond their current knowledge base, they reportedly
seek additional information when ordering tests only 30-50% of the time. Medical and
scientific advances, such as in genetic testing, will compound challenges associated with
ordering the optimal sequence of tests, correctly interpreting results, and incorporating
this information into clinical practice.

ƒ

While consultations for anatomic pathology are standard practice and reimbursed, this is
not always the case in clinical pathology. Yet, when provided, clinical pathology
interpretive consultations are well received by physicians. As reported in one study
described in this chapter, 98% of physicians found this information to be useful, 59%
perceived reduced time to diagnosis, 72% perceived that it reduced the number of tests
needed for diagnosis, and 72% believed consults helped to avoid misdiagnosis.

ƒ

The most common errors and reasons for specimen rejection during the preanalytic
phase are associated with patient and/or specimen misidentification and specimen
collection (e.g., insufficient volume, incorrect type of specimen, unusable specimen).
Other errors include missing or incorrect information on laboratory test order forms.
Among the strategies that are in place or evolving to reduce preanalytic errors are use of
barcoded labels for containers and slides, inpatient wristbands with accurate identifying
information, and CPOE.

May 2008

177

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

ƒ

Analytic errors in clinical pathology typically relate to transcription errors; failure to
reject an inadequate or damaged specimen; or bias often caused by interference,
instrument calibration error, lot variations, or lack of uniform test values across
manufacturers. Automated analyzers and results verification has decreased error rates in
clinical pathology substantially over the past decades. Errors that occur in the analytic
phase are often the result of errors that originated during preanalytic processes.

ƒ

In anatomic pathology, errors may occur during accessioning, in the gross room, or at
the microscope, and are classified as specimen/patient identification-related, cognitiverelated (e.g., inaccurate conclusions, poor descriptions, knowledge deficits), or crosscontamination-related. External, secondary consultation is common in anatomic
pathology; as summarized in one study, original diagnosis was confirmed in 70% of
consultations, but significant information was added in 16%.

ƒ

Core postanalytic challenges include improving TAT and notification of critical values.
Both are key quality measures of the testing process, but are frequently cited for ratings
of below-average to poor in customer satisfaction surveys. In clinical pathology, TAT
varies by setting, and delays are attributed to staff shortages in preanalytic and analytic
phases. In anatomic pathology, TAT varies by specimen type, with delays resulting
from difficulty in reaching the clinician for additional information.

ƒ

Better integration of laboratory automation and LIS could enable laboratories to identify
and diminish error rates in the TTP. This will require much improved communication
within and among health care institutions, including, but not limited to systematic
provisions for appropriate, timely communication between laboratorians and clinicians.

ƒ

Inappropriate test use―including overuse and underuse―can compromise case
management, result in adverse health outcomes, and increase health care costs. It arises
in tests for screening, diagnosis, and monitoring, and pertains to multiple types of
cancer, cardiovascular disease, diabetes, and other prevalent conditions with high
clinical and economic burdens and whose course can be affected by proper testing.
Sentinel studies in recent years continue to identify and call public attention to
inappropriate testing and its contribution to national shortfalls in quality of care.
Principles of appropriateness in laboratory medicine are embodied in selecting the right
test at the right time for the right patient. Multiple factors lead to inappropriate test use,
including test panels that contain unnecessary tests, poorly designed ordering forms,
delays in performing tests, failure to use earlier test results, financial incentives, and
malpractice concerns. While evidence-based practice guidelines are helping to reduce
inappropriate use, continued progress will require greater attention to how elements
across the TTP mediate test appropriateness.

Gaps, Needs, and Challenges:
ƒ

The day-to-day demands of clinical practice leave physicians with little time to acquire
knowledge of new laboratory tests. Furthermore, the average medical student receives
10 weeks or 70 hours of didactic coursework in medical genetics.

ƒ

While CPOE has been shown to reduce the frequency of medication order errors, no
research has examined the effect of CPOE on correctness of laboratory test orders. As
CPOE becomes more prevalent, this effect will need to be better understood.

May 2008

178

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

ƒ

Thirty eight states currently allow individuals to directly request that certain laboratory
tests be performed on their own blood or urine samples (some in limited capacity) and
its popularity is increasing; however, little data exists on the frequency, appropriateness,
and quality related to such orders.

ƒ

Lack of uniformity and standardization of clinical pathology test values among
manufacturers hinders implementation of laboratory-based guidelines, which require
method-dependent decision limits. Heterogeneity of test values also makes it difficult for
clinicians to work in an integrated health system using more than one testing method.

ƒ

The primary barriers to interpretive consultations in clinical pathology reports are lack of
reimbursement for such consults and the shortage of true experts with high subspecialty
expertise in coagulation, autoimmunity, and other complex areas.

ƒ

QC, performance evaluation, and test reproducibility standards to minimize diagnostic
discrepancies and errors have been better defined and applied in clinical pathology than
in anatomic pathology. Efforts should be undertaken to develop such measures for
anatomic pathology.

ƒ

The growth of innovative laboratory testing techniques is prompting changes to
laboratory report formats. Laboratory reports will need to make better use of graphical
displays to facilitate rapid assimilation and comprehension of important data by
clinicians, other laboratory professionals, and patients. Standardization of data elements
and report formats for laboratory tests is necessary to improve physician comprehension
and use of results as well as integrate report data into clinical practice IT applications.

ƒ

While the immediate availability of POCT results has the potential to significantly
enhance the quality of care, additional research is needed to identify the best methods
for integrating POCT into daily clinical processes. Operators of POCT devices must be
appropriately trained in testing practices, particularly QC. In addition, methods to
improve the accuracy of POCT results relative to those produced in the central
laboratory warrants research.

May 2008

179

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

REFERENCE LIST
1. Martin ML, Wagner WM. Improving the quality of health management through clinician
and laboratorian teamwork: the concept and the practice of quality. 1989 Institute on
Critical Issues in Health Laboratory Practice. Atlanta, GA: Centers for Disease Control and
Prevention, 1989.
2. Lundberg GD. How clinicians should use the diagnostic laboratory in a changing medical
world. Clin Chim Acta 1999;280:3-11.
3. Institute of Medicine. Medicare laboratory payment policy: now and in the future.
Washington, DC: National Academy Press, 2000.
4. Travers EM, Krochmal CF. A new way to determine test cost per instrument: part I. MLO
Med Lab Obs 1988;20(10):24-9.
5. Bonini P, Plebani M, Ceriotti F, Rubboli F. Errors in laboratory medicine. Clin Chem
2002;48(5):691-8.
6. Goldschmidt HMJ, Lent RW. Gross errors and work flow analysis in the clinical
laboratory. Klin Biochem Metab 1995;3:131-40.
7. Boone DJ, Steindel SD, Herron R, et al. Transfusion monitoring practices. A study of the
College of American Pathologists/Centers for Disease Control and Prevention Outcomes
Working Group. Arch Pathol Lab Med 1995;119:999-1006.
8. Ruby SG. Clinician interpretation of pathology reports: confusion or comprehension?
Arch Pathol Lab Med 2000;124:943-4.
9. Kratz A, Laposata M. Enhanced clinical consulting--moving toward the core competencies
of laboratory professionals. Clinica Chimica Acta 2002;319:117-25.
10. Laposata ME, Laposata M, Van Cott EM, Buchner DS, Kashalo MS, Dighe AS. Physician
survey of a laboratory medicine interpretive service and evaluation of the influence of
interpretations on laboratory test ordering. Arch Pathol Lab Med 2004;128:1424-7.
11. The laboratory and patient safety: a critical role. Joint Commission Perspectives on Patient
Safety 2004;4(7).
12. Zarbo RJ, Nakhleh RE, Walsh M. Customer satisfaction in anatomic pathology: a College
of American Pathologists Q-Probes study of 3065 physician surveys from 94 laboratories.
Arch Pathol Lab Med 2003;127:23-9.
13. Leonard M, Graham S, Bonacum D. The human factor: the critical importance of effective
teamwork and communication in providing safe care. Quality and Safety in Health Care
2004;13((Supplement 1)):i85-i90.
14. Institute of Medicine. Crossing the quality chasm: a new health system for the 21st
century. Washington, DC: National Academy Press, 2001.
15. Institute of Medicine. To err is human: building a safer health system. Washington, DC:
National Academy Press, 2000.
16. van Walraven C, Naylor CD. Do we know what inappropriate laboratory utilization is? A
systematic review of laboratory clinical audits. JAMA 1998;280:550-8.

May 2008

180

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

17. Hollensead SA, Lockwood WB, Elin RJ. Errors in pathology and laboratory medicine:
consequences and prevention. J Surg Oncol 2004;88:161-81.
18. Klein RD, Kant JA. Opportunity knocks: the pathologist as laboratory genetics consultant.
Arch Pathol Lab Med 2006;130:1603-4.
19. St.Peter RF, Reed MC, Kemper P, Blumenthal D. Changes in the scope of care provided by
primary care physicians. N Engl J Med 1999;341(26):1980-5.
20. Greendale K, Pyeritz RE. Empowering primary care health professionals in medical
genetics: how soon? How fast? How far? Am J Med Genet 2001;106:223-32.
21. Green ML, Ciampi MA, Ellis PJ. Residents' medical information needs in clinic: are they
being met? Am J Med 2000;109:218-23.
22. Standards for laboratory accreditation. Northfield, IL: College of American Pathologists,
1996.
23. McGlynn EA, Asch SM, Adams J, et al. The quality of health care delivered to adults in the
United States. N Engl J Med 2003;348(26):2635-45.
24. Standards of medical care in diabetes. Diabetes Care 2005;28:S4-S36.
25. Smith R, Cokkinides V, Eyre H. American Cancer Society guidelines for the early
detection of cancer. CA Cancer Journal of Clinicians 2003;53:27-43.
26. Ladenson P, Singer P, Ain K, Bagchi N, Levy E, et al. American Thyroid Association
guidelines for detection of thyroid dysfunction. Arch Intern Med 2000;160:1573-5.
27. Guide to clinical preventive services, 2007. Recommendations of the U.S. Preventive
Services Task Force. Rockville, MD: Agency for Healthcare Research and Quality, 2007.
www.ahrq.gov/clinic/pocketgd.htm.
28. 2006 National Healthcare Quality Report. Report No. 07-0013. Rockville, MD: Agency for
Healthcare Research and Quality, 2006.
29. Sirovich BE, Welch HG. Cervical cancer screening among women without a cervix. JAMA
2004;291:2990-3.
30. Kaplan E, Sheiner L, Boeckmann A, et al. The usefulness of preoperative laboratory
screening. JAMA 1985;253:3576-81.
31. Schein O, Katz J, Bass E, et al. The value of routine preoperative medical testing before
cataract surgery. N Engl J Med 2000;342:168-75.
32. Grant RW, Buse JB, Meigs JB. Quality of diabetes care in U.S. academic medical centers.
Diabetes Care 2005;28:337-442.
33. Hindmarsh JT, Lyon AW. Strategies to promote rational clinical chemistry test utilization.
Clin Biochem 1996;29(4):291-9.
34. Tampoia M, Fontana A, Di Serio F, Maggiolini P, Pansini N. Application of a diagnostic
algorithm in autoantibody testing: assessment of clinical effectiveness and economic
efficiency. Clinica Chimica Acta 2003;333:181-3.
35. Crump J, Corder J, Henshaw N, Reller L. Development, implementation, and impact of
acceptability criteria for serologic tests for infectious diseases. J Clin Microbiol 2004;42:881-3.

May 2008

181

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

36. Ozbeck O, Oktem M, Dogan G, Abacioglu Y. Application of hepatitis serology testing
algorithms to assess inappropriate laboratory utilization. J Eval Clin Pract 2004;10:519-23.
37. Valenstein P, Schifman RB. Duplicate laboratory orders: a College of American
Pathologists Q-Probes study of thyrotropin requests in 502 institutions. Arch Pathol Lab
Med 1996;120:917-21.
38. Bates DW, Kuperman G, Rittenberg E, et al. A randomized trial of a computer-based
intervention to reduce utilization of redundant laboratory tests. Am J Med 1999;106:144-50.
39. Solomon DH, Hashimoto H, Daltroy L, Liang MH. Techniques to improve physicians' use
of diagnostic tests: a new conceptual framework. JAMA 1998;280(23):2020-7.
40. Daucort V, Saillour-Glénisson F, Michel P, Jutand M, Abouelfath A. A multicenter cluster
randomized controlled trial of strategies to improve thyroid function testing. Med Care
2003;41:432-41.
41. van Walraven C, Goel V, Chan B. Effect of population-based interventions on laboratory
utilization: a time-series analysis. JAMA 1998;280:2028-33.
42. Neilson E, Johnson K, Rosenbloom S, et al. The impact of peer management on testordering behavior. Ann Intern Med 2004;141:196-204.
43. Valenstein P, Meier F. Outpatient order accuracy: a College of American Pathologists QProbes study of requisition order entry accuracy in 660 institutions. Arch Pathol Lab Med
1999;123:1145-50.
44. Valenstein PN, Howanitz PJ. Ordering accuracy: a College of American Pathologists QProbes study of 577 institutions. Arch Pathol Lab Med 1995;119:117-22.
45. Nakhleh RE, Zarbo RJ. Surgical pathology specimen identification and accessioning: a
College of American Pathologists Q-Probes study of 1,004,115 cases from 417 institutions.
Arch Pathol Lab Med 1996;120:227-33.
46. Lechleitner G, Pfeiffer KP, Wilhelmy I, Ball M. Cerner millennium: the Innsbruck
experience. Methods of Informatics in Medicine 2003;42:8-15.
47. Health Smart Strategy for the modernisation and replacement of information technology.
Victorian Government Department of Human Resources, 2004. (Accessed December 19,
2007, at
http://www.health.vic.gov.au/healthsmart/documents/healthsmart-strategy.pdf.)
48. Fact sheet: computerized physician order entry (CPOE). The Leapfrog Group, 2007.
(Accessed December 19, 2007, at http://leapfroggroup.org/fact_sheet.pdf.)
49. Kuperman GJ, Bobb A, Payne TH, et al. Medication-related clinical decision support in
computerized provider order entry systems: a review. J Am Med Inform Assoc
2007;14(1):29-40.
50. Georgiou A, Williamson M, Westbrook JI, Ray S. The impact of computerized physician
order entry systems on pathology services: a systematic review. Int J Med Inf 2007;76:514-29.
51. Survey of direct-to-consumer testing statutes and regulations. Washington, DC: Genetics
and Public Policy Center, 2007. (Accessed February 12, 2008, at
http://www.dnapolicy.org/resources/DTCStateLawChart.pdf.)

May 2008

182

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

52. Merlin T. Direct access testing: introduction and overview. Clinical Laboratory
Improvement Amendment Committee meeting. March 12, 2003. Atlanta, GA: Centers for
Disease Control and Prevention, 2003.
53. Wilkinson D, Pontius C. Consumer direct access to clinical laboratory testing: what are the
critical issues? Clinical Leadership and Management Review 2003;17:358-60.
54. Young D. Effects of preanalytical variables on clinical laboratory tests. 2nd ed.
Washington, DC: American Association for Clinical Chemistry, 1997.
55. Bock JL. Quality of the specimen and the analytical system. In: Clinical diagnostic
technology: the total testing process. Volume 2: the analytical phase. Ward-Cook K,
Lehman CA, Schoeff LE, Williams RH, ed. Washington, DC: American Association of
Clinical Chemistry, 2003.
56. Howanitz PJ, Steindel SJ. Digoxin therapeutic drug monitoring practices. A College of
American Pathologists Q-Probes study of 666 institutions and 18,679 toxic levels. Arch
Pathol Lab Med 1993;117:684-90.
57. Zarbo RJ, Jones BA, Friedberg RC, et al. Q-Tracks: a College of American Pathologists
program of continuous laboratory monitoring and longitudinal tracking. Arch Pathol Lab
Med 2002;126(9):1036-44.
58. Chan JC, Chu RW, Young BW, et al. Use of an electronic barcode system for patient
identification during blood transfusion: 3-year experience in a regional hospital. Hong
Kong Medical Journal 2004;10(3):166-71.
59. Howanitz P, Renner S, Walsh M. Continuous wristband monitoring over 2 years decreases
identification errors: a college of American Pathologists Q-Tracks study. Arch Pathol Lab
Med 2002;126:809-15.
60. Bologna L, Mutter M. Life after phlebotomy deployment: reducing major patient and
specimen identification errors. J Healthc Inf Manag 2002;16:65-70.
61. Patterson E, Cook R, Render M. Improving patient safety by identifying side effects from
introducing bar coding in medication administration. J Am Med Inform Assoc 2002;9:540-53.
62. Valenstein PN, Sirota RL. Identification errors in pathology and laboratory medicine. Clin
Lab Med 2004;24:979-96.
63.

Mastectomy mistake fuels debate. CBS Evening News Early Show. January 21, 2003.

64. Sazama K. Reports of 355 transfusion-associated deaths: 1976 through 1985. Transfusion
1990;30:583-90.
65. Dale J, Novis D. Outpatient phlebotomy success and reasons for specimen rejection: a QProbes study. Arch Pathol Lab Med 2002;126:416-9.
66. Valenstein PN, Raab SS, Walsh MK. Identification errors involving clinical laboratories: a
College of American Pathologists Q-Probes study of patient and specimen identification
errors at 120 institutions. Arch Pathol Lab Med 2006;130:1106-13.
67. Linden J, Wagner K, Vyotovich A, Sheehan J. Transfusion errors in New York State: an
analysis of 10 years experience. Transfusion 2000;40:1207-13.

May 2008

183

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

68. Dzik W, Murphy M, Andreu G, et al. An international study of the performance of sample
collection from patients. Vox Sang 2003;85:40-7.
69. Zarbo RJ, Meier FA, Raab SS. Error detection in anatomic pathology. Arch Pathol Lab Med
2005;129:1237-45.
70. Dzik W. Emily Cooley lecture 2002: transfusion safety in the hospital. Transfusion
2003;43:1190-9.
71. Turner C, Casvbard A, Murphy M. Barcode technology: its role in increasing the safety of
blood transfusions. Transfusion 2003;43:1200-9.
72. Marconi M, Langeberg A, Sirchia G, Sandler S. Improving transfusion safety by electronic
identification of patient, blood samples, and blood units. Immunohematology 2000;16:82-5.
73. Dzik, Corwin H, Goodnough L, et al. Patient safety and blood transfusion: new solutions.
Transfus Med Rev 2003;17:169-80.
74. Miller JJ. Specimen collection, handling, preparation, and storage. In: Clinical diagnostic
technology: the total testing process. Volume 1: the preanalytical phase. Washington, DC:
American Association of Clinical Chemistry, 2003.
75. Howanitz PJ, Schifman RB. Inpatient phlebotomy practices: a College of American
Pathologists Q-Probes quality management study of 2,351,643 phlebotomy requests. Arch
Pathol Lab Med 1994;118:601-5.
76. Plebani M, Carraro P. Mistakes in a stat laboratory: types and frequency. Clin Chem
1997;43(8):1348-51.
77. Davey D, Woodhouse S, Styer P, Stastny J, Mody D. Atypical epithelial cells and specimen
adequacy--current laboratory practices of participants in the College of American
Pathologists interlaboratory comparison program in cervicovaginal cytology. Arch Pathol
Lab Med 2000;124:203-11.
78. Jones BA, Meier F, Howanitz PJ. Complete blood count specimen acceptability. A College
of American Pathologists Q-Probes study of 703 laboratories. Arch Pathol Lab Med
1995;119:203-8.
79. Jones BA, Calam RR, Howanitz PJ. Chemistry specimen acceptability: a College of
American Pathologists Q-Probes study of 453 laboratories. Arch Pathol Lab Med 1997;
121:19-26.
80. Dale J, Pruett S. Phlebotomy--a minimalist approach. Mayo Clin Proc 1993;68:249-55.
81. Dale J, Ruby S. Specimen collection volumes for laboratory tests: a College of American
Pathologists study of 140 laboratories. Arch Pathol Lab Med 2003;127:162-8.
82. Wisser D, van Ackern K, Knoll E, Wisser H, Bertsch T. Blood loss from laboratory tests.
Clin Chem 2003;49:1651-5.
83. Foulke GE, Harlow DJ. Effective measures for reducing blood loss from diagnostic
laboratory tests in intensive care unit patients. Crit Care Med 1989;17:1143-5.
84. Hicks J. Excessive blood drawing for laboratory tests. N Engl J Med 1999;340:1690.

May 2008

184

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

85. Schifman RB, Strand CL, Meier FA, et al. Blood culture contamination. Arch Pathol Lab
Med 2007;112:216-21.
86. Valenstein P, Meier F. Urine culture contamination. Arch Pathol Lab Med 2007;122:123-9.
87. Bates D, Goldman L, Lee T. Contaminated blood cultures and resource utilization: the true
consequences of false-positive results. JAMA 1991;265:365-9.
88. Bekeris L, Tworek J, Walsh M, Valenstein P. Trends in blood culture contamination: a
College of American Pathologists Q-Tracks study of 356 institutions. Arch Pathol Lab Med
2005;129:1222-5.
89. Norberg A, Christopher N, Ramundo M, Bower J, Berman S. Contamination rates of blood
cultures obtained by dedicated phlebotomy versus intravenous catheter. JAMA
2003;289:726-9.
90. Gibb A, Hill B, Chorel B, Brant R. Reduction in blood culture contamination rate by
feedback to phlebotomists. Arch Pathol Lab Med 1997;121:503-7.
91. Shafazand S, Weinacker A. Blood cultures in the critical care unit--improving utilization
and yield. Chest 2002;122:1727-36.
92. Schifman RB, Meier F. Sputum specimen adequacy data analysis and critique. Northfield,
IL: College of American Pathologists, 1991.
93. Bartlett RC, Mazens-Sullivan M, Tetreault JZ, Lobel S, Nivard J. Evolving approaches to
management of quality in clinical microbiology. Clin Microbiol Rev 1994;7(1):55-88.
94. Peyton CL, Schiffman M, Lorincz AT, et al. Comparison of PCR- and hybrid capture-based
human papillomavirus detection systems using multiple cervical specimen collection
strategies. J Clin Microbiol 1998;36(11):3248-54.
95. Kaye SB, Rao PG, Smith G, et al. Simplifying collection of corneal specimens in cases of
suspected bacterial keratitis. J Clin Microbiol 2003;41(7):3192-7.
96. Stark A, Jones BA, Chapman D, et al. Clinical laboratory specimen rejection--association
with the site of patient care and patients' characteristics. Arch Pathol Lab Med
2007;131:588-92.
97. Lillie-Blanton M, Martinez RM, Salganicoff A. Site of medical care: do racial and ethnic
differences persist. Yale Journal of Health Policy, Law, and Ethics 2001;1:15-32.
98. Boyd JC, Felder RA. Preanalytical automation in the clinical laboratory. In: Clinical
diagnostic technology: the total testing process. Volume 1: the preanalytical phase. WardCook KM, Lehman CA, Schoeff LE, Williams RH, ed. Washington, DC: American
Association of Clinical Chemistry, 2003.
99. Fleisher M, Schwartz MK. Automated approaches to rapid-response testing. a
comparative evaluation of point-of-care and centralized laboratory testing. Am J Clin
Pathol 1995;104((4 Supplement 1)):S18-S25.
100. Arzoumanian L. Hemolysis. Tech Talk 2003;2(2):1-2.
101. Kratz A, Salem RO, Van Cott EM. Effects of a pneumatic tube system on routine and novel
hematology and coagulation parameters in healthy volunteers. Arch Pathol Lab Med
2007;131:293-6.

May 2008

185

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

102. Hindiyeh M, Acevedo V, Carroll KC. Comparison of three transport systems (Starplex
StarSwab II, the new Copan Vi-Pak Amies Agar Gel Collection and Transport Swabs, and
BBL Port-A-Cul) for maintenance of anaerobic and fastidious aerobic organisms. Journal
of Microbiology 2001;39(1):377-80.
103. Murphy JM, Browne RW, Hill L, et al. Effects of transportation and delay in processing on
the stability of nutritional and metabolic biomarkers. Nutr Cancer 2000;37(2):155-60.
104. Khoury M, Burnett L, Mackay MA. Error rates in Australian chemical pathology
laboratories. Med J Aust 1996;165:128-30.
105. Taswell HF, Galbreath JL, Harmsen WS. Errors in transfusion medicine: detection,
analysis, frequency, and prevention. Arch Pathol Lab Med 1994;118:335-464.
106. Black R, Woolman P, Kinsella J. Variation in the transcription of laboratory data in an
intensive care unit. Anaesthesia 2004;59(8):767-9.
107. Plebani M. Errors in clinical laboratories or errors in laboratory medicine? Clin Chem Lab
Med 2006;44(6):750-9.
108. Washington JA. Collection, transport, and processing of blood cultures. Clin Lab Med
1994;14:59-67.
109. Plebani M, Ceriotti F, Messeri G, Ottomano C, Pansini N, Bonini P. Laboratory network of
excellence: enhancing patient safety and service effectiveness. Clin Chem 2006;44(2):150-60.
110. Wolk D, Mitchell S, Patel R. Principles of molecular microbiology testing methods. Infect
Dis Clin North Am 2001;15(4):1157-204.
111. Chong S, Jang D, Song X, et al. Specimen processing and concentration of Chlamydia
trachomatis added can influence false-negative rates in the LCx Assay but not in the
APTIMA Combo 2 assay when testing for inhibitors. J Clin Microbiol 2002;41(2):778-82.
112. Auchinleck GF, Lines RB, Godolphin WJ. Cost-effectiveness and benefits of automation
and robotics. In: Clinical automation, robotics, and optimization. Kost GJ, ed. New York:
John Wiley and Sons, 1996.
113. Holman JW, Mifflin TE, Felder RA, Demers LM. Evaluation of an automated preanalytic
robotic workstation at two academic health centers. Clin Chem 2007;48(3):540-8.
114. Orsulak PJ. Stand-alone automated solutions can enhance laboratory operations. Clin
Chem 2000;46(5):778-83.
115. Houwen B. Make no mistake. Advance/Laboratory 2003;12:32-43.
116. Okorodudu AO. Electrochemistry. In: Clinical diagnostic technology: the total testing
process. Volume 2: the analytical phase. Ward-Cook KM, Lehman CA, Schoef LE,
Williams RH, ed. Washington, DC: American Association of Clinical Chemistry, 2005.
117. Ward-Cook KM, Lehmann CA, Schoeff LE, Williams RH. Clinical diagnostic technology:
the total testing process. Volume 2: the analytical phase. Washington, DC: American
Association of Clinical Chemistry, 2005.
118. Taylor CR. The total test approach to standardization of immunohistochemistry. Arch
Pathol Lab Med 2000;124:945-51.

May 2008

186

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

119. Howanitz P. From start to finish, how accurate are lab tests? CAP Today 1994:41-2.
120. Steele BW, Wang E, Palomaki G, Klee GG, Elin RJ, Witte DL. Sources of variability: a
College of American Pathologists therapeutic drug monitoring survey study. Arch Pathol
Lab Med 2001;125:183-90.
121. Klee GG. Analytic performance goals based on direct effect on analytic bias on medical
classification decisions. 1995 Institute on Critical Issues in Health Laboratory Practice:
Frontiers in Laboratory Practice Research. Atlanta, GA: Centers for Disease Control and
Prevention, 1995.
122. Ross JW, Miller WG, Myers GL, Praestgaard J. The accuracy of laboratory measurements
in clinical chemistry: a study of 11 routine chemistry analytes in the College of American
Pathologists Chemistry Survey with fresh frozen serum, definitive methods, and reference
methods. Arch Pathol Lab Med 1998;122(7):587-608.
123. Klee GG. Maximizing efficacy of endocrine tests: importance of decision-focused testing
strategies and appropriate patient preparation. Clin Chem 1999;45(8(B)):1323-30.
124. Jenny RW, Jackson-Tarentino KY. Causes of unsatisfactory performance in proficiency
testing. Clin Chem 2000;46(1):89-99.
125. Miller JJ, Levinson SS, Elin RJ. Interferences in laboratory tests. In: Clinical diagnostic
technology: the total testing process. Volume 2: the analytical phase. Washington, DC:
American Association of Clinical Chemistry, 2003.
126. Ismail AA, Walker PL, Barth JH, Lewandowski KC, Jones R, Burr WA. Wrong
biochemistry results: two case reports and observational study in 5310 patients on
potentially misleading thyroid-stimulating hormone and gonadotropin immunoassay
results. Clin Chem 2002;48(11):2023-9.
127. The impact of calibration error in medical decision making (planning report 04-1).
Gaithersburg, MD: National Institute of Standards and Technology, 2004.
128. Hinckley CM. Defining the best quality control systems by design and inspection. Clin
Chem 1997;46(1):89-99.
129. Steele BW, Witte DL, Whitley RJ, Klee GG, Chan DW. The effects of modifying proficiency
testing materials on thyroid function test results: a College of American Pathologists
ligand assay survey study. Arch Pathol Lab Med 1997;121:1241-6.
130. Ledue TB, Rifai N. Preanalytic and analytic sources of variations in C-reactive protein
measurement: implications for cardiovascular disease risk assessment. Clin Chem
2003;49(8):1258-71.
131. Cooper SD. The role of the laboratory information system in diagnostic services. In:
Clinical diagnostic technology: the total testing process. Volume 1: the preanalytical phase.
Ward-Cook KM, Lehman CA, Schoeff LE, Williams RH, ed. Washington, DC: American
Association of Clinical Chemistry, 2003.
132. Picco W, Pintor LP, Baruzzo G, Pistono O, Triumbari F, Pintor PP. Quality control of
laboratory results in real time. Int J Qual Health Care 1992;4:61-7.
133. Boran G, Given P, O'Moore R. Patient result validation services. Comput Methods
Programs Biomed 1996;50(2):161-8.

May 2008

187

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

134. Sheiner LB, Wheeler LA, Moore JK. The performance of delta check methods. Clin Chem
1979;25(12):2034-7.
135. Dorizzi RM, Caruso, B, Meneghelli S, Rizzotti P. The DNSev expert system in the
autoverification of tumor markers and hormone results. Accreditation and Quality
Assurance: Journal for Quality, Comparability and Reliability in Chemical Measurement
2006;11(6):303-7.
136. Pearlman ES, Bilello L, Stauffer J, Kamarinos A, Miele R, Wolfert MS. Implications of
autoverification for the clinical laboratory. Clinical Leadership and Management Review
2004;16(4):237-9.
137. Seaborg E. Reference ranges and what they mean. Washington, DC: Lab Tests Online,
2007. (Accessed December 10, 2007, at
http://www.labtestsonline.org/understanding/features/ref_ranges.html.)
138. Friedberg RC, Souers R, Wagar EA, Stankovic AK, Valenstein PN. The origin of reference
intervals. Arch Pathol Lab Med 2007;131(3):348-57.
139. How to define and determine reference intervals in the clinical laboratory. Approved
guideline: C28-A2; second edition. Wayne, PA: Clinical Laboratory Standards Institute,
2000.
140. Klee GG. Clinical interpretation of reference intervals and reference limits. A plea for
assay harmonization. Clin Chem Lab Med 2004;42(7):752-7.
141. Laboratory requirements. Code of Federal Regulations, Title 42, Section 493.1253. Subpart K-quality systems for nonwaived testing; standard: establishment and verification of
performance specifications.
142. Laboratory requirements. Code of Federal Regulations, Title 42, Section 493.1291. Subpart K-quality systems for nonwaived testing; standard: test report.
143. Pritt BS, Waters BL. Cutting injuries in an anatomic pathology department. Arch Pathol
Lab Med 2005;129:1022-6.
144. Sirota RL. Error and error reduction in pathology. Arch Pathol Lab Med 2005;129:1228-33.
145. Gephardt GN, Zarbo RJ. Extraneous tissue in surgical pathology: a College of American
Pathologists Q-Probes study of 275 laboratories. Arch Pathol Lab Med 1996;120:1009-14.
146. Coleman DV. Evaluation of automated systems for the analysis of cervical smears.
Cytopathology 1998;9(6):359-68.
147. Leong FJ, Leong AS. Digital imaging applications in anatomic pathology. Adv Anat Pathol
2003;10(2):88-95.
148. Hasegawa T, Yamamoto S, Yokoyama R, Umeda T, Matsuno Y, Hirohashi S. Prognostic
significance of grading and staging systems using MIB-1 score in adult patients with soft
tissue sarcoma of the extremities and trunk. Cancer 2002;95:843-51.
149. McLendon RE. Errors in surgical neuropathology and the influence of cognitive bias. The
psychology of intelligence analysis. Arch Pathol Lab Med 2006;130:613-6.
150. Fletcher CD, Fletcher M. Symposium on errors, error reduction, and critical values in
anatomic pathology. Arch Pathol Lab Med 2006;130:602-3.

May 2008

188

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

151. Bruner JM, Inouye L, Fuller GN, Langford LA. Diagnostic discrepancies and their clinical
impact in a neuropathology referral practice. Cancer 1997;79:796-803.
152. Raab SS. Improving patient safety by examining pathology errors. Clin Lab Med
2004;24:849-63.
153. Foucar E. Error in anatomic pathology. Am J Clin Pathol 2001;116:S34-S46.
154. Travers H. Quality assurance indicators in anatomic pathology. Arch Pathol Lab Med
1990;114:1149-56.
155. Azam M, Nakhleh RE. Surgical pathology extradepartmental consultation practices. Arch
Pathol Lab Med 2002;126:405-12.
156. Raab SS, Nakhleh RE, Ruby SG. Patient safety in anatomic pathology: measuring
discrepancy frequencies and causes. Arch Pathol Lab Med 2005;129:459-66.
157. Clary KM, Silverman JF, Liu Y, et al. Cytohistologic discrepancies: a means to improve
pathology practice and patient outcomes. Am J Clin Pathol 2002;117(4):567-73.
158. Zarbo RJ, Hoffman GC, Howanitz PJ. Interinstitutional comparison of frozen-section
consultation: a College of American Pathologists Q-Probe study of 79,647 consultations in
297 North American institutions. Arch Pathol Lab Med 1991;115:1187-94.
159. Gephardt GN, Zarbo RJ. Interinstitutional comparison of frozen-section consultations: a
College of American Pathologists Q-Probes study of 90,538 cases in 461 institutions. Arch
Pathol Lab Med 1996;120:804.
160. Nakhleh RE, Zarbo RJ. Amended reports in surgical pathology and implications for
diagnostic error detection and avoidance: a College of American Pathologists Q-Probes
study of 1,667,547 accessioned cases in 359 laboratories. Arch Pathol Lab Med
1998;122:303-9.
161. Standards for pathology and clinical laboratory services. Oakbrook Terrace, IL: The Joint
Commission on Accreditation of Healthcare Organizations, 1998.
162. Novis DA, Walsh MK, Dale JC, Howanitz PJ. Continuous monitoring of stat and routine
outlier turnaround times: two College of American Pathologists Q-Tracks monitors in 291
hospitals. Arch Pathol Lab Med 2004;128:621-6.
163. Valenstein P. Laboratory turnaround time. Am J Clin Pathol 1996;105:676-88.
164. Steindel SJ, Novis DA. Using outlier events to monitor test turnaround time: a College of
American Pathologists Q-Probes study in 496 laboratories. Arch Pathol Lab Med
1999;123:607-14.
165. Kilgore ML, Steindel SJ, Smith JA. Evaluating stat testing options in an academic health
center: therapeutic turnaround time and staff satisfaction. Clin Chem 1998;44(8 Pt 1):
1597-603.
166. Howanitz PJ. Errors in laboratory medicine: practical lessons to improve patient safety.
Arch Pathol Lab Med 2005;129:1252-61.
167. Dale JC, Steindel SJ, Walsh M. Early morning blood collections: a College of American
Pathologists Q-Probes study of 657 institutions. Arch Pathol Lab Med 1998;122:865-70.

May 2008

189

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

168. Steindel SJ, Jones BA. Routine outpatient test turnaround time 97-03: data analysis and
critique. A Q-Probes study of the College of American Pathologists. Northfield, IL:
College of American Pathologists, 1997.
169. Steindel SJ, Jones BA. Routine outpatient laboratory test turnaround times and practice
patterns: a College of American Pathologists Q-Probes study. Arch Pathol Lab Med
2002;126(1):11-8.
170. Jones BA, Valenstein PN, Steindel SJ. Gynecologic cytology turnaround time: a College of
American Pathologists Q-Probes study of 371 laboratories. Arch Pathol Lab Med
1999;123:682-6.
171. Jones BA, Novis DA. Nongynecologic cytology turnaround time: a College of American
Pathologists Q-Probes study of 180 laboratories. Arch Pathol Lab Med 2001;125:1279-84.
172. Zarbo RJ, Gephardt GN, Howanitz PJ. Intralaboratory timeliness of surgical pathology
reports: results of two College of American Pathologists Q-probes studies of biopsies and
complex specimens. Arch Pathol Lab Med 1996;120:234-44.
173. Lundberg GD. When to panic over abnormal values. MLO Med Lab Obs 1972;4:47-54.
174. Wagar EA, Stankovic AK, Wilkinson DS. Assessment of monitoring of laboratory critical
values: a College of American Pathologists Q-Tracks study of 180 institutions. Arch Pathol
Lab Med 2007;131:44-9.
175. Howanitz JH, Howanitz PJ. Evaluation of total serum calcium critical values. Arch Pathol
Lab Med 2006;130:828-30.
176. Howanitz PJ, Steindel SJ, Heard NV. Laboratory critical values policies and procedures: a
college of American Pathologists Q-Probes Study in 623 institutions. Arch Pathol Lab Med
2002;126(6):663-9.
177. Hanna D, Griswold P, Leape LL, Bates DW. Communicating critical test results. Journal
on Quality and Patient Safety 2005;31(2):68-80.
178. National Patient Safety Goals. Oakbrook Terrace, IL: The Joint Commission, 2006.
(Accessed April 19, 2007, at
http://www.jointcommission.org/PatientSafety/NationalPatientSafetyGoals/08_lab_nps
gs.htm.)
179. Hortin GL, Casko G. Critical values, panic values or alert values? Am J Clin Pathol
1990;109:496-7.
180. LiVolsi VA, Leung S. Communicating critical values in anatomic pathology. Arch Pathol
Lab Med 2006;130:641-4.
181. Silverman JF, Pereira TC. Critical values in anatomic pathology. Arch Pathol Lab Med
2006;130:638-40.
182. Pereira TC, Liu Y, Silverman JF. Critical values in surgical pathology. Am J Clin Pathol
2004;122:201-5.
183. Powsner SM, Costa J, Homer RJ. Clinicians are from Mars and pathologists are from
Venus: clinical interpretation of pathology reports. Arch Pathol Lab Med 2000;124:1040-6.

May 2008

190

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

184. Howanitz PJ, Walker K, Bachner P. Quantification of errors in laboratory reports: a quality
improvement study of the College of American Pathologists' Q-Probes program. Arch
Pathol Lab Med 1992;116:694-700.
185. Sandhaus LM, Meyer P. How useful are CBC and reticulocyte reports to clinicians? Am J
Clin Pathol 2002;118:787-93.
186. Third report of the National Cholesterol Education Program expert panel on detection,
evaluation, and treatment of high blood cholesterol in adults. Bethesda, MD: National
Heart, Lung, and Blood Institute, National Institutes of Health, 2001.
187. Warnick GR, Myers GL, Cooper GR, Rifai N. Impact of the third cholesterol report from
the Adult Treatment Panel of the National Cholesterol Education Program on the clinical
laboratory. Clin Chem 2002;48(1):11-7.
188. Gephardt GN, Baker PB. Interinstitutional comparison of bladder carcinoma surgical
pathology report: a College of American Pathologists Q-probes study of 7,234 bladder
biopsies and curettings in 268 institutions. Arch Pathol Lab Med 1995;119:681-5.
189. Nakhleh RE, Jones B, Zarbo RB. Mammographically directed breast biopsies: a College of
American Pathologists Q-Probes study of clinical physician expectations and of specimen
handling and reporting characteristics in 434 institutions. Arch Pathol Lab Med
1997;121:11-8.
190. Zarbo RJ. Interinstitutional assessment of colorectal carcinoma surgical pathology report
adequacy: a College of American Pathologists Q-Probes study of practice patterns from
532 laboratories and 15,940 reports. Arch Pathol Lab Med 1992;116:1113-9.
191. Sirota RL. Defining error in anatomic pathology. Arch Pathol Lab Med 2006;130(604):606.
192. Sharp SE. Effective reporting of susceptibility test results. Diagnostic Microbiology and
Infectious Diseases 1993;16(51):254.
193. Regeniter A, Steiger JU, Scholer A, Huber PR, Siede WH. Windows to the ward:
graphically oriented report forms. Presentation of complex, interrelated laboratory data
for electrophoresis/immunofixation, cerebrospinal fluid, and urinary protein profiles. Clin
Chem 2003;49(1):41-50.
194. Leslie KO, Rosai J. Standardization of the surgical pathology report: formats, templates,
and synoptic reports. Semin Diagn Pathol 1994;11(4):253-7.
195. Howanitz PJ, Cembrowski GS. Postanalytical quality improvement: a College of American
Pathologists Q-Probes study of elevated calcium results in 525 institutions. Arch Pathol
Lab Med 2000;124:504-10.
196. Shah B, Hux J, Laupacis A, Zinman B, van Walraven C. Clinical inertia in response to
inadequate glycemic control: do specialists differ from primary care physicians? Diabetes
Care 2005;28:600-6.
197. Laposata M. Patient-specific narrative interpretations of complex clinical laboratory
evaluations: who is competent to provide them? Clin Chem 2004;50(3):471-2.
198. Lim EM, Sikaris KA, Gill J, et al. Quality assessment of interpretative commenting in
clinical chemistry. Clin Chem 2004;50(3):632-7.

May 2008

191

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

199. Avoid missteps with clinical lab professional services--here's how. Pathology/Lab Coding
Alert 2007;8(11):81-8.
200. Price CP. Medical and economic outcomes of point-of-care testing. Clin Chem Lab Med
2002;40(3):246-51.
201. Nichols JH, Christenson RH, Clarke W, et al. Executive summary. The National Academy
of Clinical Biochemistry laboratory medicine practice guideline: evidence-based practice
for point-of-care testing. Clinica Chimica Acta 2007;379(1-2):14-28.
202. Price CP. Point of care testing. BMJ 2001;322(7297):1285-8.
203. Price CP, Saint John A, Hicks JM. Point-of-care testing. In: Clinical diagnostic technology:
the total testing process. Volume 2: the analytical phase. Ward-Cook KM, Lehman CA,
Schoef LE, Williams RH, ed. Washington, DC: American Association of Clinical
Chemistry, 2003.
204. Meier FA, Jones BA. Point-of-care testing error: sources and amplifiers, taxonomy,
prevention strategies, and detection monitors. Arch Pathol Lab Med 2005;129(10):1262-7.
205. Nichols JH. Reducing medical errors at the point of care. LabMedicine 2005;36(5):275-7.
206. Ehrmeyer SS, Laessig RH. A quality systems approach for identifying and controlling
sources of error with point of care testing devices. Accreditation and Quality Assurance:
Journal for Quality, Comparability and Reliability in Chemical Measurement 2002;7:450-4.
207. Novis DA, Jones BA. Interinstitutional comparison of bedside blood glucose monitoring
program characteristics, accuracy performance, and quality control documentation: a
College of American Pathologists Q-Probes study of bedside blood glucose monitoring
performed in 226 small hospitals. Arch Pathol Lab Med 1998;122(6):495-502.
208. Ehrmeyer SS, Laessig RH. Point-of-care testing, medical error, and patient safety: a 2007
assessment. Clin Chem Lab Med 2007;45(6):766-73.
209. Jones BA, Howanitz PJ. Bedside glucose monitoring quality control practices. A College of
American Pathologists Q-Probes study of program quality control documentation,
program characteristics, and accuracy performance in 544 institutions. Arch Pathol Lab
Med 1996;120(4):339-45.
210. ISO 22870:2006. Point-of-care testing (POCT)--requirements for quality and competence.
Wayne, PA: Clinical and Laboratory Standards Institute, 2006.
211. Sharp GH, Gibson DJ, Tindle BH. Physician office testing: the importance of ongoing
monitoring. Point of Care 2003;2:179-82.
212. Gruszecki AC, Hortin G, Lam J, et al. Utilization, reliability, and clinical impact of pointof-care testing during critical care transport: six years of experience. Clin Chem 2003;49
(6 Pt 1):1017-9.
213. Astion ML. Developing a patient safety culture in the clinical laboratory. Washington, DC:
American Association for Clinical Chemistry, 2005. (Accessed October 15, 2007, at
http://www.aacc.org/AACC/events/expert_access/2005/saftey/qanda.htm.)

May 2008

192

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

214. Still T. Electronic health records can save lives and improve medical care. Verona, WI:
Wisconsin Technology Network LLC, 2005. (Accessed October 22, 2007, at
http://wistechnology.com/printarticle.php?id=1545.)
215. Nichols JH. Point-of-care testing data management. Point of Care 2004;3(1):8-10.
216. Nichols JH, Bartholomew C, Brunton M, et al. Reducing medical errors through barcoding
at the point of care. Clin Leadersh Manag Rev 2004;18(6):328-34.
217. Kost GJ. Preventing medical errors in point-of-care testing: security, validation,
performance, safeguards, and connectivity. Arch Pathol Lab Med 2001;125:1307-15.
218. Dighe AS, Lewandrowski K. Improving point-of-care testing with automated
identification technologies. European Endocrine Review 2006:1-3.
219. Zaloga GP, Roberts PR, Black K, Santamauro JT, Klase E, Suleiman M. Hand-held blood
gas analyzer is accurate in the critical care setting. Crit Care Med 1996;24(6):957-62.
220. Zaloga GP, Dudas L, Roberts P, Bortenschlager L, Black K, Prielipp R. Near-patient blood
gas and electrolyte analyses are accurate when performed by non-laboratory-trained
individuals. J Clin Monit 1993;9(5):341-6.
221. Nichols JH. Management of point-of-care testing. Blood Gas News 1999;8(2):4-14.
222. Singh DG, Agarwal M, Bishawi B. Evaluation of a glucose meter against analytical quality
specifications for hospital use. Clin Chim Acta 2004;343(1-2):217-21.
223. Cohen M, Boyle E, Delaney C, Shaw J. A comparison of blood glucose meters in Australia.
Diabetes Res Clin Pract 2006;71(2):113-8.
224. Khan AI, Vasquez Y, Gray J, Wians FH, Kroll MH. The variability of results between
point-of-care testing glucose meters and the central laboratory analyzer. Arch Pathol Lab
Med 2006;130:1527-32.
225. Kanji S, Buffie J, Hutton B, et al. Reliability of point-of-care testing for glucose
measurement in critically ill adults. Crit Care Med 2005;33(12):2778-85.
226. Commission on Laboratory Accreditation Laboratory Accreditation Program: point-ofcare testing checklist. Northfield, IL: CAP, 2005.
http://www.cap.org/apps/docs/laboratory_accreditation/checklists/point_of_care_testi
ng_october2005.pdf.
227. Kilgore ML, Steindel SJ, Smith JA. Continuous quality improvement for point-of-care
testing using background monitoring of duplicate specimens. Arch Pathol Lab Med
1999;123(9):824-8.
228. Murphy MJ, Paterson JR. Point-of-care testing: no pain, no gain. QJM 2001;94(11):571-3.
229. Ehrmeyer SS, Laessig RH. Point-of-care testing technology. Is quality control still relevant?
MLO Med Lab Obs 1997;Suppl:10-3.
230. LaBeau KM, Simon M, Steindel SJ. Quality control of test systems waived by the Clinical
Laboratory Improvement Amendments of 1988. Perceptions and practices. Arch Pathol
Lab Med 2000;124(8):1122-7.
231. Laessig RH, Ehrmeyer SS. Quality: the next six months. Clin Chem 1997;43(5):903-7.

May 2008

193

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

232. Laboratory requirements. Code of Federal Regulations, Title 42, Section 493.15(b). Subpart A-general provisions; laboratories performing waived tests.
233. LaBeau KM, Simon M, Steindel SJ. The Pacific Northwest Laboratory Medicine Sentinel
Monitoring Network: final report of the findings of questionnaire 10, point of care testing.
Atlanta, GA: Centers for Disease Control and Prevention, 1999.
http://wwwn.cdc.gov/mlp/pdf/LMSMN/PNW/report10.pdf.
234. Howanitz PJ, Jones BA. Bedside glucose monitoring. Comparison of performance as
studied by the College of American Pathologists Q-Probes program. Arch Pathol Lab Med
1996;120(4):333-8.
235. Jacobs E, Hinson KA, Tolnai J, Simson E. Implementation, management and continuous
quality improvement of point-of-care testing in an academic health care setting. Clinica
Chimica Acta 2001;307(1-2):49-59.
236. Kost GJ, Ehrmeyer SS, Chernow B, et al. The laboratory-clinical interface: point-of-care
testing. Chest 1999;115(4):1140-54.
237. Jones JB. The PC data manager: a key component for keeping the LIS "in the mainstream"
of the broadening health care enterprise. Washington, DC: American Association for
Clinical Chemistry, 2002. http://www.aacc.org/NR/rdonlyres/7D572F53-A63E-4A5A9B5B-BB431895EC82/0/pcdata.pdf.
238. Blick KE. The essential role of information management in point-of-care/critical care
testing. Clinica Chimica Acta 2001;307(1-2):159-68.
239. Duca DJ. Autoverification in a laboratory information system. Laboratory Medicine
2002;33(1):21-5.
240. Johnson J, Stelmach D. Autoverification of test results: how to avoid mistakes and improve
efficiency in your lab. Clinical Laboratory News 2007;33(9).
241. Torke N, Boral L, Nguyen T, Perri A, Chakrin A. Process improvement and operational
efficiency through test result autoverification. Clin Chem 2005;51(12):2406-8.
242. Riebling N, Tria L. Six sigma project reduces analytical errors in an automated lab. MLO
Med Lab Obs 2005;37(6):20, 22-0, 23.
243. Colbert DL. Drug abuse screening with immunoassays: unexpected cross-reactivities and
other pitfalls. Br J Biomed Sci 1994;51(2):136-46.
244. Baden LR, Horowitz G, Jacoby H, Eliopoulos GM. Quinolones and false-positive urine
screening for opiates by immunoassay technology. JAMA 2001;286(24):3115-9.
245. Laboratory medicine practice guidelines: evidence based practice for point of care testing.
Nichols JH, ed. Washington, DC: National Academy of Clinical Biochemistry, 2006.
http://www.nacb.org/lmpg/poct/POCT_LMPG_final_rev41706.doc.
246. Tighe P. Postanalytical "error" rates in POCT: use of a quality assurance program.
Accreditation and Quality Assurance: Journal for Quality, Comparability and Reliability
in Chemical Measurement 2001;6:402-4.
247. Kadehjian LJ. Performance of five non-instrumented urine drug-testing devices with
challenging near-cutoff specimens. J Anal Toxicol 2001;25(8):670-9.

May 2008

194

Laboratory Medicine: A National Status Report

Chapter IV – Quality and the Total Testing Process

248. Louie RF, Tang Z, Sutton DV, Lee JH, Kost GJ. Point-of-care glucose testing: effects of
critical care variables, influence of reference instruments, and a modular glucose meter
design. Arch Pathol Lab Med 2000;124(2):257-66.
249. Tang Z, Du X, Louie RF, Kost GJ. Effects of pH on glucose measurements with handheld
glucose meters and a portable glucose analyzer for point-of-care testing. Arch Pathol Lab
Med 2000;124(4):577-82.
250. 2007 National Patient Safety Goals. Oakbrook Terrace, IL: The Joint Commission, 2007.
(Accessed May 9, 2007, at http://www.jointcommission.org/NR/rdonlyres/0515980F1262-438A-8DB9-C2EA28C9BB33/0/07_lab_npsgs.pdf.)
251. Potential 2008 National Patient Safety Goals and requirements, laboratory version.
Oakbrook Terrace, IL: The Joint Commission, 2007. (Accessed May 9, 2007, at
http://www.jointcommission.org/NR/rdonlyres/60B84AD5-A41B-4DE6-9C22C9DFAF99ED48/0/08_potential_LAB_NPSG.pdf.)
252. Meredith RL, Morgan BJ. Performing arterial blood gas analysis as a point-of-care test
improves turnaround time. Advance for Managers of Respiratory Care 2007;16(7):46.
253. Macfarlane I, Davis F. Instrumentation development: building blocks for the point-of-care
boom. IVD Technology 2002.

May 2008

195

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

CHAPTER V
QUALITY SYSTEMS AND PERFORMANCE MEASUREMENT
Quality generally refers to the level of performance or other attributes of interest that are achieved
by a product or service. The performance of a product or service can be defined and assessed
quantitatively or qualitatively against designated standards or goals, whether for conformance to
certain technical standards or specifications or realizing customer satisfaction. Quality
improvement programs and systems-based approaches to quality management can contribute to
achieving and sustaining high levels of quality.
As described by the IOM, quality in health care is the degree to which health services for
individuals and populations increase the likelihood of desired health outcomes and are consistent
with current professional knowledge.1 High-quality health care is: safe, effective, timely, patientcentered, efficient, and equitable.2
Laboratory medicine contributes significantly to quality of care when tests and related services
are clinically appropriate and provided in a technically competent manner with good
communication. This chapter provides an overview of the status of systematic approaches to
quality and performance measurement in laboratory medicine.

APPROACHES TO QUALITY SYSTEMS IN LABORATORY MEDICINE
Public and private sector organizations in health care, including those associated with laboratory
medicine, have described the need for patient care that consistently provides the highest levels of
quality and safety. Serious adverse events resulting in patient harm or death have prompted
consumer and provider demands for greater quality and led to many of today’s quality
improvement initiatives. Even so, most initiatives to address quality and safety issues are
undertaken with limited resources.2, 3
Quality assessment in laboratory medicine has evolved from narrowly focused activities of QC to
more comprehensive, systematic methods. For over 50 years, laboratories have instituted diverse,
evolving mechanisms to improve the quality of testing. Until 1967, efforts generally targeted
improvement in testing accuracy and precision for common analytes through the use of PT. This
is an external quality assessment process that evaluates and grades the laboratories’ analytic
performance of selected tests. Unfavorable performance rates decreased somewhat with use of
PT as the sole means of measurement. Passage of the Clinical Laboratory Improvement Act of
1967, corresponding Amendments of 1988 (CLIA), and 1992 implementation of the final CLIA
regulations standardized the approach to laboratory quality. The regulations established minimal
standards for QC; QA practices; PT; personnel qualifications and responsibilities; patient test
management; and recordkeeping. The specific requirements that laboratories must meet are
based on the complexity of the testing performed.4 Laboratories performing moderate and high
complexity testing, including hospitals, reference laboratories, and POLs, meet these standards in
order to be certified.a Laboratories performing relatively simple tests categorized as “waived” are
a

Refer to the chapter on Federal Regulatory Oversight and Appendix A of this report for additional information
about CLIA regulations.

May 2008

197

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

not subject to CLIA standards, but are required to follow manufacturers’ instructions for test
performance and QC.b (Definitions of QC and QA are provided in Box 5.1 below.)
QC and PT activities are reported to have had key roles in the reduction of errors associated with
the analytic phase of testing.5, 6 According to the American Proficiency Institute, PT failure ratesc
for chemistry and hematology laboratory tests decreased from 1994 to 2004 for eight analytes
most often tested in physicians’ office and clinical laboratories.7 PT failure rates decreased from
18.7% to 3.2% for cholesterol, from 6.3% to 1.1% for potassium, and from 5.7% to 2.4% for
creatinine. Failure rates for testing microbiology analytes decreased for both positive and
negative cultures between 1994 and 2004. Another study demonstrated decreases in deficiencies
associated with inspections. During 1995-1996, 30% of laboratories inspected to assess compliance
with CLIA failed to perform QC testing and 13% failed to follow directions outlined by
manufacturers. By 2001-2002, 18% of CLIA inspected sites failed to perform QC testing and 6%
were not following manufacturers’ directions.8
While performance in the analytic phase of testing as evaluated by basic QC, QA, and PT has
improved substantially since CLIA’s implementation, further improvements are necessary to
achieve quality and safety. The findings of CMS oversight activities, academic studies, and
accrediting organization surveys reinforce the need to move beyond analytic-focused activities to
a mechanism that supports integration of preanalytic, analytic, and postanalytic components as
described in the chapter on the TTP in this report. Such concerns arise, for example, when
inadequate specimen volumes are obtained during collection, affecting the laboratory’s ability to
test the specimen in the analytic phase. Improvements in quality and safety at each critical
control point along the path of the TTP require the extensive framework and methodologies
inherent in systems-based approaches to quality management. A critical control point is a point,
step, or procedure at which control can be exercised to prevent, eliminate, or minimize a hazard.9
The introduction of comprehensive systems-based approaches has been of interest to public and
private sector stakeholders associated with laboratory medicine. Several laboratory accrediting
organizations took the lead in promoting the adoption of systems approaches voluntarily or as a
condition of accreditation during the 1990s and early 2000s. The Joint Commission, CAP, AABB
(formerly the American Association of Blood Banks), COLA, and American Society for
Histocompatibility and Immunogenetics (ASHI) advocate the implementation of comprehensive
quality management systems (QMS), beyond the basic requirements of CLIA.10-13
In 2003, CMS restructured the QC and QA provisions in CLIA to reflect the flow of specimens
through the laboratory, and integrated QA into all phases of testing. The requirements now
correspond to the broader framework of formalized systems-based approaches to quality
management.

b
c

Some states (e.g., New York, Washington) have more stringent requirements.
Failure rate is defined as: (number of unacceptable responses/ total number of responses) x 100.

May 2008

198

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

Beyond the Basics to Quality Management Systems
The Clinical and Laboratory Standards Institute (CLSI) d adopted a hierarchical model defining
five tiers of quality.15-17 As presented in Box 5.1, QC and QA are precursor, lower-tier activities.
Mid-level tiers include the introduction of quality management systems and quality cost
management. Long-term success in all tiers is necessary to maintain the highest levels of
excellence and total quality management (TQM), generally known as continuous quality
improvement (CQI) in health care.
Most health care organizations are operating at or below the level of QA, although some
organizations, such as the Veterans Health Administration (VHA) and Latter-day Saints hospital
(Salt Lake City), are working successfully with strategic tools to implement quality management
systems (e.g., CQI, failure mode and effects analysis [FMEA]).18, 19 Efforts over the last decade to
redesign health care delivery systems have promoted adoption of the two higher-level activities:
QMS as a tool for quality, safety, and performance measurement; and pay-for-performance
programs, which, though largely unproven to date, may contribute to quality cost management.
QMS have been used in a wide variety of applications to achieve major improvements in the
quality, efficacy, safety, and/or customer-centeredness of processes, products, and services in a
full range of manufacturing and service industries.20 QMS incorporates principles of engineering,
manufacturing, and human factors science to establish an integrated infrastructure that optimizes
and continually improves health care operations. Structures, processes, and/or events are
considered together as they interact to produce an outcome.21 Whether or not system components
cross departmental or organizational boundaries, a single change anywhere in the system can
affect other parts of the system. The systems approach supports workflow design; performance
monitoring; management of organizational, personnel, and informational factors; and leveraging
technology and other contributing factors for a well-defined, constantly evolving, coordinated
mechanism to produce desired outcomes and continuously improve quality.
Organizations that implement the QMS model can greatly enhance their ability to reduce or
eliminate errors, meet customer needs, perform well on accreditation assessments, and maintain
quality objectives.22 However, the broader health care sector largely has been slower to adopt
QMS, even though the small but growing number of health care organizations that have applied
them are reporting favorable returns.23-25

d

CLSI is a non-profit standards-developing organization that promotes the development and use of guidelines and
standards within the health care community, including clinical laboratories.14

May 2008

199

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

Box 5.1: Five Levels of Quality—Definitions
Level 1: Total quality management is intended to sustain high quality by focusing on long-term success
through customer satisfaction. TQM holds quality as the driving factor behind leadership, design, planning, and
improvement. Variations from quality are artifacts of poorly designed systems, rather than the fault of one or
more individuals.19, 26
Level 2: Quality cost management (QCM) includes all activities involved in QMS, QA, and QC, along with
related economic aspects (i.e., “cost of quality”).27 QCM promotes integration of quality processes throughout
an organization subject to the constraints of the organization’s financial resources.28 Budgeting and resource
allocation are integrated within the context of the larger organization and oriented to meeting physician and
patient needs. Recent efforts to implement pay-for-performance programs may be considered a form of QCM.
Level 3: Quality management systems refers to a systematic approach to achieving quality objectives.18
QMS constitutes a coordinated and comprehensive effort to meet quality objectives using management systems
standards such as those developed by the International Organization for Standardization (ISO) and CLSI.28
Standards for QMS are implemented in laboratory medicine via such models as CQI, Six Sigma, failure mode and
effects analysis, and Toyota “lean production.”
Level 4: Quality assurance/assessment monitors the totality of components or characteristics that affect
quality and customer satisfaction.29 QA involves planned and systematic activities to provide confidence that
an organization fulfills requirements for quality. Regulatory and compliance issues are generally handled
through QA-related policies and procedures. In laboratory medicine, characteristics such as turnaround time,
patient preparation, and specimen collection may be monitored at a basic level internally and externally.
Proficiency testing is an external mechanism for QA.
Level 5: Quality control refers to laboratories’ internal procedures for day-to-day monitoring of instruments,
monitoring work processes, detecting problems, and making corrections prior to the delivery of products or
services.29 Typically, QC procedures for monitoring analytic performance rely on statistical measures (e.g.,
mean performance and within ±2 standard deviations) as an indicator of quality.28 For example, the precision
of clinical laboratory tests performed for a specific analyte is assessed relative to its reference standard. QC is
a targeted, internal mechanism.
Source: A quality management system model for health care. Approved guideline: HS1-A2; second edition. Wayne, PA:
Clinical and Laboratory Standards Institute, 2004.

Standards and Guidelines for Quality Management Systems
Formalized standards and guidelines for QMS that encompass these properties have been
developed by the International Organization for Standardization (ISO)e as well as other
institutions involved in a specific sector such as CLSI for health care and laboratory medicine. In
many instances, accreditation organizations use the standards as a framework for developing
detailed criteria and benchmarks appropriate to the respective medical discipline or subspecialty.
Standards can serve several purposes. They can establish consistency or uniformity across multiple
individuals and organizations, and set expectations of quality and safety for organizations, health
professionals, patients/consumers, and purchasers. Because the standards for QMS are based on
the day-to-day activities of health care professionals, they are an effective means for improving care
delivery.21 Measurement of performance against designated standards and feedback about that
performance are important parts of QMS-related quality improvement initiatives.

e

ISO develops international standards that outline the requirements for products, services, processes, materials, and
systems and are designed to be implemented by organizations worldwide.30 The American National Standards
Institute is responsible for endorsing consensus standards in the U.S. and for representing U.S. interests at the ISO.

May 2008

200

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

ISO and CLSI standards for QMS are internationally recognized. The ISO 9000 seriesf is
considered the world standard for establishing, verifying, and certifying QMS. It was developed
to assist diverse organizations with implementation and operation of effective QMS, regardless of
the service or product provided.32, 33 Aside from manufacturing-related industries such as
medical devices and pharmaceuticals and select high-risk specialty areas, ISO 9001:2000 has not
been implemented on a widespread scale in the U.S. health care sector. As of 2005, about 100
health care entities in North America were certified to ISO 9001:2000, including 12 hospitals and
10 medical groups in the U.S.34 ISO 9001:2000 has been more widely adopted by international
health care facilities. A 2006 survey estimated that 14,180 certificates for ISO 9001:2000 have been
issued worldwide in the health care and social work sectors.35
Historically, the standards have been employed in other sectors; about 90% of facilities in the U.S.
certified to the ISO 9001:2000 standard are involved in manufacturing.34 However, public and
private sector stakeholders are interested in shifting the largely fragmented U.S. health care
system that produces inconsistent levels of quality to more integrated, dynamic system capable of
producing and sustaining high levels of quality. Their efforts have prompted renewed interest in
the use of standardized QMS, including ISO 9001:2000, to facilitate this transformation.20 Certain
U.S. government agencies, including CMS, are requiring certain health sector contractors be
certified to ISO 9001:2000 or undergo a third-party validation process.36
Because of the broadly applicable nature of ISO 9001:2000, more targeted standards for QMS have
been developed for health care, often according to specialty area. CLSI is the chief developer of
standards for laboratory medicine and serves as the Secretariatg for the ISO Technical Committee
on Clinical Laboratory Testing (ISO/TC 212). Through the ISO/TC 212 workgroup on quality
management, CLSI led the development of ISO 15189 – Medical Laboratories—Particular
Requirements for Quality and Competence, a standard for process-based QMS specifically in clinical
laboratories. ISO 15189 addresses all aspects of laboratory operation, from patient preparation
and identification to the collection and examination of clinical samples, as well as document
control, contracts, and relationships with referral laboratories.37 Other standards target specific
uses or elements of QMS.h Since the approval of ISO 15189, CLSI has sold at least 280 copiesi of
the standard to laboratories in the U.S.38 Beyond this expression of interest, it remains to be seen
how many of these laboratories will implement the standard.
As a complement to the ISO standards, CLSI developed two guidelines that advance the
implementation of QMS. The first, GP26-A3 - Application of a Quality Management System Model for
Laboratory Services, defines the laboratory path of workflow and provides direction on the design of
policies, processes, and procedures to build required levels of quality into day-to-day operations
and reduce the potential for errors and wasted resources.39 The second guideline, HS1-A2 – A
Quality Management System Model for Health Care, supports ISO 9001:2000 and 15189 and describes
ISO 9000 was established in 1987. Its precursor was BS 5750, developed during the 1960s and 1970s by the British
Government.31
g As Secretariat for ISO/TC 212, CLSI coordinates international standardization of clinical laboratory testing in four
areas: quality management in the clinical laboratory; reference systems; IVD products; and antimicrobial
susceptibility testing.
h Other standards related to QMS include: ISO 15190 – Medical laboratories – Requirements for safety; ISO 22869 - Clinical
laboratory testing—Guidance on application of ISO 15189; ISO 22870 - Point-of-care testing—Requirements for quality and
competence; and ISO 22367 - Medical laboratories - Reduction of error through risk management and continual improvement.
i This figure does not account for copies of the ISO 15189 standard sold by ANSI, which was unavailable.
f

May 2008

201

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

the set of 12 essential components that must be in place for an organization to function in a manner
that meets QMS standards and quality objectives (see Box 5.2).18 These components, known as
quality system essentials (QSEs), provide guidelines to describe, document, integrate, measure, and
monitor the implementation and effectiveness of the work operations of any organization, service
unit, or support function in the organization. Subsequent editions of GP26-A3 also incorporated
laboratory-specific guidelines for implementing essential components of QMS identified in HS1-A2.j
The 12 QSEs are applied to every step in the path of workflow (e.g., test ordering, specimen
collection) to ensure that all quality management issues are identified and addressed in the
organization’s policies and procedures.
Box 5.2: Quality System Essentials
•

Documents and records

•

Organization

•

Personnel

•

Equipment

•

Purchasing and inventory

•

Process control

•

Information management

•

Occurrence management

•

Assessment: external and internal

•

Process improvement

•

Customer service

•

Facilities and safety

Source: A quality management system model for health care. Approved
guideline: HS1-A2; second edition. Wayne, PA: Clinical and Laboratory
Standards Institute, 2004.

Standards for QMS have been most broadly adopted in transfusion medicine. Beginning in 1990,
the AABB recognized the need for QMS specifically applicable to blood banking in order to move
from an error-detection approach to an error prevention approach.40 Initially, the AABB Quality
Program was designed to meet FDA requirements for Current Good Manufacturing Practices and
QA guidelines. To enhance user friendliness and encourage greater acceptance, AABB revised
the program in the late 1990s by relying on ISO 9001:2000 standards to develop a specific set of
QSEs for blood banking. In 1998, AABB began requiring full implementation of quality systems
for accreditation.41
Use of ISO 9000, ISO 15189, and CLSI GP26-A3 in laboratory medicine has accelerated in recent
years as CMS and other laboratory accreditation organizations incorporate QMS-related
standards and guidelines into their regulatory requirements.42, 43 In 2003, CMS used the path of
workflow as the model for restructuring the CLIA requirements. CMS also has mapped the
revised CLIA provisions to the QSEs outlined in CLSI GP26-A3 to demonstrate that laboratories
interested in using the QSEs can achieve CLIA compliance concurrently.
j

Other CLSI documents supply discipline-specific details (e.g., QC for quantitative measurements, reference
intervals) as a compliment to HS1-A2 and GP26-A3.

May 2008

202

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

Also in 2003, CAP endorsed ISO 15189 accreditation as a value-added program,k with optional
use of CLSI QSEs as a supplement.44 CAP’s Laboratory Accreditation Program General
Checklist requires that laboratories have a documented quality management program that can
be based on ISO 9000, ISO 15189, or programs designed by CAP, other organizations, or
laboratories themselves.45 At this writing, at least three laboratories are seeking accreditation to
ISO 15189 through CAP.46 Similarly, COLA announced the launch of new interactive QMS
training courses based on ISO 15189 and CLSI guidelines in 2005.47 COLA supports
implementation of QMS voluntarily; its standards for laboratory self assessment do not require
QMS as a condition for accreditation.
Among the main potential benefits of U.S. harmonization to ISO standards are:

ƒ

Laboratories can implement proven quality improvement strategies without additional
regulation

ƒ

Accreditation organizations can maximize their ability to evaluate laboratories worldwide

ƒ

Vendors can standardize instruments more easily

ƒ

Researchers can undertake cross-laboratory and cross-national comparisons of
performance48, 49

The following section provides a brief summary of available research on specific QMS-related tools.

Implementing Quality Management Systems
The strategic tools for implementing systems-based approaches to quality management and
improvement in health care and laboratory medicine include methods such as CQI, Toyota
“lean production,” Six Sigma, and FMEA. These methods have been validated as effective
quality management tools in various sectors in the U.S. and worldwide, such as the automobile
industry, telecommunications, mining, banking, and construction.20 Although use of these
methods is not widespread in laboratories at this time, an increasing number of laboratories,
from small POLs to large reference laboratories, are employing them to meet and exceed
regulatory and accreditation requirements as well as clinician and patient expectations for
quality and financial management objectives.
Among the most commonly used QMS tools, CQI, lean production, Six Sigma, and FMEA share
several key features:

k

ƒ

Scientific approach to process analysis and systems improvement

ƒ

Decision-making based on data derived from regular performance measurement

ƒ

Improvement focus

ƒ

Preventive orientation toward potential quality problems and errors

ƒ

Interdisciplinary teams9

CAP’s program supporting ISO 15189 is separate from its CLIA accreditation program.

May 2008

203

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

Continuous Quality Improvement
CQI applies the scientific methods of TQMl to process improvement in health care and may be
employed to improve clinical quality, patient satisfaction, error rates, waste, unit production
costs, productivity, and market share, among other aspects of care.19 It embodies a disciplined
approach to managing competitive challenges and enhancing customer satisfaction. CQI is
intended to reduce variation and improve performance through a four-step cycle: The teams plan
an intervention to improve the process; do the hypothesized intervention; study the results of the
intervention; and act on the results. Cross-cutting teams devise a flow chart of a work process to
be studied and use data to understand variations in performance.9
The CQI movement began in academic research institutions during the mid-1980s and gained
momentum when The Joint Commission adopted the philosophy to advance health care quality
improvement goals.50 The movement is helping to promote dynamic quality management,
establishing the patient as the true “consumer” of health care services, and motivating health care
organizations to initiate quality improvement programs. Although some health care leaders have
claimed successful use of CQI to achieve quality goals, few studies are documented in the
literature. For example, a regional group of cardiothoracic surgeons that implemented a CQI
intervention reported reduced mortality associated with coronary bypass surgery by 24%.51 A
Veterans Administration medical center used CQI to increase operating room efficiency, noting
procedure start time improvements ranging from 30 to 55 minutes.52 In a randomized controlled
trial, patients assigned to protocol-directed weaning from ventilation tubes as developed via CQI
methodology had significantly shorter durations of mechanical ventilation compared with
patients assigned to physician-directed weaning.53
Laboratories have demonstrated some successes with CQI. Implementing CQI for physicians’
ordering of blood for surgical cases at a regional hospital facilitated savings of more than 500
hours of technologist time and $350,000 in patient charges over a two-month period. Reduced
inventory and blood banking costs added further to cost savings.54 A dedicated stat laboratory
implemented CQI to improve TAT for various clinical pathology tests, after which TAT decreased
from 61 to 36 minutes for clinical chemistry tests.55 In a two-year study applying CQI to
physician ordering of preoperative tests for elective surgery, the volume of tests ordered
decreased by 50% and 60% in the first and second years, respectively, the appropriateness of test
orders improved by 81% and 86%, and overall cost savings were $76,000.56 CQI also has been
applied successfully to POCT, leading to more reliable results and helping resolve discrepancies
between test results received at the bedside versus in the core laboratory.57, 58
There are notable challenges to implementing CQI. Lack of hospital or physician administrative
and financial support were identified as barriers to CQI implementation by 4 of 9 hospital
catheterization laboratories participating in a CQI initiative during 1998-2002.59 The extensive
time involved in educating staff about CQI principles and forming and working together as
teams, along with insufficient commitment to CQI on the part of hospital management were cited
as key obstacles to implementation by a hospital-based pathology department.60

l

Originally introduced to Japanese manufacturers in the 1950s, TQM gained popularity in the U.S. in the late 1970s
and early 1980s as an alternative to QA.19

May 2008

204

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

To date, the impact of CQI on the health care sector has not been as widespread as initially
expected. This apparent lag in uptake has been attributed to such reasons as:

ƒ

The literature documenting quality improvement and clinical outcomes stemming
from CQI remains relatively sparse.

ƒ

Administrators, rather than clinicians, were the first who sought to incorporate CQI,
which may convey to some observers that it is a mechanism for cutting costs rather
than improving quality.

ƒ

Implementation was not as easy as anticipated, frustrating many of the leaders who
initially advocated CQI.19

While the concept of continuously improving performance to achieve quality remains intact,
other methods have emerged for improving quality.

Six Sigma
Developed by the Motorola Company in the early 1980s, the Six Sigma system is a means of
improving quality by identifying and taking action at the root cause of a problem, rather than on
symptoms arising from the cause.28, 61 Six Sigma identifies direct relationships between the
number of defects in a given product or process, wasted resources, and the level of customer
satisfaction.62 Six Sigma is similar to TQM but has been described as having a more aggressive
goal. The objective of Six Sigma is to reduce defect rates to low frequencies in a statistical
distribution, based on multiple standard deviations from a specified average, such as fewer than
3.4 defects per million opportunities. Table 5.1 displays the Six Sigma performance scale.
Table 5.1: Six Sigma Performance Scale
Process Sigma

Percent Accuracy

Defects per million opportunities

6

99.9997%

3.4

5

99.98%

233

4

99.4%

6,210

3.5

97.7%

22,700

3

93.3%

66,807

2

69.1%

308,537

Source: Westgard JO, Westgard SA. The quality of laboratory testing today: an assessment of sigma metrics for
analytic quality using performance data from proficiency testing surveys and the CLIA criteria for acceptable
performance. Am J Clin Pathol 2006;125(3):343-354.

Six Sigma can be used in laboratory medicine to establish tolerance limits necessary to define
good quality. Westgard has described two methods for calculating the sigma metric—one
method is based on measuring the outcome of the process, whereas the other method is based on
measuring the variation of the process directly.63 The outcome measurement approach involves
counting defects, calculating defects per million, and using a statistical table to convert the defect
rate per million to a sigma metric. This approach is applicable to any process but usually requires
extensive efforts to collect and analyze the data. As such, it is typically applied to preanalytic and
postanalytic processes. Direct measurement of process variability (i.e., standard deviation) allows
for determination of process capability. It assumes that the process distribution is stable and can
be characterized by repetitive measurements. Thus, this methodology is used to determine the

May 2008

205

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

precision and accuracy of analytic processes through experimental procedures. A sigma metric is
calculated from the defined tolerance limits and the variation observed.64
Studies using both sets of metrics have reported that laboratory testing falls short of the minimal
acceptable performance level demanded by other industries. Based on CLIA criteria for acceptable
performance in PT, the national quality of cholesterol testing is estimated at 2.9 to 3.0 sigma, glucose
at 2.9 to 3.0, calcium at 2.8 to 3.0, and PSA at 1.2 to 1.8.64 This level of quality was deemed adequate
only if performance was controlled with QC above that required by CLIA. Reported defect rates for
other common laboratory measures are approximately 2.8 sigma for Papanicolaou smears, 2.3
sigma for therapeutic drug monitoring, and 3.6 sigma for transport of red blood cells.65 Among
other factors, these lower levels resulted from missing information in test requests (preanalytic
errors) and incorrect laboratory measurement (analytic errors). However, designating a tolerance
level such as 3.4 defects per million for all processes is arbitrary; it may be unacceptably high for
some critical processes and unnecessarily difficult and costly to achieve for others.
Six Sigma provides clinical laboratories with the methodology and measurements to determine
specific QC actions necessary to achieve the desired level of quality.64 Also, Six Sigma allows for
communication of quality metrics in a format that is understood outside the health sector.61
Health care institutions and laboratories can compare performance against that of other sectors.
Six Sigma is gaining acceptance in health care and laboratory medicine as a method to generate
objective metrics and estimates of quality, though implementation is still in the early stages.66 The
following examples highlight case studies of organizations that have experienced modest
improvements in use of Six Sigma for measuring and improving performance.
In 2004, West Tennessee Healthcare’s core lab built its new laboratory using lean production and
Six Sigma principles. Focusing on phlebotomy, Six Sigma principles were used to assess the
timeliness with which phlebotomists worked and to identify best practices.67 As a result, the time
required to get phlebotomy specimens to the laboratory decreased from over 20 minutes to an
average of 5.2 minutes. Not one patient identification error was detected and documented during
the subsequent 11 months.
In another study using a combination of lean production (described in the next section) and Six
Sigma, Washington Hospital Center’s Automated Services Laboratory (Washington, DC) identified
6 changes that could improve laboratory quality and turnaround time, including decreasing
centrifuge times and taking steps to eliminate blood clotting.68 The laboratory reduced mean TAT
for blood tests from 75 to 46 minutes and decreased its annual operating cost by nearly $80,000.
North Shore-Long Island Jewish Health System (Great Neck, NY), which includes a network of
laboratories that together process 3.5 million examinations annually, used Six Sigma to reduce the
number of accessioning errors, or errors occurring when patient data is entered, tests are ordered,
or samples are labeled.69 Six Sigma methods helped staff to identify specific steps in the accession
process that were most often completed incorrectly or incompletely. Following implementation
of barcoded labels and a new training program for accessioners, the performance of the accession
department improved from 3.9 sigma to 4.5 sigma.

May 2008

206

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

Toyota (Lean) Production System
Lean production, also known as the Toyota Production System, was developed and perfected by
the Toyota Automotive Company over the latter half of the twentieth century. It is based on the
tenet that consistently eliminating waste in each step of a process leads to increased efficiency (i.e.,
the best quality and lowest cost), while improving safety and morale.70, 71 Lean production aims
to meet internal and external customer needs through a one-by-one continuous flow process with
integrated quality indicators and elimination of system waste related to materials, time, idle
equipment, and inventory. The system design highlights problems in real-time where the work is
performed and is intended to solve problems at their root cause. Front-line workers are
empowered to participate in decision-making, design changes, and problem-solving at the time
errors occur. The overarching goal is to reduce all system defects to zero.
Studies of lean production in laboratory medicine have reported successful results. One study
sought to improve chemistry test turnaround time in a central laboratory by addressing preanalytic
variables using existing resources and lean production. The redesigned preanalytic process had
fewer steps and employed the one-piece flow systemm to move specimens through the accessioning,
centrifugation, and aliquoting process. Five new workstations were added and others redesigned
for efficiency. Median preanalytic processing time was reduced from 29 to 19 minutes and the
laboratory met the goal of reporting 80% of chemistry results in less than 1 hour for 11 consecutive
months. Introduction of lean production also eliminated systemic waste, including reduction in
mislabeled and missing tubes, and elimination of the unwritten and unapproved practice of
collecting an extra tube of blood, which decreased the number of tubes used for collection, number
of tubes processed per month, and amount of biohazardous waste discarded.72
One study examined implementation of lean production in a single clinician’s office practice and
cytology laboratory to determine its impact on Pap test quality and errors.73 The clinician’s goal
was to obtain a perfect specimen for every patient, i.e., one that adequately sampled the
transformation zone in which preneoplastic lesions usually develop. Test quality improved, as
the percentage of Pap tests without a transformation zonen component decreased from 9.9% to
4.7%; the percentage of tests with a diagnosis of atypical squamous cells of undetermined
significance decreased from 7.8% to 3.9%; and the frequency of error as measured by cytologichistologic correlation decreased from 9.52% to 7.84%.
Use of lean production in process redesign was used to diminish diagnostic errors associated with
examination of thyroid gland fine-needle aspirations by cytologists and cytotechnologists at two
different hospitals.74 This procedure was chosen due to the wide variability in diagnostic
accuracy indicated in the literature.75-77 For the study, the investigators created a specimen
adequacy scoring system, used standardized diagnostic terminology, and incorporated an
immediate interpretation service. Performance changes following the use of standardized
terminology included (but were not limited to) an increase in test sensitivity from 70.2% to 90.6%,
decrease in the false-negative rate from 41.8% to 19.1%, decrease in the discrepancy rate from
31.0% to 24.2%, and increase in the uninterpretable specimen rate from 5.8% to 19.8%. With the
In a one-piece flow system, all activities involved in a process are performed on each object before work is begun
on the next object.
n On the cervix, there are two cell types, squamous cells and columnar cells, which meet at a place called the squamocolumnar junction, also known as the transformation zone. It is in this transformation zone that abnormal growth
or dysplasia develops.
m

May 2008

207

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

addition of the immediate interpretation service, the noninterpretable specimen rate decreased
significantly from 23.8% to 7.8%.
As with any QMS tool, the success of lean production depends on the commitment of the
organizational leadership and staff in adopting the methodology and managing expectations for
realizing practical gains.78

Failure Mode and Effects Analysis
Though it is not considered to be a comprehensive approach to quality management, FMEA is a
tool that supplements and contributes to QMS by providing a mechanism to systematically
evaluate and prevent errors (“failures”) that can cause harm. FMEA is a team-based, proactive
approach for identifying the ways that a process or design can fail, why it might fail, and how it
can be made safer.79 Although originally developed and used by the U.S. military and space
program, the VHA and Tenet Health System have adapted the approach for identifying and
preventing medical errors and other process-related problems.19 In 2001, the VHA’s National
Center for Patient Safety introduced Health Care Failure Mode and Effect AnalysisTM.80 That
same year, The Joint Commission established a new standard that requires all accredited hospitals
to proactively assess at least one high-risk process per year, such as the preparation of medication
or plans for infection control, using such methodologies as FMEA.10, 81 When sentinel events
occur, organizations must undertake not only the traditional retrospective root cause analysis, but
the prospective FMEA as well. The requirements were extended to home care programs in 2004.82
Typically, organizations initiate an analysis when a near miss or concern for risk occurs rather
than only when a sentinel event occurs.83
FMEA facilitates identification, evaluation, and calculation of risk for each failure mode (e.g.,
problem or defect) according to severity, frequency, and detectability. Each characteristic is
assigned a value between 1 (highest) and 10.28 The ultimate “effect” or risk priority number for a
particular failure mode is determined by multiplying the scores for each characteristic: Effect =
Severity x Frequency x Detectability. Higher risk priority numbers indicate the need to prioritize
actions to correct problems.19 Health care organizations report positive results from use of FMEA
in a range of clinical processes such as trauma care, chemotherapy, case management, blood
transfusion, and other areas of laboratory medicine.84-87
FMEA has multiple functions in laboratory medicine. First, FMEA can be used in product
development. CLSI guideline EP18-P2, Risk Management Techniques to Identify and Control
Laboratory Error Sources,o refers to FMEA as a means for manufacturers of POCT devices to
evaluate potential failure modes during the design and development of new products as well as
to ensure the ability of users to detect and remedy possible causes of error.88 Second, FMEA can
reduce risk in all phases of the laboratory testing process through evaluation and measurement
of hazards associated with process malfunctions, decision making to determine where and how to
execute improvement actions, and measurement of outcomes from improvement actions.28, 85
Several laboratories have successfully implemented FMEA. For example, in 2003, the Los
Angeles County and University of Southern California Healthcare Network used FMEA to
decrease the time in which they reported critical laboratory values to licensed caregivers of
o

EP18-P2 is a revision of a previously developed guideline EP18-A, Quality Management for Single-Use Testing.

May 2008

208

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

outpatients and non-critical care inpatients.89 Average monthly notification time declined steadily
over the 8-month period after system redesign to target areas likely to result in improved
outcomes. Another study of a children’s hospital network used FMEA to identify the preanalytic
specimen labeling phase as the key focus area for improvement.90 After laboratories adopted the
practice of accepting only those patient specimens that met Joint Commission standards for
labeling, the number of mislabeled and/or unlabeled specimens decreased by 75%. In a study of
transfusion errors in a hospital setting, laboratorians used FMEA to ascertain vulnerabilities in
day-to-day procedures and redesign processes to prevent errors.84 The new process accomplished
its goal of preventing all serious avoidable errors during the three-month pilot study and in the
eight months following laboratory-wide implementation. Although a few measures indicated the
need for occasional coaching to reinforce the procedural changes, the number of variances among
staff continued to steadily decrease.
Laboratories and other health care organizations can encounter challenges when using FMEA. As
noted by the VHA, groups that attempt to implement too many actions, fail to communicate with
team members who will actually be responsible for implementing changes, and do not gain
backing from senior management often find the FMEA process ineffective.80

Considerations for Designing Quality Management Systems
QMS implementation presents specific challenges in different laboratory settings. Hospital and
physician office laboratories often cite resistance to cultural change as a major obstacle barring
implementation of QMS. A leadership team that is competent, effective, and enthusiastic is vital
to successful implementation in POLs and hospital laboratories. In particular, team members
should possess substantial knowledge about the processes in need of improvement as well as the
QMS methodology being implemented.91-94 Commitment to change by other laboratory
personnel also is necessary and can be achieved by involving all staff in QMS deployment, data
collection and analysis.69, 91, 92 Recognition of team members’ progress is important to group
learning and continued internal support for QMS implementation.92
Some POLs, hospital, and reference laboratories have cited financial constraints and the belief that
QMS costs more than it saves as obstacles to its implementation. Strategies to minimize the cost
of QMS implementation include thorough review and selection of the QMS methodology that
most closely matches the organizational goals and budget and partnership with other health care
professionals whose work is significantly related to the laboratory (e.g., clinic administrators,
physicians, nurses).91, 95 From these strategies, laboratories have reported increasing returns on
their investment and additional opportunities for funding of QMS.96
Certain observers question the ability of QMS tools (i.e., CQI, lean production, Six Sigma, FMEA)
to improve the quality of patient care.49 Despite their face validity, the use of QMS tools may not
have been studied sufficiently to date, providing limited evidence of their effectiveness. There is
considerable variation in QMS applications, which may create challenges to comparisons of one
methodology to another. Further study of these tools in accordance with clearly defined
standards would help to build an objective evidence base.

May 2008

209

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

PERFORMANCE MEASUREMENT
The development and use of objective measures to evaluate the quality of health care is essential
for improving health care delivery.97 Information derived from measurement facilitates an
understanding of how health services, organizational factors, and financial factors affect patient
and population health, and other important effects arising from differences in patient population,
health conditions, and settings of care.98 This information can support decisions to change
resource use and delivery.
Measurement of quality serves key health care objectives, specifically to:

ƒ

Inform quality improvement efforts

ƒ

Inspect and certify that a facility or provider meets previously established standards

ƒ

Compare groups for a variety of purposes, including selective contracting by
purchasers and choice of providers and practitioners by individuals

ƒ

Support decisions by patients, families, and employees about selecting health care
providers and facilities

ƒ

Identify substandard performers, particularly those whose performance is so far below
an acceptable level that immediate actions are needed

ƒ

Highlight, reward, and disseminate best practices

ƒ

Monitor and report information about changes in quality over time98

The data from measurement allows an organization to assess performance, use that information
to improve, and increase the likelihood of achieving desired health outcomes based on current
professional knowledge.99 Performance measurement has been a core feature of improvement
programs for many public and private sector QA organizations in health care for more than 15
years (see Box 5.3 for a brief synopsis). While laboratory testing is incorporated to some extent in
most of these organizations’ performance measurement programs, the focus tends to be on
clinician ordering and patient receipt of appropriate tests for screening, diagnosis, or disease
management purposes. These organizations do not directly address measurement and reporting
of key components of quality and performance related to the total laboratory testing process.
Assessments of this nature, though very limited, are performed by laboratory sector oversight,
research, and professional organizations.
The need for a standardized set of measures to report on key aspects of quality across the TTP has
been under discussion for the last decade.100 In the last few years, CDC and other organizations
have sponsored studies that may facilitate the development of a national framework and
performance measures for laboratory medicine.

May 2008

210

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

Box 5.3: Performance Measurement Initiatives in Health Care
The importance and utility of routine, externally reported assessments of the quality of health care delivered is
widely recognized and accepted.101 Efforts to standardize quality measurement and reporting in the health care
system have been underway for more than 15 years.102 Largely driven by federal and private sector payers and
accrediting organizations, early leaders in performance measurement include: CMS, AHRQ, The Joint Commission,
and NCQA (a coalition of private sector health plans) NCQA’s Health Plan Employer Data and Information Set
(HEDIS)p is one of the most successful quality measurement initiatives to date. Initially, health plans seeking
accreditation by NCQA were required to report on HEDIS measures. However, when the measures were adapted in
the mid-1990s for use by public purchasers, CMS began to require plans participating in Medicare programs to
report on the measures as well.103 Today, the measures also are used by private purchasers in nearly 90 pay-forperformance programs.104
Other organizations have joined efforts to develop, coordinate, and harmonize performance measurement and
reporting. These include the Pacific Business Group on Health, the Leapfrog Group, American Medical Association,
Hospital Quality Alliance, Ambulatory Care Quality Alliance, the Foundation for Accountability, and National
Quality Forum (NQF). Standards and requirements for quality assessment, performance measurement, and public
reporting have extended to long-term and ambulatory care settings. Performance measures also are becoming
more specific for patient populations (e.g., children, elderly), health conditions (e.g., substance abuse disorders,
venous thromboembolism), and infrastructure (e.g., health information technology).105

Basics of Performance Measurement
Types of Performance Measures
Generally, performance measures are classified into three broad categories based on
Donabedian’s classic paradigm for evaluating quality of care.1 Each dimension has a direct
influence on the next.

ƒ

Structural measures are used to evaluate the organizational (e.g., policies and
procedures), financial (e.g., funding of health care programs), technological (e.g., use of
computerized order entry systems), and human resources (e.g., staff training and
competency) aspects of care.106-108

ƒ

Process measures are used to assess activities involved in patient care (e.g., services for
prevention, diagnosis, and treatment). These measures may reflect the interpersonal
aspects of care/service (e.g., convenience, communication) or technical aspects of care
such as the timeliness of diagnosis, appropriateness of therapy, complications, errors,
and coordination of care.98 In some frameworks, intermediate health outcomes, such
as HbA1c as a marker for diabetes control, are included in the process category.109
Process measures are the predominant quality indicators for inpatient and ambulatory
care, although they are applied only occasionally for surgery.109 The majority of
measures target quality of care for specific health conditions, usually those determined
to have high disease burden (i.e., morbidity and mortality) in the population, highest
rates of utilization of health care services, and highest costs to the health system.110
Laboratory tests are a core component of the condition-related measure sets, i.e., to
assess whether or not the patient received the appropriate test.

p

HEDIS consists of 71 measures (e.g., whether or not a patient received beta blocker treatment after a heart attack)
across 8 domains of care (i.e., effectiveness, access/availability, satisfaction, health plan stability, use of service, cost
of care, informed health care choice, health plan descriptive information). HEDIS is designed to provide purchasers
and consumers with the information they need to reliably compare the performance of health care plans.

May 2008

211

Laboratory Medicine: A National Status Report

ƒ

Chapter V – Quality Systems and Performance Measurement

Outcome measures are used to determine change in patient health status,
experiences/satisfaction with care, and cost-related analyses.111 Health-related
outcome measures include indicators of mortality (e.g., infant death rate), physiologic
state (e.g., blood pressure), clinical events (e.g., adverse drug events), symptoms (e.g.,
level of pain), functioning (e.g., disability), and patients’ and clinicians’ satisfaction
with services (e.g., Consumer Assessment of Health Plan survey).112, 113 Outcome
measurement increasingly includes assessment of patient quality of life. Laboratory
values are important clinical endpoints in outcomes measurement.
The measures are employed to document differences in medical practices, variations in
the quality of care, and the efficacy or effectiveness of particular interventions.114, 115 In
addition, cost-related outcome measures are increasingly applied to determine
efficiency and effectiveness in the use of scarce health care resources.

This quality paradigm is an important part of the framework espoused by the IOMq and adopted
by AHRQ in the National Health Care Quality Report.116, 117 The desirable attributes of a measure
are importance in the clinical sense to the general population or the quality of care; scientific
soundness in terms of reliability, validity, and explicitness of evidence base; usability in that it has
been successfully used in the past and is compatible with other measures; feasibility to implement
with existing prototypes and considerations pertaining to data availability and cost; alignment
with leading measure sets; and comprehensiveness as part of a measure set that can reflect quality of
care for a given condition. (Refer to Appendix A for further information.)

Quality Indicators
Quality indicators are specific attributes of structure, processes, or outcomes whose measurement is
capable of providing accurate estimates of the degree to which designated standards were achieved.
Although the term “performance indicator” is often used interchangeably with “quality indicator,”
it is possible to make inferences about performance without making inferences about quality.118
Activity indicators measure the frequency of an event (e.g., screenings for cancer).97
Indicators are often based on clinical guidelines and performance standards issued by specialty
societies, government agencies, or others, but require greater specificity to be implemented.110
They are designed to convey a finding about the quality of care being provided or health
outcomes.119 In particular, they can identify problems that may need to be addressed, usually
identified by low indicator scores, statistical outliers, or unexplained variations in care.
To the extent possible, indicators should link processes and outcomes in quality assessment.
When cause-and-effect relationships or other associations are established, improvement on
process measures can be linked confidently to improvement in patient and population health.101
Linking process measures to outcome measures can demonstrate the capability of clinical
approaches to improve patient and population outcomes. Establishing such linkages between
indicators and quality or outcomes can be challenging.120-122 Table 5.2 summarizes AHRQ’s

q

Other components of the framework include the IOM’s six aims of quality (safety, effectiveness, patient-centeredness,
timeliness, efficiency, equity); the Foundation for Accountability’s four domains of consumer needs (staying healthy,
getting better, living with illness or disability, and coping with end of life); and the priority areas for quality
improvement identified by the IOM, the National Quality Forum, and CMS.

May 2008

212

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

application of performance measure characteristics to evaluate potential quality indicators for its
programs as well as that of other organizations.
Indicators can be defined by various metrics such as a rate, ratio, index, or percentage that
contributes to qualitative interpretation of a contribution to health care or outcomes.123, 124 For
dichotomous metrics, an indicator is presented as a proportion where a numerator and
denominator are defined,125 such as the proportion of diabetic patients who received HbA1c
testing. Other metrics may be continuous measures that can be averaged, such as time to an event
(e.g., TAT for a clinical chemistry test), a rate defined as a proportion within a given timeframe, or
scores on a pain or patient satisfaction scale.
Table 5.2: AHRQ application of criteria for evaluating quality indicators
Quality indicator evaluation framework criteria
1. Importance

ƒ
ƒ
ƒ
ƒ

Assesses an important leverage point for improving quality; significant to target audiences; impact on health
Opportunity for improvement, considerable variation in quality of care exists
Aspect of quality is under provider or health system control
Should not create incentives or rewards to improve without truly improving quality of care

2. Scientific acceptability

ƒ
ƒ
ƒ
ƒ
ƒ
ƒ
ƒ

Relationship to quality is based on scientific evidence
Well defined and precisely specified
Valid, measures the intended aspect of quality; accurately represents the concept being evaluated; data sources are comparable
Adequate proportion of total variation is explained by provider performance and amount of variation in measurement
is small after provider performance and patient characteristics are taken into account
Reliable, producing the same results a high proportion of time in the same population*
Precise, adequately discriminating between real differences in provider performance and reasonable sample size exists
to detect actual differences; captures all possible cases and bias related to case exclusion or limited data is minimal.
Risk adjustment is adequate to address confounding bias

3. Usability

ƒ
ƒ
ƒ
ƒ

Effective (understandable and clear) presentation and dissemination strategies exist
Statistical testing can be applied to communicate when differences in performance levels are greater than would be
expected by chance*
Has been used effectively in the past and/or have high potential for working well with other indicators currently in use
Compelling content for stakeholder decision making

4. Feasibility

ƒ
ƒ
ƒ
ƒ

Consistent construction and assessment of the measure*
Feasible to calculate; benefits exceed financial and administrative burden of implementation*
Confidentiality concerns are addressed*
Audit strategy can be implemented, quality of data is known*

* Indicates the criterion was not in the initial evaluation framework but has been addressed through incorporation in subsequent
work such as quality indicator software.
Source: Guidance for using the AHRQ quality indicators for hospital-level public reporting or payment. Rockville, MD: Agency for
Healthcare Research and Quality, 2004.

May 2008

213

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

Collecting and Reporting Data on Performance
Various instruments are employed to collect and report performance data. Some common
approaches to data collection include surveys of a targeted population; focus groups; interviews;
data abstraction during chart reviews or claims reviews; use of disease registries; reporting
through performance monitoring, patient safety, and surveillance systems; and competency
assessments. Advances in health information technology are enabling more of these data to be
collected electronically. The data derived from measurement often are used to describe patterns
of care, detect variations and deficiencies in quality and/or utilization, and perform intra-and
interorganizational comparisons. Data may be analyzed and reported at the patient, population,
and system levels depending on the purpose of the measurement.r
Measurement data are reported publicly through assessment tools such as benchmark comparisons,
report cards, and balanced scorecards. Benchmark comparisons assess a technical aspect of
performance against a designated standard or indicator. Report cards grade performance according
to established criteria and indicators. Balanced scorecards give a multidimensional view of
performance including technical quality of care, cost, access, patient satisfaction, health status,
waiting time in clinics, TAT for laboratory and radiology reports, efficiency, and clinical pathway
variances.126 Report card data pertaining to laboratory testing are generally limited to utilization.
For example, NCQA collects data on whether or not appropriate screening and other laboratory
tests for selected medical conditions were performed as an indicator of quality of care. The data are
used in annual reports on clinician and health plan performance.127

Performance Measurement Initiatives in Laboratory Medicine
A key part of performance improvement in laboratory medicine is the generation of data
substantiating the level of practice and defining where practice improvements can have the
greatest impact. For clinical and anatomic pathology, this involves collection of data on practices
at points along the TTP where risks to quality and safety are likely to arise and where corrective
action can result in learning, recovery, and improvement. Historically, this has entailed detecting
and correcting errors. However, errors are only one type of shortfall in quality, and the methods
used to select quality indicators do not appear to conform to the AHRQ criteria.128 Today’s
quality improvement initiatives have incorporated a more patient-centric, systems-based
approach desired by many stakeholders.
As noted above, formal measurement and reporting on quality in laboratory medicine have been
limited largely to QC and PT activities to meet regulatory obligations. The effort dedicated to
evaluating performance and finding and avoiding errors in the analytic phase has been greater
than that for the preanalytic and postanalytic phases. In particular, extensive data on analytic
performance has been generated for clinical pathology and certain cytology tests to comply with
regulatory and accreditation requirements. These focus on accuracy and consistency of reagents,
equipment, and/or methods through internal process control, external PT, and on-site
r

Federal, state, and private sector initiatives in performance measurement are increasingly concerned with data
collection at the level of the individual patient, which can be aggregated along three important dimensions: (1)
composite measures of the care provided to the individual patient that may document whether that person
received recommended care; (2) population-oriented results defined by socioeconomic status, race, and ethnicity;
(3) system-oriented results to identify gaps in performance and accountability at the level of the provider,
provider group, hospital, and community, and the use of composite measures.102

May 2008

214

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

inspections.129 CMS and accreditation organizations use the data generated from these activities
to determine compliance with standards for analytic processes; accreditation organizations also
use the data in training sessions to assist laboratory staff in improving their performance.130
Private and public sector organizations have sponsored research and quality improvement efforts
to address specific issues in the pre- and postanalytic phases of clinical and anatomic pathology.
Most of the studies measure specific variables (e.g., patient and specimen identification errors,
results reports correction rates) in order to discern ways to reduce errors, improve safety, and
identify best practices.41, 131-133 These research studies were not conducted with indicators that
have been evaluated against the AHRQ criteria presented previously, and are not included in
AHRQ’s National Quality Measures Clearinghouse. Nevertheless, these studies may offer useful
opportunities to assess the characteristics of quality indicators and practices that warrant further
research and development. Brief descriptions of the studies undertaken by CAP through its QProbes and Q-Tracks programs, The Joint Commission’s initiatives, and activities of other
organizations and government agencies are provided below.

College of American Pathologists
CAP launched the Q-Probes program in 1989 to establish performance measures for laboratory
quality.134 Targeted short-term studies in these programs assess the effects of specific practices
and attempt to establish benchmarks for measuring performance. Typically completed within 4
months, these studies aim to provide a snapshot of participants’ performance on QA variables,
which can be used for external peer-comparison, goal setting, and improvement. Most of the
variables are process-oriented, although a few are structural or outcome oriented. To date, CAP
has published more than 120 studies involving Q-Probes in peer-reviewed journals on a limited
number of indicators and topics. As noted above, they have not been evaluated against AHRQ’s
criteria for inclusion in the National Quality Measures Clearinghouse.135
The same holds true for the CAP Q-Tracks program initiated in 1998 for longitudinal tracking of
laboratory performance on key indicators. The program was designed to satisfy accreditation
requirements for continuous monitoring and benchmarking in clinical and anatomic pathology.
In 1999, the Q-Tracks program was approved by The Joint Commission for inclusion in its ORYXs
quality indicator monitoring system. Q-Tracks provide continuous monitoring of 12 indicators
(see Appendix B) and variables previously defined in Q-Probes studies (e.g., patient identification
accuracy, blood culture contamination, laboratory specimen acceptability). Users subscribe to the
program for a 12-month period during which they are provided with quarterly reports about
their performance as well as the practices and policies of other participants associated with
improved performance based on the non-standardized, self-reported data.133 Since the program’s
inception, about one-third of Q-Tracks indicators have been retired.t Given current efforts to
establish national, standardized performance measures across the health system that can be used
for external public reporting, the applicability of CAP Q-Tracks indicators remains unclear. CAP
measures were designed for internal process improvement purposes, and may continue to be
useful in this regard.

ORYX is a performance monitoring system developed by The Joint Commission. It was designed to collect data on
standardized core and non-core performance measures approved by The Joint Commission. In order to maintain
Joint Commission accreditation, hospitals are required to collect and submit data on some of the measures.
t
Since the inception of Q-Tracks, 17 indicators have been developed; 12 are in use and 5 have been retired.
s

May 2008

215

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

Many institutions participating in Q-Probes and Q-Tracks studies have documented performance
improvements. Even so, such improvements were based largely on self-reported, longitudinal
data of participating institutions, as opposed to comparisons to control groups, characteristic of
more rigorous scientific studies.133, 136, 137 Another factor that may have a modest effect on
performance comparisons is the degree of variability among participating laboratories in such
aspects as test menus, employee skill levels, and utilization by physicians. CAP programs adjust
for some of the differences by restricting interlaboratory comparisons to peer groupings of
laboratories that are similar with respect to unmanageable characteristics. Even so, the
adjustments are likely insufficient to counter the lack of standardization, the most relevant issue
pertaining to validity of laboratory performance measures. As a result, laboratories participating
in CAP studies are reporting error rates and other measurss that may have been determined
differently. This may in part explain the large variation in error rates among the various studies.
There appear to be no published studies evaluating CAP or other measures according to criteria
appropriate for quality indicators (e.g., health importance, scientific acceptability, usability,
feasibility) as defined by AHRQ. Similarly, the CAP measures do not appear to have been
assessed for generalizability across laboratory settings or studied for applicability and feasibility
in POLs.138 Although CAP measures were not designed for the purpose of accountability and
public reporting, they have established face validity, feasibility for data collection, and that they
are actionable.138 The CAP measures are in need of further testing to ascertain their validity,
accuracy and reliability for potential use in external reporting.
Along similar lines, in 1993, CAP initiated the Laboratory Management Index Program, a fiscal
management tool that provides peer comparisons to assist laboratories with the annual budget
process, contract negotiations, and daily operations management.139, 140 The program uses
management ratios and total standard billable tests as performance indicators to assess the three
main management-related factors affecting laboratory performance: productivity, utilization, and
cost-effectiveness. Their favorable impact notwithstanding, CAP indicators of management quality
also are based on self-reported, non-standardized data.

The Joint Commission
In addition to the ORYX monitoring system, The Joint Commission issues National Patient Safety
Goals (NPSGs) annually to advance specific improvements in patient safety by highlighting
problematic areas and prescribing expert-based solutions.141 Four of the NPSGs for health care
organizations apply directly to the laboratory (see Table 5.3). For example, based on expert opinion,
The Joint Commission states that the goal of improving patient identification accuracy can be
reached by using two patient identifiers when administering medication, collecting blood samples,
or providing other treatments. It suggests not using the patient’s room number or physical location
for identification, labeling containers for blood and other specimens in the presence of the patient,
and maintaining a sample’s identity throughout all stages of laboratory testing as methods to reach
this goal.142 Because these actions recommended by The Joint Commission are considered to be
viable means of reducing errors, they warrant further study and evaluation.
A few studies that examined areas important to The Joint Commission’s work cite modest
decreases in error rates based on high-level interventions; however, it is difficult in these studies
to establish causal relationships or evaluate an explicitly defined measure. For example, from
2002 to 2004, UCLA Clinical Laboratories studied patient identification and specimen labeling

May 2008

216

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

errors, a priority area for The Joint Commission. Laboratories implemented three patient safety
improvement strategies: (1) increased the number of phlebotomists on staff and expanded
services to 24 hours daily, (2) implemented an online electronic event reporting system, and (3)
instituted an automated processing system.143 Errors associated with mislabeled specimens
decreased significantly. In another example, three microbiology laboratories assessed the impact
of requiring “read-back” from the recipient of telephone results reports and noted reduced risk of
medical errors.144 The measures used in such studies are not necessarily validated, and it is
difficult to determine which of the interventions, if any, had a causal effect on error rates, or
whether there were other factors that influenced the observed changes in error rates.
Some researchers have experienced challenges when incorporating NPSGs into their information
systems. For example, staff at Massachusetts General Hospital analyzed its reporting of critical
values, i.e., those laboratory results indicating the patient is in imminent danger and requires
immediate initiation of appropriate therapy. The hospital uses computer applications (e.g.,
CPOE, clinical decision support systems, electronic results reporting) that apply rule-based logic
to determine which values are truly critical and which are within acceptable reference limits for
certain medical conditions and given the patient’s previous test results. However, the researchers
concluded that these more nuanced approaches to critical value reporting are constrained by the
Joint Commission’s requirement that all critical values be reported.145
In addition, results of a recent study funded by the CDC may contradict current and long-held
conventional wisdom underlying critical values reporting. For example, preliminary findings
presented at the September 2007 CLIAC meeting indicate that reporting of non-critical
potassium values (within a certain range) had a greater impact on patient treatment and
outcomes than critical values.146 This implies the need to re-evaluate traditional beliefs about
effective measures, practices and requirements to improve safety and quality.
Table 5.3: 2008 National Patient Safety Goals, Laboratory Services Program
Goal 1

Improve the accuracy of patient identification. (Introduced in 2004)

1A

Use at least two patient identifiers when providing care, treatment or services.

1B

Prior to the start of any invasive procedure, conduct a final verification process (such as a “time out”) to
confirm the correct patient, procedure and site, using active—not passive—communication techniques.

Goal 2

Improve the effectiveness of communication among caregivers. (Introduced in 2004)

2A

For verbal or telephone orders or for telephonic reporting of critical test results, verify the complete
order or test result by having the person receiving the information record and "read-back" the
complete order or test result.

2B

Standardize a list of abbreviations, acronyms, symbols, and dose designations that are not to be used
throughout the organization.

2C

Measure and assess, and if appropriate, take action to improve the timeliness of reporting, and the
timeliness of receipt by the responsible licensed caregiver, of critical test results and values.

2E

Implement a standardized approach to “hand off” communications, including an opportunity to ask
and respond to questions.

Goal 7

Reduce the risk of health care-associated infections. (Introduced in 2004)

7A

Comply with current World Health Organization (WHO) Hand Hygiene Guidelines or Centers for Disease
Control and Prevention (CDC) hand hygiene guidelines.

7B

Manage as sentinel events all identified cases of unanticipated death or major permanent loss of
function associated with a health care-associated infection.

May 2008

217

Laboratory Medicine: A National Status Report

Goal 13
13A

Chapter V – Quality Systems and Performance Measurement

Encourage patients’ active involvement in their own care as a patient safety strategy.
(Introduced in 2008)
Define and communicate the means for patients and their families to report concerns about safety and
encourage them to do so.

Note: Gaps in the numbering indicate that the Goal is inapplicable to the program or has been “retired,” usually because the
requirements were integrated into Joint Commission standards.
Source: 2008 National Patient Safety Goals, laboratory services program. Oakbrook Terrace, IL: The Joint Commission, 2007.
(Accessed November 14, 2007, at
http://www.jointcommission.org/PatientSafety/NationalPatientSafetyGoals/08_lab_npsgs.htm.)

Other Accreditation Organizations and Government Initiatives
While other laboratory accrediting organizations do not collect and evaluate specific measures
in the manner of CAP or The Joint Commission, some are developing alternative methods of
tracking performance. In 2007, ASHI developed an online database of the deficiencies reported
from inspections to assist laboratories it accredits with fulfillment of CMS requirements. AABB
requires blood banks to implement QMS with internal processes for monitoring and addressing
transfusion practices as a condition for accreditation. Monitored performance indicators are
related to ordering practices, patient identification, sample collection and labeling, infectious
and noninfectious adverse events, near-miss events, usage and discard practices,
appropriateness of use, blood administration policies, ability to meet patient needs, and
compliance with peer-review recommendations.42
Several federal health agencies have undertaken efforts to support development of laboratory
performance measures, including AHRQ, CDC, and CMS. AHRQ funded the creation of a
national anatomic pathology errors database in 2002 to track the frequency and severity of
errors detected by cytologic-histologic correlation for both gynecologic and nongynecologic
cytologic and histologic diagnoses.147
CDC launched a project in 2006 to develop an evidence-based process to identify and evaluate
laboratory medicine practices and to recommend “best practices.” The first phase of the project
(2006-2007) completed initial development of the Laboratory Medicine Best Practices review and
evaluation methods, and the second phase (2007-2008) is intended to refine and improve methods
to review these practices, including development of a new investigational component to obtain
unpublished studies to supplement the available published studies. The investigational
component will rely on establishing a network of laboratories who have previously completed
practice evaluations to pilot test the process. These methods are being developed under a contract
with Battelle Memorial Institute with the guidance of a 14-member multi-disciplinary expert
advisory workgroup that will also make recommendations relating to an organizational structure
and other requirements for future implementation and sustainability of a laboratory medicine
best practices evaluation and identification process, among other things.u CDC also announced a
funding opportunity in 2007 to evaluate clinical laboratory practice by developing evidence-based
laboratory medicine quality and performance measures associated with the pre- and postanalytic
u

Specifically, phase I of the project (2006-2007) established definitions of best practices, developed inclusion and
exclusion criteria for candidate practices, created a classification scheme, established methods for a systematic
review, created a framework for making best practices recommendations, conducted a proof of concept exercise,
and defined strategic organizational and implementation constructs. Phase II (2007-2008) aims to refine and
develop process review and evaluation methods, pilot test the process, develop and test the investigational
component, and evaluate alternative organizational mechanisms for implementing the process.

May 2008

218

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

stages of the TTP. These measures are to be based on important gaps and opportunities for
improvement in laboratory medicine quality consistent with available evidence and national
health care priorities to improve public health.148 To date, cooperative agreements have been
awarded to the following entities to develop measures for the pre- and postanalytic stages:

ƒ

Texas Department of State Health Services—linking the quality of patient care with the
quality of pre- and postanalytic stages of the newborn screening process

ƒ

Kaiser Permanente Center for Health Research Research Institute—evaluating laboratory
medicine quality in chronic kidney disease associated with improved patient outcomes

ƒ

University of Colorado—associated with specific pathology and laboratory processes
affecting clinical and patient-related outcomes149

CMS maintains the OSCAR database of information collected by surveyors during laboratory
inspections.150 Another database houses information on laboratory PT results. The database
systems are not accessible to the public; however, CMS will provide information by request.
While CMS has not undertaken performance measurement initiatives directly assessing the
quality of laboratory medicine, it recently launched the Physician Quality Reporting Initiative,
which provides a financial incentive for physicians to participate in this voluntary quality
reporting program. For example, physicians reporting from July 1 through December 31, 2007
were eligible for an incentive payment in 2008. One of the measures pertains to physician
ordering of laboratory tests.151

A Model for a National Laboratory Performance Measurement System
In 2006, CDC funded an initiative led by Raj Rehal in association with the National Quality Forum
(NQF) to develop a framework for collecting, measuring, and reporting performance data on
laboratory tests associated with priority health conditions, processes of care, and infrastructure.138
As a first step, the study examined the strength of evidence for health condition-specific
guidelines and associated laboratory-related performance measures recommended by
professional societies.v The priority health conditions include diabetes, obesity, heart failure,
ischemic health disease, stroke, kidney disease, hypertension, depression and mental illness,
pneumonia, cervical and colon cancer, pregnancy and childbirth, asthma, and patient safety.
For example, the clinical guidelines published in the Seventh Report of the Joint National
Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure recommended
a set of laboratory tests prior to initiating therapy: blood glucose, hematocrit, potassium,
creatine and estimated glomerular filtration rate, calcium, low-density lipoprotein cholesterol,
high-density lipoprotein cholesterol, triglycerides, and urinalysis. Tests for urinary albumin
excretion or albumin/creatine ratio are described as optional.152 However, according to the
NQF study, laboratory-based performance measures to assess the quality of laboratory testing
and services have not been specified by professional societies. The lack of such measures was
consistent across each health condition reviewed in the study.

v

The professional societies included in the NQF study are: the American Diabetes Association, American College of
Cardiology, American Heart Association, The Joint Commission, Hospital Quality Alliance, National Committee for
Quality Assurance, Physician Consortium for Performance Improvement of the American Medical Association.

May 2008

219

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

Subsequent to this review, NQF commissioned the development of a framework based on the
structure established for the AHRQ National Health Care Quality Report. In addition, the
framework is consistent with the IOM reports, Priority Areas for National Action: Transforming
Health Care Quality (2003) and Performance Measurement: Accelerating Improvement (2006). NQF
drew from the design principles espoused in the latter report, including:
(1) Comprehensive measurement that advances the six aims identified in the IOM Quality
Chasm report
(2) Longitudinal measurement that spans care settings over time
(3) Individual patient-level, population-level, and system-level measurement instead of a
provider-specific or silo-of-care focus
(4) Shared accountability instead of focusing on individuals’ actions
The framework is depicted in Figure 5.1. The intention of this approach is to address needs of
multiple stakeholders and link evidence-based laboratory tests with national initiatives in
performance measurement. It allows for data collection on specific tests associated with priority
conditions at the level of the individual patient, which can be rolled up to the levels of the
populations and systems. The framework addresses laboratory performance in key dimensions
of the TTP: ordering, specimen, analysis, reporting, and follow-up.
Figure 5.1: A Framework for performance measurement in laboratory medicine.

Source: Behal R. Identification of performance measures of importance for quality in laboratory medicine.
Washington, DC: National Quality Forum, 2007.

The framework was designed with flexibility for evaluation of both clinical and anatomic pathology
testing processes. However, several limitations of the current framework have been identified.138
First, all but one of the laboratory tests associated with the health conditions are clinical pathology
tests. The exception is cancer screening for women, which includes gynecologic cytology tests.
Anatomic pathology tests may not be sufficiently represented. However, the extensive research on

May 2008

220

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

anatomic pathology tests associated with cancer could provide sufficient evidence base for selecting
specific indicators. Second, there are few, if any, measures for children with special needs, frailty
due to old age, mental illness, self management, care coordination, care at the end of life, and pain
management. Third, few laboratory tests that are considered evidence-based are included in the
performance measures. Fourth, measures to evaluate appropriateness of laboratory tests (e.g.,
overuse, underuse, misuse) have not been developed. Generally, HEDIS and other conditionrelated measures are used as substitutes to determine underuse; only a few studies of clinical
decision support systems relative to physician test ordering and payer utilization reviews have been
employed to assess overuse or misuse.153, 154

Current Measures Used in Laboratory Medicine
Specific areas for evaluation and development of performance measures in laboratory medicine
are depicted in Figure 5.2. Structural measures are affiliated with core environmental factors that
may affect quality. Process measures assess the preanalytic, analytic, and postanalytic
components of the TTP, also considered to be the critical control points at which errors and other
problems can be corrected and/or prevented. Outcome measures tie the laboratory testing
process to patient health and costs of care. The facets identified in the structural, process, and
outcome categories may be used for internal or external assessment and quality improvement
programs, as depicted by the arrows in the diagram. Each structural, process, and outcome
measure also may be evaluated independently. The status of the measures is discussed more
fully in the next section. Appendix B provides examples of specific structural, process, and
outcome measures identified in the literature.

Structural Measures
With the exception of PT, most of the regulatory provisions to support quality systems mandated
under CLIA are structural requirements. The majority of provisions outline basic requirements for
policies, procedures, and documentation that must be in place for preanalytic, analytic, and
postanalytic phases as a condition of certification/accreditation.
A few studies have examined other structural measures such as workforce productivity and staffing
ratios and use of web-based error reporting systems. A study examined the impact of workforce on
quality and factors associated with favorable staffing ratios. They measured productivity of
technical staff and management staff span-of-control ratios. The study found wide variability in
staffing levels among institutions, suggesting opportunities to improve staff productivity in many
facilities.155 In studies of health or laboratory information technology infrastructure, basic metrics
are developed to determine the rates of implementation, often delineated by initiation of specific
software programs, cross-departmental data exchange networks, and database systems.9
Another study of POCT systems evaluated the implementation of specific error-prevention
systems and safeguards, most of which are structural factors influencing quality because they are
associated with technology systems and workforce training. The study produced a summary of
actions for preventing medical errors: (1) adopt operator certification and validation in POCT
testing; (2) implement security, validation, performance, and emergency override systems on
existing and new devices; (3) require flexible, user-defined error prevention system options on
instruments as a prerequisite to federal licensing of new diagnostic tests and devices; (4) integrate
connectivity standards for bidirectional information exchange; (5) preserve fast TAT of POCT

May 2008

221

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

Figure 5.2: Diagram of Categories for Performance Indicators in Laboratory Medicine
Structural
Measures

Outcome
Measures

Process
Measures
Total Testing Process

•
•
•
•
•
•
•

Policies
Procedures
Practices
Volume
Workforce
Access
Technology

PREANALYTIC
• Physician test
orders
• Patient
identification
• Specimen
collection
• Specimen
labeling
• Specimen
delivery

ANALYTIC
• Accessioning
• Specimen
preparation
• Specimen
analysis
(PV, PT, False
negative/
positive) 1
• Report
verification

POSTANALYTIC
• TAT
• Critical value
reporting
• Report accuracy
&completeness
• Report delivery
• Physician
follow-up

HEALTH
• Mortality
• Morbidity
• Adverse
events
• Biomarkers

HUMANISTIC
& OTHER
• Quality of
life
• Functional
status
• Patient
satisfaction
• Provider
satisfaction

ECONOMIC
• Cost per test
• Budget impact
• Cost
effectiveness
• Cost utility
(e.g., cost per
QALY)
• Cost benefit

Internal Assessment

External Assessment
• CAP
(QT&QP)2
• AABB
• ASHI
• COLA
1
2
3

•
•
•
•
•

AHRQ
CMS
CDC
JC
ARCs3

PV-Predictive Value, PT-Proficiency Testing
QT-QTracks, QP-QProbes
ARCs-Academic research centers

May 2008

222

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

results; (6) monitor invalid use, operator competence, quality compliance, and other performance
improvement indices to reduce errors.156
Most structural measures are insufficient or too general to serve as indicators of performance or
quality of care .116 Links between structure and processes of care and structure and outcomes
tend to have adequate evidence-based support only in limited areas (e.g., mortality at high
volume hospitals is up to 10% lower than at lower volume facilities).157, 158 Furthermore, structural
measures have limited use outside the hospital setting, are not readily actionable, and do not
adequately discriminate performance among individual clinicians. With the exception of
laboratory medicine, national initiatives assessing quality have shifted focus to outcome-validated
process measures and disease- or process-specific outcome measures.116

Process Measures
Process-related performance measures in laboratory medicine are associated with the TTP as
presented in Figure 5.2 and Appendix B, and discussed at length in the previous chapter of this
report (Quality and the Total Testing Process). Thus far, the non-standardized methods
employed to evaluate quality and estimate error rates have formed a growing body of research on
all three phases of the laboratory testing cycle. Error rates and a few other dimensions for certain
aspects of pre- and postanalytic phases of testing have been evaluated through the CAP programs
and other research studies.159 Studies of the preanalytic processes typically measure performance
via error rates tied to specimen collection (e.g., phlebotomy success, specimen acceptability,
specimen contamination), followed by specimen labeling (e.g., ABO typing or various errors), and
patient identification (e.g., wristband error).160-165 A few published studies have examined errors
associated with clinician ordering, in terms of the accuracy and completeness of requisition slips
and/or the appropriateness of test orders (e.g., duplicate orders).166
For postanalytic processes, performance measures have targeted measurement of TAT in various
capacities (i.e., routine TAT, critical value TAT, and stat TAT) and report accuracy (e.g.,
completeness of descriptors, discrepancies, amended report rates).134, 167-172 More recent studies
measured interpretive consultation rates and report delivery errors.173-175 Very few studies have
examined appropriate interpretation of laboratory test results and ensuing laboratory-driven
clinical/preventive actions and related patient outcomes.
Most published studies of preanalytic and postanalytic factors calculated total error rates, but not
all collected sufficient data to determine the nature of and specific causes of the errors for
identification of potentially effective quality improvement strategies. For example, in a Q-Tracks
study of specimen acceptability, the overall specimen rejection rate served as the primary
performance indicator, with secondary data submitted on specific reasons for rejection such as for
clotted specimen, container leaking, specimen contamination, hemolyzed specimen, insufficient
volume, tube over/underfilled, specimen lost/not received, and improper container.133
In contrast to pre- and postanalytic measures, analytic phase process measures are better
developed and accompanied by a stronger evidence base, such as for error rates as measured by
internal QC and PT. Routine QC checks can provide important data about the potential for
accuracy versus the potential for error in testing and sources of variation for each analyte. These
data are tracked over time to evaluate within-laboratory factors as well as to ensure achieving
specified levels of performance. Similarly, PT is a standard measure of analytic performance for

May 2008

223

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

purposes of external evaluation and regulatory oversight. PT data, including root causes of
errors, serve as key measures to provide insight into how well a laboratory is performing
compared to other laboratories. (Refer to the Federal Regulatory Oversight chapter of this report
for additional discussion of QC and PT.)
Figure 5.3 summarizes the process-related performance measures. Experts participating in CDC’s
Institute for Quality in Laboratory Medicine in 2005 expanded on the measures initially employed
by CAP through its Q-Tracks program. CDC recently undertook an evaluation of process
measures published in the literature and found a general lack of evidence supporting health
importance, scientific soundness, relevance, and usefulness.176 Significant variability and gaps
were identified in terms of standardized terminology, measurement specifications, data collection
methods, and relationships to process, clinical, health, and economic outcomes. For example,
although specimen adequacy can affect the accuracy and usefulness of laboratory test results,
there is no standard definition or measure to evaluate adequacy aside from overall rejection
rates.160-162, 177 A systematic study that relates specimen adequacy or rejection to other outcomes
has not been conducted.
When CDC assessed the process measures for their applicability to the IOM’s six domains of
health care quality, they fell short, especially in the areas of patient-centeredness and equity.
Other areas not adequately addressed include laboratory testing for preventive care and the use
of health information technology (e.g., order entry and decision support) as a tool to enhance
quality and safety.178-181
Overall, an important limitation of current laboratory process measures is the variable extent to
which they link to outcome measures, along with sample size constraints and potential to be
affected by such confounding factors as patient compliance. Some measures can be linked to
outcomes (e.g., false-positive and false-negative results) supported with scientific evidence, while
the relationship for others is weak (e.g., patient identification, specimen collection, TAT).109
Additional work is needed to standardize the indicator metrics (i.e., numerators and
denominators, or other methods for calculation) for pre-and postanalytic process-related
performance measures. As most research to date has focused on the needs of larger, hospitalbased laboratories, further research also is needed to ascertain the specific challenges that smaller
POLs may confront when implementing process-related performance measurement and quality
improvement programs.
In efforts to address these issues, the Behal/NQF framework presents a different approach to
performance measurement. The particular indicators for the preanalytic and postanalytic phases
of the total testing process have yet to be evaluated using AHRQ criteria; however, the paper
identified general areas for additional evaluation that included test ordering, specimen collection,
proficiency testing, results reporting, and physician actions. Rather than measure performance in
the broad sense, evaluations are proposed based on laboratory tests associated with the national
priority health conditions.138 This approach may facilitate understanding of the relationships
between laboratory testing and outcomes.

May 2008

224

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

Table 5.3: Potential Areas for Performance Measure Development and Standardization
CAP (2002), Howanitz (2005),
and IQLM (2005)

NQF (2007)

Preanalytic

Preanalytic Phase

ƒ
ƒ
ƒ

Patient identification

ƒ
ƒ

Specimen rejection (chemistry &
hematology)

Analytic Phase

Test order accuracy and appropriateness

Test order
Specimen collection

ƒ

Blood product wastage(transfusion
medicine)

ƒ

ƒ

Blood culture contamination
(microbiology)

Postanalytic Phase

ƒ

Adequacy of specimen container
information

Analytic Phase

ƒ
ƒ
ƒ

Cervical cytology-biopsy correlation
Accuracy of POCT
Proficiency testing

Postanalytic Phase

ƒ
ƒ
ƒ

Test TATs (chemistry & hematology)

ƒ

Clinician satisfaction (all lab disciplines)

Critical values reporting
Clinician follow-up (abnormal Pap test,
hypercalcemia)

Additional special areas

ƒ
ƒ
ƒ
ƒ

Diabetes monitoring
Hyperlipidemia screening

ƒ
ƒ

Specimen analysis (proficiency testing)

Results reporting (TAT, critical values)
Physician follow-up

Specific health condition-related tests

ƒ
ƒ
ƒ
ƒ
ƒ
ƒ
ƒ
ƒ
ƒ
ƒ
ƒ
ƒ
ƒ

Diabetes
Obesity
Heart failure
Ischemic heart disease
Stroke
Kidney disease
Hypertension
Depression and mental illness
Pneumonia
Cervical and colon cancer screening
Pregnancy and childbirth
Asthma
Patient safety

Timing of therapeutic drug monitoring
Intraoperative consultations

Sources: Zarbo RJ, Jones BA, Friedberg RC, Valenstein PN, Renner SW, Schifman RB, et al. Q-tracks: a College of American
Pathologists program of continuous laboratory monitoring and longitudinal tracking. Arch Pathol Lab Med 2002;126(9):1036-1044.
Howanitz PJ. Errors in laboratory medicine: practical lessons to improve patient safety. Arch Pathol Lab Med 2005;129:1252-1261.
Hilborne L. Quality Indicators Workgroup presentation. 2005 Institute for Quality in Laboratory Medicine Conference: Recognizing
Excellence in Practice. April 29, 2005. Atlanta, GA: Centers for Disease Control and Prevention, 2005.
Behal R. Identification of performance measures of importance for quality in laboratory medicine. Washington, DC: National Quality
Forum, 2007.

May 2008

225

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

Outcome Measures and Indicators
As is the case for other types of health care interventions, laboratory testing can be assessed for its
impact on health outcomes, humanistic outcomes, and economic outcomes.138 Health outcomes
include mortality, morbidity, adverse events, and biomarkers (e.g., laboratory values). Some
biomarkers are used as intermediate or surrogate outcomes, in that they are known to be
predictive of health outcomes. Humanistic outcomes typically include quality of life, functional
status, patient satisfaction, and other patient-reported outcomes. Among the many types of
economic outcomes are cost per test, patient, treatment and episode of care; budget impact; and
analyses of tradeoffs, including cost-effectiveness, cost-utility, and cost-benefit analysis.
Outcome studies in laboratory medicine address questions of the form: Is use of test X associated
with outcome Y?182 For example, does fecal occult blood testing decrease the incidence of colorectal
cancer? These studies have focused on the usefulness of screening tests in the prevention and
early detection of disease, such as various cancers, and comorbidities associated with certain
conditions (e.g., diabetes), among others.183-185 For example, a systematic review examined the
evidence that screening and earlier treatment are effective in reducing morbidity and mortality
associated with Type II diabetes. The review found that, for high-risk populations, screening tests
can detect diabetes in its preclinical phase, and that over the 10-15 years after clinical diagnosis,
tight glycemic control can improve patient outcomes by reducing the risk for blindness and endstage renal disease.186 The USPSTF relies on outcome studies of this nature to develop evidencebased guidelines for priority health conditions.
Outcome studies also have identified adverse events arising from incorrect test results. These
studies assess the relationships between analytic accuracy (i.e., false-negative or false-positive
results) to detrimental effects on patient health (e.g., disease progression that would have otherwise
been discovered or unnecessary surgery).187-189 An example concerns a study in which 12 women
were diagnosed with having postgestational choriocarcinoma on the basis of false-positive test
results for human chorionic gonadotropin.190 The errors had adverse consequences for patient
outcomes, as most of these women were subject to unnecessary surgery and chemotherapy.
Other outcome studies examined provider or patient satisfaction with aspects of care. For
example, an ongoing Q-Tracks study is using two high-level measures, i.e., overall patient
satisfaction score and percentage of patients more than satisfied, to assess satisfaction with
outpatient specimen collection.191 A Q-Probes study relied on more detailed data collection to
assess patient satisfaction, including aggregate scores, percentage of excellent/good ratings,
below average/poor ratings, and satisfaction for 10-13 specific aspects of laboratory service.192
While patient outcome studies are gradually becoming more common in laboratory medicine,
they are not yet a regular part of ongoing quality improvement practices. Laboratory-related
outcome measurement is underused for several reasons, including the high cost of capturing
outcomes data, lack of standardization of data collection and reporting methods, and lack of
agreement regarding appropriate analysis of data (e.g., whether or not to risk-adjust data).
Furthermore, outcome measurement can be severely constrained by sample size, test results
missing from patients’ medical records, limited risk adjustment capability with data abstracted
from administrative records, and higher cost to abstract data from medical records.109 Other
problems consistently identified in the literature include the inability to conceal the identity of

May 2008

226

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

tested versus non-tested patients, the relative remoteness of an outcome to the test itself, and the
number of patients or volunteers required for a study to achieve statistical significance.114, 115, 182, 193
Intermediate outcomes association with biomarkers, such as HbA1c and glucose for diabetes, are
generally easier to measure than patient outcomes to assess the impact of health care
interventions and thus, are more commonly used. A biomarker is a variable that is objectively
measured as an indicator of normal biological processes, pathogenic processes, or
pharmacological response to a therapeutic intervention. A surrogate (or intermediate) endpoint is
a biomarker or physiological marker that is intended to act as a substitute for or predict a patient
outcome. They are relatively quickly or easily measured and their use is based on evidence that it
is a reliable predictor of the patient outcome of interest. Certainly, some biomarkers are better
predictors of patient outcomes than others. Examples are: blood pressure or cholesterol levels for
predicting the incidence and course of cardiovascular disease, T-cell counts for predicting survival
of AIDS patients, and PSA levels for predicting the incidence and course of prostate cancer.

Cost-related Measures and Indicators
Cost-related outcome assessment is growing in use, although relatively little research has been
conducted as pertains to laboratory testing.102, 194 An example of such studies in laboratory
medicine is one that assessed the cost-effectiveness of periodic, population-based dipstick
screening for early detection of urine protein in adults with neither hypertension nor diabetes and
in adults with hypertension. Using a Markovw decision analytic model, the study compared a
strategy of annual screening with no screening (usual care) for proteinuria at age 50 years
followed by treatment with an angiotensin-converting enzyme inhibitor or an angiotensin IIreceptor blocker. Among the findings, for persons with neither hypertension nor diabetes, the
cost-effectiveness ratio for screening vs. no screening (usual care) was an unfavorable $282,818 per
QALY,x but $53,372 for persons age >60 years, and a highly favorable $18,621 per QALY for
persons with hypertension. The analysis showed that factors that tend to improve costeffectiveness of screening for the general population include a greater incidence of proteinuria,
age >60 years at screening, and lower frequency of screening. The investigators concluded that
early detection of urine protein to slow progression of chronic kidney disease and decrease
mortality is not cost-effective unless selectively directed toward high-risk groups (older persons
and persons with hypertension) or conducted at an infrequent interval of 10 years.198
Another study modeled the cost-effectiveness of routinely using tandem mass spectrometry to
screen newborns for inborn errors of metabolism based on data from a large health maintenance
organization. Depending upon assumptions in three scenarios, the cost per QALY ranged from
$736 to $11,419. These findings led the investigators to conclude that costs per QALY saved by

The Markov model is a statistical state-transition model in which the transition probabilities depend only on the
current state and not on previous states or the pathway by which the state was entered.195
x The QALY is a composite unit of length of life and utility for particular levels of quality of life. Ones’ utility for quality
of life is measured on a scale from 1.0 (utility for perfect health) to death (0.0). For example, 1 year of life lived in
perfect health (utility of 1.0) health yields 1.0 QALYs, 0.5 years of life lived in perfect health yields 0.5 QALYs, and 1
year of life lived in a lesser state of health (e.g., bedridden) with a utility of 0.5 is also equivalent to 0.5 QALYs. The
QALY may be used as the unit of patient/user outcomes in a cost-effectiveness (or cost-utility) analysis.196, 197 In
developed economies, a health care intervention is considered to be cost effective if its cost per QALY falls under a
certain threshold, (e.g., $30,000 per QALY or $50,000 per QALY or $100,000 per QALY). In the U.S., such thresholds
are generally regarded as informal only, and do not have bearing on payment decisions.
w

May 2008

227

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

tandem mass spectrometry for inborn errors of metabolism compare favorably with other mass
screening programs such as for breast and prostate cancer.199
Several studies have examined the cost of POCT relative to central laboratory testing using
metrics such as cost-per-test (including estimates for the cost of equipment, supplies, labor, and
other variables) and laboratory test TAT.200, 201 For example, a study reported in 2004 compared
the analytical costs of central laboratory glucose testing and semiautomated bedside glucose
testing (BGT) among 445 institutions enrolled in the Q-Probes program. Results showed different
distributions of costs across three main types of sites. The median (10th-90th percentile range)
analytical costs per glucose test were $1.18 dollars ($0.36-$5.59) for central laboratories, $1.96
($0.77-$9.51) for high-volume BGT sites, and $4.66 ($1.02-$27.54) for low-volume BGT sites. In
addition to being higher than costs for central laboratories, costs for BGT were highly variable and
dependent on volume. The investigators observed that data that would be used for financial
justification for BGT were widely aberrant and in need of improvement.202

CONCLUSIONS
To achieve consistently high levels of quality in laboratory medicine requires moving beyond
analytic-focused QC and PT activities to more comprehensive, systematic approaches to quality
management that support the integration of preanalytic, analytic, and postanalytic components,
external assessment and accountability. An important tool of QMS and improvement programs is
the use of performance measurement to assess achievement of quality standards for structural
features that support quality, processes of care, and health outcomes.

ƒ

Relative to the five levels of quality,y most health care organizations, including
laboratories, are operating at or below the stage of QA, although some have
implemented and are working successfully at the level of QMS. Organizations that
implement QMS are better equipped to reduce or eliminate errors, meet customer
needs, score well on government or accreditation assessments, and maintain quality
objectives. CLSI and ISOz have developed standards for QMS.

ƒ

By adopting ISO 9000 standards, QMS has been most broadly adopted in transfusion
medicine to meet FDA requirements for Current Good Manufacturing Practices and
Quality Assurance Guidelines. Use of ISO 9001:2000, ISO 15189, and CLSI guidelines in
laboratory medicine has accelerated in recent years as CMS and laboratory accreditation
organizations incorporate QMS-related standards into their regulatory requirements.

ƒ

CQI, Toyota “lean” production, Six Sigma, and FMEA are strategic tools for
implementing QMS that are realizing benefits among early adopters, from small
physician office laboratories to large reference laboratories. Use of CQI and Six Sigma
has contributed to financial savings and decreased TAT, lean production has improved
test quality and reduced errors, and FMEA has decreased time to report critical
laboratory values.

The CLSI five tiers of quality are: (1) QC; (2) QA, including PT; (3) QMS; (4) quality cost management; and (5) TQM,
known as CQI in health care.
z International Organization for Standardization (ISO) has developed two standards for QMS: the more general ISO
9000 series and ISO 15189, which is focused on health care. CLSI guidelines HS1-A2 and GP26-A3 facilitate the
implementation of these standards.
y

May 2008

228

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

ƒ

The great bulk of effort on formal performance measurement and improvement has been
devoted substantially to the analytic phase, rather than the pre- and postanalytic phases.
Public and private sector organizations have sponsored research and other initiatives to
address some aspects of the deficit of information on pre- and postanalytic quality.
However, the indicators used to date have not been uniformly defined or assessed for
generalizability, and are subject to wide variation in their implementation.

ƒ

The emphasis of provisions under CLIA is largely structural, outlining policies,
procedures, and documentation requirements as a condition for accreditation and
certification. Aside from PT and corresponding false-negative and positive rates, process
measures to assess the quality in the TTP remain relatively underdeveloped and are
selected based on expert opinion rather than evidence-based outcomes and gaps in
quality. Typically, research studies rely on high-level calculations of error rates (e.g.,
specimen rejection, specimen labeling, TAT). The evidence pertaining to the impact of
laboratory testing on outcome measures― health outcomes, humanistic outcomes, and
economic outcomes― remains sparse. A small body of research has examined such areas
as the costs of the analytic phase of particular tests and the adverse consequences of
incorrect test results.

Gaps, Needs, and Challenges:
ƒ

Stakeholders in laboratory medicine must address organizational obstacles to the
implementation of QMS in order to achieve higher levels of quality, including
resistance to culture change, lack of leadership and staff commitment to QMS, and
insufficient funding of QMS activities.

ƒ

Most research on the adoption and results of laboratory quality to date has focused on
the larger, hospital-based laboratories. Further research is needed to examine the
specific challenges that smaller laboratories and POLs may confront when implementing
process-related performance measurement and quality improvement programs.

ƒ

Substantial work is needed to support the selection, development and standardization
of pre- and postanalytic process-related performance measures that are important to
health care quality and patient-related outcomes, and satisfy minimum criteria such as
those used by AHRQ’s National Quality Measures Clearinghouse. Data collection,
analysis, and reporting methods also need to be standardized.

ƒ

The evidence for the impact of laboratory medicine on patient outcomes, humanistic
outcomes, and economic outcomes must be augmented. The lack of a substantial and
evolving body of such evidence diminishes the ability to assess and demonstrate the
value of laboratory medicine. Taking on this effort is essential in a health care market
that increasingly demands evidence of value for adoption, use, and payment,
especially for new health care technologies.

May 2008

229

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

REFERENCE LIST
1. Institute of Medicine. Medicare: a strategy for quality assurance. Washington, DC:
National Academy Press, 1990.
2. Institute of Medicine. Crossing the quality chasm: a new health system for the 21st
century. Washington, DC: National Academy Press, 2001.
3. Brennan TA, Leape LL, Laird NM, et al. Incidence of adverse events and negligence in
hospitalized patients: results of the Harvard medical practice study I. Quality and Safety
in Health Care 2004;13:145-52.
4. Laboratory requirements. Code of Federal Regulations, Title 42, Part 493.
5. Shahangian S. Proficiency testing in laboratory medicine: uses and limitations. Arch
Pathol Lab Med 1998;122(1):15-30.
6. Johnson PR. The contribution of proficiency testing to improving laboratory performance
and ensuring quality patient care. Clin Leadersh Manag Rev 2004;18(6):335-41.
7. Edson D, Massey LD. Proficiency testing performance in physician's office, clinic, and
small hospital laboratories, 1994-2004. Laboratory Medicine 2007;38(4):237.
8. Ehrmeyer SS, Laessig RH. Has compliance with CLIA requirements really improved
quality in US clinical laboratories? Clinica Chimica Acta 2004;346(1):37-43.
9. Institute of Medicine. Patient safety: achieving a new standard of care. Washington, DC:
National Academy Press, 2004.
10. O'Leary D. Patient safety: instilling hospitals with a culture of continuous improvement.
Testimony before the Senate Committee on Governmental Affairs, 108th Congress, 1st
Session. 2003.
http://www.jointcommission.org/NewsRoom/OnCapitolHill/testimony_061104.htm.
11. Quality management systems (QMS): the premiere accreditation program for medical
laboratories. COLA, 2004. (Accessed March 24, 2008, at http://www.cola.org/qms.html.)
12. Quality management in clinical laboratories: promoting patient safety through risk reduction
and continuous improvement. Northfield: College of American Pathologists, 2005.
13. Greer N, Munk L, Brown T. AABB briefs members on new quality systems template.
AABB, (Accessed March 24, 2008, at
http://www.aabb.org/Content/Meetings_and_Events/Annual_Meeting_and_TXPO/60
amonline/60amtuetem.htm.)
14. About CLSI. Wayne, PA: Clinical and Laboratory Standards Institute, 2007. (Accessed
May 8, 2007, at http://www.clsi.org/AM/Template.cfm?Section=About_CLSI.)
15. Cianfrani CA, Tsiakals JJ, West JE. The ASQ ISO 9000:2000 handbook. Milwaukee, WI:
American Society for Quality, 2002.
16. Deming WE. Out of the crisis. Cambridge, MA: MIT Press, 1986.
17. Juran JM. Quality by design: the new steps for planning quality into goods and services.
New York, NY: Free Press, 1992.

May 2008

230

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

18. A quality management system model for health care. Approved guideline: HS1-A2;
second edition. Wayne, PA: Clinical Laboratory Standards Institute, 2004.
19. McDonough JE, Solomon R, Petosa L. Quality improvement and proactive hazard analysis
models: deciphering a new Tower of Babel. In: Patient safety: achieving a new standard
for care. Aspden P, Wolcott J, Erickson SM, et al., ed. Washington, DC: National Academy
Press, 2004.
20. Institute of Medicine. Building a better delivery system: a new engineering/health care
partnership. Washington, DC: National Academy Press, 2005.
21. Wieman TJ, Wieman EA. A systems approach to error prevention in medicine. J Surg
Oncol 2004;88:115-21.
22. Continuous quality improvement: integrating five key quality system components.
Approved guideline: GP22-A2; second edition. Wayne, PA: Clinical and Laboratory
Standards Institute, 2004.
23. Feistritzer NR, Keck BR. Perioperative supply chain management. Semin Nurse Manag
2007;8(3):151-7.
24. Fone D, Hollinghurst E, Temple M, et al. Systematic review of the use and value of
computer simulation modeling in population health and health care delivery. Journal of
Public Health in Medicine 2003;25(4):325-35.
25. Leatherman S, Berwick D, Iles D, et al. The business case for quality: case studies and an
analysis. Health Aff (Millwood) 2003;22(2):17-30.
26. Padhi N. The eight elements of TQM. iSixSigma.com, 2002. (Accessed May 4, 2007, at
http://www.isixsigma.com/library/content/c021230a.asp.)
27. Wilson L, Goldschmidt P. Quality management in health care. Sydney, Australia:
McGraw-Hill Book Company, 1995.
28. Garber CC, Kaufman HW. Quality systems for the clinical laboratory in the 21st century.
In: Clinical diagnostic technology: the total testing process. Volume 3: the postanalytical
phase. Ward-Cook K, Lehman CA, Schoeff LE, Williams R.H., ed. Washington, DC:
American Association for Clinical Chemistry, 2006.
29. Westgard JO. Basic planning for quality: training in analytical quality management for
healthcare laboratories. Madison, WI: Westgard QC, Inc., 2000.
30. ISO in brief. Geneva, Switzerland: International Organization for Standardization, 2006.
(Accessed May 8, 2007, at http://www.iso.org/iso/en/prodsservices/otherpubs/pdf/isoinbrief_2006-en.pdf.)
31. Seddon J. Chapter 1: a brief history of ISO 9000. Buckingham, UK: Vanguard Education,
2006. (Accessed May 16, 2007, at http://www.lean-service.com/3-1-article.asp.)
32. ISO 9001:2000; quality management systems--requirements. Geneva, Switzerland:
International Organization for Standardization, 2005. (Accessed May 17, 2007, at
http://www.iso.org/iso/en/CatalogueDetailPage.CatalogueDetail?CSNUMBER=21823&
ICS1=3&ICS2=120&ICS3=10.)

May 2008

231

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

33. Understanding ISO/IEC 17025:2005, A2LA specific applications of additions to the
standard. Frederick, MD: American Association for Laboratory Accreditation, 2007.
(Accessed May 10, 2007, at http://www.a2la.org/faq/faqfinder170252005.cfm.)
34. Levett JM. Implementing an ISO 9001 quality management system in a multispecialty
clinic. Physician Exec 2005;31(6):46-51.
35. The ISO survey 2006. Geneva, Switzerland: International Organization for
Standardization, 2007. http://www.iso.org/iso/survey2006.pdf.
36. Crago MG. Centers for Medicare and Medicaid (formerly HCFA) now require Medicare
program safeguard contractors to attain ISO certification. American Society for Quality
Health Care Division Newsletter 2002:3-4.
37. ISO 15189;2007; medical laboratories--particular requirements for quality and competence.
Geneva, Switzerland: International Organization for Standardization, 2007. (Accessed
May 16, 2007, at
http://www.iso.org/iso/en/CatalogueDetailPage.CatalogueDetail?CSNUMBER=42641&
showrevision=y.)
38. Sterry D. Clinical and Laboratory Standards Institute. Personal communication.
March 10, 2008.
39. Application of a quality management system model for laboratory services. Approved
guideline: GP26-A3; third edition. Wayne, PA: Clinical Laboratory Standards Institute,
2004.
40. Smith DM, Otter J. Performance improvement in a hospital transfusion service. Arch
Pathol Lab Med 1999;123:585-91.
41. Roseff SD. Regulatory and quality initiatives--trends in transfusion medicine.
LabMedicine 2005;36(2):108-14.
42. Standards for blood banks and transfusion services. 24th ed. Bethesda, MD: American
Association of Blood Banks, 2006.
43. Yost J. CLIA and quality systems. COLA Symposium for Clinical Laboratories--an
interactive experience with quality systems. May 2, 2007. Columbia, MD: COLA, 2007.
44. Commission building bridges and turning out tools. CAP Today 2003.
45. Commission on Laboratory Accreditation Laboratory Accreditation Program: laboratory
general checklist. Northfield, IL: College of American Pathologists, 2006.
http://www.cap.org/apps/docs/laboratory_accreditation/checklists/laboratory_general
_october2006.pdf.
46. College of American Pathologists offers ISO accreditation for the nation's medical
laboratories. Northfield, IL: College of American Pathologists, 2008. (Accessed March 7,
2008, at
http://www.cap.org/apps/cap.portal?_nfpb=true&cntvwrPtlt_actionOverride=%2Fportl
ets%2FcontentViewer%2Fshow&_windowLabel=cntvwrPtlt&cntvwrPtlt%7BactionForm.c
ontentReference%7D=laboratory_accreditation%2Fiso%2Fnewsrel_iso.html&_state=maxi
mized&_pageLabel=cntvwr.)

May 2008

232

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

47. COLA launches new interactive e-learning courses for QMS training, 14 new QMS courses
offered. Columbia, MD: Commission on Laboratory Accreditation, 2005. (Accessed May 8,
2007, at http://www.cola.org/news.html?NewsID=124.)
48. Southwick K. Creating a common language for labs. CAP Today 2003.
49. Doris AK. Standardization and accreditation of clinical laboratories in the U.S. The
Biochemist 2004:44-45.
50. Blumenthal D, Kilo CM. A report card on continuous quality improvement. Milbank
Quarterly 1998;76(4):625-48, 511.
51. O'Connor GT, Plume SK, Olmstead EM, et al. A regional intervention to improve the
hospital mortality associated with coronary artery bypass graft surgery. The Northern
New England Cardiovascular Disease Study Group. JAMA 1996;275(11):841-6.
52. Cantwell R, Mirza N, Short T. Continuous quality improvement efforts increase operating
room efficiency. Journal of Healthcare Quality 1997;19(6):32-6.
53. Kollef MH, Shapiro SD, Silver P, et al. A randomized, controlled trial of protocol-directed
versus physician-directed weaning from mechanical ventilation. Crit Care Med
1997;25(4):567-74.
54. Austin T, Jarvis C. How to save time, money, and a precious resource via CQI. MLO Med
Lab Obs 1998;30(1):50-2.
55. Saxena S, Wong ET. Does the emergency department need a dedicated stat laboratory?
Continuous quality improvement as a management tool for the clinical laboratory. Am J
Clin Pathol 1993;100(6):606-10.
56. Nardella A, Pechet L, Snyder LM. Continuous improvement, quality control, and cost
containment in clinical laboratory testing. Effects of establishing and implementing
guidelines for preoperative tests. Arch Pathol Lab Med 1995;119(6):518-22.
57. Kilgore ML, Steindel SJ, Smith JA. Continuous quality improvement for point-of-care
testing using background monitoring of duplicate specimens. Arch Pathol Lab Med
1999;123(9):824-8.
58. Jacobs E, Hinson KA, Tolnai J, Simson E. Implementation, management and continuous
quality improvement of point-of-care testing in an academic health care setting. Clinica
Chimica Acta 2001;307(1-2):49-59.
59. Moscucci M, Rogers EK, Montoye C, et al. Association of a continuous quality
improvement initiative with practice and outcome variations of contemporary
percutaneous coronary interventions. Circulation 2006;113(6):814-22.
60. Wooten MJ. Integrating CQI and QA--continuous quality improvement; complete quality
assessment. MLO Med Lab Obs 1994;26(1):33-6.
61. Garber CC. Six sigma: its role in the clinical laboratory. Clinical Laboratory News 2004.
62. Nevalainen D, Berte L, Kraft C, Leigh E, Picaso L, Morgan T. Evaluating laboratory
performance on quality indicators with the six sigma scale. Arch Pathol Lab Med
2000;124(4):516-9.

May 2008

233

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

63. Westgard JO. Six Sigma basics: outcome measurement of process performance. Madison,
WI: Westgard QC, 2001. (Accessed November 12, 2007, at
http://www.westgard.com/lesson66.htm.)
64. Westgard JO, Westgard SA. The quality of laboratory testing today: an assessment of
sigma metrics for analytic quality using performance data from proficiency testing
surveys and the CLIA criteria for acceptable performance. Am J Clin Pathol
2006;125(3):343-54.
65. Nevalainen DE. The quality systems approach. Arch Pathol Lab Med 1999;123(7):566-8.
66. Schroeder R. Six sigma, the breakthrough management strategy revolutionizing the
world's top corporations. 1st ed. New York, NY: Currency, 1999.
67. Lusky K. Lean, mean--the evergreen hunt for efficiency. CAP Today 2004.
68. Hospital lab uses Six Sigma to reduce turnaround time and improve quality. Rochester,
NY: ValuMetrix Services, 2007. (Accessed May 7, 2007, at
http://www.orthoclinical.com/Products/pdf/WHC_CaseStudy.pdf.)
69. Riebling NB, Condon S, Gopen D. Toward error free lab work. ASQ Six Sigma Strategy
Forum Magazine 2004;4(1):23-29.
70. Liker JK. The Toyota Way: 14 management principles from the world's greatest
manufacturer. New York, NY: McGraw-Hill, 2004.
71. Ohno T. Toyota production system: beyond large-scale production. Portland, OR:
Productivity Press, 1988.
72. Persoon TJ, Zaleski S, Frerichs J. Improving preanalytic processes using the principles of
lean production (Toyota Production System). Am J Clin Pathol 2006;125(1):16-25.
73. Raab SS, Andrew-Jaja C, Condel JL, Dabbs DJ. Improving Papanicolaou test quality and
reducing medical errors by using Toyota production system methods. Am J Obstet
Gynecol 2006;194(1):57-64.
74. Raab SS, Grzybicki DM, Sudilovsky D, Balassanian R, Janosky JE, Vrbin CM. Effectiveness
of Toyota process redesign in reducing thyroid gland fine-needle aspiration error. Am J
Clin Pathol 2006;126(4):585-92.
75. Gharib H, Goellner JR. Fine-needle aspiration biopsy of the thyroid: an appraisal. Ann
Intern Med 1993;118(4):282-9.
76. Miller JM, Hamburger JI, Kini S. Diagnosis of thyroid nodules. Use of fine-needle
aspiration and needle biopsy. JAMA 1979;241(5):481-4.
77. Caruso D, Mazzaferri EL. Fine needle aspiration biopsy in the management of thyroid
nodules. Endocrinologist 1991;1:194-202.
78. Novis DA, Konstantakos G. Reducing errors in the practices of pathology and laboratory
medicine: an industrial approach. Am J Clin Pathol 2006;126(Suppl. 1):S30-S35.
79. The launch of Shared Visions-New Pathways. Jt Comm Perspect 2004;24(1):1-3.

May 2008

234

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

80. DeRosier J, Stalkhandske E, Bagian JP, Nudell T. Using health care failure mode and effect
analysis: the VA National Center for Patient Safety's Prospective Risk Analysis System.
Journal on Quality Improvement 2002;28(5):248-67.
81. Traynor K. Safety analyses take on engineer's perspective at JCAHO's urging. HealthSystem Pharmacy News 2007.
82. Brown DS, Bonacum D, Vonderheide-Liem D. The Kaiser Permanente FMEA model-simplified for healthcare personnel. Journal of Healthcare Quality 2005;27(1):48-55.
83. Day S, Dalto J, Fox J, Turpin M. Failure mode and effects analysis as a performance
improvement tool in trauma. J Trauma Nurs 2006;13(3):111-7.
84. Burgmeier J. Failure mode and effect analysis: an application in reducing risk in blood
transfusion. Jt Comm J Qual Improv 2002;28(6):331-9.
85. Capunzo M, Cavallo P, Boccia G, Brunetti L, Pizzuti S. A FMEA clinical laboratory case
study: how to make problems and improvements measurable. Clinical Leadership and
Management Review 2004;18(1):37-41.
86. Sheridan-Leos N, Schulmeister L, Hartranft S. Failure mode and effect analysis: a
technique to prevent chemotherapy errors. Clin J Oncol Nurs 2006;10(3):393-8.
87. FMEA (failure mode analysis): a new QI tool to help improve case management processes.
Hosp Case Manag 2003;11(3):33-6.
88. Krouwer JS, Hatjimihail AT, Jacobs E, et al. Risk management techniques to identify and
control laboratory error sources. Proposed guideline: EP18-P2, second edition. Wayne:
Clinical and Laboratory Standards Institute, 2007.
89. Saxena S, Kempf R, Wilcox S, et al. Critical laboratory value notification: a failure mode
effects and criticality analysis. Joint Commission Journal on Quality and Patient Safety
2005;31(9):495-506.
90. Dock B. Improving the accuracy of specimen labeling. Clin Lab Sci 2005;18(4):210-2.
91. Gras JM, Philippe M. Application of the Six Sigma concept in clinical laboratories: a
review. Clin Chem Lab Med 2007;45(6):789-96.
92. Zarbo RJ, D'Angelo R. Transforming to a quality culture: the Henry Ford production
system. Am J Clin Pathol 2006;126(Suppl 1):S21-S29.
93. Estioko-Taimuri T. The makeover of the Lakeshore General Hospital laboratories. Clinical
Leadership and Management Review 2006;20(1):E5.
94. Westgard S. Quality management cocktail: ISO, Lean, and Six Sigma. Madison, WI:
Westgard QC, 2005. (Accessed July 11, 2007, at http://www.westgard.com/guest30.htm.)
95. Landek D. Con: six sigma not always the right answer in the clinical laboratory. Clinical
Leadership and Management Review 2006;20(5):E3.
96. Daley AT. Pro: lean six sigma revolutionizing health care of tomorrow. Clinical
Leadership and Management Review 2006;20(5):E2.

May 2008

235

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

97. Marshall MN, Roland MO, Campbell SM, et al. Measuring general practice: a
demonstration project to develop and test a set of primary care clinical quality indicators.
Santa Monica, CA: RAND Corporation, 2003.
98. Institute of Medicine. Measuring the quality of health care. Washington, DC: National
Academy Press, 1999.
99. Institute of Medicine. To err is human: building a safer health system. Washington, DC:
National Academy Press, 2000.
100. Silverstein M. An approach to medical errors and patient safety in laboratory medicine.
Institute for Quality in Laboratory Medicine: Making the Laboratory a Key Partner in
Patient Safety. April 13, 2003-April 15, 2003. Atlanta, GA: Centers for Disease Control and
Prevention, 2003.
101. McGlynn EA. Choosing and evaluating clinical performance measures. The Joint
Commission Journal on Quality Improvement 1998;24(9):470-9.
102. Institute of Medicine. Performance measurement: accelerating improvement.
Washington, DC: National Academy Press, 2006.
103. NCQA timeline. Washington, DC: National Committee for Quality Assurance, 2005.
(Accessed May 23, 2007, at http://www.ncqa.org/about/timeline.htm.)
104. Rosenthal MB, Fernadopulle R, Song HR, Landon B. Paying for quality: providers'
incentives for quality improvement. Health Aff (Millwood) 2004;23(2):127-41.
105. National Quality Forum: publications. Washington, DC: The National Quality Forum,
2007. (Accessed May 10, 2007, at http://www.qualityforum.org/.)
106. Donabedian A. Evaluating the quality of medical care. Milbank Mem Fund Q 1966;
44:166-203.
107. Aday LA, Begley CE, Lairson DR, Balkrishman R. Evaluating the healthcare system:
effectiveness, efficiency, and equity. Third ed. Washington, DC: Academy Health, 2004.
108. What does Leapfrog ask hospitals? Washington, DC: The Leapfrog Group, 2007. (Accessed
March 14, 2007, at
http://www.leapfroggroup.org/for_consumers/hospitals_asked_what.)
109. Birkmeyer JD, Ker EA, Dimick JB. Improving the quality of quality measurement. In:
Performance measurement: accelerating improvement. Washington, DC: National
Academy Press, 2006.
110. McGlynn EA, Asch SM. Developing a clinical performance measure. Am J Prev Med
1998;14(3S):14-21.
111. Donabedian A. The quality of care: how can it be assessed? JAMA 1988;260(12):1743-8.
112. Outcomes research. Fact sheet. AHRQ publication no. 00-P011. Rockville, MD: Agency for
Healthcare Research and Quality, 2000. (Accessed July 11, 2007, at
http://www.ahrq.gov/clinic/outfact.htm.)
113. Aller RD. The construction and usage of clinical databases. In: Laboratory-related
measures of patient outcomes: an introduction. Bissell MG, ed. Washington, DC:
American Association for Clinical Chemistry, 2000.

May 2008

236

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

114. Bissell MG, Jones H. The laboratory's role in outcome assessment. MLO Med Lab Obs
1998;30(1):54, 56-4, 58.
115. Bissell MG. Introduction: what's in a laboratory outcome? In: Laboratory-related measures
of patient outcomes: an introduction. Bissell MG, ed. Washington, DC: American
Association for Clinical Chemistry, 2000.
116. Institute of Medicine. Envisioning the National Health Care Quality Report. Washington,
DC: National Academy Press, 2001.
117. 2006 National Healthcare Quality Report. Report No. 07-0013. Rockville, MD: Agency for
Healthcare Research and Quality, 2006.
118. Buck D, Godfrey C, Morgan A. Performance indicators and health promotion targets.
York, UK: Center for Health Economics, University of York, 1996.
119. Lawrence M, Olesen F. Indicators of quality in health care. European Journal of General
Practice 1997;3:103-8.
120. Eddy DM. Performance measurement: problems and solutions. Health Aff (Millwood)
1998;17(4):7-25.
121. Gross PA, Braun BI, Kritchevsky SB, Simmons BP. Comparison of clinical indicators for
performance measurement of healthcare quality: a cautionary note. Clin Perform Qual
Health Care 2000;8(4):202-11.
122. James BC, Hammond EH. The challenge of variation in medical practice. Arch Pathol Lab
Med 2000;124:1001-3.
123. Hoeizer S, Waechter W, Stewart A, Liu R, Schweiger R. Towards case-based performance
measures: uncovering deficiencies in applied care. J Eval Clin Pract 2001;7(4):355-63.
124. Galvin RS. Are performance measures relevant? Health Aff (Millwood) 1998;17(4):29-31.
125. Rubin HR, Pronovost P, Diette GB. From a process of care to a measure: the development
and testing of a quality indicator. Int J Qual Health Care 2001;13(6):489-96.
126. Luttman RJ. Next generation quality, part 2: balanced scorecards and organizational
improvements. Top Health Inf Manage 1998;19(2):22-9.
127. Report cards. Washington, DC: National Committee for Quality Assurance, 2007.
(Accessed November 14, 2007, at http://web.ncqa.org/tabid/60/Default.aspx.)
128. Howanitz PJ, Walker K, Bachner P. Quantification of errors in laboratory reports: a quality
improvement study of the College of American Pathologists' Q-Probes program. Arch
Pathol Lab Med 1992;116:694-700.
129. Bock JL. Quality of the specimen and the analytical system. In: Clinical diagnostic
technology: the total testing process. Volume 2: the analytical phase. Ward-Cook K,
Lehman CA, Schoeff LE, Williams RH, ed. Washington, DC: American Association of
Clinical Chemistry, 2003.
130. Clinical lab quality: CMS and survey organization oversight should be strengthened.
Report No. GAO-06-416. Washington, DC: Government Accountability Office, 2006.
www.gao.gov/cgi-bin/getrpt?GAO-06-416.

May 2008

237

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

131. Hollensead SA, Lockwood WB, Elin RJ. Errors in pathology and laboratory medicine:
consequences and prevention. J Surg Oncol 2004;88:161-81.
132. Stankovic AK. The laboratory is a key partner in assuring patient safety. Clin Lab Med
2004;24:1023-35.
133. Zarbo RJ, Jones BA, Friedberg RC, et al. Q-Tracks: a College of American Pathologists
program of continuous laboratory monitoring and longitudinal tracking. Arch Pathol Lab
Med 2002;126(9):1036-44.
134. Novis DA, Walsh MK, Dale JC, Howanitz PJ. Continuous monitoring of stat and routine
outlier turnaround times: two College of American Pathologists Q-Tracks monitors in 291
hospitals. Arch Pathol Lab Med 2004;128:621-6.
135. QPC publications listing. Northfield, IL: College of American Pathologists, 2007.
136. Novis DA, Walsh M, Wilkinson D, St LM, Ben-Ezra J. Laboratory productivity and the rate
of manual peripheral blood smear review: a College of American Pathologists Q-Probes
study of 95,141 complete blood count determinations performed in 263 institutions. Arch
Pathol Lab Med 2006;130(5):596-601.
137. Howanitz PJ. Errors in laboratory medicine: practical lessons to improve patient safety.
Arch Pathol Lab Med 2005;129:1252-61.
138. Behal R. Identification of performance measures of importance for quality in laboratory
medicine. Washington, DC: National Quality Forum, 2007.
http://216.122.138.39/pdf/projects/lab-med/txlabpaper_behal_final%2005-21-07.pdf.
139. Reilly B. LMIP: the next generation in productivity measurement. MLO Med Lab Obs 1993.
140. About LMIP. Northfield, IL: College of American Pathologists, 2006. (Accessed
July 16, 2007, at
http://www.cap.org/apps/cap.portal?_nfpb=true&cntvwrPtlt_actionOverride=%2Fportl
ets%2FcontentViewer%2Fshow&_windowLabel=cntvwrPtlt&cntvwrPtlt%7BactionForm.c
ontentReference%7D=lmip%2Flmip_desc.html&_state=maximized&_pageLabel=cntvwr.)
141. 2007 National Patient Safety Goals. Oakbrook Terrace, IL: The Joint Commission, 2007.
(Accessed May 9, 2007, at http://www.jointcommission.org/NR/rdonlyres/0515980F1262-438A-8DB9-C2EA28C9BB33/0/07_lab_npsgs.pdf.)
142. Potential 2008 National Patient Safety Goals and requirements, laboratory version.
Oakbrook Terrace, IL: The Joint Commission, 2007. (Accessed May 9, 2007, at
http://www.jointcommission.org/NR/rdonlyres/60B84AD5-A41B-4DE6-9C22C9DFAF99ED48/0/08_potential_LAB_NPSG.pdf.)
143. Wagar EA, Tamashiro L, Yasin B, Hilborne L, Bruckner DA. Patient safety in the clinical
laboratory: a longitudinal analysis of specimen identification errors. Arch Pathol Lab Med
2006;130(11):1662-8.
144. Barenfanger J, Sautter RL, Lang DL, Collins SM, Hacek DM, Peterson LR. Improving
patient safety by repeating (read-back) telephone reports of critical information. Am J Clin
Pathol 2004;121(6):801-3.
145. Dighe AS, Rao A, Coakley AB, Lewandrowski KB. Analysis of laboratory critical value
reporting at a large academic medical center. Am J Clin Pathol 2006;125(5):758-64.

May 2008

238

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

146. Grzybicki DM. Assessment of best practices for standardized quality assurance activities
in pathology and laboratory medicine. Atlanta: Centers for Disease Control and
Prevention, 2007.
147. National anatomic pathology errors database can reduce hospital pathology errors.
Rockville, MD: Agency for Healthcare Research and Quality, 2007. (Accessed July 13, 2007,
at http://www.ahrq.gov/research/jan06/0106RA1.htm.)
148. Evidence-based laboratory medicine: quality/performance measure evaluation. Atlanta,
GA: Centers for Disease Control and Prevention, 2007. (Accessed December 12, 2007, at
http://www.cdc.gov/od/pgo/funding/CI07-710.htm.)
149. Tracking accountability in government grants system: award actions (CFDA number
93064). Department of Health and Human Services, 2007. (Accessed December 13, 2007, at
http://taggs.hhs.gov/CFDAAwardsList.cfm?&WhereString=ACTION_CFDA_LINK.CF
DA_NUM%20IN('93064')%20AND%20AWD_BUD_ACT.DATA_FY=2007&WhereText=C
FDA%20Number%20%3D%2093064%3Cbr%3EFiscal%20Year%20%3D%202007.)
150. Jasmin K. Centers for Medicare and Medicaid Services. Personal communication. July 13,
2007.
151. Medicare announces measure specifications for the physician quality reporting
initiative. Baltimore, MD: Centers for Medicare and Medicaid Services, 2007. (Accessed
July 11, 2007, at
http://www.cms.hhs.gov/apps/media/press/release.asp?Counter=2103&intNumPerPa
ge=10&checkDate=&checkKey=2&srchType=2&numDays=90&srchOpt=0&srchData=ph
ysician%20quality%20reporting&keywordType=All&chkNewsType=1%2C+2%2C+3%2C
+4%2C+5&intPage=&showAll=1&pYear=1&year=2007&desc=&cboOrder=date.)
152. Chobanian AV, Bakris GL, Black HR, et al. The seventh report of the Joint National
Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure.
Bethesda: National Heart, Lung, and Blood Institute, National Institutes of Health, 2003.
http://www.nhlbi.nih.gov/guidelines/hypertension/jnc7full.pdf.
153. van Walraven C, Naylor CD. Do we know what inappropriate laboratory utilization is? A
systematic review of laboratory clinical audits. JAMA 1998;280:550-8.
154. Bates DW, Kuperman G, Rittenberg E, et al. A randomized trial of a computer-based
intervention to reduce utilization of redundant laboratory tests. Am J Med 1999;106:144-50.
155. Valenstein PN, Souers R, Wilkinson DS. Staffing benchmarks for clinical laboratories: a
College of American Pathologists Q-Probes study of staffing at 151 institutions. Arch
Pathol Lab Med 2005;129:467-73.
156. Kost GJ. Preventing medical errors in point-of-care testing: security, validation,
performance, safeguards, and connectivity. Arch Pathol Lab Med 2001;125:1307-15.
157. Dudley RA, Johansen KI, Brand R, Rennie DJ, Milstein A. Selective referral to high-volume
hospitals: estimating potentially avoidable deaths. JAMA 2000;283(9):1159-66.
158. Halm EA, Lee C, Chassin MR. Is volume related to outcome in health care? A systematic
review and methodologic critique of the literature. Ann Intern Med 2002;137(6):511-20.

May 2008

239

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

159. Novis DA. Detecting and preventing the occurrence of errors in the practices of laboratory
medicine and anatomic pathology: 15 years' experience with the College of American
Pathologists' Q-Probes and Q-Tracks programs. Clin Lab Med 2004;24:965-78.
160. Dale J, Novis D. Outpatient phlebotomy success and reasons for specimen rejection: a QProbes study. Arch Pathol Lab Med 2002;126:416-9.
161. Jones BA, Calam RR, Howanitz PJ. Chemistry specimen acceptability: a College of
American Pathologists Q-Probes study of 453 laboratories. Arch Pathol Lab Med
1997;121:19-26.
162. Jones BA, Meier F, Howanitz PJ. Complete blood count specimen acceptability. A College
of American Pathologists Q-Probes study of 703 laboratories. Arch Pathol Lab Med
1995;119:203-8.
163. Howanitz PJ, Schifman RB. Phlebotomists' safety practices: a College of American
Pathologists Q-Probes study of 683 institutions. Arch Pathol Lab Med 1994;118:957-62.
164. Valenstein PN, Raab SS, Walsh MK. Identification errors involving clinical laboratories: a
College of American Pathologists Q-Probes study of patient and specimen identification
errors at 120 institutions. Arch Pathol Lab Med 2006;130:1106-13.
165. Howanitz P, Renner S, Walsh M. Continuous wristband monitoring over 2 years decreases
identification errors: a college of American Pathologists Q-Tracks study. Arch Pathol Lab
Med 2002;126:809-15.
166. Nakhleh RE, Zarbo RJ. Surgical pathology specimen identification and accessioning: a
College of American Pathologists Q-Probes study of 1,004,115 cases from 417 institutions.
Arch Pathol Lab Med 1996;120:227-33.
167. Steindel SJ, Novis DA. Using outlier events to monitor test turnaround time: a College of
American Pathologists Q-Probes study in 496 laboratories. Arch Pathol Lab Med
1999;123:607-14.
168. Steindel SJ, Jones BA. Routine outpatient laboratory test turnaround times and practice
patterns: a College of American Pathologists Q-Probes study. Arch Pathol Lab Med
2002;126(1):11-8.
169. Valenstein P, Walsh M. Five-year follow-up of routine outpatient test turnaround time: a
College of American Pathologists Q-Probes study. Arch Pathol Lab Med 2003;127:1421-3.
170. Jones BA, Valenstein PN, Steindel SJ. Gynecologic cytology turnaround time: a College of
American Pathologists Q-Probes study of 371 laboratories. Arch Pathol Lab Med
1999;123:682-6.
171. Gephardt GN, Baker PB. Interinstitutional comparison of bladder carcinoma surgical
pathology report: a College of American Pathologists Q-probes study of 7,234 bladder
biopsies and curettings in 268 institutions. Arch Pathol Lab Med 1995;119:681-5.
172. Zarbo RJ. Interinstitutional assessment of colorectal carcinoma surgical pathology report
adequacy: a College of American Pathologists Q-Probes study of practice patterns from
532 laboratories and 15,940 reports. Arch Pathol Lab Med 1992;116:1113-9.
173. Lim EM, Sikaris KA, Gill J, et al. Quality assessment of interpretative commenting in
clinical chemistry. Clin Chem 2004;50(3):632-7.

May 2008

240

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

174. Laposata ME, Laposata M, Van Cott EM, Buchner DS, Kashalo MS, Dighe AS. Physician
survey of a laboratory medicine interpretive service and evaluation of the influence of
interpretations on laboratory test ordering. Arch Pathol Lab Med 2004;128:1424-7.
175. Sirota RL. Error and error reduction in pathology. Arch Pathol Lab Med 2005;129:1228-33.
176. Shahangian S, Snyder SR. Laboratory medicine quality indicators: a review of the
literature. Centers for Disease Control and Prevention, In preparation.
177. Stark A, Jones BA, Chapman D, et al. Clinical laboratory specimen rejection--association
with the site of patient care and patients' characteristics. Arch Pathol Lab Med
2007;131:588-92.
178. Sheridan SL, Harris RP, Woolf SH. Shared Decision-making Workgroup of the U.S.
Preventive Services Task Force. Am J Prev Med 2004;26:56-66.
179. Shahangian S. Laboratory-based health screening: perception of effectiveness, biases,
utility, and informed/shared decision making. MLO Med Lab Obs 2006;37:210-6.
180. Bu D, Pan E, Walker J, et al. Benefits of information technology-enabled diabetes
management. Diabetes Care 2007;30:1137-42.
181. Rothschild JM, Mcgurk S, Honour M, et al. Assessment of education and computerized
decision support interventions for transfusion practices. Transfusion 2007;47:228-39.
182. Bruns DE. Laboratory-related outcomes in healthcare. Clin Chem 2001;47(8):1547-52.
183. Scheid DC, McCarthy LH, Lawler FH, Hamm RM, Reilly KE. Screening for
microalbuminuria to prevent nephropathy in patients with diabetes: a systematic review
of the evidence. J Fam Pract 2001;50(8):661-8.
184. Harris R, Lohr KN. Screening for prostate cancer: an update of the evidence for the U.S.
Preventive Services Task Force. Ann Intern Med 2002;137(11):917-29.
185. Allison JE, Lawson M. Screening tests for colorectal cancer: a menu of options remains
relevant. Current Oncology Reports 2006;8(6):492-8.
186. Harris R, Donahue K, Rathore SS, Frame P, Woolf SH, Lohr KN. Screening adults for type
2 diabetes: a review of the evidence for the U.S. Preventive Services Task Force. Ann
Intern Med 2003;138(3):215-29.
187. False-negative Pap test rate high. Newsline (People with AIDS Coalition of New York)
1999;31.
188. Sawaya GF, Sung HY, Kinney W, Kearney KA, Miller MG, Hiatt RA. Cervical cancer after
multiple negative cytologic tests in long-term members of a prepaid health plan. Acta
Cytol 2005;49(4):391-7.
189. Leyden WA, Manos MM, Geiger AM, et al. Cervical cancer in women with comprehensive
health care access: attributable factors in the screening process. J Natl Cancer Inst
2005;97(9):675-83.
190. Rotmensch S, Cole LA. False diagnosis and needless therapy of presumed malignant
disease in women with false-positive human chorionic gonadotropin concentrations.
Lancet 2000;355:712-5.

May 2008

241

Laboratory Medicine: A National Status Report

Chapter V – Quality Systems and Performance Measurement

191. QT7--satisfaction with outpatient specimen collection. Northfield, IL: College of American
Pathologists, 2005. (Accessed November 14, 2007, at http://www.cap.org.)
192. Zarbo RJ, Nakhleh RE, Walsh M. Customer satisfaction in anatomic pathology: a College
of American Pathologists Q-Probes study of 3065 physician surveys from 94 laboratories.
Arch Pathol Lab Med 2003;127:23-9.
193. Beck JR. Pathologist as a designer of process improvement. CAP Foundation Conference
VIII. Patient-Centered Pathology Practice: Outcomes and Accountability. February 2,
1997. St. Petersburg, FL: College of American Pathologists, 1997.
194. Institute of Medicine. Valuing health for regulatory cost-effectiveness analysis.
Washington DC: National Academy Press, 2006.
195. Haddix AC, Teutsch SM, Corso PS. Prevention effectiveness: a guide to decision analysis
and economic evaluation. Oxford, United Kingdom: Oxford University Press, 2003.
196. Patrick DL, Erickson P. Health status and health policy. New York, NY: Oxford University
Press, 1993.
197. Drummond MF, O'Brien B, Stoddart GL, Torrance GW. Methods for the economic
evaluation of health care programmes. Oxford, England: Oxford Medical Publications, 1997.
198. Boulware LE, Jaar BG, Tarver-Carr ME, Brancati FL, Powe NR. Screening for proteinuria
in US adults: a cost-effectiveness analysis. JAMA 2003;290(23):3101-14.
199. Schoen EJ, Baker JC, Colby CJ, To TT. Cost-benefit analysis of universal tandem mass
spectrometry for newborn screening. Pediatrics 2002;110(4):781-6.
200. Felder RA. Robotics and automated workstations for rapid response testing. Am J Clin
Pathol 1995;104(4 Suppl 1):S26-S32.
201. De Cresce RP, Phillips DL, Howanitz PJ. Financial justification of alternate site testing.
Arch Pathol Lab Med 1995;119(10):898-901.
202. Howanitz PJ, Jones BA. Comparative analytical costs of central laboratory glucose and
bedside glucose testing: a College of American Pathologists Q-Probes study. Arch Pathol
Lab Med 2004;128(7):739-45.

May 2008

242

Laboratory Medicine: A National Status Report

Chapter VI – Laboratory Information Systems

CHAPTER VI
LABORATORY INFORMATION SYSTEMS
Laboratory information systems (LIS) have evolved over the past 30 years from simple systems
designed to generate accurate reports to complete systems that can link laboratory data “end to
end” across the TTP, including clinician-related pre- and postanalytic activities (e.g., test selection,
interpretive consultation). Health information technology (HIT) and Web-based applications
have enabled dramatic improvements in the ways in which laboratories communicate, provide
services, educate their workforce and clients, market themselves to clients, and track clinical data
and information.1 Health care organizations have played a key role in advancing such
communication by linking the LIS with hospital information systems, pharmacy database
systems, etc.
As a strategic tool, the LIS should enhance quality and efficiency of health care professionals,
allowing them to deliver high quality, cost-effective service.2 In fact, several studies of urban and
rural facilities have reported a direct association between the implementation of an LIS and
improved financial performance.3-5
This chapter provides a brief overview of the basic structure of LISs in integrated delivery systems
(IDS) and POLs. Particular attention was given to data management capabilities and clinical
practice applications of interest to public and private sector stakeholders as they pursue
comprehensive health information systems integration. In addition, the chapter discusses
important informatics issues that continue to inhibit data exchange between applications both
internal and external to clinical laboratories.

STRUCTURE OF LIS
Today, the LIS is a complex computer system of clinical and administrative applications programs
with widely varying configurations in different types of laboratories (see Figure 6.1).6 An LIS may
be a single, integrated software package running on one or more database systems. It also may be
comprised of different modules (e.g., specimen accessioning, chemistry tests, and microbiology
tests), often supplied by different vendors. Larger organizations may operate several LISs. For
example, one LIS may be designated for general laboratory services, another for transfusion
medicine, and a third for anatomic pathology. The traditional preference for interfaced,a
heterogeneous, best-of-breed systems appears to be shifting to integrated, single-source systems.7

a

An interface is a point of interconnection between a computer terminal and a network or between two networks.

May 2008

243

Laboratory Medicine: A National Status Report

Chapter VI – Laboratory Information Systems

Figure 6.1: Integrated Comprehensive Laboratory Information System
Medical Reference
Database

Pharmaceutical
Manufacturer

Biotech Company

Human Genome
Database Provider

In Vitro Diagnostic
Manufacturer

Secured Virtual Private Network
and Wide Area Network
Hospitals
Reference
Laboratories

Physicians’
Offices
Remote
Laboratories

Core
Laboratory

Group Clinics,
HMOs

Servers and Integrated LIS Software
Order Entry/CPOE/Accessioning

Automated Systems Interfacing

Links to Pharmacy Database

Barcode Specimen Labeling

POCT

Billing Systems

Specimen Tracking

Digital Pathology

Supplies Management

Applications For:

Links to Knowledge Systems

Patient Safety/Error Reporting

Results Reporting

Public Health Surveillance

Links to Electronic Health Records

Online Education

•
•
•
•

Clinical Pathology
Microbiology
Transfusion Medicine
Anatomic Pathology

Interface/Exchange Engine
Adapted from: Cooper SD. The role of the laboratory information system in diagnostic services. In: Clinical diagnostic
technology: the total testing process. Volume 1: the preanalytic phase. Ward-Cook KM, Lehmann CA, Schoeff LE, Williams RH,
ed. Washington, DC: AACC Press, 2003.

Integrated Systems and Large Laboratories
An IDS is a group of health care service units (e.g., hospitals, outpatient clinics, ambulatory
surgery centers, long-term care facilities, physicians’ offices, and hospital, POL and reference
laboratories) that provide a spectrum of health care services across a geographic region.8 The LIS
may operate across the network or group of networks in an IDS, although occasionally some
operate in a closed or proprietary environment.
Within the IDS, a “core” laboratory serves as the primary testing site for the network. Smaller,
rapid response laboratory systems operate locally where needed.9 While networking models vary
by testing menus and location, the core laboratory usually performs most non-stat (non-urgent)
and high complexity testing, including outpatient and outreach settings.8 In essence, the core
laboratory functions as a reference laboratory serving both internal and external users.

May 2008

244

Laboratory Medicine: A National Status Report

Chapter VI – Laboratory Information Systems

The inter-laboratory connections are established by either a single LIS with multifacility function or
multiple LISs that interface with the core laboratory.8 Multifacility systems typically rely on the
computing hardware at one main data center and a common application interface standard such as
health level 7 (HL7) for data exchange. Although all users can access the system, restrictions
according to patient, provider, and facility identifier can be applied. Hardware standardization can
increase efficiency in multifacility systems; however, this must be weighed against costs of replacing
legacy systems. The multi-LIS model incorporates a central application interface engine in the IT
architecture that routes and translates messages among the disparate systems based on predefined
rules. Each site preserves its own security and access rules. While this option allows organizations
to retain legacy systems, integration (via standardization and consolidation) requires major efforts
to redefine certain database elements and standards across the different systems.
Linkages between hospital information systems and LISs are critically important to institutional
systems and integrated delivery systems. The most “wired” interconnections are ordering of tests
and reporting of results, claims and billing for tests, and links to the inpatient pharmacy database.
The American Hospital Association (AHA) reported that, in 2007, 78% of hospitals had electronic
reporting of laboratory results and 72% had laboratory test electronic order entry, either by the
physician or within the laboratory.10 According to AHA, connectivity for laboratory reporting
and ordering functions ranked on par with those for radiology (77% for reporting and 70% for
ordering respectively), and ahead of pharmacy-related order entry (61%).
The VHA provides a real-world example of an integrated system, among the many in the public
and private sector. In 1996, the VHA reorganized its 173 hospitals into 22 Veterans Integrated
Service Networks (VISNs); each VISN is a regional health care system that provides a continuum
of services to veterans residing in a specific geographic area.11, 12 Laboratory Electronic Data
Interchange software consolidates electronic laboratory test ordering and results reporting
throughout all VHA medical care facilities within and between VISNs.13 The VHA and Military
Health System (i.e., TRICARE) are redesigning their networks to allow health information to be
shared between the systems. Two of their demonstration projects specifically involve the LIS: the
Bidirectional Health Information Exchange allows two-way exchange of health information,
including laboratory results on shared patients in text format; and the Laboratory Data Sharing
Interface application facilitates electronic transfer and sharing of laboratory orders and results.14

Modules
Most hospital and reference laboratory LISs are composed of multiple, separate clinical and
administrative hardware and software modules (e.g., order entry, specimen tracking, report
generation, accounts receivable, QC and QA, and automated instrument monitoring) that can
operate either independently or be used as a system component. The main functions of hospital
LISs are identified in Table 6.1. Currently, most LISs in reference laboratories do not link
directly to clinical practice applications such as CPOE, POCT devices, EHRs, and pharmacy
systems; however, data from their systems can be sent directly to providers electronically (e.g.,
results reporting) or be made available through regional health information organizations.b
b

A regional health information organization (RHIO) is a group of organizations that are capable of electronic health
care data exchange within a defined geographic region, usually via interoperable electronic health records.
RHIOs are the building blocks of the National Health Information Network initiative proposed by the DHHS
Office of the National Coordinator for Health Information Technology.

May 2008

245

Laboratory Medicine: A National Status Report

Chapter VI – Laboratory Information Systems

Table 6.1: Functions of a Laboratory Information System
LIS-related Application

Characteristics

Preanalytic

Order entry/CPOE/accessioning

Barcode specimen labeling

•

Built-in functions for test ordering from standardized menus; may include automatic confirmation of medical
necessity for performing the test

•

Clinicians may enter orders in hospital information system that will automatically transfer to the LIS, including
appropriate laboratory location, time of day, priority status, or client

•

Algorithms may be used to assess appropriateness of test order in light of information contained in the LIS and in the
patient’s medical record

•

Electronic identification of the health care provider, the patient, the specimen container, what specimen to collect,
and the order in which the specimens were drawn

•

Barcoded labels with unique identifiers for each specimen allow specimen to be tracked through the highly
automated areas of the laboratory or from station to station during specimen examination

•

Tracking may be accomplished via scanning, either automatically or manually by the technician

•

Clinical pathology modules support specimen collection and tracking, order entry, rule-based decision making,
results reporting, and interfacing with automated instruments

Analytic
Specimen tracking

Applications for clinical
pathology, microbiology,
transfusion medicine, and
anatomic pathology

Automated systems interfacing

May 2008

ƒ

Hematology modules allow for manual differential counts to be added, deleted, or masked to automated results
when sent to the LIS and for the creation of histograms

ƒ
ƒ

Immunology modules transmit data from enzyme-linked immunosorbent assay (ELISA) readers to LISs
Toxicology modules automatically rerun positive results, generate complex random drug testing schedules for
organ donors, and produce utilization reports to study testing trends

•

Microbiology modules allow for recording observations, additional test ordering, and creation of antibiograms (used
by hospitals in the review of antibiotic usage)

•

Transfusion medicine modules automate transfusion services, including inventory, distribution of blood, quality
control, and emergency release capabilities

•

Anatomic pathology modules provide access to patient clinical histories, links to clinical pathology department,
correlation studies, and links gross and/or microscopic images to case worksheet and patient reports

•

Allows laboratorians to manage and review large volumes of data and support QC requirements

•

QC functions track and review QC data from all workstations and perform statistical analyses to ensure that
laboratory testing accuracy and precision are maintained at acceptable levels

•

Automatic functions for repeat testing on the same sample for either the same test, a different test, or different
instrument, following an abnormal result or specific decision criteria

•

Enables manual results entry or automated result generation followed by automated results review and release
(autoverification)

•

Blocking function prevents autorelease of results based on flags for abnormal results, delta check failure, panic
value, improbable results, results failing user-defined criteria

246

Laboratory Medicine: A National Status Report
LIS-related Application
POCT

Digital pathology

Links to knowledgebase systems

Chapter VI – Laboratory Information Systems
Characteristics

•

Supports POCT devices that link to the LIS via docking devices or infrared data transmission

•

Capable of receiving POCT data, creating and storing appropriate records, and generating final results reports

•

Digital photography, storage, cataloging, archiving, and dissemination of pathology images at the gross and
microscopic levels

•

Supports electronic, multimedia communication of digital images between laboratories and clinicians, including
telemedicine

•

Links in LISs provide laboratorians access to knowledge bases containing clinical, genetic and molecular information
(e.g., clinical definitions, gene ontology, protein structure and sequencing, mass spectrometry)

•

Report formatting

•

Results reporting communications via fax, email, or Internet (usually through a virtual private network)

•

Links between LISs and EHRs being developed to allow laboratory results to be directly deposited into an EHR;
eventually will include the ability to order tests directly from the EHR

•

Supports linkages of pharmacy database systems to LISs for selection and management of medications and
prevention of medication errors; links are generally from pharmacy system to LIS, not vice-versa

•

Allows for generation of bills with minimal effort, automated coding of tests, generated from test orders

•

Supports accounts receivable to assist with management, receipt, collections, banking, and reporting

•

Manages use, ordering, and inventory of supplies in the laboratory

•

Tracks lot number, quantity, and date received, opened, closed, and expired for reagents and other laboratory
supplies

•

Alerts laboratorians when critical value has occurred and provides protocol to be followed, contact information for
clinician, and log of time that message was delivered and name of person receiving it

•

Automation of quality assurance functions (e.g., documentation of abnormal results, TAT, list of corrected
laboratory reports, comparison to standards set by CAP and other professional bodies)

•

Links between LISs and laboratory error databases in development to allow tracking of diagnostic errors made in the
laboratory

•

Surveillance and management reporting of infectious diseases and hospital acquired infections.

•

Communication functions with public health networks in the event of chemical or biological threats, emerging
infectious diseases, and natural disasters

•

Online laboratory manuals and handbooks, most often in Web-based form, to support knowledge and regulatory
requirements

Postanalytic
Results reporting
Links to EHR
Links to pharmacy database
Other
Billing systems

Supplies management

Patient safety and error reporting

Public health surveillance

Online education

Sources: Adapted from Cooper 2003; Goh 2003; Li 2004; Grzybicki 2005; Asare 2000; Yuan 2005; Cimino 2004; Marchevsky 2002.

May 2008

247

Laboratory Medicine: A National Status Report

Chapter VI – Laboratory Information Systems

For hospitals and reference laboratories, certain more advanced modules such as digital pathology
and telepathology already have been implemented. In addition, many hospitals are creating
interconnections from LISs to clinical practice applications, such as CPOE, POCT devices, EHRs,
and patient safety reporting systems. These advanced functions are vital components to the
development of comprehensive information systems in hospital and ambulatory care settings.
Automated instruments for each subspecialty area (e.g., chemistry, hematology, microbiology)
are linked to the LIS architecture. The combination of innovative automated equipment and IT
has dramatically improved throughput, precision, convenience, and data handling.15 Linkages
between modular systems and the LIS support QC and QA, consolidation of work and
personnel, and more efficient integration and management of laboratory operations. Process
control software allows the operator to monitor hardware, software, and work cell modules
from one PC workstation.16
The LIS architecture is designed around workflow of analytic processes in order to enhance
efficiency, decrease errors, and improve the overall quality of testing. Workflow analysis
measures the impact of change in relation to laboratory costs and efficiencies.9 For each
workstation involved in producing analytical test data, several factors are evaluated, including
the age of analyzers, capital cost, throughput, service cost, supplies, and the labor associated
with producing results. The data summary provides an estimate of the cost for each assay that
is performed at each workstation. Computer simulation programs facilitate workflow analyses.
The programs can imitate and capture dynamic system behavior, support workflow design, and
evaluate alternative ways of improving efficiency and management.17 A good simulation model
assists laboratory staff in determining the best configuration to reduce risks, costs, and
turnaround time without disrupting the working system.

Interconnectivity
Each module or application can be used independently as a stand-alone unit or component,
although some also have the capacity to import, export, imbed, or exchange data with other
modules or applications. There are challenges to attaining optimum interconnectivity when the
LIS modules are developed by different vendors using different data standardsc (e.g., data
exchange, terminologies, document architectures, knowledge representation). As a result, many
health care institutions must depend on middlewared for connectivity and enhanced data
management, especially for new, more complex laboratory testing technologies. Using “if-then”
rule-based algorithms, newer middleware systems not only process data but offer flexibility in
filtering, sorting, and displaying data.19 Development of middleware for data management stems
from the need to resolve problems between instruments and the LIS (i.e., via workarounds) and to
address the shortage of laboratorians available to manage and evaluate data.20 Middleware serves
the functions needed for today’s clinical laboratory that have not been met in the designs of
Data standards encompass methods, protocols, terminologies, and specifications for the collection, exchange,
storage, and retrieval of information associated with health care applications, including EHRs, medications,
radiological images, laboratory information, payment and reimbursement, medical devices and monitoring
systems, and administrative processes.18
d Middleware is software that facilitates communication between laboratory instruments and the LIS, or two
applications, and management of workflow within a workstation or the system as a whole.19 Functions may
include autoverification of results, addition of new orders to test files, insertion of comments, or take other actions
for specific patients or physicians.20
c

May 2008

248

Laboratory Medicine: A National Status Report

Chapter VI – Laboratory Information Systems

automated systems and interconnections with the LIS. A challenge arising from this third party
approach is the integration of multiple, additional vendor applications and standards to the
already complex pool, increasing opportunities for error.
Another important feature of LIS interconnectivity is the growing adoption of Web-based
mechanisms to support electronic test ordering and results reporting executed from a hospital,
physicians’ office, reference laboratory, or other health care facility.21 Internet browsers running
on personal computers allow direct data exchange between these facilities while portals provide
gateways to the hospital LIS to access online ordering functions and retrieve results reports. Some
Web-based applications allow results to be reported directly into a patient’s EHR or a physician’s
practice management system.

Physician Office Laboratories
Increased regulatory requirements, testing costs, and coding complexity following the
implementation of CLIA in 1992 accelerated adoption of certain LIS functions by some POLs.22
Similar to IDS, these systems help POLs comply with QC, PT, QA, and patient test management
requirements. For QC, the LIS receives data from each instrument and evaluates whether values
are within the control range. Along with PT data, the QC data are then stored for retrieval and
review during inspections and for troubleshooting. LIS applications also are used to comply with
CLIA patient test management requirements by automatically loading key data points (e.g.,
patient identifiers, lab name, address, age-specific normal ranges, units of measure, testing
analyst, tests ordered).
For most POLs, use of other LIS applications is quite limited, because the majority of these
laboratories perform waived tests and non-waived microscopy tests. Regarding interconnectivity,
POLs that perform certain moderately complex chemistry and hematology tests (e.g., CBC) tend to
structure analyzers as independent modules connected only to printers for report generation. If the
POL has an electronic labeling function, it is usually connected to the office administrative
application or the patient’s EHR. Printed test results are added to a paper chart or manually entered
into the EHR. Often, the EHR application includes an electronic ordering/reporting function that
can link via the Internet to a larger reference laboratory with which the POL has an established
business relationship. Generally, the need for broader access and more cost-effective networking
solutions is boosting adoption of Web-based ordering and reporting capabilities in POLs.8
There is sizable cost involved in implementing and linking EHRs to other functions. While more
physician offices are implementing them, it has become standard practice for reference
laboratories to directly provide and/or finance basic electronic connectivity for ordering tests and
reporting results. In doing so, however, external data exchange is limited to the “sponsoring”
reference laboratory.23 As this limitation inhibits compliance with open standards and
development of health care data exchange, efforts are underway to require interoperability with
other vendor systems. For example, MedPlus, Inc., an HIT subsidiary of Quest Diagnostics, has
developed a program (now available) called “Universal Laboratory Orders and Results” that
allows physicians’ offices to electronically order and receive results and track patient laboratory
and prescription data from both Quest laboratories and other hospital laboratories.24

May 2008

249

Laboratory Medicine: A National Status Report

Chapter VI – Laboratory Information Systems

ADVANCED APPLICATIONS FOR COMPREHENSIVE SYSTEMS
Clinicians must have access to potentially pivotal patient information (e.g., laboratory results,
medication list, and medical history) and to current scientific information (e.g., infectious
diseases, medical evidence, guidelines) in order to provide high quality care. A comprehensive
IT infrastructure is essential for:

ƒ

Managing the expanding volumes of clinical and administrative data generated from
patient encounters

ƒ

Transforming clinical data into information that can be used to improve the safety,
effectiveness, and efficiency of health care delivery25

The infrastructure must ensure that laboratory and other data are accessible through efficient
networks and clinical practice applications that link to the EHR.
Public and private sector initiatives to build the health information infrastructure have started
with the ancillary services that generate the highest volume of data critical to patient care and
safety—laboratory, pharmacy, and radiology. These services also tend to rely on sophisticated
data management systems (e.g., LIS or pharmacy database) and innovative technologies (e.g.,
automated analyzer, robotic dispensing) for day-to-day operations. Links between the ancillary
disciplines are necessary for accurate clinical diagnosis, treatment, and disease management.
For example, links between clinical pathology and pharmacy data are needed for therapeutic
drug monitoring, and links between anatomic pathology and radiology data are needed for
diagnosis of cancer metastases and guidance of complex surgical procedures.

UNRESOLVED ISSUES RELATED TO ADVANCED APPLICATIONS
Automating Data Management
Effectively automating data management capabilities is an important factor in the evolution of the
LIS and automation systems for clinical and anatomic pathology and requires informatics
standardization, expanded computing power for new testing technologies, and other support for
networked systems.
Automation of data management already has yielded several advantages to the clinical laboratory,
including higher productivity and efficiency (e.g., higher test volume, fewer instruments), more
streamlined workflows and easily reproducible processes, and decreased costs.26
Current data management system designs have progressed from a hardware-based approach to a
software-based approach that incorporates middleware and/or process control software. These
applications generally automate such features as repeat testing, dilutions, reflex orders, and
adjudication of instrument errors. e, 16, 27 For a middleware system to release results automatically,
it must select from a range of actions based on various criteria and the desired scope of the
e

Repeat testing will be automated ultimately so that a specimen yielding a specific result can be returned to the
system for additional testing using an alterative method or confirmatory testing on the same or different
instrument. “Reflex testing” refers to additional testing performed automatically when a specific rule is applied
to the result of the first test.

May 2008

250

Laboratory Medicine: A National Status Report

Chapter VI – Laboratory Information Systems

application which can differ by vendor, the type of test, and the nature of the patient population.28
More sophisticated integrated systems incorporate knowledge-based process control software
features and expert rules, such as algorithm testing, disease pattern checks, specimen integrity
checks, clinical range checks, and delta checks.29
Process control automates routine tasks and employs standards of practice to support the human
operator. Although most common in large hospital and reference laboratories, process control
software for automating data management is expected to increase sharply over the next several
years. Process control features in development will facilitate the transportation, storage, and
retrieval of laboratory specimens; physically identify and track specimens throughout the system;
and support automated test cancellations and workload management. Next generation laboratory
automation software will consolidate and integrate all aspects of laboratory, data, and instrument
management from sample logistics to results management, archiving, and retrieval.30 Successful
integration of these process control data management features requires multidirectional,
coordinated communication that links the LIS, preanalytic processing components, the specimen
transportation system, analyzers, and the postanalytic archiving system.28

Data Management for New Testing Technologies
The LIS must be flexible enough to handle the demands posed by new laboratory tests and
technologies. In particular, the quantity of specimens involved in genetic, proteomic, and PGx
testing pose significant challenges for LISs.31 The complete mapping of the human genome will
continue to enable creation of new laboratory tests and LIS applications programs to support them.6
Greater use of genetic testing is increasing the need for LIS applications that support cytogenetic
and molecular diagnostic testing. Examples of such applications include increased capacity for
data storage, ability to detect genetic rearrangements and other abnormalities associated with
malignant disease and hereditary genetic abnormalities, and ability to access patient records that
contain personal gene databank and family tree information.32-34 Decision support, specimen
tracking and automation of QA and QC documentation features also must accommodate the
demands of genetic testing. In order to accomplish these goals, additional software applications
and more sophisticated, high-speed computer processing capabilities are required of the LIS.35
Extension of data management to genetic findings requires an LIS to standardize information and
support genomics-based inference.31 Current medical vocabularies are insufficient to describe the
findings generated by some new molecular tests.31, 36 Vocabularies such as the Clinical
Bioinformatics Ontology have been designed for clinical molecular diagnostics and cytogenetics.37
In order for LISs to handle the nature and volume of genomic tests, current discrepancies between
how genetic mutations are described in the literature and how they are described in internet
databases must be resolved.f, 36 Use of separate, specialized modules or systems in the laboratory
that transmit summarized information to EHRs has been proposed as another means to allow
LISs to meet the needs of genetic testing.39
The upswing in genomic testing is speeding the integration of patient-specific genomic
information into EHRs. Entering and formatting molecular diagnostic and cytogenetic laboratory
f

One example of an online database is RefSeq, a collection of genomic DNA, transcript, and protein products for
major research organisms compiled by the National Center for Biotechnology Information.38

May 2008

251

Laboratory Medicine: A National Status Report

Chapter VI – Laboratory Information Systems

findings into EHRs is necessary to support clinical decision-making and to automatically flag
PGx-based risks.31
The volume and complexity of data generated from proteomic profiling, especially from highthroughput analyses and increased reliance on automation, demands that LISs be capable of
storing large quantities of data.40, 41 In particular, LISs need to be able to search large internal and
external databases containing information about proteins, which requires greater computing
power than is currently available in these settings.40, 42
PGx testing presents particularly difficult challenges to the LIS due to the potential enormity of its
data requirements—molecular biology (e.g., sequences, structures, pathways), clinical medicine
(e.g., medications, diseases, side effects) and pharmacology (e.g., pharmacokinetics and
pharmacodynamicsg).45 Among other roles, PGx entails linking genetic information and clinical
data in order to identify genotype-phenotype associations.
Patients also will need support when seeking, interpreting, and acting upon personal geneticbased data. The 2006 NACB draft guidelines on the application of PGx to the clinical laboratory
recommends that laboratories develop educational resources to recipients of test results and that
interpretative reports include a summary of the patient’s metabolic status and its effect on the
drug regimen being considered.46

Computerized Physician Order Entry
Growing awareness of prescription errors and their associated costs provide some of the rationale
for development and implementation of CPOE systems. These are online computer applications
that allow physicians to order laboratory, pharmacy, and radiology services and therefore have
the potential to prevent common medication ordering errors (e.g., selecting the wrong drug or
dose, overlooking drug allergies).47, 48 Laboratory data are a vital component of CPOE systems
and are often displayed for physician review prior to ordering.49 CPOE systems must be able to
exchange data with LISs using standardized communication protocols.47, 50
CPOE adoption remains relatively low. A study reported in 2006 estimated that national
adoption of CPOE is 15% in integrated delivery systems,h 9% in stand-alone hospitals, and 1% in
skilled nursing facilities and rehabilitation hospitals.51 Despite the use of interfacing standards
such as HL7, messaging between the CPOE and LIS still requires customized programs and close
collaboration between the laboratory and the team implementing the CPOE system.52 LISs face
additional significant challenges in enabling CPOE, including how to provide real-time access to
data stored in the LIS and how to ensure that the software-based rules governing the CPOE are
appropriate and do not result in unacceptable rates of false positive and false negative alerts.53
In 2000, the Ohio State University Health System implemented a CPOE system that linked
bidirectionally with its LIS, automating the laboratory process from order entry to results
reporting.54 Compared to areas of the hospital in which CPOE had not been implemented,
Pharmacokinetics refers to the activity of drugs in the body over a period of time, including the processes by which
the body absorbs, distributes, localizes, and excretes drugs.43 Pharmacodynamics refers to the reactions between
drugs and living systems.44
h An integrated delivery system is an aggregation of acute care hospitals, rehabilitation hospitals, and ambulatory
care practices.
g

May 2008

252

Laboratory Medicine: A National Status Report

Chapter VI – Laboratory Information Systems

laboratory TAT decreased by 25% and both work flow accuracy and efficiency increased. Extensive
modifications made to their commercially available vendor-based CPOE system allowed the health
system to interface with its other information systems and be tailored to physician needs. The
ability to customize or adapt a CPOE system to an organization’s needs is cited as a major
consideration in a consensus statement on successful CPOE implementation published in 2003 by a
group of 13 international CPOE experts.55
The Leapfrog Group established an initiative to increase adoption of CPOE. Hospitals can meet
Leapfrog’s CPOE standard by assuring that physicians enter at least 75% of medication orders via
a computer system with prescribing-error prevention software. In addition, the inpatient CPOE
system must be able to alert physicians of at least 50% of common, serious prescribing errors, as
well as allow physicians to electronically document a reason for overriding an interception prior
to actually doing so.56 Some vendors have offered CPOE systems as part of their information
system products, making readily integrated CPOE systems more accessible to organizations that
cannot develop their own systems.50

Clinical Decision Support Systems
CDSSs rely on software algorithms designed to improve clinical decision making by matching
characteristics of individual patients to a computerized knowledge base and generating patientspecific clinical options, recommendations and other support.57 Such support may include alerts,
reminders, order sets, reference information, and education.58 Although currently available
CDSSs require providers to enter patient data, forthcoming applications will automatically link to
patient data held in EHRs and generate support features without duplicate entry.59
Laboratory data are a key component of CDSSs. Currently, the most common laboratory-related
feature of CDSSs is flagging abnormal test results.59 Another feature in use among early adopters
alerts physicians if they order additional, potentially redundant laboratory tests before results of
previous tests can be obtained.60 Eventually, CDSSs will include laboratory information that aids
provider interpretation of medical symptoms, signs, and diagnoses. For example, systems
developed at the Massachusetts General Hospital and Kaiser Permanente in Escondido,
California, use symptoms, signs, laboratory data, and other clinical findings to produce a ranked
differential diagnosis list.61, 62
To supply laboratory data to a CDSS, an LIS must generate data interpretable by the decision
support application. Laboratory data must be transparent enough to be understood,
computationally unambiguous, and capable of being linked or adapted to multiple platforms.63
Once laboratory data are transmitted to the CDSS, they may need to be reformatted to ensure
their maximal clinical efficiency and efficacy.52 A major obstacle to the ability of a LIS to interact
with and provide data to a CDSS is lack of interoperability between laboratory and clinical
applications. The Clinical Context Object Workgroup (CCOW) of HL7 developed a standard to
facilitate the integration of applications from many different systems at the point of use to give the
clinical user the experience of interacting with a single system.i, 64 Widespread adoption of the

i

The CCOW standard is based on context management, or the coordination and synchronization of applications so
that they are mutually updated with the set of their common data objects (e.g., patients or medical encounters)
that frame the user’s interactions with applications.64

May 2008

253

Laboratory Medicine: A National Status Report

Chapter VI – Laboratory Information Systems

CCOW standard would enable laboratories to seamlessly and efficiently exchange information
with CDSS and other clinical tools.52
To support CDSS, an LIS must have several complex capabilities. Decision making relies in part
on rule-based systems or algorithms programmed into the LIS to automate processes such reflex
testing and alerts. While most modern LISs have rule-based capabilities sufficient for results
autoverification, the capacity necessary to program a rule-based system for CDSS and the clinical
and pharmacy data necessary to support these systems (e.g., QC and QA data, results of other
laboratory tests, and historical test performance data) are not always available.52 Even once a
rule-based system has been programmed; it requires regular updating to ensure that the
knowledge base is current.

Point-of-Care Testing
Innovative sensor technologies, microprocessor-based analyzers, and disposable test cartridges
have enabled the creation of small and portable testing devices that support minimally-invasive
collection techniques, followed by rapid analysis and results presentation.1 Examples of the many
tests that can be conducted at the point of care include measurements of blood gases, blood count,
glucose, drugs of abuse, and fecal occult blood.65 More recently, technologies are enabling new
and less-invasive POCT, such as minimally-invasive, laser-based skin perforators that collect
interstitial fluid for measurements of glucose levels, and infrared sensors that measure glucose
and other analytes through the skin.66 Data captured in these devices can be downloaded via a
docking station to a database that is linked to the laboratory or hospital information system,
billing, and generation of summary data reports.67
POCT devices in day-to-day patient care are expected to increase substantially in the near term,
perhaps more so than any other laboratory-related technology.67 However, one of the key
unresolved issues in clinical settings is the lack of interoperability between POCT devices and the
LIS.65 The Connectivity Industry Consortium was formed in 1999 to develop a base-level
interoperability standard for data exchange between POCT devices and EHRs and LISs.67, 68 The
standard was transferred to CLSI and approved as POCT1-A2: Point-of-Care Connectivity (Second
edition, 2006).j, 69, 70 It provides the framework for engineers to create medical devices,
workstations, and interfaces that allow different types and brands of POCT systems to
communicate with each other and LISs.71 CLSI developed additional guidelines for POCT device
interoperability.k In particular, POCT4-A2: Point-of-Care In Vitro Diagnostic Testing (2006) provides
general definitions, procedures, and recommendations for POCT, including guidance on quality
assurance, alternative QC, and specimen collection and identification.72 Two other guidelines
target specific testing devices.l

POCT4-A2 replaced its predecessor, AST2-A, published in 1995.
CLSI developed guidelines to provide users of POCT devices, including nonlaboratory personnel, with information
and suggestions for good clinical testing practice and for producing reliable results regardless of where or by
whom the test is performed.
l C30-A2: Point-of-Care Glucose Testing in Acute and Chronic Care Facilities (2002) provides instructions and
recommendations on the administration of a POCT blood glucose monitoring program, persons who perform the
tests, selection of methods, reporting of results, and quality assurance.73 Guideline H49-A: Point-of-Care Monitoring
of Anticoagulation Therapy. (2004) provides information on how to proceed in the evaluation, implementation, and
monitoring of heparin and warfarin therapy.74
j

k

May 2008

254

Laboratory Medicine: A National Status Report

Chapter VI – Laboratory Information Systems

Technical issues (i.e., analytic performance, interconnectivity) are being addressed through
international standards development initiatives; however, they have not yet been fully resolved.
Many discrepancies between results from POCT devices and central laboratory values have been
reported. For example, in a 2006 study comparing seven POCT glucose meters from four
manufacturers to a central laboratory analyzer, differences greater than 10% were found 61% of
the time for POCT hyperglycemic values, and differences of 20% or greater were found 57% of the
time in the hypoglycemic range.75 Some of these disagreements have been attributed to differences
in testing methodologies between POCT and central laboratory testing.76 Among these, POCT
typically involves multiple instruments, each of which is operated by several people, as opposed
to central laboratory testing, which generally involves one instrument that is operated by only a
few people.77
The lack of standardized hardware and software continues to challenge the integration of POCT
devices with health information systems and applications. Until recently, vendors manufactured
their respective devices with different data management features, data manager systems, and
interface standards, resulting in high levels of redundancy in laboratory testing as well as significant
additional costs.67 As POCT becomes more common in inpatient and outpatient settings, the
devices must be designed for interoperability with LISs and EHRs to support electronic storage,
retrieval, and comparability of patient data for current care and future reference.

Electronic Medical and Health Records
Although often used interchangeably, electronic medical records (EMRs) and electronic health
records (EHRs) are different. An EMR is a local (single provider) system installed in an
organization to support secure, comprehensive, and electronic patient medical record information.
An EHR is a shared information system that aggregates patient information from various systems
and sites, including multiple patient EMRs, on a scale that is regional or broader. Because EHRs
include EMR data, yet offer more extensive linkages outside the provider organization, U.S.-based
initiatives have focused on building the health information infrastructure for the EHR.
Estimates of EMR and EHR adoption rates vary widely, often due to ambiguities in the definitions
of EMRs and EHRs. The 2007 AHA survey on hospital use of IT reported that 11% of U.S. hospitals
had fully implemented EHRs (mostly larger, urban, and teaching institutions).10 The Healthcare
Information and Management Systems Society (HIMSS) has developed a database that maintains
assessments of EMR implementation in approximately 4,000 hospitals using a scale of stage 0
(information systems for laboratory, pharmacy, and radiology not implemented) to stage 7 (the
hospital has a paperless EMR environment).78 December 2007 data indicate that nearly 39% of
hospitals were in stage 2 and 28% had reached stage 3 or higher (see Table 6.2). However, the
HIMSS definition of an EMR appears to fit the AHA definition of an EHR noted above.
EMRs and EHRs also are being implemented outside of hospital settings. A 2006 report and
meta-analysis of quality-ranked studies sponsored by the Robert Wood Johnson Foundation
estimated that 17-25%of physician offices, 13-16% of solo practitioners, and 9-57% of large

May 2008

255

Laboratory Medicine: A National Status Report

Chapter VI – Laboratory Information Systems

physician officesm had implemented EHRs.79 Despite relatively widespread support, three main
barriers continue to inhibit widespread implementation:
(1) Lack of adoption of common data standards for EHRs
(2) Quick turnover of HIT companies
(3) Cost of installing and converting to EHR systems80
Table 6.2: EMR Adoption Model Trend Q3 2007
Stage of
Adoption

Description of EMR Capabilities

Percent of
U.S. Hospitals
(N=4,381)

7

ƒ
ƒ

Medical record fully electronic
CDO able to contribute to EHR as byproduct of EMR

0.0%

6

ƒ
ƒ
ƒ

Physician documentation (structured templates)
Full CDSS (variance and compliance)
Full Picture Archiving and Communications System (PACS)

0.6%

5

ƒ

Closed loop medication administration

1.4%

4

ƒ
ƒ

CPOE
CDSS (clinical protocols)

2.2%

3

ƒ
ƒ
ƒ

Clinical documentation (flow sheets)
CDSS (error checking)
PACS available outside radiology

24.1%

2

ƒ
ƒ
ƒ
ƒ

Clinical data repository
Controlled medical vocabulary
CDSS inference engine
May have document imaging

39.1%

1

ƒ

Ancillaries- laboratory, radiology, pharmacy

15.0%

0

ƒ

All three ancillaries not installed

17.6%

Adapted from: HIMSS Analytics Databases (derived from the Dorenfest IHDS+ Database™). ©2007 HIMSS Analytics™

Because laboratory results are a major component of patient records (electronic or paper),
interoperability between LISs and EHRs can reduce medical errors, increase appropriate and
reduce unnecessary testing, and improve quality and efficiency of health care. Interoperability
allows authorized clinicians (i.e., ordering and non-ordering) to electronically access laboratory
results, view historical results, receive automatic alerts for critical values and abnormal results,
and integrate laboratory data with other EHR-linked functions (e.g., CPOE, CDSS, disease
management).81, 82
Interoperability of EHRs and LISs is still in early stages of development, and most laboratory
results cannot be directly integrated into an EHR.83 Data transfer between systems has been
hindered by incompatible programming platforms, components that prevent successful
authentication and authorization of patients and providers, and security protocols. Existing LISs
m

Large physician office was defined as greater than 20 physicians by one study and as greater than 50 physicians in
another study.

May 2008

256

Laboratory Medicine: A National Status Report

Chapter VI – Laboratory Information Systems

are devoted primarily to optimizing performance across internal system modules; data exchange
with external systems is not a priority.84 As part of a 2005 survey conducted by CAP, LIS vendors
were asked about their products and the types of settings in which they were installed.85 A
majority of respondents indicated that a portion of their LIS installations were stand-alone and
had not been integrated with other clinical applications (e.g., EHR).
The main barriers to EHR-LIS interoperability include technical complexity of developing data
standards and costs. To date, initiatives to develop national standards for EHR-LIS
interoperability have targeted results reporting for patient care and disease management. Several
public and private sector initiativesn are underway to address these constraints, including
activities to develop data standards for EHR-LIS interoperability, including the following:

ƒ

The Health Information Technology Standards Panel (HITSP) is a multi-stakeholder
coordinating body established in 2005 by the American National Standards Institute
(ANSI). The EHR Interoperability Specifications are based on the DHHS Office of the
National Coordinator for Health Information Technology Use Case for EHRs
initiative.o Designed as national standards, the specifications were developed using
the data standards approved for the national HIT infrastructure.

ƒ

The EHR-Lab Interoperability and Connectivity Standards project (ELINCS), created
by the California HealthCare Foundation (CHCF),p developed a standard and detailed
specifications for coding and formatting laboratory results messages delivered in realtime from the LIS to an EHR.88 The standard can be used by commercial and hospital
laboratories to send test results electronically and by developers/vendors in technical
design of EHR systems. In 2006, CHCF funded pilot testing of ELINCS at five clinical
sites, including three hospital-based laboratories and two national commercial
laboratories.89 Four of the five sites are currently operational,q two of which have
begun to extend ELINCS-based connectivity requirements to their data exchange
partners (e.g., independent laboratories, other local hospital laboratories). Ownership
of ELINCS is currently being transferred to HL7.

Digital Pathology
Recent advances in tissue labeling/processing techniques, imaging sensors, digital image
processing, image analysis, and personal computers are transforming anatomic pathology and
traditional microscopy. Digital microscopy is a form of digital pathology that integrates digital
imaging and light microscopy to capture pathological images at the gross and microscopic levels.90
In October 2006, the federal government finalized new HIT safe harbor regulations for the Stark and anti-kickback
regulations that allow for the provision of HIT below cost by a medical organization as long as the HIT increases
patient safety and is interoperable with other information systems.86
o The Interoperability Specifications were developed from an assessment of the current practices in electronic
laboratory results reporting, as well as from the EHR Use Case, which was created to describe interoperability
between EHRs and laboratory systems from patient and provider perspectives.84 The recommended standards
put forth by HITSP were chosen to meet the requirements identified in the Use Case and to reflect current
practice and future HIT needs and were chosen with the assumption that the standards will become more broadly
used over the next several years.
p CHCF has also led the development of CALINX, a standard to retrospectively communicate batch reporting of
laboratory test results to data warehouses and disease registries to support population-level quality-improvement
programs.87 CALINX has been created to meet the specific needs of California.
q The fifth site withdrew from the ELINCS pilot project after switching laboratory vendors.
n

May 2008

257

Laboratory Medicine: A National Status Report

Chapter VI – Laboratory Information Systems

The current rapid transition to digital media affords many advantages to pathologists, laboratory
staff, and patients. The images may be electronically stored, replicated, cataloged, employed for
educational purposes, transmitted for further interpretation (i.e., telepathology), analyzed for salient
features (e.g., medical vision/image analysis), or used to support a wider digital pathology strategy.
Digital images of pathologic slides also can support quality assurance by eliminating the need to cut
and disseminate multiple sections for analysis. Following the substantial initial investment, digital
technology costs less to operate than photographic prints. Memory cards, CD-ROMs, and other
storage media that capture the images are inexpensive and can be reused. In addition, digital
pathology shortens image production times.
The combination of automation technology with digital pathology has facilitated the development
of automated cytology screening.91, 92 Cytology automation reduces the false-negative detection rate
of manual, microscopic cytology examinations that rely solely on human evaluation. Computerassisted systems can enhance human performance by identifying highly suspicious areas.
Digital technology is playing an important role in telepathology. The advent of telemedicine, i.e.,
the exchange of medical information electronically from one site to another to improve patients’
health, has significantly changed the way pathologists compare and share diagnoses between
institutions.93 Traditionally, information shared includes stained and unstained slides, tissue
blocks, wet tissue, and pathology reports.94 Telepathology supports electronic multimedia
communication between pathologists (and other authorized, trained laboratory staff) concerning
primary diagnoses and second opinion consultations as well as consultations with other
clinicians.94 Information for telepathology consultations, including photographic and video
images,r is shared in three different ways: statically (information is stored and forwarded),
dynamically (information is shared synchronously), and via a hybrid mode, which incorporates
both static and dynamic sharing.
Educational institutions are incorporating digital pathology into their curricula as a user-friendly,
interactive teaching method. Instructional databases include pathology slides scanned at highmagnification along with three-dimensional simulations of organs and links to other relevant
literature, information, and diagnostic tools.95 Some institutions also provide Web access to
digital pathologic images.
Prominent issues in digital pathology pertain to image quality (e.g., optics and calibration of
system components), standardization of image sizes, and Web-based connectivity.90 High-power
computation, large storage capacity, new image formats, and novel processing algorithms are
needed to advance digital microscopy from multiple single-field images to whole-tissueprocessing. High-content screening technology is necessary to meet genomic imaging
requirements. Most pathology systems are currently designed as department-based applications;
however, efficient and timely expansion of capacity for interpretive consultations requires broad
implementation of Web-enabled, interactive telepathology.96

r

In primary diagnosis telepathology, specimens at a referring site are diagnosed remotely by pathologists, and the
pathologist at the remote site is solely responsible for the diagnosis. In second opinion telepathology,
responsibility for the diagnosis belongs to the pathologist at the local site (known as the referring pathologist) and
the remote pathologist (the consulting pathologist).

May 2008

258

Laboratory Medicine: A National Status Report

Chapter VI – Laboratory Information Systems

DATA STANDARDS FOR COMPREHENSIVE SYSTEMS
Lack of harmonized data standards is the greatest barrier to laboratories’ ability to integrate data
within the laboratory as well as to exchange data with external “trading” partners (e.g., hospital
and ambulatory clinicians, other laboratories, pharmacy department, radiology department,
public health entities, payers). Harmonization of data standards is at varying stages of
completion and adoption for each application. However, further progress in building the health
information infrastructure and integrating laboratory data with clinical practice applications
cannot be realized unless the standards issues are resolved. Main components of data
standardization are identified in Box 6.1.
Box 6.1: Components of Data Standardization
•

Definition of data elements: determination of the data content to be collected and exchanged

•

Data interchange formats: standard formats for electronically encoding the data elements. Interchange
standards include sequencing and error handling, document architectures for structuring data elements,
information models that define relationships among the data elements, user interface, and patient data links

•

Terminologies: medical terms and concepts used to describe, classify, and code the data elements

•

Knowledge representation: standard methods for electronically representing medical literature, clinical
guidelines, and the like for decision support

Source: Institute of Medicine. Patient safety: achieving a new standard for care. Washington DC: National Academy Press, 2004.

The challenge of data standards for the broader health and laboratory information systems is not
their absence, but their proliferation. The lack of uniform, common data standards creates great
difficulty in establishing interoperability to transfer data easily and economically from one system
or application to another. Although most standards are publicly available and developed by
international standards organizations, some standards are proprietary and developed by private
sector organizations. Traditionally, use of a standard in the U.S. has been voluntary and left to the
discretion of the vendor or institution. As a result, an obstacle to achieving interoperability
among information systems and applications has been the haphazard adoption of data standards
across the board.97 Historically, most laboratory-related systems and applications were designed
to operate independently. Thus, lack of common data standards in laboratory medicine has
inhibited information sharing between laboratories and health care providers.
Standards development organizations (e.g., CLSI), professional societies (e.g., HIMSS), and state
and federal government agencies are collaborating to harmonize data standards specifications.
Laboratory-specific technology integration initiatives include the following:

ƒ

May 2008

The American Health Information Community (AHIC) was established in 2005 by the
DHHS Office of the National Coordinator for Health Information Technology to
provide the Secretary with recommendations for accelerating the development and
adoption of HIT. To begin, the program will target standards harmonization and
technology adoption in four areas: consumer empowerment, chronic care,
biosurveillance, and electronic health records. This federal advisory committee is
being converted to a public-private sector partnership. Several documents on
laboratory-related integration can be found at:
http://www.hhs.gov/healthit/community/background/

259

Laboratory Medicine: A National Status Report

Chapter VI – Laboratory Information Systems

ƒ

HITSP, discussed above, develops harmonized interoperability specifications,
implementation guides, code sets, terminologies, integration profiles, and information
policies. This panel provides input to the AHIC initiative. A summary of the
recommended standards for interoperability can be found at:
http://www.hhs.gov/healthit/ahic/materials/02_07/ce/hitsp.doc

ƒ

The Laboratory Exchange meeting of public and private sector experts was convened
by AHRQ in 2006 as part of the AHIC initiative to address challenges to data exchange
between the LIS and EHR for test orders and results reporting. The meeting identified
relevant business, financial, regulatory, data security, privacy and confidentiality,
technical, and patient identification issues.98 See:
http://healthit.ahrq.gov/portal/server.pt/gateway/PTARGS_0_217663_0_0_18/AHR
Q_Lab_Meeting_Summary.pdf

ƒ

The HIMSS’ Integrating the Healthcare Enterprise initiative was extended to clinical
laboratories in 2003.99 The international, private-sector initiative developed a technical
framework that defines exchange partners, data exchange standards, and guidelines
for integration of laboratory information and automation systems with the larger
health care enterprise (i.e., hospital or IDS). See:
http://www.himss.org/ASP/topics_News_item.asp?cid=66712&tid=10

Efforts to harmonize standards and connectivity requirements through these initiatives have
focused on data interchange formats and terminologies. Data interchange standards include
encoding formats for data exchange as well as document architectures for structuring data
elements, information models that define relationships among the data elements, user interface,
and patient data links, all of which are needed to support high-performance interoperability.
Because the HL7 standard for data interchange has been selected as the core standard for the
health information infrastructure, the standard also is serving as the primary exchange and
interface standard for LIS system-to-system, automation-to-LIS, and application-application
connectivity. HL7 clinical document architecture is used for claims attachments to administrative
transactions that use the ANSI American Standards Committee X12 standard for transmission.
Other interchange standards address specific attributes or applications, such as the Unified Code
for Units of Measure and Digital Imaging and Communications in Medicine standard.
Even though these standards are well integrated in stand-alone systems and first-phase data
exchange, they are not sufficiently developed for next-level integration and interoperability for
clinical practice applications. New means must be developed to represent data sets that are
exchanged directly among EHRs or other end-user systems, consistent with CLIA requirements
(e.g., where test was performed and methods used), and to accommodate ongoing changes and
updates to laboratory reports in the EHR.98 Also to be developed is the ability to associate
laboratory test results with test orders, and patients with their providers in the EHR. The ELINCS
project may offer a viable vehicle for specifying message standards to the rigorous degree
necessary and to facilitate certification of conformance.
Important integration issues also include: record linkage and patient identification, data quality,
data synchronization, business rules management, and real-time access.100 Detailed descriptions
and analyses of the informatics challenges of relating laboratory data to the EHR and other
functions (e.g., biosurveillance) are provided in reports of HITSP, AHIC, and HIMSS cited above.

May 2008

260

Laboratory Medicine: A National Status Report

Chapter VI – Laboratory Information Systems

Controlled vocabularies facilitate data collection at the point of care, retrieval of relevant data,
information, and knowledge, along with data reuse for multiple purposes such as automated
surveillance and clinical decision support. The two vocabularies used in data interchange are the
Logical Observation Identifiers Names and Codes (LOINC) and the Systematized Nomenclature
of Medicine- Clinical Terms (SNOMED-CT). Unlike data interchange standards, many other code
sets are used for different types of data. Examples include the Current Procedural Terminology®
Fourth Edition and Healthcare Common Procedure Coding System.
Although LOINC is an appropriate terminology for laboratories and their coding, it is not readily
applicable to health care providers.98 Also, there is not a standard vocabulary for providers that
maps unambiguously to LOINC. Wide variability in the way that common tests are named
among the different terminologies and vendors (e.g., “serum sodium” versus “NA_S”), and codes
for test batteries have not been fully developed for all local patterns that may be selected by
providers. Another obstacle to the adoption of a common vocabulary is that most clinical data is
stored in a natural language (free text) context that must be converted into an electronic format
(structured, predetermined phrases and codes) prior to transmission.100
Near-term LIS priorities for the laboratory medicine sector include achieving and implementing:

ƒ

Standardized ways of representing orders for laboratory tests

ƒ

Standardized means for reporting laboratory information in terms of both messaging
format and test names

ƒ

Standardized approaches for correlating laboratory information with the correct patient

ƒ

Systematic, acceptable methods for maintaining privacy and confidentiality of
laboratory data for public health surveillance and quality improvement purposes98

CONCLUSIONS
LISs have evolved from systems to facilitate clinical laboratory workflow and generate results
reports to complete systems capable of linking laboratory data through the entirety of the TTP,
including clinician-related pre- and postanalytic activities. The extent of LIS adoption and
capability varies; while IDS and large laboratories rely on LISs for many aspects of laboratory
testing (e.g., test ordering, results reporting, links to the inpatient pharmacy database), POLs and
smaller laboratories primarily use the LIS to facilitate compliance with QC, PT, QA, and patient
test management requirements.
To improve quality and safety, manage the increasing volume of clinical and administrative
information, and support emerging laboratory tests and clinical practice applications, LISs must
increase computing power and conform to common data standards. All laboratories must develop
Wed-based connectivity to becoming fully integrated into the health information infrastructure.

ƒ

May 2008

Successful integration of enhanced data management features requires
multidirectional, coordinated communication that links the LIS, preanalytic processing
components, the specimen transportation system, analyzers, and the postanalytic
archiving system.

261

Laboratory Medicine: A National Status Report

Chapter VI – Laboratory Information Systems

ƒ

The volume and complexity of data generated from genetic, proteomic, and
pharmacogenetic testing, especially from high-throughput analyses and increased
reliance on automation, requires that LISs be capable of storing and retrieving large
quantities of data.

ƒ

Enabling CPOE, CDSS, and EHR applications with laboratory data in real-time
requires continued development of rule-based algorithms capable of generating and
integrating accurate alerts, reminders, order sets, results reports, and a list of
differential diagnoses based on patient signs, symptoms, and characteristics.

ƒ

Digital pathology systems requires further advances in high-power computation, data
storage capacity, image formatting, and processing algorithms to facilitate the shift
from single-field images to whole-tissue-processing.

ƒ

Lack of harmonized data standards is the single greatest barrier to laboratories’ ability
to integrate data within the laboratory as well as exchange data with external “trading”
partners. Further progress in building the health information infrastructure and the
capabilities for integrating laboratory data more fully with clinical practice applications
cannot be realized unless laboratories, health care organizations, vendors, and other
stakeholders resolve data interchange and terminology standards issues.

May 2008

262

Laboratory Medicine: A National Status Report

Chapter VI – Laboratory Information Systems

REFERENCE LIST
1. Institute of Medicine. Medicare laboratory payment policy: now and in the future.
Washington, DC: National Academy Press, 2000.
2. Friedman BA. Using the LIS as a strategic tool. Comput Healthc 1990;11(9):38-40.
3. Eggert AA, Emmerich KA, Quam EF, Bowers KL. The LIS as process manager. Save
money and reduce tech fatigue. MLO Med Lab Obs 2000;32(7):32-40.
4. Jacobs R. Big gains, small budget. A rural hospital lab implements a hosted LIS that yields
big improvements in lab productivity, client service and patient care with a low total-costof-ownership. Health Manag Technol 2003;24(9):32, 35.
5. Workman RD, Lewis MJ, Hill BT. Enhancing the financial performance of a health system
laboratory network using information systems. Am J Clin Pathol 2000;114:9-15.
6. Cooper SD. The role of the laboratory information system in diagnostic services. In:
Clinical diagnostic technology: the total testing process. Volume 1: the preanalytical phase.
Ward-Cook KM, Lehman CA, Schoeff LE, Williams RH, ed. Washington, DC: American
Association of Clinical Chemistry, 2003.
7. Skjei E. Pulling out all the stops in the LIS marketplace. CAP Today 2003.
8. Henricks WH. Information system issues facing clinical laboratories serving complex
integrated delivery systems. J Healthc Inf Manag 2000;14(3):55-67.
9. Lehmann CA, Cappello SC. Workflow analysis: an overview. In: Clinical diagnostic
technology: the total testing process. Volume 1: the preanalytical phase. Ward-Cook K,
Lehman CA, Schoeff LE, Williams RH., ed. Washington, DC: American Association of
Clinical Chemistry, 2003.
10. Continued progress: hospital use of information technology. Washington, DC: American
Hospital Association, 2007.
11. Kizer KW, Garthwaite TL. Vision for change: an integrated service network. In:
Computerizing large integrated health networks: the VA success. Kolodner RM, Douglas
JV, ed. New York, NY: Springer, 2007.
12. Statement of Frances M. Murphy, MD, MPH, acting deputy undersecretary for health for
policy and management, Department of Veterans Affairs: Hearing before the
Subcommittee on National Security, Veterans Affairs, and International Relations of the
House Committee on Government Reform, 106th Congress, 2nd session. 2000. 2000.
http://www.va.gov/OCA/testimony/hgrc/10ap00FM.asp.
13. VistA monograph: 2005-2006. Washington, DC: Office of Information, Veterans Health
Administration, Department of Veterans Affairs, 2005. (Accessed October 25, 2007, at
http://www.va.gov/vista_monograph/docs/vista_monograph2005_06.pdf.)
14. Statement of Linda D. Koonta, director, information management issues: testimony before
the Subcommittee on Federal Financial Management, Government Information, and
International Security of the Senate Committee on Homeland Security and Governmental
Affairs, 109th Congress, 2nd session. 2006. http://www.gao.gov/new.items/d06905t.pdf.

May 2008

263

Laboratory Medicine: A National Status Report

Chapter VI – Laboratory Information Systems

15. Orsulak PJ. Stand-alone automated solutions can enhance laboratory operations. Clin
Chem 2000;46(5):778-83.
16. Markin RS, Newcomb MC. Selection of laboratory automation technology: instruments,
workcells, and systems. In: Clinical diagnostic technology: the total testing process.
Volume 2: the analytical phase. Ward-Cook KM, Lehman CA, Schoeff LE, Williams RH,
ed. Washington, DC: American Association of Clinical Chemistry, 2005.
17. Mohammad AA, Okorodudu AO, Petersen JR, Sciacca RD. Assessment of processes in the
clinical laboratory through computer simulation. In: Clinical diagnostic technology: the
total testing process. Volume 2: the analytical phase. Ward-Cook KM, Lehman CA, Schoef
LE, Williams RH, ed. Washington, DC: American Association of Clinical Chemistry, 2005.
18. Institute of Medicine. Patient safety: achieving a new standard of care. Washington, DC:
National Academy Press, 2004.
19. Vail GR. Middleware: what's the buzz? MLO Med Lab Obs 2006:50-1.
20. Paxton A. Smooth operator in the lab: middleware. CAP Today 2006.
21. Friedman BA. The total laboratory solution: a new laboratory e-business model based on a
vertical laboratory meta-network. Clin Chem 2001;47(8):1526-35.
22. Duncan H, Williams M, Hetsko CM. Challenges of the POL. Advance for Medical
Laboratory Professionals 2006:26-35.
23. Simpson B. Laboratory Corporation of America. Personal communication.
November 15, 2007.
24. Lemmerbrock S. MedPlus, Inc. Personal communication. December 10, 2007.
25. Institute of Medicine. Crossing the quality chasm: a new health system for the 21st
century. Washington, DC: National Academy Press, 2001.
26. Zakowski J, Powell D. The future of automation in clinical laboratories. IVD Technology 1999.
27. Markin RS, Whalen SA. Laboratory automation: trajectory, technology, and tactics. Clin
Chem 2000;46(5):764-71.
28. Melanson SE, Lindeman NJ, Jarolim P. Selecting automation for the clinical chemistry
laboratory. Arch Pathol Lab Med 2007;131:1063-9.
29. Middleton SR. Developing an automation concept that is right for your laboratory. Clin
Chem 2000;46(5):757-63.
30. Herzog DP. Emerging trends in lab automation and instrumentation. IVD Technology 2006.
31. Hoffman MA. The genome-enabled electronic medical record. J Biomed Inform
2007;40(1):44-6.
32. Promoting safe and effective genetic testing in the United States: final report of the Task
Force on Genetic Testing. Holtzman NA, Watson MS, ed. Bethesda, MD: National Human
Genome Research Institute, National Institutes of Health, 1997.
http://www.genome.gov/10001733.
33. Asare AL, Caldwell CW. An information system for improving clinical laboratory
outcomes. Proc AMIA Symp 2000:22-6.

May 2008

264

Laboratory Medicine: A National Status Report

Chapter VI – Laboratory Information Systems

34. Sembiante T. Lab info systems ease workflow. MLO Med Lab Obs 2004.
35. Tuthill JM, Klatt EC. Information technology in the laboratory. Laboratory Medicine
2001;7(32):356-60.
36. Hoffman M, Arnoldi C, Chuang I. The clinical bioinformatics ontology: a curated semantic
network utilizing RefSeq information. Pac Symp Biocomput 2005:139-50.
37. Clinical Bioinformatics Ontology: frequently asked questions. Kansas City, MO: Cerner
Corporation, 2007. (Accessed August 3, 2007, at
https://www.clinbioinformatics.org/cbopublic/faq.aspx.)
38. NCBI Reference Sequences. Bethesda, MD: National Center for Biotechnology
Information, National Library of Medicine, National Institutes of Health, 2007. (Accessed
August 3, 2007, at http://www.ncbi.nlm.nih.gov/RefSeq/.)
39. Sullivan E. To the middle and beyond: exploring the expanding universe of laboratory
software. Laboratory Medicine 2006;37(7):407-11.
40. Yu U, Lee SH, Kim YJ, Kim S. Bioinformatics in the post-genome era. J Biochem Mol Biol
2004;37(1):75-82.
41. Alterovitz G, Liu J, Chow J, Ramoni MF. Automation, parallelism, and robotics for
proteomics. Proteomics 2006;6(14):4016-22.
42. Goh CS, Lan N, Echols N, et al. SPINE 2: a system for collaborative structural proteomics
within a federated database framework. Nucleic Acids Res 2003;31(11):2833-8.
43. Dictionary of cancer terms. Bethesda, MD: National Cancer Institute, National Institutes of
Health, 2007. (Accessed August 7, 2007, at http://www.cancer.gov/dictionary/.)
44. Pharmacodynamics. Bethesda, MD: Medline Plus, National Library of Medicine, National
Institutes of Health, 2007. (Accessed August 7, 2007, at http://www2.merriamwebster.com/cgi-bin/mwmednlm?book=Medical&va=pharmacodynamic.)
45. Altman RB, Klein TE. Challenges for biomedical informatics and pharmacogenomics.
Annu Rev Pharmacol Toxicol 2002;42:113-33.
46. Guidelines and recommendations for laboratory analysis and application of
pharmacogenetics to clinical practice. Washington, DC: National Academy of Clinical
Biochemistry, 2006. http://www.aacc.org/NR/rdonlyres/AE772314-08C0-4F75-9895D40B7181E4F8/0/LMPG_Pharmacogenetics.pdf.
47. A primer on physician order entry. Oakland, CA: California HealthCare Foundation,
2000. http://www.chcf.org/documents/hospitals/CPOEreport.pdf.
48. Koppel R, Metlay JP, Cohen A, et al. Role of computerized physician order entry systems
in facilitating medication errors. JAMA 2005;293(10):1197-203.
49. Metzger J, Turisco F. Computerized physician order entry: a look at the vendor
marketplace and getting started. Long Beach, CA: First Consulting Group, 2001.
http://www.leapfroggroup.org/media/file/Leapfrog-CPOE_Guide.pdf.
50. Computerized physician order entry: costs, benefits and challenges--a case study
approach. Long Beach, CA: First Consulting Group, 2003.

May 2008

265

Laboratory Medicine: A National Status Report

Chapter VI – Laboratory Information Systems

http://www.hospitalconnect.com/ahapolicyforum/resources/content/AHAFAHCPOE
ReportFINALrevised.pdf.
51. Poon EG, Jha AK, Christino M, et al. Assessing the level of healthcare information
technology adoption in the United States: a snapshot. BMC Med Inform Decis Mak 2006;6:1.
52. Blechner M, Dighe AS. Clinical decision support: how laboratorians can provide clinicians
with effective tools. Clinical Laboratory News 2006:10-2.
53. Skjei E. Helping clinicians cope with CPOE systems. CAP Today 2002.
54. Mekhjian HS, Kumar RR, Kuehn L, et al. Immediate benefits realized following
implementation of physician order entry at an academic medical center. J Am Med Inform
Assoc 2002;9(5):529-39.
55. Ash JS, Stavri PZ, Kuperman GJ. A consensus statement on considerations for a successful
CPOE implementation. J Am Med Inform Assoc 2003;10(3):229-34.
56. Factsheet: computer physician order entry. Washington, DC: The Leapfrog Group, 2007.
(Accessed August 15, 2007, at http://www.leapfroggroup.org/media/file/LeapfrogComputer_Physician_Order_Entry_Fact_Sheet.pdf.)
57. Garg AX, Adhikari NK, McDonald H, et al. Effects of computerized clinical decision
support systems on practitioner performance and patient outcomes: a systematic review. J
Am Med Inform Assoc 2005;293(10):1223-38.
58. Osheroff JA, Teich JM, Middleton BF, Steen EB, Wright A, Detmer DE. A roadmap for
national action on clinical decision support. Bethesda, MD: American Medical Informatics
Association, 2006. http://www.amia.org/inside/initiatives/cds/cdsroadmap.pdf.
59. Longhurst C ,Hahn JS. Clinical decision-support systems in pediatrics. Reston, VA: Isabel
Healthcare, 2005. (Accessed March 20, 2007, at
http://www.isabelhealthcare.com/info/images/COCITnewsletterCDSSinPediatrics(Longhurst,2005).pdf.)
60. Bates DW, Kuperman GJ, Wang S, et al. Ten commandments for effective clinical decision
support: making the practice of evidence-based medicine a reality. J Am Med Inform
Assoc 2003;10(6):523-30.
61. DXplain: using decision support to help explain clinical manifestations of disease. Boston,
MA: Laboratory of Computer Science, Massachusetts General Hospital, 2007. (Accessed
March 20, 2007, at http://lcs.mgh.harvard.edu/projects/dxplain.html.)
62. Singer A. Book reviews: DiagnosisPro 5.0 CD-ROM by MedTech USA, Inc. The
Permanente Journal 2002;6(3):84.
63. Greenes RA. Why clinical decision support is hard to do. AMIA Annu Symp Proc
2006:1169-70.
64. Seliger R. Overview of HL7's CCOW standard. Ann Arbor, MI: Health Level Seven, 2001.
http://www.hl7.org/library/committees/sigvi/ccow_overview_2001.doc.
65. Price CP, Saint John A, Hicks JM. Point-of-care testing. In: Clinical diagnostic technology:
the total testing process. Volume 2: the analytical phase. Ward-Cook KM, Lehman CA,

May 2008

266

Laboratory Medicine: A National Status Report

Chapter VI – Laboratory Information Systems

Schoef LE, Williams RH, ed. Washington, DC: American Association of Clinical
Chemistry, 2003.
66. Felder RA, Graves S, Mifflin T. Reading the future: the increased relevance of laboratory
medicine in the next century. MLO Med Lab Obs 1999;31(7):20-6.
67. Nichols JH. Point-of-care testing data management. Point of Care 2004;3(1):8-10.
68. Connectivity Industry Consortium organization plan. Dusseldorf, Germany: European
Round Table Initiative, 1999. http://www.poct.fraunhofer.de/pdfs/cic_plan.pdf.
69. Point-of-care connectivity. Approved guideline: POCT1-A2; second edition. Wayne, PA:
Clinical Laboratory Standards Institute, 2006.
70. Implementation guide of POCT1 for healthcare providers. Proposed guideline: POCT2-P.
Wayne, PA: Clinical Laboratory Standards Institute, 2007.
71. DuBois JA. Challenges for implementing connectivity standards for point-of-care testing.
Point of Care 2002;1(2):111-3.
72. Point-of-care in vitro diagnostic (IVD) testing. Approved guideline: POCT4-A2; second
edition. Wayne, PA: Clinical Laboratory Standards Institute, 2006.
73. Point-of-care blood glucose testing in acute and chronic care facilities. Approved guideline:
C30-A2; second edition. Wayne, PA: Clinical Laboratory Standards Institute, 2002.
74. Point-of-care monitoring of anticoagulation therapy. Approved guideline: H49-A. Wayne,
PA: Clinical Laboratory Standards Institute, 2004.
75. Khan AI, Vasquez Y, Gray J, Wians FH, Kroll MH. The variability of results between
point-of-care testing glucose meters and the central laboratory analyzer. Arch Pathol Lab
Med 2006;130:1527-32.
76. Luley C, Ronquist G, Reuter W, et al. Point-of-care testing of triglycerides: evaluation of
the Accutrend triglycerides system. Clinical Chem 2000;46(2):287-91.
77. Hortin GL. Beyond traditional quality control. How to check costs and quality of point-ofcare testing. MLO Med Lab Obs 1997;29(9):31-7.
78. Garets D, Davis M. Electronic medical records versus electronic health records: yes, there
is a difference. Chicago, IL: HIMSS Analytics, 2006.
http://www.himssanalytics.org/docs/WP_EMR_EHR.pdf.
79. Health information technology in the United States: the information base for progress.
Princeton, NJ: Robert Wood Johnson Foundation, 2006.
http://www.rwjf.org/files/publications/other/EHRReport0609.pdf.
80. Middleton B, Hammond WE, Brennan PF, Cooper GF. Accelerating U.S. EHR adoption:
how to get there from here. Recommendations based on the 2004 ACMI retreat. J Am Med
Inform Assoc 2005;12(1):13-9.
81. French DH. The laboratory's role in the universal electronic health record. MLO Med Lab
Obs 2006;38(4):10-5.
82. HITSP interoperability specifications: electronic health record laboratory results reporting,
biosurveillance, and consumer empowerment. Executive overview. Chicago, IL:

May 2008

267

Laboratory Medicine: A National Status Report

Chapter VI – Laboratory Information Systems

Healthcare Information and Management Systems Society, 2006.
http://www.himss.org/content/files/HITSP/ExecutiveSummary.pdf.
83. Harmonized use case for electronic health record (laboratory result reporting).
Washington, DC: Office of the National Coordinator for Health Information Technology,
Department of Health and Human Services, 2006.
http://www.hhs.gov/healthit/documents/EHRLabUseCase.pdf.
84. Use case #3, electronic health record: accessing laboratory results. Washington, DC: Office
of the National Coordinator for Health Information Technology, Department of Health
and Human Services, 2006.
85. Only time will tell, but LIS experts offer a forecast. CAP Today 2005:24-56.
86. Medicare and state health care programs: fraud and abuse; safe harbors for certain
electronic prescribing and electronic health records arrangements under the anti-kickback
statute; final rule. Fed Regist 2006;71(152):45110-37.
87. CALINX vs. ELINCS: what's the difference? Oakland, CA: California Health Care
Foundation, 2006. (Accessed March 14, 2007, at
http://www.chcf.org/topics/chronicdisease/index.cfm?itemID=118636.)
88. ELINCS: developing a national lab data standard for EHRs. Oakland, CA: California
Health Care Foundation, 2006. (Accessed March 14, 2007, at
http://www.chcf.org/topics/chronicdisease/index.cfm?itemID=108868&subsection=rep
orts?redirectSource=elincs%2Eorg.)
89. Implementing the ELINCS standard: technical experience from the field. Oakland, CA:
California HealthCare Foundation, 2007.
http://www.chcf.org/documents/chronicdisease/ImplementingTheELINCSStandardLes
sonsIB.pdf.
90. Leong FJ, Leong AS. Digital imaging applications in anatomic pathology. Adv Anat Pathol
2003;10(2):88-95.
91. Cenci M, Nagar C, Giovagnoli MR, Vecchione. The PAPNET system for quality control of
cervical smears: validation and limits. Anticancer Res 1997;17:4731-4.
92. Wilbur DC, Prey MU, Miller WM, Pawlick GF, Colgan TJ. AutoPap system detection of
infections and benign cellular changes: results from primary screener clinical trials. Diagn
Cytopathol 1999;21:355-8.
93. About telemedicine. Washington, DC: American Telemedicine Association, 2007.
(Accessed March 12, 2007, at http://www.atmeda.org/news/library.htm.)
94. Clinical guidelines for telepathology. Tucson, AZ: American Telemedicine Association
Special Interest Group for Telepathology, 1999. (Accessed March 12, 2007, at
http://telepathology.upmc.edu/ata/guideline.htm.)
95. Kayser K, Kayser G, Radziszowski D, Oehmann A. From telepathology to virtual
pathology institution: the new world of digital pathology. Rom J Morphol Embryol
1999;45:3-9.
96. McCullough B, Ying X, Monticello T, Bonnefoi M. Digital microscopy imaging and new
approaches in toxicologic pathology. Toxicol Pathol 2004;32 Suppl 2:49-58.

May 2008

268

Laboratory Medicine: A National Status Report

Chapter VI – Laboratory Information Systems

97. Hammond WE. Overview of health care data standards. Commissioned paper for the
Institute of Medicine's Committee on Data Standards for Patient Safety. Washington, DC:
The National Academy, 2002.
98. Laboratory exchange meeting: final report. Rockville, MD: Agency for Healthcare
Research and Quality, 2006.
99. Integrating the Health Care Enterprise: laboratory technical framework. Volume 1
(LTF-1)—integration profiles. Chicago, IL: Healthcare Information and Management
Systems Society, 2004.
100. 18th annual 2007 HIMSS leadership survey: CIO results final report. Chicago, IL:
Healthcare Information and Management Systems Society, 2007.
http://www.himss.org/2007Survey/DOCS/18thAnnualLeadershipSurvey.pdf.

May 2008

269

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

CHAPTER VII
FEDERAL REGULATORY OVERSIGHT OF LABORATORY MEDICINE
In the U.S. health care system, the purposes of regulation include one or more of: protect and
promote personal and public health, advance personal and public health, and ensure that the
public has access to sufficient, accurate information for using regulated products and services to
improve their health. Regulation is designed to protect consumers by assuring certain levels of
quality, such as acceptable performance, safety, and effectiveness of products and services; and to
establish market conditions that will generate validated innovations for improving health, such as
controlling elements of monopoly power, and minimizing fraud and abuse.1, 2 The purpose of
oversight by designated agencies and organizations is to enforce and otherwise achieve adherence
to the rules and standards comprising regulation.
Three main elements of oversight are: information development and synthesis, standards setting,
and compliance mechanisms.3 Information development and synthesis refers to the scientific
studies, data collection, and reporting requirements for identifying and measuring potential
benefits and harms of a product or service. Standards setting is a process for identifying and
describing the attributes that a product or service should have in order to offer an acceptable mix
of benefits and risks. Compliance mechanisms refer to the manner and extent to which standards
are followed and enforced, varying from traditional “command and control” regulatory
approaches to informal or voluntary approaches.
In its 2001 report, Crossing the Quality Chasm, the IOM characterized U.S. regulation of health care
as a patchwork of laws, regulations, agencies, and accreditation processes through which each
care delivery system must navigate at the federal, state, and local levels.4 The regulatory
frameworks at the various levels are often inconsistent, contradictory, and duplicative, in part
because of their often disparate origins, jurisdictions and evolution, as well as because their
respective needs, priorities, and available resources are poorly articulated and inadequately
coordinated. Moreover, current regulatory frameworks often lag behind changes in the health
system, including those related to advances in scientific and medical knowledge and
technological innovation. This chapter provides an overview of the federal regulatory oversight
in laboratory medicine by CMS and FDA.

REGULATORY OVERSIGHT SYSTEMS
Regulatory oversight of the laboratory medicine sector is conducted at multiple levels by
numerous governmental and non-governmental bodies. These include federal and state
legislatures, federal and state regulatory agencies, professional and private sector organizations,
and federal and state courts.

Legislative Oversight
The U.S. Congress establishes provisions for oversight through the passage of legislation that
governs various aspects of laboratory medicine, including the Federal Food, Drug, and Cosmetic
Act (FDCA) and CLIA. At the federal and state levels, legislatures can delegate authority to
regulatory agencies to interpret, apply, and enforce the statutory standards.3 Federal and state

May 2008

271

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

agencies providing oversight generally have clearly defined jurisdictions for which they may
exercise regulatory powers and controls.

Federal Oversight
Several DHHS agencies share oversight responsibility for health care products and services at the
federal level. In particular, CMS, FDA, and CDC have prominent oversight roles for clinical
laboratories that generally complement one another. CMS is responsible for oversight of clinical
laboratories and their testing services under CLIA. This includes implementation of the CLIA
program and provisions that govern PT, QC, personnel requirements and training; laboratory
inspections; enforcement of CLIA regulations; and approval of PT providers, accrediting
organizations, and CLIA-exempt states.5 CMS’ authority extends to oversight of tests developed
”in-house” by a laboratory, i.e., laboratory-developed tests (LDTs), that may or may not have
sought FDA approval. FDA is responsible for test categorization and for regulating diagnostic
tests and certain software used in laboratory information systems under the provisions of the
FDCA for medical devices. These tests are developed by manufacturers and regulated as
products, e.g., test kits. CDC’s responsibilities include conducting CLIA-related studies, convening
CLIAC, and providing scientific and technical support and consultation to CMS.
The DHHS OIG also has a role in oversight of clinical laboratories. The OIG conducts audits,
investigations, inspections, and other evaluations to protect the integrity of DHHS programs,
including FDA and CMS oversight, as well as the health and welfare of consumers and
beneficiaries. OIG provides reports to the Secretary of DHHS and Congress on findings and
recommendations for corrective action, as appropriate.
Other federal agencies regulating specific types of clinical laboratories include the DoD and the
VHA. Laboratories in the DoD Military Health System are subject to CLIA requirements.
However, a memorandum of understanding with DHHS permits MHS to operate independently
following a successful review giving it “deemed status” for CLIA certification. In lieu of CLIA,
separate legislative requirements govern laboratories in the VHA.

State Oversight
States can take a prominent regulatory role in laboratory medicine, although not all states
uniformly do so. Currently, 26 states have some degree of statutory authority for oversight of
clinical laboratories.3 A state may also apply to CMS for exempt status. If CMS approves, the
state is recognized to be the primary regulatory authority of clinical laboratories within that state,
and is determined to be exempt under CLIA. At this time, only two states, New York and
Washington, are CLIA-exempt.

Private Sector Oversight
Other important sources of formal and informal regulatory oversight include professional and
private sector organizations such as The Joint Commission and CAP. In a formal capacity, a
federal oversight body may delegate regulatory oversight to selected organizations. Informally,
many of these organizations set professional standards to which compliance is voluntary or a
condition for accreditation.

May 2008

272

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

Judicial Decisions
Legal remedies sought through state and, to a lesser degree, federal courts influence regulatory
oversight. Tort actions brought to the courts can facilitate definition of standards, clarify
interpretation of laws, and enforce compliance with regulations and standards of conduct among
those that can be held accountable, including manufacturers, laboratories, health care providers,
and other parties.

CLIA PROGRAM
Overview of Clinical Laboratory Improvement Amendments of 1988
Efforts to decrease laboratory error rates and improve the quality of laboratory testing led to the
passage of the Medicare, Medicaid, and Clinical Laboratories Improvement Act of 1967
Programs (Public Law 90-174).a To obtain a license under that law, laboratories were required
to maintain records and equipment, perform QC, participate in PT, and set qualifications for
laboratory directors and other supervisory personnel. However, the legislation did not define
quality or set protocols to enforce performance standards, such as QC limits or PT criteria.
Furthermore, laboratories receiving fewer than 100 specimens per year (mostly POLs) were
exempt.b Thus, as many as 25% of all tests performed on patient specimens were not subject to
minimum quality standards.6
In the mid-1980s, the news media began to publicize the high incidence of laboratory errors in
many of the nation’s clinical laboratories, particularly reports of inaccurate test results from Pap
smears.7 These media reports generated public and congressional concerns about the quality of
clinical laboratory services in the U.S., prompting Congress to expand regulatory oversight of
clinical laboratories.8 The resulting action was passage of CLIA 1988 (Public Law 100-578), which
amended the 1967 law and revised the authority for the regulation of clinical laboratories
conducting testing on “human specimens for health assessment or for the diagnosis, prevention
or treatment of disease.”9
The CLIA regulations, published on February 28, 1992, established explicit minimum standards
for QA, QC, PT, record maintenance, certification, inspection, and personnel qualifications for all
laboratories,c including POLs.10 States and private organizations can have standards that are
more expansive and/or more stringent than CLIA. Since its implementation, CLIA has served as
the primary regulatory program governing the U.S. laboratory system. A revised final rule
published on January 24, 2003, included a restructuring of QC and QA requirements in a quality
systems framework.

The Medicare, Medicaid, and Clinical Laboratories Improvement Act of 1967 regulated laboratories engaged in
interstate commerce by declaring that “no person may solicit or accept in interstate commerce, directly or
indirectly, any specimen for laboratory examination or other laboratory procedures unless there is in effect a
license for such laboratory issued by the Secretary under this section applicable to such procedures.”
b The 1967 law did not apply to laboratories whose operations were so small or infrequent as not to constitute a
significant threat to the public health.
c The regulations exclude three types of testing: forensic testing; research testing for which patient-specific results are
not reported; and drug testing that is performed in laboratories certified and governed by the SAMHSA.
a

May 2008

273

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

The requirements are organized according to test complexity and the level of risk associated with
reporting erroneous results, rather than the type of laboratory in which testing is performed.
Waived tests follow a different regulatory pathway than non-waived tests, e.g., moderate
complexity tests, and high complexity tests. The regulatory requirements for each category are
considered the minimum requirements necessary to ensure accuracy, reliability, and timeliness of
laboratory test results.11 Appendix C of the final rule, Survey Procedures and Interpretive
Guidelines for Laboratories and Laboratory Services, is used to determine compliance with the
regulatory requirements of CLIA.12

Administration of CLIA
Chief authority for CLIA is delegated to the CMS, which conducts the program in conjunction
with CDC and FDA under an interagency agreement. CMS has responsibility for all financial
management and administrative operations of the program, including certification and fee
collection, inspections, enforcement, accreditation and state exemption approvals, PT program
approvals, and rulemaking. The CLIA program is funded entirely through user fees managed
by CMS.
CDC conducts assessment studies; provides scientific and technical support and consultation to
CMS in developing and revising technical standards, evaluating accreditation/exemption
applications, reviewing PT programs, and developing technical information and educational
materials; and manages the work of CLIAC.
FDA is responsible for laboratory test categorization, including test complexity and waiver
determinations. The transfer of responsibility for test categorization from CDC to FDA allowed
manufacturers of in vitro diagnostics (IVDs) to work primarily with one agency in pursuing
requests to market their devices. Manufacturers now submit IVDs for premarket review (via the
510(k) clearance process or the pre-market approval (PMA) process, as appropriate) and requests
for complexity categorizations solely to FDA.
In addition, professional and private sector organizations may apply to CMS for accreditation
status and, if approved, are provided with the authority to accredit clinical laboratories, evaluate
compliance with CLIA regulations, and educate laboratory staff to improve performance.
Currently, six accreditation organizations have received such approval from CMS;d
approximately 25% of all laboratories receive accreditation through one of these organizations.14

d

Accreditation organizations may be granted “deemed status” for up to six years. Designees include: AOA, AABB,
ASHI, CAP, COLA, and The Joint Commission.13

May 2008

274

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

WAIVED TESTS
The original CLIA statute required laboratories performing one or more of a given set of nine
types of tests or examinations (e.g., certain dipstick or tablet reagent urinalyses, fecal occult blood
tests, ovulation tests) to obtain a CW from CMSe and pay biennial fees. In a proposed rule
published in 1995, DHHS clarified the criteria and process for applying for waived status.15
Under the proposed rule, waived status can be obtained if a test employs methods that are so
simple and accurate as to render the likelihood of erroneous results by the user negligible or that
are determined by the Secretary of DHHS to pose no unreasonable risk of harm to the patient if
performed incorrectly.16, 17 The FDA Modernization Act of 1997 contained provisions further
amending these CLIA requirements, stating that a test will automatically be considered waived if
FDA has approved it for home use.17
As a result of simplification of testing procedures and advances in instrumentation, the number of
waived tests has increased dramatically. Tests that were once considered moderate complexity
are being redesigned, and when they meet certain criteria for being simple and accurate, FDA
may grant them waived status. These new waived tests are expected to make analysis easier,
faster and more accessible to the public while maintaining reliability and accuracy.
Since implementation of CLIA, the proportion of laboratories performing only waived testing has
increased from 20% to 58% of the more than 200,000 laboratories.18 From 1995 to 2005, the number
of laboratories performing waived tests increased by more than 70% in clinical laboratories affiliated
with non-exempt states and in POLs (see Table 7.1). Whereas there were approximately 200 waived
tests associated with 9 analytes available in the U.S. in 1993, these numbers had increased
dramatically by 2004 to more than 1,600 waived test systems that test for 76 analytes.18
Table 7.1: Number of Facilities that hold Certificates of Waiver
1995 to 2005
1995

2000

2005

2007

Labs in non-exempt states

65,031

85,944

113,445

119,839

POLs

28,951

40,990

52,632

54,467

Source: CLIA update—June 2007. Centers for Medicare and Medicaid Services. CLIA
database information, 2000. Centers for Medicare and Medicaid Services.

e

Still in the waived category, these tests include: (1) dipstick or tablet reagent urinalysis (nonautomated) for
bilirubin, glucose, hemoglobin, ketone, leukocytes, nitrite, pH, protein, specific gravity, and urobilinogen; (2) fecal
occult blood; (3) ovulation tests (visual color comparison tests for human luteinizing hormone); (4) urine
pregnancy tests (visual color comparison tests); (5) erythrocyte sedimentation rate (nonautomated); (6)
hemoglobin (copper sulfate, nonautomated); (7) blood glucose by glucose monitoring devices cleared by FDA
specifically for home use; (8) spun microhematocrit; and (9) hemoglobin by single analyte instruments providing
direct measurement and readout. See: 42 Code of Federal Regulations 493.15(c).

May 2008

275

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

OUTSTANDING ISSUES IN WAIVED TESTING
Risk-benefit Tradeoffs
One of the factors contributing to the growth in waived testing is the increasing number of
laboratories that are certified to perform only waived testing (Table 7.1).19 However, broad access to
some tests in this category poses evolving risk-benefit tradeoffs to patients and other stakeholders.
An example that presents such tradeoffs is the OraQuick Rapid HIV-1 Antibody Test (OraQuick), a
single-use qualitative immunoassay that detects antibodies to HIV-1 in a finger stick sample or whole
blood. An IVD, OraQuick was originally approved via the PMA route by FDA in November 2002 as a
moderate complexity test under CLIA. In January 2003, FDA granted CLIA waived status to
OraQuick.20 This made the test potentially available through many more health care providers and
about five times as many clinical laboratories. In March 2004, FDA approved the test for detection of
HIV-2 (a variant of HIV that is prevalent in parts of Africa but rarely found in the US) in whole blood,
with continued waived status. Some in the public health community lauded the easier access to HIV
testing and greater opportunity to reduce the number of individuals unaware of their HIV status. On
the other hand, many in the laboratory community expressed strong reservations about the
implications of waived status due to concerns about specimen adequacy, availability of counseling for
HIV positive results when testing is conducted in a waived setting, and the reliability of the device.
These concerns address whether specific waived tests really are so “accurate as to render the
likelihood of erroneous results by the user negligible” and present “no unreasonable risk of harm.”21, 22

Oversight for Certificate of Waiver Facilities
Facilities that perform only waived testing are not subject to many of the CLIA regulations
applied to those that perform non-waived testing, including QC, routine inspections, PT, and
personnel qualifications and training. Although there is no routine oversight of these facilities,
they are required to follow manufacturer’s instructions when performing testing and they must
permit inspections by designated authorities as part of random compliance evaluations and
complaint investigations.23 Due to the large number of waived facilities and this corresponding
lack of oversight, the government became concerned about conditions that have the potential to
contribute to errors and patient harm. Several studies were undertaken to determine the extent to
which waived facilities were meeting their obligation to follow manufacturers’ instructions and
complying with other federal regulations requiring safe work practices.
During 1999-2001, CMS conducted preliminary on-site surveys of a representative sample of CW
sites in 10 states. The pilot surveys detected certain quality concerns that had the potential to
result in medical errors. In 2001, the OIG published a report following its investigation of CLIA
certification and enrollment processes, identifying quality deficiencies in approximately 50% of
CW sites.24
In response to these findings, CMS reported results of nationwide on-site surveys of 4,214
laboratory sites during 2002-2004. (Such surveys are ongoing.) POLs comprised the largest
percentage (47%) of CW facilities surveyed, followed by skilled nursing facilities (14%).18 About
90% of facilities performed no more than 5 different waived tests, and another 9% performed no

May 2008

276

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

more than 10 different tests.f The CMS surveys indicated that the majority of CW sites were aware
of and followed manufacturer instructions; however, lapses in quality were identified at certain
sites. About 5% of sites were conducting tests with a degree of complexity that exceeded their CW
CLIA certification, which may compromise the accuracy of these tests and patient safety.g The most
frequently performed non-waived tests (72%) were direct microscopic examinations (e.g.,
potassium hydroxide preparations, wet mounts, or urine sediment examinations), although several
other types of non-waived tests also were reported. In addition to conducting these non-waived
tests, the sites were found to be non-compliant with CLIA requirements for laboratories performing
non-waived testing.
For those facilities who were conducting only waived testing, 12% were not using the most recent
manufacturer’s instructions and 21% did not routinely check the product’s package insert or
instructions for changes to the information.18 Relative to manufacturer’s instructions, 21% of CW
sites did not perform QC testing and 18% did not use the correct terminology or units of measure
when reporting results. Table 7.2 summarizes the prevalence of quality-related deficiencies in
waived facilities. Data from the CMS surveys are similar to the findings of CDC-funded studies
conducted from 1999 to 2003 by the Laboratory Medicine Sentinel Monitoring Network.h These
findings indicate a need for greater oversight of waived facilities, more stringent enforcement of
manufacturer’s requirements, and training of testing personnel. CMS has taken several measures
to support more frequent site visits to CLIA-waived laboratories in order to monitor compliance
of CW sites. Follow-up studies that evaluate the effect of strengthened enforcement on quality
and safety in testing have not yet been conducted.
CMS has data from follow-up visits to CW laboratories indicating that the initial educational
visits by CLIA surveyors contributed to improvements in these laboratories performance of at
least 70%. CMS plans to continue this program of annual educational visits to a sample of
waived laboratories.25

The most common waived tests performed were glucose, dipstick urinalysis, fecal occult blood, urine human chorionic
gonadotropin (visual color comparison), and group A streptococcal antigen (direct test from throat swabs).
g Conducting non-waived testing can be potentially hazardous to public health since these facilities do not have the
same QC measures as those certified for non-waived tests.
h Laboratory Medicine Sentinel Monitoring Network is a CDC network established through CDC Cooperative
Agreements with the state health departments of Arkansas, New York, and Washington.
f

May 2008

277

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

Table 7.2: Number and Percentage of Quality Deficiencies Related to
Following Manufacturer’s Instructions and Documentation in
Certificate of Waiver Sites from CMS Surveyed Sites*
2002-2004
Quality Deficiencies

No. of sites

Following manufacturer’s instructions

% of sites

£

The site did not:
Have current manufacturer’s instructions

485

12

701

21

Perform quality control testing

866

21

Report test results with terminology or units described in package insert

744

18

Adhere to proper expiration dates

267

6

Perform required confirmatory tests

265

6

Routinely check new product insert for changes

¤

Based on manufacturer’s instructions, the site did not:

Perform function checks or calibration

195

5

Adhere to storage and handing instructions

135

3

Perform instrument maintenance

125

3

Use appropriate specimen for each test

81

2

Add required reagents in the prescribed order

24

1

1,493

45

Documentation¶
The site did not:
Document the name, lot number and expiration date for all tests performed¤
Maintain a quality-control log

¤

Maintain a log of tests performed
Require test requisition (or patient chart) before performing a test¤
Keep the test report in the patient’s chart¤
Check patient identification

¤

1,151

35

1,318

31

304

9

56

2

31

1

Source: Howerton D, Anderson N, Bosse D, et al. Good laboratory practices for waived testing sites: survey findings from testing
sites holding a certification of waiver under the Clinical Laboratory Improvement Amendments of 1988 and recommendations for
promoting quality testing. MMWR Recommendations and Reports 2005;54(RR-13):1-25.
*

N= 4,214 sites

£

Required for waived testing under CLIA

¤

2003-2004 data only (n=3,317)

¶

Not required for waived testing under CLIA.

May 2008

278

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

NON-WAIVED TESTS
Diagnostic tests categorized by FDA as moderate and/or high complexity are considered nonwaived. Laboratories performing these tests must comply with the regulatory requirements
specified in CLIA for non-waived testing. Regulatory requirements include, but are not limited
to, QC and QA, participating in periodic PT, and meeting qualification requirements for
personnel. FDA categorizies the level of test complexity using specific CLIA regulatory criteriai
(see Box 7.1). Together, these requirements facilitate a laboratory’s ability to ensure quality
testing.
Box 7.1: Test Categorization
To categorize a test system, assay or examination, a grade of 1, 2 or 3 is assigned for each of seven criteria,
and scores are totaled. A score of <12 indicates categorization of moderate complexity, and a score of >13
indicates high complexity. Criteria include: (1) knowledge needed to perform test; (2) training and
experience required; (3) reagents and materials preparation; (4) characteristics of operational steps; (5)
calibration, quality control and proficiency testing materials; (6) test system troubleshooting and equipment
maintenance; and (7) degree of interpretation and judgment required.26
A subset of the moderate complexity category, PPM tests are specific microscopy procedures performed by a
physician, mid-level practitioner or dentist during the course of a patient’s visit (e.g., direct wet mount
preparations for presence or absence of bacteria). Under CLIA, separate certification requirements were
developed for PPM.j

State Exceptions
A few states and territories expressed interest in applying for exemption from CLIA, notably
California, New York, Oregon, Puerto Rico, and Washington. Washington was the first state to
have its clinical laboratory licensure program judged equivalent to CLIA and therefore was
granted an exemption from the federal regulation.k New York State holds an exemption for all of
its laboratories, except POLs. Oregon received exemption status on June 13, 1996, but formally
notified CMS of its decision not to renew its application. Oregon’s exemption period ended
December 31, 1999.27 Puerto Rico’s request for exemption was denied on October 28, 1996.
When a state is granted an exemption from CLIA, CMS may no longer charge a fee to the
laboratories in the exempt state. However, CMS does assess the state for its share of costs associated
with CLIA. The regulation indicates that exempt states must pay for costs incurred through federal
oversight investigations and surveys, their share of general overhead costs, and costs associated
with follow-up complaints.28 The weight of this cost for California, estimated at $2.4 million
annually, was the reason the state withdrew its initial application for exemption (after meeting all
requirements to be granted an exemption).29

In accordance with CLIA, CDC categorized more than 25,000 test systems before the responsibility for categorization
was transferred to FDA in 2000.
j Tests in the PPM category include: (1) all direct wet mount preparations for the presence or absence of bacteria, fungi,
parasites and human cellular elements; (2) all potassium hydroxide preparations; (3) pinworm examinations; (4) fern
tests; (5) post-coital direct, qualitative examinations of vaginal or cervical mucous; (6) urine sediment examinations;
(7) nasal smears for granulocytes; (8) fecal leukocyte examinations; (9) qualitative semen analysis (limited to
appearance of sperm and detection of motility).16
k Washington State Department of Health. Office of Laboratory Quality Assurance.
i

May 2008

279

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

CLIA Standards
The CLIA technical requirements were designed to establish quality standards for clinical
laboratory testing through the assurance of accuracy, reliability, and timeliness of patient test
results, regardless of where the test was performed.30 As noted above, the 2003 final rule
reorganized the technical standards for laboratories performing non-waived testing to reflect the
flow of a patient specimen through the laboratory (i.e., from receipt of the specimen with the test
request through test performance and test result reporting). However, the core technical
standards related to analytic phase QA and QC remain intact, including PT.l Although CLIA
always required monitoring of all phases of testing, the reorganization more clearly aligns the
regulations toward a comprehensive QMS-based approach. Laboratories must establish and
follow written policies and procedures for an ongoing mechanism to monitor, assess and, when
indicated, correct problems identified in the preanalytic and postanalytic phases.32
Data are limited regarding the impact of preanalytic and postanalytic QA. The next section
provides a brief description of unresolved issues associated with QC and PT. Challenges
concerning personnel qualifications and provisions for surveys and sanctions also are covered.
Issues in regulatory oversight of genetic testing are discussed in a separate section. More extensive
discussion on the new CLIA framework for systems-based approaches is provided in the Quality
Systems and Performance Measurement chapter of this report. In a forthcoming report, CDC will
publish recommendations of an expert workgroup for enhancing the effectiveness of PT.

OUTSTANDING ISSUES ASSOCIATED WITH CLIA STANDARDS
Methods for Analyzing PT Data
PT is an objective means of evaluating analytical test performance in a laboratory. It assesses
whether compliance with CLIA has improved intra-laboratory quality.33 All laboratories that
perform non-waived testing are required to participate in PT. For most regulated analytes, each
participating laboratory receives specimens three times a year from external, CLIA-approved PT
providers.m,34 The laboratory tests these samples in the same manner that other tests are
performed and reports the test results to their PT provider. The PT provider grades the results to
determine their accuracy and compares them to peer groups or reference laboratory performance,
i.e., inter-laboratory comparisons. PT is required for each specialty, subspecialty, analyte, or test
listed in the regulations for which the laboratory is certified.
Failure to attain an overall testing event score of at least 80% is considered unsatisfactory
performance for all specialties and subspecialties, except for gynecologic cytology (90%), ABO
QA refers to the internal and external planned and systematic activities to monitor all components or characteristics
that affect quality and customer satisfaction, including policies and procedures to ensure compliance with
regulatory requirements. PT is an external QA mechanism. QC refers to the internal procedures and statistical
analyses for day-to-day monitoring of analytic instruments and work processes, detecting problems, and
implementing corrections prior to the delivery of products or services.31
m Approved PT provider programs for 2007 include: Accutest, Inc., American Academy of Family Physicians,
American Association of Bioanalysts, American Proficiency Institute, California Thoracic Society, COLA, CAP,
Medical Laboratory Evaluation Program, New Jersey Department of Health and Senior Services, Commonwealth
of Pennsylvania, Puerto Rico Proficiency Testing Service, Wisconsin State Laboratory of Hygiene, Maryland
Department of Health and Mental Hygiene, ASCP, and New York State Department of Health.
l

May 2008

280

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

group and D (Rho) typing (100%), and compatibility testing (100%). Unsatisfactory performance
on two consecutive or two out of three testing events constitutes unacceptable performance for
the laboratory.35 A laboratory with an unacceptable performance rating must cease testing the
failed analyte until it passes two consecutive “remedial” PT events. PT results are transmitted by
the PT providers to a CMS database. According to the GAO, PT is the one available data source
that can be used to uniformly compare laboratory quality across survey organizations.36
Few studies have examined the long-term impact of PT on laboratory performance. A report
published in 2007 examined PT performance in physician office, clinic, and small hospital
laboratories during the ten-year period 1994-2004, using data from a PT provider, the American
Proficiency Institute. The data showed failure rates for chemistry and hematology analytes
declined significantly over the ten-year period. For example, failure rates for cholesterol testing
dropped from 18.7% in 1994 to 3.2% and failure rates for glucose testing declined from 15.6% to
2.4%. Although failure rates for microbiology analytes also declined, they still exceeded 5% for
certain tests in 2004, including positive genital/gonorrhea cultures, positive urine cultures, and
Gram stains. Several limitations of the study were noted by investigators: the data were not
stratified by type of laboratory and thus do not reflect the performance of laboratories that are
newly-regulated under CLIA; PT focuses on the analytical phase of testing and cannot detect
errors in the pre- and postanalytical phases; and the data reflect trends in a changing population
of laboratories but not individual laboratory trends, making it difficult to determine if most
laboratories improved or if poor performing laboratories ceased testing.37 Studies by CDC and
the State of California also have been instrumental in generating key assessments of PT success
and failure rates in POL and non-POL settings.38, 39
Similarly, the PT results do not necessarily indicate that a laboratory’s proficiency is sufficient to
meet the needs of the clinician.33 CMS would increase transparency of PT if it were to consider
new ways to code certain tests for PT purposes, including disaggregation of tests within specialty
areas. For example, as noted by the authors of the report referenced above on PT performance
during 1994-2004, CMS aggregates all bacteriology tests (cultures, Gram stains, and susceptibility
studies) under one code, which means that statistics depicting unsatisfactory performance in
bacteriology (5.1% of laboratories) are somewhat misleading.37 This is particularly the case in
bacteriology, since methods of analysis can differ substantially, as some of these tests do not have
analytes while others do. Instead, analyzing failure rates for individual tests would show more
clearly those areas posing the greatest challenges in order to focus improvement activities.
Laboratories can interpret and use their PT results to improve their practices, along with their QC
and QA data, whether or not grades are satisfactory.37, 40, 41 A recent study of detection and
correction of systematic laboratory problems found that, although accredited laboratories
generally perform well in PT, having the PT provider identify clusters of PT failures assists the
laboratory in correcting the problems.42 Additional studies on the use of feedback from PT could
facilitate further improvements in performance.

May 2008

281

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

Cytology PT
CLIA mandates that all laboratorians involved in the testing of gynecologic cytology specimens
participate annually in a CMS-approved PT program.n,o,43, 44 Cytology PT consists of 10
gynecologic cytology slides, which test-takers must examine and diagnose within two hours. Of
the 12,831 individuals who took the initial test in 2005, 93% of cytotechnologists passed the exam,
67% of pathologists working without cytotechnologist passed, and 90% working with
cytotechnologists passed.p,46
Concerns have been raised about the structure of cytology PT programs. CLIA requires
unanimous agreement among at least three anatomic pathologists in order for a slide to be
included in a cytology PT program.47 However, the regulations do not require that the slides be
subject to the rigorous review usually undertaken during field validation studies. Some
organizationsq have found that slides selected by expert pathologists as good examples of
cytodiagnostic abnormalities ultimately fail field validation. A 2005 study of the CAP’s
Interlaboratory Comparison Program in Cervicovaginal Cytology reported that, of more than
10,000 conventional smears and ThinPrep cases selected by an expert panel of 3 cytopathologists,
19% of conventional smears and 15% of ThinPrep specimens failed field validation.48
It is not apparent whether cytology PT programs lead to improvement in the quality of Pap
testing. A 1999 study compared accuracy of diagnosis of Pap smear slides from 40,245 women
examined by 81 cytology screeners with the PT scores received by the same screeners.49
Correlation between the accuracy of diagnosis of the Pap smear slides and PT results led the
researchers to conclude that performance on a 10-slide test gives only some indication of the true
performance of screeners. A study published in 2000 found a low correlation between cytology
PT and actual work performance. Researchers concluded that cytology PT should be considered
just one measure of performance and should be evaluated in conjunction with other quality
assessment monitors, such as re-screening studies, discrepancy rates, and workload patterns.50
Several efforts could alter the structure of cytology PT. CLIAC convened a workgroup to
consider changes to the regulations in several areas: participation in educational programs, use
of new technology (virtual media), testing frequency, number of challenges, categories of
challenges, number of challenges per category, grading scheme, validation, test site, retesting,
and confidentiality.51 CMS has been developing a Notice of Proposed Rulemaking based on the
CLIAC’s recommendations. In 2006, the Cytology Proficiency Improvement Act was
introduced in the Senate and the House (H.R. 6133, S. 4056), which would have amended CLIA
to require all individuals involved in screening and interpreting cytological preparations to
participate in annual continuing medical education programs in gynecologic cytology, rather

Laboratories performing only nongynecologic cytology are not required to enroll in PT programs.
Three organizations are currently approved by CMS to conduct cytology PT: CAP, the State of Maryland Cytology PT
Program, and ASCP.
p Individuals have up to four opportunities to receive a passing score. After not passing on the fourth opportunity, they
must cease examining Pap smears and must obtain at least 35 hours of continuing education in diagnostic cytology.45
q Slides that are donated to CAP’s Interlaboratory Comparison Program in Cervicovaginal Cytology are reviewed by a
panel of three expert cytopathologists; slides that are determined to be good examples of a single cytodiagnostic entity
enter the PT program, after which they undergo a field validation process in order to designate each slide as “graded,”
a designation that marks that slide as being regularly and reliably identified by CAP cytology PT participants.48
n
o

May 2008

282

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

than in PT programs.52, 53 The bill, never enacted into law, was re-introduced in the House in
2007 (H.R. 1237).54

Guidance on QC protocols, particularly for innovative testing technologies
QC is designed to ensure that instruments perform as expected by controlling factors that affect
test quality during the analytic phase, including technical and methodological variables,
environment, and personnel performance.r Generally, laboratories are required to run at least
two levels of control per day, but the CLIA final rule considered technological changes in altering
the frequency of QC testing in certain specialty and subspecialty areas of testing.56 The CLIA
interpretative guidelines provide laboratories with the flexibility to determine control procedures
that are equivalents to the traditional QC frequency of two levels of control per day, i.e.,
“equivalent QC.”12 The decision whether to implement equivalent QC is the responsibility of
laboratory directors, not manufacturers or regulators, but can occur only if all of the laboratory’s
quality systems are functioning properly.
Implementation of the CLIA requirements has resulted in significant decreases in the number of QC
deficiencies (see Figure 7.1). A study of the impact of QC implementation in enzyme immunoassay
testing for HIV-1 antibodies found that laboratories not using QC were at increased risk of
systematic error, but adhering to CLIA requirements for QC was more protective against error.57
However, several factors indicate a need for greater guidance in the area of QC, including the
emergence of increasingly complex, innovative testing technologies (e.g., genomics, proteomics,
PGx) and the need for immediate detection of errors and monitoring of ongoing test performance
through QC protocols. Specifically, additional QC guidance is needed in light of the diversity of
technology used to perform genetic and molecular-based tests, the rate at which this technology
evolves, regional differences in the tests that are offered and the populations that are tested, and the
lack of standardization of laboratory-developed genetic tests.58 Given these factors, CLIA could fall
short in assuring quality in laboratory testing in the future. CMS is working with the genetics
community to develop specific guidance for laboratories to address these QC needs.

Under CLIA, laboratories also must implement and monitor quality systems for the pre- and postanalytic phases,
thereby emphasizing the role of QA throughout the entire testing process.55
s The interpretive guidelines allow a laboratory to analyze two external controls per day for 10, 30, or 60 consecutive
days to evaluate equivalent QC. If the comparison of internal and external controls is acceptable during this full
testing period, the laboratory may adopt equivalent QC and increase the interval for analyzing external controls to
every 7 or 30 days, depending on the equivalent QC option selected.
r

May 2008

283

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

Figure 7.1: Percent of Labs with Major Quality Control Deficiencies

Source: Memorandum from Mark McClellan, CMS Administrator, to Leslie Aronovitz,
GAO, May 17, 2006. Baltimore, MD: Centers for Medicare and Medicaid Services, 2006.

During the phase-in, FDA was to establish a process to review and clear manufacturers’ instructions
for use in QC protocols.56 Once approved, laboratories would follow manufacturers’ instructions
for QC rather than develop their own QC protocols.59 The expected date for phasing-in this QC
requirement was extended four times (December 6, 1994, May 12, 1997, October 14, 1998, and
December 29, 2000) to allow completion of the phase-in period. In the final rule, laboratoriest
retained responsibility for establishing and adhering to written QC procedures for monitoring and
evaluating the quality of the analytical testing process for each method of testing.56
Another concern is use of the interpretive guidelines in deciding whether to implement equivalent
QC. CLIA states that the laboratory director must consider the laboratory’s clinical and legal
responsibility for providing accurate and reliable patient test results versus the potential cost
savings of reducing the QC testing frequency.56 Laboratory directors are likely to be aware of such
tradeoffs in fulfilling their responsibilities; however, the lack of a framework for implementing QC
for the wide variety of test systems may create inconsistencies in implementing QC.23 Factors that
contribute to these inconsistencies may include the lack of adequate risk management information
from manufacturers, the different types and levels of QC required by each device and method, and
the unique considerations of individual laboratories (e.g., personnel).60

t

Although not part of the QC requirements, laboratories with unmodified FDA-cleared systems must include
accuracy, precision, reportable range, and reference intervals in their procedures. For modified, non-FDA-cleared
systems, analytical sensitivity, analytical specificity, and interference must be addressed. Validation studies must
be provided for new methods.23

May 2008

284

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

To address these concerns, CLSI is working on the development of evaluation protocols that will
outline principles for validation and provide laboratories with scientific guidance on the
development of QC procedures for specific testing technologies and environments.61 In addition,
CLIAC members have reinforced the importance of laboratory directors’ understanding of their
responsibilities in implementing equivalent QC.60
While flexibility in methods for QC (both general and equivalent) may be desirable, more
comprehensive information is required than is currently produced by instruments.62
Technological advances have led to test systems that contain internal monitoring systems, but
these newer types of control often monitor only certain elements of the test system.63 For
example, electronic controls may indicate the status of the test systems’ electrical components, but
may not alert the laboratory to trouble with environmental interactions. Similarly, systems may
now include tests that cross specialty areas, making standardized QC impractical.
In some instances, the need to revise QC requirements was accomplished through stakeholder
collaboration. For example, the American Society for Microbiology (ASM) conducted two
surveys to determine the rates of QC failures for 24 microbiology reagents. ASM shared the
resulting data with DHHS and recommended that, for commercial reagents with a 98% or greater
success rate, only new lots need to be tested.64 This resulted in a change to the CLIA QC
requirements for microbiology reagents, as published in the 2003 final rule. A CLSI workshop
held in 2005 discussed potential approaches for future QC, convening representatives from the
laboratory, industry and government. As a result of the workshop, a proposal to revise the CLIA
QC requirements for laboratories, using the CLSI consensus process, was developed by
stakeholders and accepted by CMS.
In the absence of amending CLIA, some ways in which the laboratory community can improve
QC include the following:

ƒ

Select QC procedures commensurate with the quality required for the test and the
precision and accuracy observed for the method.65

ƒ

Encourage manufacturers to include in their instruction explanations of the
components of internal monitoring systems.

ƒ

Encourage manufacturers that want to diverge from traditional QC practices to
demonstrate performance characteristics (improved stability of methods, better QC
technology, etc.) and include such data in their dossiers submitted to FDA.

ƒ

Eliminate confusion on proper QC use.66

ƒ

Collect and share among laboratories data on specific test system performance.

Personnel Qualifications to Meet Technical Requirements of Advanced Testing
Methods
CLIA specifies personnel requirements for experience, education, and training, along with
detailed corresponding responsibilities, that must be met by laboratories performing PPM,
moderate complexity testing, high complexity testing, or any combination of these tests.
Laboratories performing only waived testing do not have specific personnel qualifications.67 A

May 2008

285

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

summary of the CLIA personnel requirements for high complexity testing is provided in Box 7.2.
The categories and requirements for moderate complexity are different.
Box 7.2: Summary of CLIA Personnel Requirements for High Complexity Testing
•

Director. A director in a laboratory performing high-complexity testing has essentially the same responsibilities
as a moderate-complexity testing laboratory director, including the overall operation and administration of the
laboratory. A director must hold a doctoral degree in medicine, osteopathy, or one of the sciences and have
appropriate board certification or one to two years of laboratory training or experience directing or supervising
high complexity testing. Directors are limited to overseeing five laboratories at one time.

•

Technical Supervisor. A technical supervisor is responsible for the scientific and technical supervision of highcomplexity testing in specialties or subspecialties in which they are trained or have experience. Other duties
are similar to those indicated for the technical consultant in laboratories conducting moderate-complexity
testing. The requirements for a technical supervisor vary depending on the specialty or subspecialty in which
the laboratory conducts high-complexity testing. Doctoral-level degrees are required for some subspecialties,
including cytology, histopathology, dermatopathology, ophthalmic pathology, oral pathology,
histocompatibility, clinical cytogenetics, and immunohematology.

•

Clinical Consultant. The clinical consultant provides consultations to the laboratory’s clients in matters
related to reporting and interpreting test results. The qualifications and duties are similar to those specified
for laboratories performing moderate complexity testing.

•

General Supervisor. Unlike moderate testing facilities, high-complexity laboratories also have at least one
general supervisor who provides day-to-day supervision of testing personnel and reporting of test results at the
discretion of the laboratory director and technical supervisor. In order to qualify as a general supervisor, an
individual must have a doctoral, master’s or bachelor’s degree in medicine, osteopathy or one of the sciences
and one year of applicable laboratory training or experience, or have an associate’s degree and two years of
training or experience in high complexity testing. Subspecialties in histopathology require board-certified
pathologists for the role of general supervisor.

•

Testing personnel. Responsibilities of testing personnel include specimen handling and processing, test
analyses, and reporting and maintaining records of patient test results. In order to conduct testing without
direct supervision, testing personnel in high complexity labs are expected to have at minimum the education
and training equivalent to an associate degree in a laboratory science or medical laboratory technology.

Note: Personnel qualifications may vary with test complexity as well as with date and year an individual was hired into a
laboratory position.
Source: Laboratory requirements. Code of Federal Regulations, Title 42, Section 493. Subpart M--personnel for non-waived
testing.

Technologists/scientists and technicians
The matter of determining personnel requirements for clinical laboratories preceded CLIA and
remains controversial today. Two prominent laboratory professional societies and their members,
ASCLS and ASCP, are collaborating to resolve their differing views concerning appropriate
personnel qualifications and nomenclature (e.g., medical technologist vs. clinical laboratory
scientist, medical laboratory technician vs. clinical laboratory technician). (Refer to the Workforce
chapter of this report for a more discussion of the personnel qualifications advocated by these
organizations.)
CLIA prompted some convergence among the organizations on these issues. The CLIA rule
outlined minimum qualifications needed for individuals to work in a laboratory and offered
pathways for an individual to perform high- and moderate-complexity testing. Rather than
recognizing the titles of medical technologist or clinical laboratory scientist as approved testing

May 2008

286

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

personnel, CLIA relied instead on education and training experience of personnel. Phase-ins
permitted some individuals to continue their testing responsibilities until they could achieve
additional education and training, and grandfather clauses allowed individuals with certain
qualifications to continue testing without additional education, training, or experience.68 Testing
personnel responsibilities were outlined based on the complexity level of the tests.69, 70
One concern for the professions was how the regulation would affect the National Labor
Relations Board (NLRB) decision that technologists/scientists are considered professional
employees. Nine amicus briefs were filed with the NLRB by laboratory professional
organizations, including CAP, American Association for Clinical Chemistry, ASCP, and ASCLS,
that highlighted the educational and training experience required for technologists/scientists.
The briefs confirmed that, while automation allows technologists/scientists to work more
efficiently, accurately, and productively, it does not replace the intellectual and analytical nature
of their work. The NLRB agreed and upheld the educational and training requirements.71

Laboratory Directors
The matter of qualifications for laboratory directors prompted a different debate among
laboratory organizations. Since laboratory directors are responsible for communicating
laboratory data to physicians, including the interpretation for patient diagnosis and management,
some believe that only a pathologist be qualified to fill this role, while others find it acceptable to
permit doctoral scientists to oversee the laboratory. In its 2003 final rule, CLIA required that, after
February 28, 2003, individuals with a doctoral degree in the chemical, physical, biological or
clinical laboratory sciences seeking employment as a director of a laboratory performing high
complexity testing must be certified by a DHHS-approved board.72 Physicians that are board
certified in clinical and/or anatomic pathology continue to serve predominantly as laboratory
directors, although some directors are certified by other boards.

Certification and Personnel Requirements
CLIA has provided impetus for change in the laboratory community. Laboratory organizations
offering professional certification use CLIA as a minimum standard, which in some instances has
meant increasing their personnel requirements. Other certifying bodies have used CLIA to build
new career ladder opportunities, noting concern for the placement and retention of skilled
personnel. For example, ASCLS is working toward development of advanced practice scientists,
representing a doctoral degree in clinical laboratory science, to serve in consultant roles and
manage patient laboratory data.73 CLIA and its personnel provisions have prompted
considerations to unite the ASCP Board of Registry and the NCA. In July 2006, the ASCLS Board
of Directors, ASCP Board of Registry-Board of Governors, ASCP Board of Directors, NCA Board
of Directors, and Association of Genetic Technologists Board of Directors reviewed and voted on
a document outlining the rationale for such a union.74
Personnel requirements for performing genetic testing are also at issue. A 2003 study of hospitalbased, independent, and research-based biochemical genetic testing laboratories in the U.S.
examined personnel qualifications using a mail survey of laboratory directors. Using survey
responses, investigators assigned QA scores, which served as the main outcome measure of the
study and were based on the standards defined by the American College of Medical Genetics
Laboratory Practice Committee. The study found that, although all directors had doctoral

May 2008

287

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

degrees, personnel qualifications varied. The mean QA score was 77%, with a range of 28-100%.
Higher scores were associated with: laboratory director having a PhD degree versus MD degree;
director board certification in biochemical genetics; research and hospital laboratory versus
independent laboratory setting; and participation in a PT program. The investigators concluded
that personnel qualifications and laboratory practice standards may need improvement to ensure
quality in clinical biochemical genetic testing laboratories and appropriate use of test results.75

Oversight Mechanisms Employed Through Surveys and Sanctions
Non-waived laboratories must permit biennial inspections to assess their compliance with the
regulations as well as permit non-routine inspections to validate and investigate complaints.
During the survey process, surveyors may require the laboratory to test samples and will observe
testing, interview all levels of personnel, review copies of records, and tour all areas of the
laboratory.76 As noted above, laboratories with CWs are not subject to biennial inspections, but
may be examined at any time to evaluate a complaint or assure compliance with CLIA.
CLIA provides CMS with the discretion to select the appropriate corrective action or sanction for
identified laboratory deficiencies.77 Enforcement mechanisms range from severe monetary
penalties or onsite monitoring to cancelling a laboratory’s approval for obtaining Medicare
payments for its services or suspending or limiting a laboratory’s CLIA certificate. Often,
regulators take an educational approach and work collaboratively with laboratories to correct
problems in lieu of imposing sanctions.
CMS also has implemented the Alternate Quality Assessment Survey (AQAS). The goal of this
self-assessment survey tool was to streamline the CLIA inspection process and reward good
performers. The AQAS form is consistent with the onsite survey process with a focus on QA.
Laboratories with no deficiencies and satisfactory PT performance may use AQAS instead of
having an inspection on their next two-year cycle. Approximately 5% of laboratories using the
AQAS are inspected on-site to validate their self-assessment process.78
The initial CLIA on-site surveys of laboratories in 1992 revealed quality problems in as many as
35% of laboratories; however, by 2004, fewer than 7% of 12,000 laboratories surveyed were
considered to have quality problems. In 2004, CMS reported that it had proposed enforcement
action in 6,084 cases since 1999, and implemented such action in 487 of them.79 However, despite
oversight mechanisms provided through CLIA requirements, laboratory quality problems in
several states have raised questions about the adequacy of laboratory oversight.36 For example, in
2003, the laboratory at Maryland General Hospital was among those identified with serious
quality and management problems. Accredited by CAP, the laboratory was the subject of several
inspections, arising from State inspections and complaints. A complaint by a laboratory
employee alleged that the equipment used for HIV and hepatitis testing was not adequately
maintained and that erroneous test results were possible. Oversight, management, and
communication concerns in this case triggered congressional interest and a GAO investigative
report on the status of CMS and survey organization oversight of clinical laboratories.79, 80
In 2003, CMS regional offices followed suit and initiated a program to review and monitor state
agency surveyors against 13 performance standards, including the training and qualifications of
surveyors. However, the extent of serious quality problems at laboratories remained unclear
because of incomplete data on condition-level deficiencies identified by state survey agencies
May 2008

288

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

prior to 2004 and the lack of direct linkage between CLIA requirements and CLIA-equivalent
requirements of some survey organizations.36
The GAO’s 2006 report to Congress concluded that oversight of clinical laboratory quality is
inadequate to ensure that laboratories are meeting CLIA requirements, noting weaknesses in survey
methods, complaint processes, and enforcement.36 GAO stated that, aside from PT scores, there is
lack of standardized means for measuring and reporting laboratory quality. Among its concerns,
GAO suggested that the announcement of forthcoming inspections up to 12 weeks in advance may
result in an unrealistic assessment of laboratory performance. The report observed that variability
in reported survey deficiencies suggests that laboratories are not surveyed consistently. GAO also
observed that CMS and survey organizations appear to stress education over regulation in the
implementation of the 2003 QC requirements and PT for gynecologic cytology. Gaps in complaint
processes compromise whistle-blower protection to those filing complaints about quality problems.
GAO perceived that sanctions are not being used effectively as an enforcement tool to promote
compliance even in laboratories with serious, consecutive, condition-level deficiencies. CLIA
regulations require PT three times per year on analytes listed in Subpart I rather than the statutorily
originally defined rate of four times per year. The report found that the length of time (2-4 years)
allowed for implementation of new requirements is unnecessarily excessive and compromises
quality. GAO noted that CMS was late in ensuring CLIA equivalence of exempt states’ and
accrediting organization inspection requirements, and that many of CMS’ validation reviews lack
independence and reviews skip some state survey agencies.
To address these issues, the GAO report provided 13 recommendations (see Box 7.3). CMS, CAP,
COLA, and the Joint Commission were provided with an opportunity to comment on the report.
CMS agreed with 11 of the 13 recommendations, the exceptions being the frequency of PT
assessments and the extent of simultaneous accrediting organization validation reviews.36 CMS
also provided an alternative assessment of laboratory quality, disagreed with the GAO conclusion
about the lengthy education phase-in for new CLIA requirements, and expressed concern about
identifying and sanctioning laboratories with repeat condition-level deficiencies. CMS restated its
intention to retain an educational approach to CLIA compliance, commitment to cite deficiencies
appropriately when found, and provide additional training to state agency surveyors. CAP,
COLA, and the Joint Commission agreed with some of the findings of the report, but disagreed
with others.36 Since the report’s publication, CAP has implemented mandatory training for its
surveyor team leaders, beginning on July 1, 2006.81 In response to GAO’s recommendations, CMS
has undertaken and completed certain actions and efforts intended to augment the CLIA program
and strengthen oversight.
In 2006, CMS issued a guidance document, Partners in Laboratory Oversight, which identifies
elements of an effective survey process that partner organizations are to incorporate into their
respective survey protocols and suggests communication protocols for information sharing
among partners. The guidance applies to CLIA activities among the CMS central and regional
offices, state agencies (including those with licensure requirements), accreditation organizations,
and CLIA-exempt states.82

May 2008

289

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

Box 7.3: Summary of GAO Recommendations to
Improve Regulatory Oversight of Clinical Laboratories
Recommendation 1: To enable CMS to track the nature and extent of lab quality problems across survey
organizations, the CMS Administrator should:
•

Work with exempt-state programs and accrediting organizations to standardize their categorization and
reporting of survey findings in a way that tracks to CLIA inspection requirements and allows for meaningful
comparisons across organizations, such as the analysis of trends in the citation of condition-level deficiencies.

Recommendations 2-5: To ensure consistency in the oversight of labs by survey organizations, the CMS
Administrator should:
•

Ensure that the advance notice of upcoming surveys provided to physician office labs is consistent with
CMS's policy for advance notice provided by state survey agencies.

•

Ensure that regulation of labs is the primary goal of survey organizations and that education to improve lab
quality does not preclude the identification and reporting of deficiencies that affect lab testing quality.

•

Impose appropriate sanctions on labs with consecutive condition-level deficiencies in the same
requirements.

•

Require all survey organizations to develop, and require labs to prominently display, posters instructing lab
workers on how to file anonymous complaints.

Recommendations 6-13: To improve oversight of labs and survey organizations, the CMS Administrator should:
•

Consistent with CLIA, require quarterly proficiency testing, except when technical and scientific
considerations suggest that less frequent testing is appropriate for particular examinations or procedures.

•

Ensure that evaluations of exempt-state and accrediting organization inspection requirements take place
prior to expiration of the period for which they are approved in order to ensure the continued equivalency
of their requirements with CLIA's.

•

Ensure that changes to the inspection requirements of exempt states and accrediting organizations be
reviewed prior to implementation, as required by regulation, to ensure that individual changes do not
affect the overall CLIA equivalency of each organization.

•

Allow the CLIA program to utilize revenues generated by the program to hire sufficient staff to fulfill its
statutory responsibilities.

•

Ensure that federal surveyors validate a sufficient number of inspections conducted by each state survey
agency to allow a reasonable estimate of their performance, including a minimum of one independent
validation review for each state survey agency surveyor.

•

Require that almost all validation reviews of each accrediting organization’s surveys be an independent
assessment of performance.

•

Collect and routinely review standardized survey findings and other available information for all survey
organizations to help ensure that CLIA requirements are being enforced and to monitor the performance of
each organization.

•

Establish an enforcement database to monitor actions taken by state survey agencies and regional offices
on labs that lose their accreditation.

Source: Clinical Lab Quality: CMS and Survey Organization Oversight Should Be Strengthened. Government Accountability Office.
June 2006.

May 2008

290

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

GENETIC TESTING
Genetic testing is becoming an increasingly important component of health care delivery. In
federal and state legislation, a genetic test is considered a DNA-based test, though the term is also
used to refer to tests of gene products (proteins and metabolites), chromosomes, and acquired
somatic cell mutations.83 However, no single definition of a genetic test is universally accepted by
all stakeholders.
Genetic tests can be used to “diagnose existing disease, to predict future risk of disease, to identify
carriers of mutations that might cause disease in one’s offspring, or to identify particular traits in a
fetus or embryo such as gender or human leukocyte antigen type.”84 At present, genetic tests for
at least 1,430 diseases are available for clinical use, and the number and availability of new tests
continues to rise.85 Given the life-altering influence that genetic test results can have, it is
imperative that they be subject to adequate regulatory oversight.
PGx testing is a relatively new form of genetic testing. To date, only a few PGx tests are used to
support decisions for selecting and dosing therapies.86 Some early applications of PGx include
HER2/neu testing to guide use of trastuzumab (Herceptin) for metastatic breast cancer, and
thiopurine methyltransferase genotyping to manage the use of thiopurine drugs to treat acute
lymphoblastic leukemia in children. Other PGx tests include CYP2C9 and VKORC1 testing to
manage the use of warfarin for those at risk of harmful blood clots, the Roche Amplichip for
CYP450 mutations, and the UGT1A1 test for irinotecan (Camptosar) sensitivity.
Currently, there are few laws and regulations that specifically address the complexity of genetic
testing. At the federal level, oversight authority is provided through application of CLIA, FDA, and
the Federal Policy for the Protection of Human Subjects. Specifically, while FDA has the regulatory
authority for all tests, genetic tests developed as in-house (i.e., LDTs) are currently subject only to
CLIA requirements, while tests developed by manufactures are subject to FDA requirements. In
addition, some state legislatures have introduced laws and regulations pertaining specifically to
genetic testing. Reports by SACGHS and others have concluded that serious gaps in the regulatory
framework for genetic testing could compromise patient and public health.83

OUTSTANDING ISSUES IN GENETIC TESTING
Regulatory Oversight of Laboratory-Developed Genetic Testing
For the specialty of cytogenetics,u CLIA provides specific requirements to address QC, PT, QA,
analytical validity, and personnel.83 However, the broader scope of genetic testing is subject only
to CLIA’s general requirements for non-waived testing. Public and private sector stakeholders
are working together to address outstanding concerns about the oversight of laboratorydeveloped genetic tests and personnel requirements of those performing genetic tests.
The CLIA Quality Systems final rule, published in 2003, restructured and updated certain
requirements that are relevant to genetic testing, such as facility requirements for unidirectional
workflow for molecular amplification procedures (§493.1101), quality system requirements for
u

Cytogenetics is the examination of chromosomes and the conditions caused by numerical and structural
chromosomal abnormalities.

May 2008

291

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

confidentiality of patient information (§493.1231), and QC procedures for PCR (493.1256).87, 88
These latest requirements do not include tailored QC, personnel, or PT requirements for
molecular genetic, biochemical genetic, or PGx testing. Notwithstanding that genetic tests are
high complexity tests, most genetic testing laboratories are not required by CLIA to perform
formal PT, unless they are offering any one of the 83 regulated analytes. According to CLIA
regulations, alternative performance assessment must be conducted for all other tests.
Consistent with earlier support by a National Institutes of Health-Department of Energy task
force and CLIAC in 2000, SACGHS recommended that CMS develop regulations for genetic
testing and that FDA review all genetic tests. In September 2006, CMS announced that it would
not pursue a Notice of Proposed Rule Making for genetic testing. In August 2007, in response to a
petition from the Genetics and Public Policy Center, CMS stated that supportive evidence does
not justify rulemaking to establish a new genetics specialty under CLIA. Included in its rationale,
CMS noted:
“Various tests that we would all regard as ‘genetic tests’ are in actuality dispersed throughout
different operational sections of the laboratory and many are found in different existing CLIA
specialties. Creation of a new genetic testing specialty would require not only greater precision
in the current definitions, but would also require a teasing out of certain tests from some
existing specialties, and cause some disruption to existing regulatory and payment structures.”
Instead, CMS stated that it would implement an action plan for enhanced oversight of genetic tests
under the existing CLIA authority.89 CMS is working with SACGHS, FDA, and other experts to
address current gaps in oversight of laboratories that conduct genetic testing and to otherwise
support or augment its action plan.90 CMS has begun to implement its action plan, which includes:

ƒ

Training of surveyors in how to assess genetic testing laboratories’ compliance with
regulatory obligations

ƒ

Collaboration with CDC to publish educational materials for genetic testing
laboratories (e.g., Morbidity and Mortality Weekly Report Recommendations and Reports)

ƒ

Development of alternative PT mechanisms for genetic testing laboratories, such as
inter-laboratory comparisons

ƒ

Collaboration with CDC and FDA on ongoing oversight activities91, 92

CLIAC acknowledged CMS’ decision not to proceed with the rulemaking, but cited the need to
examine the regulatory framework further in order to achieve enhanced oversight of genetic
testing.92
Some accreditation organizations also are addressing genetic issues related to laboratorydeveloped genetic tests. In 2007, CAP began an Internet-based registry service intended to
connect genetic testing laboratories that perform low-volume genetic tests. When three
laboratories are identified as testing for the same genetic disorder, CAP will facilitate an exchange
of specimens for alternative assessment.

May 2008

292

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

Enhancement of FDA’s role in regulating laboratory-developed genetic testing
FDA has statutory authority to fully regulate all LDTs but has not done so because of resource
constraints.3 With genetic tests of increasing complexity continuing to be marketed, often
unaccompanied by direct evidence of clinical utility, there is a need to clarify or extend regulatory
oversight of these tests.84
In September 2006, FDA issued two draft guidance documents to address regulatory oversight of
more complex laboratory-developed test devices that are used in genetic testing. One guidance
document pertains to the marketing of in vitro diagnostic multivariate index assays (IVDMIAs)
and the other pertains to the marketing of analyte specific reagents (ASRs), which are the active
ingredients used by clinical laboratories in developing LDTs.

In Vitro Diagnostic Multivariate Assays
Certain complex genetic and proteomic tests are IVDMIA test systems―tests that employ data
from one or more in vitro assays, in some cases demographic data, and an algorithm that usually,
but not always, runs on software to generate results for diagnosis and treatment. A great concern
with these test systems is that the results cannot be independently derived and confirmed by
another laboratory without access to proprietary information used in the development of the test.
In addition, the results cannot be interpreted by well-trained health care practitioners without
information from the developer of the test regarding its clinical performance and effectiveness.
According to the 2006 draft guidance, FDA proposed to actively regulate these test systems as
medical devices and classify them according to their intended use and level of control necessary
to assure their safety and effectiveness, including requiring premarket review as class II or III
devices, where applicable.94
After receiving comments on the initial draft guidance for IVDMIAs, FDA issued revised draft
guidance for public comment in July 2007. The revisions clarified the definition of an IVDMIA
and provided examples of tests that the agency does and does not recognize as IVDMIAs. The
agency noted that these clarifications did not alter the scope or intent of the definition of an
IVDMIA that appeared in the initial draft guidance document.95
FDA approved the first IVDMIA PGx test system, MammaPrint, in February 2007. Marketed in The
Netherlands since 2005, MammaPrint is a gene expression profiling test for predicting whether an
existing cancer will metastasize in women with early stage breast cancer.96 As genetic testing and
PGx, in particular, evolve, there will be ongoing need for guidance from CMS and FDA.

Analyte Specific Reagents
FDA regulates certain components of genetic tests that are developed and performed by
laboratories, but that are not marketed as test kits. ASRs include antibodies, receptor proteins,
nucleic acid sequences, and other biological or chemical reagents which, through specific
binding or chemical reactions with substances in a specimen, are used to identify or quantify an
individual chemical substance or ligand in biological specimens. Among other regulatory
requirements, ASR manufacturers must list with FDA, follow quality system regulation, and
restrict the sale of these reagents to high-complexity laboratories. FDA does not regulate how
ASRs are used to create a new test. The great majority of genetic tests performed by
laboratories are based on FDA-approved ASRs. FDA issued draft guidance in September 2006,
May 2008

293

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

made final in September 2007, which clarifies the definition of ASRs and related regulatory
requirements. Specifically, a single ASR that is (1) combined, or promoted for use, with another
product such as other ASRs, general purpose reagents, controls, laboratory equipment,
software, etc., or (2) promoted with specific analytical or clinical performance claims,
instructions for use in a particular test, or instructions for validation of a particular test using
the ASR, is considered by FDA to be a test system and, thus, is not exempt from premarket
notification requirements.97 The guidance addresses industry efforts to market increasingly
complex combinations of ASR-based products (which might be considered test kits subject to
premarket FDA review, rather than analytes) under the less demanding requirements of single
ASRs. Indeed, there has been an increase in LDTs for simultaneous detection of multiple
genetic variants. A related concern involves claims of multiple functions for a single ASR when
selling it to a laboratory.98, 99

Regulatory Oversight of Direct Access Testing for Genetic Tests
Some tests are being sold directly to consumers via Internet web sites and retail stores without the
involvement of a health care provider in ordering the test or interpreting the results. The
popularity of direct access testing is likely to increase given the rapid pace of genetic research
availability of services via the Internet, and the growing interest of consumers in self-care.100
When genetic tests are advertised and sold over the Internet or directly to consumers, significant
clinical information may be missing or misleading, such as the clinical validity and utility of the
test.3 For example, advertisements may de-emphasize the uncertainty of genetic test results,
exaggerate the influence of a particular genetic polymorphism on health (e.g., likelihood of
acquiring diabetes, heart disease, breast cancer, obesity), and exaggerate the positive influence the
test can have on an individual’s health. Reports of misleading information related to direct access
genetic testing were published in a July 2006 GAO report.101 In that same month, the Federal
Trade Commission issued a consumer alert on at-home genetic tests.
Consumers often lack the requisite knowledge to make informed decisions about whether to get
genetic tests or how to interpret test results. Without the aid of a health care provider who can
explain the advantages and disadvantages of being tested and implications of the results, and
make sound recommendations on next steps, there is considerable potential for physical and
emotional harm.
The wide variability in policies for direct access genetic testing among laboratories adds to the
concerns. Although some laboratories require patients to provide the name of a physician to
whom they may send the test results, others provide test results directly to the consumer.
Similarly, although some laboratories have readily accessible genetic counselors to provide
information and answer any questions that consumers may have about testing or their results,
some laboratories do not.
Stakeholders have proposed ways to regulate advertising of and limit access to genetic testing.
Careful regulation of advertisements could minimize misleading or exaggerated claims made by
test manufacturers and providers, as well as limit the channels through which these
advertisements are introduced. Regulating access to such tests would make it more difficult for
consumers to obtain tests or results without authorization from a health care provider. This

May 2008

294

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

measure would help prevent consumers from misinterpreting tests due to a lack of knowledge.
Even so, efforts to impose more stringent restrictions on direct access for genetic testing may be
challenged by consumers’ desire for greater autonomy over their health care, including direct
access to services and control of personal health information.3

Contrasting the FDA and CLIA Routes
FDA requirements for the 510(k) notification and PMA review processes for tests as medical
devices and CLIA requirements for LDTs serve different purposes and rely on different data. In
general, FDA emphasizes safety and efficacy of testing devices and CLIA emphasizes a quality
testing process.
FDA has an agency-level responsibility for oversight via the 510(k) or PMA processes.102, 103 This
oversight involves considerations of analytical validity and clinical validity for tests to establish
their safety and efficacy, particularly those subject to premarket review via the PMA route.
However, resources are inadequate for review of analytical validity and clinical validity for many
tests, and there is little or no agency oversight of clinical utility for nearly all tests. As noted above,
due to resource constraints, FDA has not exercised its statutory authority to regulate all LDTs.
Among other aspects of quality described elsewhere in this report, laboratories must
demonstrate analytical validity of tests through an initial inspection and subsequent biennial on
site inspections. However, as standards of evidence for these inspections are determined by the
individual laboratories, not CLIA, the quality of assessments of analytical validity for given
tests can vary across laboratories. CLIA specifies (Subpart M—Personnel for Nonwaived
Testing) that the laboratory director must “Ensure that … [t]he test methodologies selected have
the capability of providing the quality of results required for patient care.”67 As such, CLIA
vests considerable oversight responsibility in the individual laboratory directors. When a
testing service is offered by a laboratory, its director is responsible for having determined that
the testing service, as ordered by clinicians, is capable of providing the quality of results
required for patient care. Further, the laboratory director is responsible for ensuring that the
test is performed and reported properly and is subject to appropriate QC. LDTs can only be
performed by the laboratories that develop those tests. In contrast to the national scope of FDA
oversight, this is more decentralized, testing service-specific form of oversight.
Tests that are cleared or approved for market by FDA are available for use by any laboratories, and,
like LDTs, are subject to PT and other CLIA oversight provisions. Most tests that are performed by
laboratories are either cleared or approved by FDA or are LDTs. Some LDTs are based on ASRs
that are regulated as medical devices by FDA. (As described above, recent FDA guidance has
clarified that ASRs marketed in combination or alone, but with specific indications are, by
definition, IVDs and subject to FDA regulatory oversight, accordingly.)97 For the remaining LDTs
that are not derived from these FDA-regulated products, the laboratory directors are still
responsible for ensuring capability of providing the quality of results required for patient care.
Certainly, many common LDTs that were not subject to FDA oversight have become standards of
care over decades and have demonstrated, in practice, clinical validity and clinical utility.
Notwithstanding the means and extents of their respective oversight of analytical validity,
clinical validity, and clinical utility, both the FDA and CLIA provisions are incomplete in their
oversight of these attributes of tests. Whether for FDA-cleared or approved tests or LDTs, the
May 2008

295

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

national capacity for establishing clinical validity and clinical utility, in particular, in field
testing remains limited.
Market factors influence decisions to develop tests and can influence the regulatory route
pursued for marketing a test or testing service. The likelihood of a manufacturer developing a
test and collecting data sufficient to gain FDA clearance or approval is influenced by the size of
the potential commercial market for that test. Pursuing an FDA pathway for a low-volume test
may be economically prohibitive. Manufacturers may not find it commercially viable to pursue
development and FDA review of tests for certain rare inheritable conditions or other “orphan”
conditions affecting very small populations. Given the lower hurdles for reaching the market,
laboratories may pursue such tests as LDTs and offer them as specialty or “niche” testing
services. Manufacturers that choose to devote the necessary resources to generate data for
gaining FDA approval to market a test may encounter competition from laboratories that
develop and perform their own in-house version of the test. Compared to CLIA-regulated
genetic testing, FDA-regulated tests generally are subject to greater scrutiny for clinical validity
and, in some instances, clinical utility.
The current regulatory framework poses incentives for offering genetic tests as LDTs rather than
marketing them via the 510(k) or PMA routes associated with FDA-regulated tests. While
compliance with CLIA provides certain assurances of quality, launch of a genetic test (including,
e.g., a PGx test) in the form of an LDT offers the advantage of more rapid access to market than
launch of the test in the form of an IVD test kit or system that must be approved by FDA.
Furthermore, even when a manufacturer has gained FDA approval for a genetic test, laboratories
can develop their own version of the test and add it to their menu of CLIA-only regulated
laboratory services. As noted above, depending on the benefit-risk tradeoffs of particular genetic
tests to patients and clinicians, these tests may be more or less suitable for premarket clearance or
approval by FDA.

TRANSFUSION-RELATED SERVICES
Transfusion-related services are critically important to the health and survival of millions of patients
each year. More than 15 million units of blood were collected and more than 14 million units of
blood were transfused in the U.S. in 2004, the most recent year for which data are available.104
Since the early 1980s, the concept of blood banking has changed from being a laboratory
discipline to the clinical and consultation specialty of transfusion medicine.105 Concerns about
transfusion-transmitted diseases contributed to this change, along with recognition of the success
of transfusion-related laboratory services in addressing problems such as antibody identification
and record keeping.
Protection of the blood supply is a multi-step process. Following collection, donated blood is
tested in a clinical laboratory to detect the presence of pathogens and contaminants such as
hepatitis B and C viruses, HIV, human T-lymphotropic virus,v and syphilis.108 If no pathogens or

v

Human T-lymphotropic virus can cause infections that can lead to leukemia or to a variety of neurological
diseases.107

May 2008

296

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

contaminants are found and the specimen is approved, the blood undergoes further testing for
ABO blood type, Rh factor, and unexpected red blood cell antibodies.
Clinical laboratories of large blood banking organizations are devoted exclusively to testing
donated blood. The American Red Cross, which collects approximately 43% of all blood donated
in the U.S., maintains five national testing laboratories and 32 blood manufacturing facilities.109,w
The laboratories test all blood collected by the American Red Cross, as well as some collected by
other organizations. Laboratories providing transfusion-related services are subject to both CLIA
and FDA oversight, and, as such must be certified by CMS and registered with FDA.

CLIA Requirements
CLIA regulations apply to the blood testing processes related to transfusion services. These
laboratories must be certified and in compliance with the requirements for personnel, QC, QA,
recordkeeping, and PT associated with performing high complexity testing and, when applicable,
the specialty of immunohematology. For laboratories that are part of transfusion services but not
involved in processing blood components, CLIA and FDA have a memorandum of
understanding under which CMS assumes responsibility for regulatory oversight.110
CMS or CMS-approved accrediting agencies can assess compliance with CLIA regulations as well
as with their own standards. Most transfusion service centers are accredited and surveyed by
AABB. AABB currently accredits 221 laboratories and almost 1,600 organizations.111 CAP inspects
laboratories using its transfusion medicine checklist, providing on-site inspections on a biennial
basis with facilities completing interim-self assessments during alternate years.112 The Foundation
for the Accreditation of Cellular Therapy runs a voluntary standard setting, inspection, and
accreditation program for facilities involved in cellular therapy and cord blood banking.113 The Joint
Commission also assesses transfusion services and donor centers within hospitals.114
AABB is not a CMS-approved PT provider, but administers PT for laboratories seeking
accreditation as an immunohematology reference laboratory.111 CAP offers two PT programsx for
blood testing laboratories, one for laboratories involved in transfusion medicine (J-survey) and
one for automated transfusion medicine laboratories (JAT-survey), in which more than 4,500
laboratories currently participate.116 CLIA’s PT requirements for laboratories performing
transfusion related testing is more stringent than for most other specialties. A score of 100% is
required for a satisfactory performance for ABO grouping, Rh D typing, and cross matching and
80% for antibody detection and antibody identification. (Eighty percent is a satisfactory PT score
for most other specialties.)

FDA Requirements
FDA seeks to ensure that blood and blood products are safe for transfusion. FDA regulations
apply to the components of blood product processing, including blood collection and testing, and

The American Red Cross’ national testing laboratories are located in Charlotte, NC; Portland, OR; Detroit, MI; St.
Louis, MO; and Philadelphia, PA.109
x The J-survey is mailed to subscribing participants three times each year and tests participants on determination of
ABO group, Rh D type, antibody detection and identification, and cross-matching.115
w

May 2008

297

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

mandate that blood testing laboratories implement QA and QC functions. Laboratories
performing tests on donated blood must use only FDA-approved tests.117
Blood is regulated as a drug, as defined in section 201(g) of the FDCA, and a biological product, as
defined in section 351(a) of the Public Health Service Act.118 FDA’s Center for Biologics
Evaluation and Research holds primary statutory authority to regulate and set standards for all
aspects of blood banking, including collection procedures, blood components used for transfusion
or manufacturing of pharmaceutical products (e.g., clotting factors), and products that ensure the
safety of the blood supply (e.g., cell separation devices, blood collection containers, and HIV
screening tests).119, 120 The Center for Biologics Evaluation and Research also has authority to
conduct regular unannounced inspections of blood banking centers and investigate reports of
blood-related errors, accidents, and adverse clinical events.
Title 21 of the Code of Federal Regulations (CFR) outlines the specific provisions to which
laboratories and manufacturing facilities must comply, e.g., licensing requirements, QA
procedures, product standards, and current good manufacturing practices. Each laboratory must
designate an individual who is responsible for ensuring compliance with regulations by all
personnel and who will serve as the facility’s representative to FDA.120 Each facility’s QA
operational framework must provide designated individual(s) with the authority to report serious
infractions directly to FDA and, if applicable, stop production immediately.121
At a minimum, FDA conducts biennial inspections of blood testing laboratories and
manufacturing facilities, usually unannounced, to assess compliance with the CFR. More
frequent inspections may be undertaken if compliance problems are identified. Inspections are
based on a multi-layered set of safeguards highlighted in Box 7.4.117 Advisory, administrative,
and/or judicial enforcement mechanismsy are available to FDA in the event that inspection
findings demonstrate operational deficiencies and/or management is unwilling or unable to
implement corrective actions in a timely fashion.
Box 7.4: Safeguards for Inspection of Blood Banking Laboratories
•

Donor screening: donors are informed about potential risks and are required to answer questions about
factors that may influence the safety of their blood

•

Blood testing: each unit of donated blood is tested for infectious diseases

•

Donor lists: blood establishments must keep a current list of deferred donors and must ensure that blood
is not collected from anyone on that list

•

Quarantine: donated blood is quarantined until it is tested and proven to be free of infectious agents

•

Problems and deficiencies: blood centers must investigate manufacturing problems and correct all
deficiencies; they must also notify FDA when product deviations occur in any distributed products107

Source: Keeping blood transfusions safe: FDA's multi-layered protections for donated blood. Rockville: Food and Drug
Administration, 2002.

Industry stakeholders perceive several regulatory constraints in the area of blood banking. Many
of these issues are related to manufacturing (e.g., cellular therapies, plasma preparation) and
tissue storage and are, therefore, outside the scope of this report. Those issues related directly
y

Mechanisms include warning letters, license revocation or suspension, seizure of products, injunction and prosecution.

May 2008

298

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

with laboratory testing (e.g., recruitment of donors, health history questionnaire and emerging
threats to blood supply) are discussed below.

OUTSTANDING ISSUES FOR TRANSFUSION-RELATED SERVICES
Simplification of Donor History Questionnaire
Physical assessment and a medical history interview are tools used to determine a donor’s
eligibility to donate blood and blood components.122 Since the first formal uniform donor
screening questionnaire was developed in 1953, the number of questions and the amount of
information that it captures as a part of donor screening has increased, reflecting advances in
identifying risks of disease transmission.
In 2000, AABB organized a multi-organizational task force to address concerns related to the
donor assessment process with the goal of reducing the complexity and improving
comprehension of the questionnaire and supplementary education materials. The task force
produced the Uniform Donor History Questionnaire (DHQ), a new set of documents that
includes a full-length questionnaire (of 17 specific questions), user brochure, medication deferral
list, and blood donor educational materials. While FDA does not mandate the use of a particular
screening tool for donors to assess risk of communicable disease, it does recognize the DHQ as an
effective donor screening tool through which licensed and unlicensed facilities can meet
regulatory obligations.123
An abbreviated DHQ (aDHQ) was developed as an alternative for a specific subset of repeat
donors.124 The aDHQ originally proposed by the AABB task force consolidated the 17 questions
about medications and medical events on the full-length DHQ into one medical history capture
question that asked donors whether they had any new medical problems, diagnoses, or treatments,
including vaccination, since their last donation.124 However, FDA expressed concern about the
ability of one question to capture important information. The aDHQ was revised to include two
medical questions that ask whether donors have had any new medical problems or diagnoses and
whether they have had any new medical treatments since their last donation. However, unlike the
full-length DHQ, FDA has not officially recognized the aDHQ.125,z Additional work is needed to
improve the ability of abbreviated forms to account for the medical history of donors.

Mechanisms to Handle Emerging Threats to Blood Supply
FDA’s ability to handle emerging threats to the blood supply, such as new HIV variants, new
hepatitis agents, Creutzfeldt-Jakob Disease, bacterial contamination of blood products, and others,
has significant impact on the public health.127 In many cases, blood collection organizations
choose to implement specific screening tests prior to FDA issuing guidance. For example, FDA
does not currently require blood collection agencies to test for West Nile Virus or Chagas’ disease,
two diseases that are increasingly prevalent in the U.S. However, the American Red Cross
currently tests all of its blood for contamination with both of these diseases. This decision was
based primarily on the recognition that blood collected by the organization in one part of the U.S
may be shipped to another area for transfusion, rendering the geographic clustering of the
z

Licensed blood establishments that plan to implement a version of the DHQ materials that have not been officially
FDA-recognized must submit a formal request to FDA for approval.126

May 2008

299

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

diseases less meaningful. According to some reports, approximately 70% of all blood centers in
the U.S. are testing blood for Chagas’ disease.128
A 2006 IOM report on the future of drug safety provided recommendations to FDA related to
protecting and improving the safety of the blood supply.129 In response to IOM’s
recommendation that FDA strengthen the scientific base that supports their medical product
safety team, and as a result of FDA’s self assessments, FDA began working with CDC in 2006 to
identify new threats to the blood supply and to develop, evaluate, and deploy modern
technologies to address them.130

CONCLUSIONS
CLIA 1988 has served as the primary regulatory program governing the U.S. laboratory system
since its implementation in 1992. The final rule addressing quality systems and certain personnel
qualifications was published in 2003. The CLIA program is administered as a tri-agency effort
that involves CMS, which has primary oversight of the program, CDC and FDA.
The current CLIA regulatory framework for clinical laboratory testing, including its goal to ensure
high standards of quality, promote access, and improve patient outcomes, is limited in certain
important ways by its language and by the time frame required for development and amending
relevant regulation. Rapid technological advances, demographic shifts, lower tolerance for error,
and higher expectations for personal data security are challenging and, perhaps, outstripping
certain aspects of the current regulatory framework for clinical laboratories. Population and
epidemiological trends are increasing the incidence and prevalence of chronic diseases; broader
screening benefits and coverage of prescription drugs for Medicare beneficiaries are contributing
to the demand for rapid and patient accessible laboratory testing for screening, diagnosis, and
therapeutic monitoring.

ƒ

CLIA’s interpretative guidelines that allow laboratories the flexibility to determine control
procedures that are equivalent to the traditional frequency of two levels of control per day
lack a framework for implementing QC across the wide variety of test systems. While
flexibility in implementing QC is desirable, additional information is required to
implement the guidelines effectively.
•

ƒ

May 2008

Several factors may contribute to the inconsistencies in implementing
equivalent QC. These include the lack of adequate risk management
information from manufacturers, the different types and levels of QC required
by each device and method, and the unique considerations of individual
laboratories.60 CMS, CLSI, and other stakeholders are developing evaluation
protocols that will outline principles for validation and provide laboratories
with scientific guidance on the development of QC procedures for specific
testing technologies and environments.61

Available evidence on the long-term impact of PT on laboratory performance is limited,
and findings of existing studies are confounded by limited comparable data from CMS
and survey organizations and other methodological shortcomings. Existing studies
indicate generally improved performance in recent years.

300

Laboratory Medicine: A National Status Report

ƒ

•

Laboratories with PT performance scores of 80% or higher for most specialties
and subspecialties are considered successful. As such, PT statistics suggesting
satisfactory laboratory performance may fail to detect significant proficiency
problems in particular areas. Similarly, the PT results do not necessarily
indicate that a laboratory’s proficiency is sufficient to meet the needs of the
clinician. CMS would increase transparency of PT if it were to consider new
ways to code certain tests for PT purposes, including disaggregation of tests
within specialty areas, such as microbiology.

•

Several efforts are underway to improve the quality of cytology PT programs.
CLIAC and CMS are emphasizing the importance of rigorous review and field
validations studies of cytology slides used by PT programs. CLIAC
recommended changes to the regulations in several areas, such as use of new
technology (virtual media), testing frequency, number of challenges, and a
grading scheme.51 Legislation (H.R. 1237) also has been reintroduced.

Consistent with GAO recommendations, CLIA oversight is being improved by
standardizing reporting of survey deficiencies and strengthened enforcement of
regulatory obligations.
•

ƒ

The large proportion of clinical laboratories that are certified have wide access to
tests in this category. Tradeoffs include easier access to recommended testing,
with benefits for individual and population health, versus the potential for
waived tests to fall short on specimen adequacy, test reliability and accuracy.

Traditional oversight of LDTs under CLIA for analytical validity may be insufficient for
genetic tests for which clinicians and patients seek assurances of efficacy and protections
against potential harms. CLIA requirements for LDTs and FDA requirements for the
510(k) and PMA review processes serve different purposes and rely on different data. In
general, CLIA emphasizes testing accuracy (i.e., analytical validity, not clinical validity)
and FDA emphasizes safety and efficacy (clinical validity and, in some instances, clinical
utility). Most genetic tests are LDTs and therefore are subject solely to CLIA regulations.
Only a small number of genetic tests are regulated as IVD test kits or systems subject to
premarket review by FDA for safety and efficacy via the 510(k) or PMA processes.
•

May 2008

Many laboratories have more stringent quality management programs than are
required by CLIA. These programs provide opportunities for laboratories to
adapt and validate quality improvement interventions or aspects of these
programs. In the short term, these may be incorporated into improved voluntary
guidelines and perhaps eventually into an improved regulatory framework.

Technological advances have made laboratory tests easier to use and less subject to user
error, resulting in considerable growth in the number of waived tests since the advent of
CLIA. From the nine tests or examinations waived in 1993, approximately 1,600 waived
test systems that measure 76 analytes are now waived under CLIA.
•

ƒ

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

The extent to which FDA has actively regulated certain LDTs is changing.
Guidance documents issued by FDA in 2006 and 2007 pertaining to ASRs and
IVDMIAs indicate a noteworthy shift of regulatory oversight for a small, yet
growing number of complex tests. The guidance is likely to expose these tests to
increased scrutiny similar to premarket review via the 510(k) or PMA processes.
301

Laboratory Medicine: A National Status Report

ƒ

May 2008

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

Although they account for a small proportion of all tests performed by clinical laboratories,
more than 1,000 genetic tests are available for clinical use, with hundreds of others used in
research settings. PGx testing is a relatively new form of genetic testing; to date, only a few
such tests are used to support decisions for selecting and dosing therapies.
•

CMS announced in September 2006 that it would not pursue a Notice of
Proposed Rule Making for genetic testing, but that it would implement an
action plan for enhanced oversight of genetic testing under existing CLIA
authority. Elements of the action plan include expanded training of surveyors,
collaboration with CDC to publish educational materials, development of
alternative PT mechanisms, and coordination with CDC and FDA on oversight
activities. CMS also is working with SACGHS, FDA, and other experts to
address current gaps in oversight of laboratories that conduct genetic testing
and to otherwise support or augment its action plan.

•

As genetic testing and PGx, in particular, evolve, there is an ongoing need for
guidance from CMS and FDA regarding respective scopes of regulatory
oversight pertaining to these tests. Manufacturers of tests and therapies whose
safety and effectiveness are mediated by genetic factors will seek ongoing
guidance from FDA for co-development of drugs and diagnostics and related
regulatory matters pertaining to PGx.

302

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

REFERENCE LIST
1. Brennan T, Berwick D. New rules, regulation, markets, and the quality of American health
care. San Francisco, CA: Jossey-Bass Publishers, 1996.
2. Challenge and opportunity on the critical path to medical products. Rockville, MD: Food
and Drug Administration, 2004.
http://www.fda.gov/oc/initiatives/criticalpath/whitepaper.html.
3. Oversight of genetic tests. Bethesda, MD: Secretary's Advisory Committee on Genetics,
Health, and Society, 2007.
4. Institute of Medicine. Crossing the quality chasm: a new health system for the 21st
century. Washington, DC: National Academy Press, 2001.
5. Program descriptions/projects. Baltimore, MD: Centers for Medicare and Medicaid
Services, 2008. (Accessed March 26, 2008, at
http://www.cms.hhs.gov/CLIA/07_Program_Descriptions_Projects.asp#TopOfPage.)
6. What is CLIA? Answers to frequently asked questions. Washington, DC: American
Society for Microbiology, 2007. (Accessed September 15, 2007, at
http://www.asm.org/Policy/index.asp?bid=2698.)
7. Bogdanich W. Medical labs, trusted as largely error-free, are far from infallible. Wall Street
Journal. February 2, 1987: 1.
8. Otten A. The influence of the mass media on health policy. Health Aff (Millwood)
1992;11(4):111-8.
9. Clinical Laboratory Improvement Amendments of 1988. Public Law 100-578. 42 U.S. Code 263a.
10. CLIA program. Baltimore, MD: Centers for Medicare and Medicaid Services, 2005.
(Accessed May 1, 2006, at http://www.cms.hhs.gov/clia.)
11. Rivers PA, Dobalian A, Germinario FA. A review and analysis of the Clinical Laboratory
Improvement Amendment of 1988: compliance plans and enforcement policy. Health
Care Management Review 2005;30(2):93-102.
12. Clinical Laboratory Improvement Amendments, appendix C: survey procedures and
interpretive guidelines for laboratories and laboratory services. Baltimore, MD: Centers
for Medicare and Medicaid Services, 2003.
http://www.cms.hhs.gov/CLIA/03_Interpretive_Guidelines_for_Laboratories.asp .
13. CLIA program; simplifying CLIA regulations relating to accreditation, exemption of
laboratories under a state licensure program, proficiency testing, and inspection. Fed
Regist 1998;63(93):26722-38.
14. CLIA update--December 2006, laboratories by type of facility. Baltimore, MD: Centers for
Medicare and Medicaid Services, 2006. (Accessed January 3, 2007, at
http://www.cms.hhs.gov/CLIA/17_CLIA_Statistical_Tables_Graphs.asp#TopOfPage.)
15. Public health service; CLIA program; categorization of waived tests. Fed Regist
1995;60(177):47534-43.

May 2008

303

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

16. Laboratory requirements. Code of Federal Regulations, Title 42, Section 493.15(b). Subpart A-general provisions; laboratories performing waived tests.
17. Food and Drug Administration Modernization Act of 1997. Public Law 105-115. 21 U.S. Code
31 et. seq.
18. Howerton D, Anderson N, Bosse D, Granade S, Westbrook G. Good laboratory practices
for waived testing sites: survey findings from testing sites holding a certificate of waiver
under the clinical laboratory improvement amendments of 1988 and recommendations for
promoting quality testing. MMWR Recommendations and Reports 2005;54(RR-13):1-25.
19. Yost J. CLIA and quality systems. COLA Symposium for Clinical Laboratories--an
interactive experience with quality systems. May 2, 2007. Columbia, MD: COLA, 2007.
20. OraQuick ADVANCE rapid HIV-1/2 antibody test. Rockville, MD: Office of In Vitro
Diagnostic Device Evaluation and Safety, Food and Drug Administration, 2007. (Accessed
June 10, 2007, at http://www.fda.gov/cdrh/oivd/CLIA-oraquick.html.)
21. Meeting minutes and testimony. Washington, DC: Presidential Advisory Council on
HIV/AIDS, 2002. (Accessed October 19, 2007, at http://www.pacha.gov/.)
22. Testimony before the FDA Blood Products Advisory Committee. June 14-15, 2001.
Rockville, MD: Centers for Biologics Evaluation and Research, U.S. Food and Drug
Administration, 2001.
http://www.fda.gov/ohrms/dockets/ac/cber01.htm#Blood%20Products.
23. Laboratory requirements. Code of Federal Regulations, Title 42, Part 493.
24. Enrollment and certification processes in the Clinical Laboratory Improvement
Amendments Program. Washington, DC: Office of the Inspector General, Department of
Health and Human Services, 2001. http://oig.hhs.gov/oei/reports/oei-05-00-00251.pdf.
25. Yost J. Centers for Medicare and Medicaid Services. Personal communication.
November 26, 2007.
26. CLIA categorization criteria. Rockville, MD: Center for Devices and Radiological Health,
Food and Drug Administration, 2001. (Accessed October 18, 2007, at
http://www.fda.gov/cdrh/CLIA/categorization.html.)
27. Medicare, Medicaid, and CLIA programs; Clinical Laboratory Improvement Amendments
of 1988 removal of exemption of laboratories in the State of Oregon. Fed Regist
2000;65(85):25492-3.
28. Medicare, Medicaid, and CLIA programs; Clinical Laboratory Improvement Amendments
of 1988 exemption of laboratories in the State of Oregon. Fed Regist 1996;61(115):30072-5.
29. Richman K. An act to add Section 1300.5 to the Business and Professions code, relating to
clinical laboratories. California Assembly Bill No. 2452. 2006.
30. Clinical Laboratory Improvement Amendments (CLIA). Baltimore, MD: Centers for
Medicare and Medicaid Services, 2006.
http://www.cms.hhs.gov/MLNProducts/downloads/CLIABrochure.pdf.
31. A quality management system model for health care. Approved guideline: HS1-A2;
second edition. Wayne, PA: Clinical Laboratory Standards Institute, 2004.

May 2008

304

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

32. Laboratory requirements. Code of Federal Regulations, Title 42, Section 493.1253. Subpart K-quality systems for nonwaived testing; standard: establishment and verification of
performance specifications.
33. Ehrmeyer SS, Laessig RH. Has compliance with CLIA requirements really improved
quality in US clinical laboratories? Clinica Chimica Acta 2004;346(1):37-43.
34. CLIA approved proficiency testing programs--2007. Baltimore, MD: Centers for Medicare
and Medicaid Services, 2007. http://www.cms.hhs.gov/CLIA/downloads/ptlist.pdf.
35. Laboratory requirements. Code of Federal Regulations, Title 42, Section 493. Subpart H-participation in proficiency testing for laboratories performing nonwaived testing.
36. Clinical lab quality: CMS and survey organization oversight should be strengthened.
Report No. GAO-06-416. Washington, DC: Government Accountability Office, 2006.
www.gao.gov/cgi-bin/getrpt?GAO-06-416.
37. Edson D, Massey LD. Proficiency testing performance in physician's office, clinic, and
small hospital laboratories, 1994-2004. Laboratory Medicine 2007;38(4):237.
38. Stull TM, Hearn TL, Steindel SJ, Collins CL. Clinical laboratory performance on
proficiency testing samples--United Sates, 1994. Morbidity and Mortality Weekly Report
1996;45(9):193-6.
39. Hurst J, Nickel K, Hilborne LH. Are physicians' office laboratory results of comparable
quality to those produced in other laboratory settings? JAMA 1998;279(6):468-71.
40. Cembrowski G, Carey RN. Adding value to proficiency testing programs. Clin Chem
2000;46:7-8.
41. Jenny RW, Jackson-Tarentino KY. Causes of unsatisfactory performance in proficiency
testing. Clin Chem 2000;46(1):89-99.
42. Hoeltge GA, Phillips MG, Styer PE, Mockridge P. Detection and correction of systematic
laboratory problems by analysis of clustered proficiency testing failures. Arch Pathol Lab
Med 2005;129(2):186-9.
43. Clinical Laboratory Improvement Amendments (CLIA). Atlanta, GA: Centers for Disease
Control and Prevention, 2006. (Accessed December 14, 2007, at
http://wwwn.cdc.gov/clia/default.aspx.)
44. Cytology proficiency testing. Baltimore, MD: Centers for Medicare and Medicaid Services,
2007. (Accessed December 14, 2007, at
http://www.cms.hhs.gov/CLIA/02_CytologyProficiencyTesting.asp.)
45. Medicare, Medicaid, and CLIA programs: statutory and regulatory requirements related
to laboratory participation in proficiency testing and the availability of proficiency testing
programs in cytology. Baltimore, MD: Centers for Medicare and Medicaid Services, 2006.
http://www.cms.hhs.gov/CLIA/downloads/Informational_Supplement.pdf.
46. Final 2005 national cytology proficiency testing results. Baltimore, MD: Centers for
Medicare and Medicaid Services, 2005.
http://www.cms.hhs.gov/CLIA/downloads/2005FinalTestingResults080906MDMIME.pdf.

May 2008

305

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

47. Laboratory requirements. Code of Federal Regulations, Title 42, Section 493.945. Subpart H-participation in proficiency testing for laboratories performing nonwaived testing;
standard: cytology: gynecological examinations.
48. Renshaw AA, Wang E, Mody DR, Wilbur DC, Davey DD, Colgan TJ. Measuring the
significance of field validation in the College of American Pathologists interlaboratory
comparison program in cervicovaginal cytology: how good are the experts? Arch Pathol
Lab Med 2005;129:609-13.
49. Keenlyside RA, Collins CL, Hancock JS, et al. Do proficiency test results correlate with the
work performance of screeners who screen Papanicolaou smears? Am J Clin Pathol
1999;112(6):769-76.
50. Davey DD, McGoogan E, Somrak TM, et al. Competency assessment and proficiency
testing. Acta Cytol 2000;44(6):939-43.
51. Clinical Laboratory Improvement Advisory Committee. Summary report. Atlanta:
Centers for Disease Control and Prevention, 2006.
http://www.cdc.gov/cliac/pdf/cliac0606.pdf.
52. S. 4056--109th Congress (2006): Cytology Proficiency Improvement Act of 2006.
GovTrack.us, 2007. (Accessed December 15, 2007, at
http://www.govtrack.us/congress/bill.xpd?bill=h109-6133.)
53. H.R. 6133--109th Congress (2006): Cytology Proficiency Improvement Act of 2006.
GovTrack.us, 2007. (Accessed December 15, 2007, at
http://www.govtrack.us/congress/bill.xpd?bill=h109-6133.)
54. H.R. 1237--110th Congress (2007): Cytology Proficiency Improvement Act of 2007.
GovTrack.us, 2007. (Accessed December 15, 2007, at
http://www.govtrack.us/congress/bill.xpd?bill=h109-6133.)
55. Laboratory requirements. Code of Federal Regulations, Title 42, Section 493.1200. Subpart K-quality systems for nonwaived testing; introduction.
56. Medicare, Medicaid, and CLIA programs; laboratory requirements relating to quality
systems and certain personnel qualifications. Fed Regist 2003;68(16):3640-714.
57. Astles JR, Lipman HB, Schalla WO, et al. Impact of quality control on accuracy in enzyme
immunoassay testing for human immunodeficiency virus type 1 antibodies. Arch Pathol
Lab Med 1998;122(8):700-7.
58. DynCorp Health Research Services. General recommendations for quality assurance
programs for laboratory molecular genetic tests. Atlanta, GA: Centers for Disease Control
and Prevention, 1999. http://wwwn.cdc.gov/dls/pdf/genetics/dyncor.pdf.
59. Medicare, Medicaid and CLIA programs; regulations implementing the Clinical
Laboratory Improvement Amendments of 1988 (CLIA). Fed Regist 1992;57(40):7002-134.
60. Yost J. CLIA update. Clinical Laboratory Improvement Advisory Committee meeting.
September 8, 2005. Atlanta, GA: Centers for Disease Control and Prevention, 2005.
61. Projects in development. Wayne, PA: Clinical and Laboratory Standards Institute, 2007.
(Accessed December 17, 2007, at

May 2008

306

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

http://www.clsi.org/Content/NavigationMenu/CommitteesandProjects/AreaCommitte
eonEvaluationProtocols/ProjectsinDevelopment/default.htm.)
62. Westgard JO. CLIA final rule: equivalent quality control practices. Westgard QC, 2004.
(Accessed October 21, 2007, at http://www.westgard.com/cliafinalrule7.htm.)
63. Westgard JO. Electronic quality control, the total testing process, and the total quality
control system. Clinica Chimica Acta 2001;307(1-2):45-8.
64. Survey to ASM clinical microbiologists on QC. Washington, DC: American Society for
Microbiology, 1997. (Accessed October 18, 2007, at
http://www.asm.org/policy/index.asp?bid=13175.)
65. Westgard JO. Internal quality control: planning and implementation. Ann Clin Biochem
2003;40:593-611.
66. LaBeau KM, Simon M, Steindel SJ. Quality control of test systems waived by the Clinical
Laboratory Improvement Amendments of 1988. Perceptions and practices. Arch Pathol
Lab Med 2000;124(8):1122-7.
67. Laboratory requirements. Code of Federal Regulations, Title 42, Section 493. Subpart M-personnel for nonwaived testing.
68. Laboratory requirements. Code of Federal Regulations, Title 42, Section 493.1489. Subpart M-personnel for nonwaived testing; laboratories performing high complexity testing; testing
personnel qualifications.
69. Laboratory requirements. Code of Federal Regulations, Title 42, Section 493.1495. Subpart M-personnel for nonwaived testing; laboratories performing high complexity testing; testing
personnel responsibilities.
70. Laboratory requirements. Code of Federal Regulations, Title 42, Section 493.1425. Subpart M-personnel for nonwaived testing; laboratories performing moderate complexity testing;
testing personnel qualifications.
71. Group Health Association, Inc. and Christopher X Fedor, Petitioner and Office and
Professional Employees International Union Local 2, AFL-CIO. Case 5-RD-1102.
Washington, DC: National Labor Relations Board, 2002.
http://www.nlrb.gov/shared_files/Board%20Decisions/317/317-37.pdf.
72. Laboratory requirements. Code of Federal Regulations, Title 42, Section 493.1443. Subpart M-personnel for nonwaived testing; laboratories performing high complexity testing;
laboratory director qualifications.
73. Advanced practice: doctorate in clinical laboratory science. Bethesda, MD: American
Society for Clinical Laboratory Science, 2005.
http://www.ascls.org/position/AdvPractice.asp.
74. Ellinger P. Chair's message. American Society for Clinical Pathology BOR Newsletter 2006.
75. McGovern MM, Benach M, Wallenstein S, Boone J, Lubin IM. Personnel standards and
quality assurance practices of biochemical genetic testing laboratories in the United States.
Arch Pathol Lab Med 2003;127(1):71-6.

May 2008

307

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

76. Laboratory requirements. Code of Federal Regulations, Title 42, Section 493.1773. Subpart Q-inspection; basic inspection requirements for all laboratories issued a CLIA certification
and CLIA-exempt laboratories.
77. Laboratory requirements. Code of Federal Regulations, Title 42, Section 493.1832. Subpart R-enforcement procedures; directed plan of correction and directed portion of a plan of
correction.
78. Feld R, Schwabbauer M, Olson J. The inspection process. Iowa City, IA: Continuing
medical education, University of Iowa College of Medicine, 2006. (Accessed September 20,
2007, at http://www.medicine.uiowa.edu/cme/clia/modules.asp?testID=5.)
79. Tunis S. Laboratory operations at Maryland General Hospital. Testimony before the U.S.
House of Representatives Government Reform Subcommittee on Criminal Justice, Drug
Policy and Human Resources, 108th Congress, 2nd session. 2004.
80. O'Brien D. Labs face tougher oversight. Baltimore Sun. December 2, 2005.
81. Mandatory inspector training frequently asked questions. Northfield, IL: College of
American Pathologists, 2006. (Accessed November 9, 2007, at
http://www.cap.org/apps/cap.portal?_nfpb=true&cntvwrPtlt_actionOverride=%2Fportl
ets%2FcontentViewer%2Fshow&_windowLabel=cntvwrPtlt&cntvwrPtlt%7BactionForm.c
ontentReference%7D=laboratory_accreditation%2Fmandatory_faq.html&_state=maximiz
ed&_pageLabel=cntvwr#strength.)
82. Partners in laboratory oversight. Guidance for coordination of CLIA activities among CMS
central office, CMS regional offices, state agencies (including states with licensure
requirements, accreditation organizations and
states with CMS approved state laboratory programs). Baltimore, MD: Centers for
Medicare and Medicaid, 2006.
83. Sternsky L. Oversight of genetic testing. Genetics Brief 2002(11).
84. Javitt GH. Policy implications of genetic testing: not just for geneticists anymore.
Advances in Chronic Kidney Disease 2006;13(2):178-82.
85. GeneTests. Seattle, WA: GeneTests.org, 2007. (Accessed July 30, 2007, at
http://www.genetests.org/servlet/access?id=8888891&key=Z1o3AV3PRzDMu&fcn=y&f
w=apWV&filename=/.)
86. Realizing the promise of pharmacogenomics: opportunities and challenges. Draft report
of the Secretary's Advisory Committee on Genetics, Health, and Society. Bethesda, MD:
Secretary's Advisory Committee on Genetics, Health, and Society, 2007.
87. Boone JD. Development of a genetic testing specialty under CLIA. Secretary's Advisory
Committee on Genetics, Health, and Society meeting. October 22, 2003. Atlanta, GA:
Centers for Disease Control and Prevention, 2003.
88. Laboratory requirements. Code of Federal Regulations, Title 42, Section 493.1231. Subpart K-quality systems for nonwaived testing; confidentiality of patient information.
89. Letter to Dr. Kathy Hudson from the Centers for Medicare and Medicaid Services.
Washington, DC: Genetics and Public Policy Center, 2007. (Accessed November 1, 2007, at
http://www.dnapolicy.org/resources/CMSresponse8.15.07.pdf.)

May 2008

308

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

90. Centers for Medicare and Medicaid Services update. Clinical Laboratory Improvement
Advisory Committee meeting. September 21, 2006. Atlanta, GA: Centers for Disease
Control and Prevention, 2006.
91. U.S. system of oversight of genetic testing: a response to the charge of the Secretary of
DHHS. Draft report of the Secretary's Advisory Committee on Genetics, Health, and
Society (available for public comment November 5-December 21, 2007). Bethesda, MD:
Secretary's Advisory Committee on Genetics, Health, and Society, 2007.
http://www4.od.nih.gov/oba/SACGHS/reports/SACGHS%20Draft%20Report%20on%
20the%20Oversight%20of%20Genetic%20Testing%2011-5-2007.pdf.
92. CLIA oversight of genetic testing. Clinical Laboratory Improvement Amendment
Committee meeting. February 14, 2007. Atlanta, GA: Centers for Disease Control and
Prevention, 2007.
93. Tort. Wikipedia, 2007. (Accessed October 22, 2007, at
http://en.wikipedia.org/wiki/Tort_law.)
94. Draft guidance for industry, clinical laboratories, and FDA staff: in vitro diagnostic
multivariate index assays. Rockville, MD: Center for Devices and Radiological Health,
Food and Drug Administration, 2006.
http://www.fda.gov/cdrh/oivd/guidance/1610.pdf .
95. Draft guidance for industry and Food and Drug Administration staff; in vitro diagnostic
multivariate index assays; availability. Fed Regist 2007;72(143):41081-3.
96. FDA clears breast cancer specific molecular prognostic test. Rockville, MD: Food and Drug
Administration, 2007. (Accessed May 17, 2007, at
http://www.fda.gov/bbs/topics/NEWS/2007/NEW01555.html.)
97. Draft guidance for industry and FDA staff: commercially distributed analyte specific
reagents (ASRs): frequently asked questions. Rockville, MD: Center for Devices and
Radiological Health, Food and Drug Administration, 2006.
http://www.fda.gov/cdrh/oivd/guidance/1590.pdf .
98. Center sees "new era in oversight" of genetic tests in two new FDA draft guidances.
Washington, DC: Genetics and Public Policy Center, 2006. (Accessed June 10, 2007, at
http://www.dnapolicy.org/news.release.php?action=detail&pressrelease_id=56.)
99. Gibbs JN. Regulations and standards: the past, present, and future of ASRs. IVD
Technology 2003.
100. Javitt GH, Hudson K. Federal neglect: regulation of genetic testing. Issues Sci Technol
2006;22(3):59-66.
101. Nutrigenetic testing: test purchased for four Web sites mislead consumers. Washington,
DC: Government Accountability Office, 2006.
102. Information on premarket approval applications. Rockville, MD: Center for Devices and
Radiological Health, Food and Drug Administration, 2008. (Accessed March 27, 2008, at
http://www.fda.gov/cdrh/pmapage.html.)

May 2008

309

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

103. Information on releasable 510(k)s. Rockville, MD: Center for Devices and Radiological
Health, Food and Drug Administration, 2008. (Accessed March 27, 2008, at
http://www.fda.gov/cdrh/510khome.html#1.)
104. Whitaker BI, Sullivan M. The 2005 nationwide blood collection and utilization survey
report. Rockville, MD: Department of Health and Human Services, 2005.
105. Menitove JE. Controversies in transfusion medicine. The recent emphasis on good
manufacturing practices and the pharmaceutical manufacturing approach damages blood
banking and transfusion medicine as medical care activities. Transfusion 1993;33(5):439-42.
106. The Rh factor: how it can affect your pregnancy. San Francisco, CA: Medem, Inc., 2005.
(Accessed October 29, 2007, at
http://www.medem.com/MedLB/article_detaillb.cfm?article_ID=ZZZAM52C67C&sub_
cat=2005.)
107. Keeping blood transfusions safe: FDA's multi-layered protections for donated blood.
Rockville, MD: Food and Drug Administration, 2002.
108. Blood frequently asked questions. Bethesda, MD: AABB, 2007. (Accessed October 26, 2007,
at http://www.aabb.org/AABBContent/Templates/AABBSub.aspx?NRMODE=
Published&NRORIGINALURL=%2fContent%2fAbout_Blood%2fFAQ%2fbloodfaq%2eht
m&NRNODEGUID=%7b3A25F398-BFFA-4909-9257B673AD063A2C%7d&NRCACHEHINT=NoModifyGuest#a9.)
109. Introduction: national testing labs. Washington, DC: American Red Cross, 2007. (Accessed
October 26, 2007, at http://www.redcross.org/services/biomed/0,1082,0_494_,00.html.)
110. Memorandum of understanding between the Health Care Financing Administration and
the Food and Drug Administration. MOU number: 225-80-4000. Rockville, MD: Food and
Drug Administration, 1983. http://www.fda.gov/oc/mous/domestic/225-80-4000.html.
111. Sullivan J. AABB. Personal communication. October 25, 2007.
112. Commission on Laboratory Accreditation Laboratory Accreditation Program: transfusion
medicine checklist. Northfield, IL: College of American Pathologists, 2007.
http://www.cap.org/apps/docs/laboratory_accreditation/checklists/transfusion_medic
ine_sep07.pdf.
113. About FACT. Omaha, NE: Foundation for the Accreditation of Cellular Therapy, 2007.
(Accessed October 29, 2007, at http://www.factwebsite.org/main.aspx?id=72.)
114. Laboratory services: facts about laboratory accreditation. Oakbrook Terrace, IL: The Joint
Commission, 2007. (Accessed May 10, 2007, at
http://www.jointcommission.org/AccreditationPrograms/LaboratoryServices/lab_facts.htm.)
115. Shulman IA, Maffei LM, Downes KA. North American pretransfusion testing practices,
2001-2004: results from the College of American Pathologists Interlaboratory Comparison
Program survey data, 2001-2004. Arch Pathol Lab Med 2005;129(8):984-9.
116. Transfusion medicine. Northfield, IL: College of American Pathologists, 2007. (Accessed
October 29, 2007, at
http://www.cap.org/apps/cap.portal?_nfpb=true&cntvwrPtlt_actionOverride=%2Fportl
ets%2FcontentViewer%2Fshow&_windowLabel=cntvwrPtlt&cntvwrPtlt%7BactionForm.c

May 2008

310

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

ontentReference%7D=disciplines%2Ftransfusion_medicine%2Ftransmed_index.html&_st
ate=maximized&_pageLabel=cntvwr.)
117. Compliance program guidance manual. Chapter 42. Blood and blood products.
Inspection of licensed and unlicensed blood banks, brokers, reference laboratories, and
contractors--7342.001. Rockville, MD: Center for Biologics Evaluation and Research, Food
and Drug Administration, 2006. http://www.fda.gov/cber/cpg/7342001bld.pdf.
118. Gustafson M. Blood safety. The Food and Drug Administration's role. Arch Pathol Lab
Med 1999;123(6):475-7.
119. Blood. Rockville, MD: Centers for Biologics Evaluation and Research, Food and Drug
Administration, 2007. (Accessed October 22, 2007, at
http://www.fda.gov/cber/blood.htm.)
120. Roseff SD. Regulatory and quality initiatives--trends in transfusion medicine.
LabMedicine 2005;36(2):108-14.
121. Guideline for quality assurance in blood establishments. Docket no. 91N-0450. Rockville,
MD: Center for Biologics Evaluation and Research, Food and Drug Administration, 1995.
http://www.fda.gov/cber/gdlns/gde_qa.pdf.
122. Guidance for industry: implementation of acceptable full-length donor history
questionnaire and accompanying materials for use in screening donors of blood and blood
components. Rockville, MD: Center for Biologics Evaluation and Research, Food and
Drug Administration, 2006.
123. Donor History Questionnaire for allogeneic HPC, apheresis, and HPC, marrow (HPCDHQ). Bethesda, MD: AABB, 2007. (Accessed October 25, 2007, at
http://www.aabb.org/Content/Donate_Blood/Donor_History_Questionnaires/HPC_D
onor_History_Questionnaire/dhqhpc.htm.)
124. Topic III: study design for an abbreviated uniform donor history questionnaire. Blood
Products Advisory Committee Meeting. October 25, 2007. Rockville, MD: Food and Drug
Administration, 2005.
125. AABB donor history questionnaire documents. Rockville, MD: Center for Biologics
Evaluation and Research, Food and Drug Administration, 2006. (Accessed October 30,
2007, at http://www.fda.gov/cber/dhq/dhq.htm.)
126. Blood donor history questionnaire. Bethesda, MD: AABB, 2007. (Accessed October 30, 2007,
at http://www.aabb.org/Content/Donate_Blood/Donor_History_Questionnaires/
AABB_Blood_Donor_History_Questionnaire.)
127. Blood action plan. Rockville, MD: Center for Biologics Evaluation and Research, Food and
Drug Administration, 2001. (Accessed October 29, 2007, at
http://www.fda.gov/cber/blood/bap.htm.)
128. Blood donations in U.S. testing positive for Chagas' disease. New York, NY: PR Newswire,
2007. (Accessed October 30, 2007, at http://www.prnewswire.com/cgibin/stories.pl?ACCT=109&STORY=/www/story/10-20-2007/0004686177&EDATE=.)
129. Institute of Medicine. The future of drug safety: promoting and protecting the health of
the public. Washington, DC: National Academy Press, 2006.

May 2008

311

Laboratory Medicine: A National Status Report

Chapter VII – Federal Regulatory Oversight of Laboratory Medicine

130. The future of drug safety--promoting and protecting the health of the public. FDA's
response to the Institute of Medicine's 2006 report. Rockville, MD: Food and Drug
Administration, 2007. http://www.fda.gov/oc/reports/iom013007.pdf.

May 2008

312

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

CHAPTER VIII
REIMBURSEMENT FOR LABORATORY MEDICINE
Reimbursement for most health care in the U.S. is administered by government and private sector
third parties using multiple systems of coverage, coding, and payment. Third-party payment has
enabled patients to access and benefit from health care products and services, including
laboratory testing, and ensures broad markets for these. However, difficulty in acquiring
coverage, appropriate coding, and adequate payment can pose significant hurdles to laboratory
testing. The consequences can include reduced patient access to laboratory testing and decreased
incentives for laboratories and test manufacturers to engage in further test development.
As health care continues to usurp a larger proportion of national spending, it places greater
burdens on employers, patients, and the health care system itself, with broader implications for
industry, competitiveness, and other aspects of the economy. These pressures are leading to
proposals to modify and even broadly reform health care payment systems.1, 2, 3
Many of the concerns pertaining to Medicare payment for laboratory services were identified in
the 2000 IOM report, Medicare Laboratory Payment Policy: Now and in the Future.4 Yet, little has
changed since publication of that report. This chapter provides an overview of the current public
and private sector payment systems for laboratory services and an analysis of payment issues that
affect laboratory testing access, effectiveness, efficiency, and innovation.

PUBLIC AND PRIVATE SECTOR PAYERS
The U.S. health system has multiple public and private sector payers. CMS is the largest
purchaser of health care in the U.S. CMS administers Medicare, a federal program, and the
federal portion of the Medicaid program and the State Children’s Health Insurance Program,
which are funded jointly with the states. In fiscal year 2006, CMS net outlays were approximately
$515.2 billion for these programs,a about 19.4% of total federal outlays.5
Medicare provides benefits for 43.2 million beneficiaries, including 36.3 million elderly and 7 million
disabled enrollees, comprising 14% of the population. By 2031, the number of beneficiaries will be
an estimated 77 million.6 This includes the baby-boom generation, the last of which will turn 65 in
2029. The Medicaid program covers 50.3 million beneficiaries, about 17% of the U.S. population.
More than 75% of Medicaid enrollees are low-income children and their parents.6
Other government payers, i.e., the Military Health System (MHS), VHA, and Indian Health
Service, operate independent health care systems that provide services directly to their
beneficiaries or through negotiated contracts with private sector providers. In fiscal year 2006, the
MHS and VHA programs were estimated to spend $70 billion on health care for approximately 16
million enrolled veterans, active duty, and retired military personnel and their beneficiaries.7, 8

a

The other federal outlays included Social Security $586 billion (22%), defense $499 billion (18.8%), treasury $464
billion (17.5%), and other $590 billion (22.2%).

May 2008

313

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

Private sector payers include insurance companies and commercial health plans such as Aetna,
Blue Cross and Blue Shield plans, CIGNA, Kaiser Permanente, UnitedHealthcare, and WellPoint.
Some of these plans also act as contractors to Medicare and Medicaid, processing claims and
administering payments to laboratories. In 2005, approximately 200 million Americans were
enrolled in managed care plans, including 15 million enrolled in Medicaid programs, and 7
million in Medicare programs.9, 10
Based on 1999 data reported by the IOM, clinical laboratories derive their revenues from fee-forservice payments (42%), followed by Medicare payments (29%), Medicaid payments (12%),
consumers’ out-of pocket payments (10%), and health maintenance organization (HMO)
capitation payments (7%).4

Components of Reimbursement Systems
Three main components of payment systems are coverage, payment, and coding:

ƒ

Coverage decisions establish the conditions under which third-party payment is
provided, including the range of benefits provided under particular plans or contracts,
which items and services can be reimbursed under those benefits, clinical indications
for which these items and services (e.g., laboratory tests) will be reimbursed, and the
circumstances or settings in which the items and services will be reimbursed. Medical
necessity and appropriateness determinations affect payer coverage decisions.

ƒ

Payment methodologies establish payment levels for tests and services provided and
methods for calculating these amounts. Payment levels typically are tied to the codes
for these tests and services. These levels may be provided in the form of prospective
payment systems (PPSs), fee schedules, or negotiated contracts with payment rates for
particular codes. The type of payment methodology of choice may vary by insurer.

ƒ

Coding systems involve the alphanumeric nomenclatures assigned to particular health
conditions, services, or products and the processes for assigning and updating these.
The coding systems that apply to laboratory medicine include Current Procedural
Terminology® (CPT) codes and Healthcare Procedural Coding System (HCPCS) codes
for laboratory tests and services and International Classification of Diseases, Ninth
Revision, Clinical Modification (ICD-9-CM) codes for diagnoses.

Coverage and coding policies shape the operation of payment systems. Whether for laboratory
testing or other types of health care, the design and implementation of payment systems can
influence patient access, provider decision-making, and innovation.

COVERAGE DECISIONS
Public and private sector payers make coverage decisions independently, often with
consideration of the financial impact resulting from specific determinations. However, legislative
actions may mandate coverage of specific tests by all payers.

May 2008

314

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

Medicare Coverage Decisions
Medicare’s authorizing legislation in 1965 established broad categories of coverage for hospital,
physician, and laboratory services, but limited payment to expenses deemed reasonable and
necessary for the diagnosis or treatment of illness or injury or to improve the functioning of a
malformed body member.11 Coverage decisions are left to Medicare officials who, in turn, have
delegated most of their authority to local contractors, which are typically private sector insurance
companies that administer the program in specific geographic jurisdictions, subject to national
Medicare provisions and requirements.12
The contractors that administer Part A of Medicare are known as fiscal intermediaries (FIs) and
those that administer Part B are known as carriers. About 90% of all Medicare coverage reviews
and decisions still occur at the local level while 10% occur at the national level. In 2007, there
were 16 carriers and 20 FIs. However, for laboratory services, there are 56 carriers for
corresponding geographic areas (see payment section below). Carriers handling other Part B
services can serve as one of the 56 carriers handling laboratory services.
Most Medicare coverage policies for new tests and services under Part B are established by local
carrier advisory committees and are known as local coverage determinations (formerly referred to
as local medical review policies). In this decentralized system, there is substantial variation
among coverage policies of different carriers. Aside from the uncertainty and inconsistency that
arise from this arrangement are long-standing concerns regarding the lack of openness,
transparency, predictability, and length of time involved in these coverage processes.11 In
addition, the Medicare statute restricts payment for preventive and screening services and
technologies, unless otherwise specified by Congress.
Several legislative initiatives, along with various policy reports, have been directed at some of these
issues. The Balanced Budget Act of 1997 provided the legislative vehicle to consolidate the number
of CMS contractors making coverage decisions and processing laboratory claims from 56 to 5.13
However, the consolidation has not been implemented to date. In 1999, CMS established the
Medicare Coverage Advisory Committee (now called the Medicare Evidence Development and
Coverage Advisory Committee) and restructured the national coverage processes in order to
facilitate more systematic and transparent national coverage determinations for Medicare. The
Medicare Evidence Development and Coverage Advisory Committee provides recommendations
based on literature reviews, technology assessments, and expert advice, though CMS retains control
over final decisions.11 National coverage determinations take precedence over local carrier
decisions. Although it continues to improve, CMS has failed frequently to meet its standard for
prompt assessment of six to nine months, depending on the type of test or technology. Review
times for some decisions frequently exceed nine to twelve months.14
In 2001, the Medicare Payment Advisory Committee (MedPAC) recommended the elimination of
local coverage policies and payment schedules in order to reduce complexity, inconsistency, and
uncertainty associated with Medicare reimbursement.15 (MedPAC is an independent federal
body established by the Balanced Budget Act of 1997 to advise the U.S. Congress on issues
affecting the Medicare program.) However, this proposal was resisted by many stakeholders,
including those affiliated with laboratory medicine. In response to concerns from the laboratory
community, CMS published a negotiated proposed rule making in 2001 to implement 23 national

May 2008

315

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

coverage policies for 66 CPT codes that represent about 60% of all laboratory tests billed to the
Medicare program (see Box 8.1).16 The national policies describe the medical conditions for which
a laboratory test is covered and set frequency limits on coverage of the same test for a patient.
The rule making also established opportunities for stakeholder involvement in the decisionmaking process during annual reviews of coverage policies. Even so, some stakeholders continue
to call for further transparency in this process.
Under provisions in the MMA 2003, CMS began reforming the carrier-based approach to
administration of its programs. This initiative is expected to have a direct impact on the number
of carriers affiliated with coverage and processing of laboratory claims. CMS is transitioning to
the Medicare Administrative Contractor (MAC) system that will eventually replace all carriers
and FIs. The first MAC contract was awarded in July 2006 to Noridian Administrative Services
(Fargo, ND) to implement consolidated claims processing for Medicare Part A and Part B in
Jurisdiction 3 (Arizona, Montana, North and South Dakota, Utah, and Wyoming).17 However, at
present, the use of MACs will not change the geographic-oriented payment rates or number of fee
schedules for laboratory services.18
In 2003, GAO issued a report that also recommended elimination of local coverage and expansion
of the national coverage system. However, as noted in the GAO report’s summation of public
comments, DHHS and certain stakeholders, including some in industry, opposed doing so,
stating that the elimination of local coverage would result in net increases in expenditures, mainly
due to spending on additional resources to manage national coverage decisions for all services
and products.19 MMA 2003 expanded Medicare coverage of certain screening tests, including for
cardiovascular disease and diabetes.20 These tests and other preventive services are
recommended for certain population groups by authoritative groups such as the USPSTF and the
Advisory Committee on Immunization Practices.21
Box 8.1: 23 National Coverage Determinations for Medicare, Effective 2001
Alpha-fetoprotein (AFP)
Blood counts
Blood glucose testing
Carcinoembryonic antigen (CEA)
Collagen crosslinks, any method
Digoxin therapeutic drug assay
Fecal occult blood test (FOBT)
Gamma glutamyl transferase (GGT)
Glycated hemoglobin/glycated protein
Hepatitis panel/acute hepatitis panel
Human chorionic gonadotropin (hCG)
Human immunodeficiency virus (HIV) testing
(diagnosis)

Human immunodeficiency virus (HIV) testing
(prognosis including monitoring)
Lipid testing
Partial thromboplastin time (PTT)
Prostate specific antigen (PSA)
Prothrombin time (PT)
Serum iron studies
Thyroid testing
Tumor antigen by immunoassay – CA 125
Tumor antigen by immunoassay – CA 15-3/
CA 27.29
Tumor antigen by immunoassay – CA 19-9
Urine culture, bacterial

Source: Lab NCDs. Baltimore, MD: Centers for Medicare and Medicaid Services, 2007.

May 2008

316

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

Denials and Advanced Beneficiary Notice
Medicare denies claims for laboratory tests when they fail to meet required criteria.b According to
MedPAC, the national claims denial rate for laboratory services in 2002 was 15%, with much
higher rates in certain geographic areas.22 A 2006 analysis found that Medicare denial rates for
laboratory testing peaked in 1998 (15%) and have been declining since then to 8.8% in 2004.23
However, this analysis is confounded by the definition of claims denial, which includes both true
denials (i.e., services for which payment was never made) and “paperwork” denials (i.e., denials
that are ultimately resolved and paid).
Documentation of reasons for claims denials associated with medical necessity determinations
have been inconsistent among carriers. To remedy this problem, CMS introduced additional
documentation requirements for medical necessity in the late 1990s. However, inconsistencies
have persisted. The IOM concluded that regional variations in denial rates for the top 20
laboratory tests (by dollar value) by carrier were attributed to geographic patterns of fraud and
abuse, varying interpretation of Medicare rules by local carriers, and low numbers of tests in a
particular region that might skew the proportion of denied claims. Thus, while denials associated
with fraud and abuse are certainly appropriate, some proportion of denials are associated with
unresolved inconsistencies among carriers.4
In a fee-for-service reimbursement system, denied claims are the financial responsibility of the
laboratories, rather than the physician ordering the test.4 While justifiable claims denials can save
costs and improve efficiency, those that are subject only to inconsistent decisions or policies, or
that are otherwise unjustified, can contribute to the total cost of providing laboratory services
because they may increase administrative costs, decrease aggregate revenue for laboratories, and
create bad debt expense. Because the laboratory does not have direct contact with the patient in
most situations, it must depend on the clinician to recognize that: (1) the laboratory test ordered
is subject to medical review, (2) the medical necessity of the test is indeterminate, and (3) the
patient’s signature on an Advanced Beneficiary Noticec (ABN) is needed.4 In cases where a
patient receives a laboratory test that is not covered by Medicare and for which an ABN was not
signed, the laboratory must absorb the cost.25
In 2002, CMS issued an ABN form specific to laboratory testing that allowed laboratories to list
tests that could be denied as well as specify the possible reasons for denial.26 To simplify ABN
further, in 2007, CMS proposed combining the general use ABN and the laboratory ABN.27
However, many clinical laboratory professional societies stated their preference to retain the
separate laboratory ABN form as it provides beneficiaries with a clear understanding of the
reasons for the denial of coverage of specific laboratory tests.28, 29 As of December 2007, CMS
maintained both forms; however, laboratory experts anticipate that the agency will introduce and
require use of a new, combined form in 2008.30
Medicare denies claims for laboratory tests when such claims are for tests that Medicare does not cover, do not meet
medical necessity requirements, are for individuals who are not Medicare beneficiaries, are from laboratories that
are not Medicare-providers or are not certified by CLIA to perform that particular test, are not documented
sufficiently, and are for patients whose primary coverage is from another payer.4
c Medicare requires that physicians notify beneficiaries of the possibility that Medicare might not deem a laboratory
test medically appropriate. Beneficiaries must sign an ABN acknowledging their understanding of this policy
and personal responsibility for payment.24
b

May 2008

317

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

Medicaid Coverage Decisions
Medicaid beneficiaries receive a comprehensive set of medical benefits, with some services
required by the federal government and others offered at the discretion of the state.31 Individual
states have substantial decision making authority over which benefits their Medicaid programs
will cover. Within the broad national guidelines established by federal statutes, regulations and
policies, each state: (1) establishes its own eligibility standards; (2) determines the type, amount,
duration, and scope of services; (3) sets the payment rates for services; and (4) administers its own
program.32 About 35% of spending in each state is for federally mandated benefits (e.g.,
laboratory services) while 65% is for optional services (e.g., prescription drugs).33
Federal guidelines apply to coverage and payment for laboratory services under Medicaid.
Medicaid programs are required to cover professional and technical laboratory services that are:

ƒ

Ordered and provided by or under the direction of a physician or other licensed
practitioner, or ordered by a physician and provided by a referral laboratory

ƒ

Furnished by a laboratory meeting the requirements outlined in CLIA33

This includes laboratory testing conducted in the spectrum of inpatient and ambulatory care
settings in which services can be provided to Medicaid beneficiaries.33 Also, Medicaid must cover
screening, diagnostic, and treatment services for beneficiaries under age 21.
All states participating in the Medicaid program must designate one state agency that is
responsible for administering and supervising the state’s Medicaid program.34 However, federal
rules do not dictate a specific administrative structure for state Medicaid programs or how
coverage decisions must be made, including those for new laboratory testing technologies. In
New York State, for example, various offices within the Department of Health, Governor’s Office,
the Legislature, and other agencies all play a role in determining what benefits the state’s
Medicaid program will cover and what models of service delivery it will employ.

TRICARE and Veterans Health Administration Coverage Decisions
Both the TRICARE and VHA significantly expanded their benefit programs in the 1990s.
Independently, the programs were reorganized to incorporate policies and procedures used by
preferred provider and managed care organizations. For TRICARE, coverage of health care
services, including laboratory services, has remained constant and generous. Because TRICARE
provides care to family members of military personnel, a full spectrum of diagnostic services are
available as part of the benefits package, including many genetic tests used to screen and
diagnose newborns through adults.
The VHA provides a standard health benefits plan to enrollees; however, unlike TRICARE, it does
not provide benefits to family members. The plan emphasizes preventive and primary care, and
offers a full range of inpatient and ambulatory care services within the VA health care system,
including laboratory tests for screening, diagnosis and treatment. Funding of the program is
discretionary and the system is expected to meet the needs of its beneficiaries within its budget,
although Congress may authorize additional funds if needed.

May 2008

318

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

Private Sector Coverage Decisions
Most private payers maintain their own process for making decisions regarding coverage,
although they often follow the coverage decisions made by Medicare and some of the larger
private health plans.10, 35-37 Similar to government payers, coverage decisions are made at the local
level; however, in contrast, private payers are not obligated to establish advisory committees or
engage in publicly open processes.38 Private payers may choose to adjust policies following
introduction of new technologies to the market, with the impetus for such changes coming from a
variety of sources, such as state or federal mandates, consumer preference, or financial concerns.d
In addition, payers may negotiate specific coverage plans with the groups or employers
purchasing the plan.39
Constraints on health care costs, demand for better outcomes and higher quality, and the
unprecedented rate at which new technologies are being introduced to the market have driven
many health care payers to use evidence-based decision-making to determine the most
appropriate services and technologies to cover.35 Public and private sector payers increasingly
draw upon health technology assessments (HTAs) to inform their coverage decisions. HTAs
typically involve using a systematic approach to assembling and interpreting available evidence
to determine whether a test or medical service is safe, effective, and, sometimes, cost-effective, for
particular patients and health care settings. Several payers have conducted health technology
assessments for screening, diagnostic, and monitoring laboratory tests.
Some health insurance plans and purchasers have created thorough processes for conducting
HTAs internally. Those that do not conduct formal reviews of new technologies may purchase
assessments from HTA vendors. Examples of private health plans that conduct formal reviews of
new technologies include Aetna, CIGNA, UnitedHealthcare, and WellPoint.10 Various HTA
vendors in the U.S. generate proprietary assessments that are available to payers and other
subscribers. For example, the Technology Evaluation Center (TEC), established by the Blue Cross
Blue Shield Association, produces evidence-based technology assessments of the clinical
effectiveness and appropriateness of a specific medical procedure, device, or drug.40, 41 TEC is
guided by its Medical Advisory Panel, which consists of 19 independent, nationally recognized
experts in HTA, clinical research, and medical specialties.42 ECRIe and HAYES, Inc.f are two other
examples of U.S.-based HTA vendors whose reviews are used by payers in support of their
coverage decisions.43, 44

Health insurance plans are required by law to provide information to plan members about what services are
covered and how coverage decisions are made and can be appealed. Denied claims must be accompanied by a
reason for the denial, including citations of any policy or criteria on which the coverage decision is based.35
e ECRI Institute is a nonprofit organization that provides consulting services related to, e.g., patient safety, quality,
risk assessment and management, and technology assessment. It is also an AHRQ-designated Evidence-based
Practice Center.
f HAYES, Inc., provides technology assessment reports for health plans, managed care companies, hospitals, and
health networks and offers training programs to facilitate participants’ understanding of the HTA process.
d

May 2008

319

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

Laboratory tests that have been assessed by the TEC include:

ƒ

Serial endpoint testing for the diagnosis and treatment of allergic disorders

ƒ

High-sensitivity C-reactive protein measurement for coronary heart disease risk
stratification

ƒ

iFOBT vs. gFOBT

ƒ

Use of intermittent or continuous interstitial fluid glucose monitoring in patients with
diabetes mellitus

ƒ

Use of epithelial cell cytology in breast cancer risk assessment and high-risk patient
management

ƒ

Chemotherapy sensitivity and resistance assays40

In addition to HTA findings, private payers also may consider clinical practice guidelines, cost,
and the availability of an appropriate CPT code.35 Private payers are becoming more open to
meeting with manufacturers to discuss what data they need to make coverage decisions for new
technology.39 In general, there is consistency in coverage among private sector payers in routine
laboratory testing associated with standard of care, although some remaining variations in
coverage may inhibit an individual’s access to certain tests and services.10 The 2006 SACGHS
report on coverage and payment for genetic tests and services found that, of those private payers
whose coverage policies are publicly available, most cover genetic testing for chromosomal
abnormalities, prenatal and neonatal diagnosis, and pre-implantation genetic diagnosis in some
cases.35 These payers also generally cover genetic testing for certain rare diseases.
Less information is available regarding denial rates for private payers. One indication of denial
rates comes from Athenahealth, Inc., a provider of web-based services and software to medical
practices, whose “PayerView” program reviews health insurance performance from the
perspective of physicians.45 In 2006, Athenahealth published its first “PayerView” for of Texas,
which analyzed claim performance data for more than 330 providers and 59 medical practices.
Medicare Part B in Texas denied 5.6% of claims submitted, which was below the rate for the
state’s private insurers, which ranged from 6.5 to 10.4% among the top eight companies.

Outstanding Issues in Coverage
Limited Medicare Coverage of Screening Tests
Medicare is limited by statute to providing coverage only for items or services that are “reasonable
and necessary for the diagnosis or treatment of illness or injury or to improve the functioning of a
malformed body member.” In order for Medicare to cover screening or preventive interventions
(i.e., in patients without signs, symptoms, complaints or personal history of disease or injury),
Congress must pass new legislation mandating coverage of those specific interventions. Examples
of preventive tests approved by Congress to date include tests to screen for cardiovascular disease,
cervical and vaginal cancer, colorectal cancer, breast cancer, prostate cancer , glaucoma,
osteoporosis, abdominal aortic aneurysm, and diabetes.46 In addition, Congress has also approved
Medicare coverage of smoking cessation counseling, medical nutrition therapy for beneficiaries
with diabetes or renal disease, vaccinations to prevent influenza, pneumonia, and hepatitis B virus,
and a one-time “Welcome to Medicare” physical examination for new enrollees. In contrast,

May 2008

320

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

coverage of preventive services and screening tests is quite extensive for private sector payers. The
Medicare Preventive Services Coverage Act of 2007 (S. 2115), introduced in Congress in September
2007 by Senator Ben Cardin (D-MD), includes provisions for expand Medicare coverage for certain
preventive tests and related services. (See Box 8.2.)47
Box 8.2: Medicare Preventive Services Coverage Act of 2007 (S. 2115)
The bill’s core provisions call for Congress to:
•

Extend the eligibility period of the “Welcome to Medicare” visit, a preventive physical examination,
from 6 months to 1 year

•

Authorize the Secretary of DHHS to expand Medicare coverage to include services deemed reasonable
and necessary for the prevention or early detection of illness or disability, taking into account
evidence-based recommendations by the USPSTF and other appropriate organizations

•

Eliminate coinsurance rates for the “Welcome to Medicare” visit and other preventive services47, 48

Source: S. 2115: The Medicare Preventive Services Coverage Act of 2007.

Inconsistencies in Coverage of Genetic Tests among All Payers
Some emerging tests (e.g., those used in PGx) may not fit neatly within Medicare’s coverage
criteria. Medicare considers most predictive and predispositional genetic tests to be screening
tests, and as such, congressional authorization is necessary for coverage. Most genetic tests are
not covered by Medicare unless they are performed on symptomatic individuals or are used to
identify treatment-responsive populations.49 Unresolved issues pertaining to coverage of
genetic tests include: (1) the extent to which genetic tests used in broader metabolic profiles, or
for more limited profiles targeted to specific biomarkers meet current medical necessity criteria;
and (2) the extent to which genetic tests can be tied to clinical decision making and patient
health outcomes.50
In 2006, SACGHS recommended that Medicare cover predictive and predispositional genetic tests
and their accompanying services (e.g., genetic counseling) that meet appropriate evidence
standards.35 SACGHS also recommended that CMS clarify that a “personal history” of disease
can include having a family history of a disease, thereby making it possible for beneficiaries with
a family history of a disease to meet Medicare’s standard of “reasonable and necessary.”
With the exception of newborn genetic screening and follow up, which are federally mandated,
states are responsible for Medicaid coverage decisions for genetic tests and services.35 Given
individual state differences in policy, there is significant disparity in patient access to these tests
and services. States’ decisions regarding whether to cover specific genetic tests is determined in
large part by financial and political factors. Although Medicaid programs account for, on
average, about 22% of state budget, states sometimes restrict Medicaid spending by limiting
coverage and/or payment rates, particularly on those items and services outside the scope of
federal requirements.51, 52 In 2006, SACGHS recommended that DHHS ensure that all states
receive information about genetic tests and services in order to inform their Medicaid coverage
decision making processes.35

May 2008

321

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

Coverage for newly developed tests, especially molecular, genetic, and PGx tests, varies widely
among private sector payers. Often, there are fewer research studiesg available on their clinical
utility and costs.10, 35, 39, 53 In 2004, the Congressional Research Service reviewed the coverage
decisions of 27 private payers and found that 16 had developed policies for genetic testing to
detect hereditary colon cancer.53 However, coverage decisions for this testing varied widely
across individual payers.h
The variability in coverage has arisen in part because neither public nor private payers have
developed uniform methods for obtaining information from laboratories about the new tests.54
The lack of a uniform process for making coverage decisions can lead to inappropriate use (e.g.,
overuse and underuse) of new laboratory testing technologies that otherwise have the potential to
improve the quality of health care.10 Recently, some public and private sector stakeholders have
recognized the need to provide guidance to private payers in the areas of PGx and genetic testing.
In 2006, SACGHS recommended the establishment of a task group that includes public and
private sector representatives to create a set of guiding principles for coverage decisions,
addressing issues such as economic evaluation/cost-effectiveness, prevention, rare disease tests,
therapeutic benefit, and informational utility, and make available all scientific evidence needed to
support private payer coverage decisions for individual patient populations served by the
respective payer.

PAYMENT METHODOLOGIES
Although private sector insurance accounts for higher revenues, it is the Medicare program that
exerts the strongest influence on laboratory services payment for all payers. According to a
microcosting study performed for an IOM report, 66.7% of private insurance plans evaluated for
the study used the Medicare payment rates as the basis for their own.55 The Medicare payment
rates also affect state Medicaid programs and other federal payer programs. As such, various
strengths and weaknesses of the Medicare payment system extend beyond Medicare to other
payers, providers, and beneficiaries.

Medicare Payment Methodologies
The current payment structure of the Medicare program comprises multiple, often complex
payment methodologies for Part A (inpatient care), Part B (hospital outpatient and ambulatory
care), and Part C (private sector options), depending on site of care and services provided. Part A
pays for inpatient hospital, skilled nursing facility, home health, and hospice care. (See Appendix
C for additional information on the development of the Medicare payment system.) In 2006, Part
A accounted for 41% of Medicare spending, Part B accounted for 35%, Part C accounted for 16%,
and the new Part D prescription drug benefit accounted for 8%.56

Although demonstrating improved health outcomes is a primary factor in coverage determinations, the effect of
molecular and genetic test results on a patient’s health may not be realized for many years, making it difficult to
perceive and measure the immediate benefits and long term cost effectiveness of such tests.39
h Four payers covered genetic testing without specifying the genes, five covered mutation analysis in three common
genes, four covered microsatellite instability analysis in addition to the standard genetic tests, one covered genetic
testing of only one gene, and two did not cover genetic testing at all.
g

May 2008

322

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

Rising costs of care and pending retirement of the baby boom generation suggest accelerated
growth in Medicare outlays in 2010 and beyond. Over the past 40 years, costs per beneficiary
under Medicare and Medicaid have increased about 2.5% faster per year than has per capita gross
domestic product.57 Medicare spending is projected to grow at a rate of 7.3% over the next
decade.58 Congress has directed federal agencies, particularly payers, to study possible
alternatives to the current payment methodology (such as competitive bidding) as well as more
aggressive strategies to cut costs.

Prospective Payment Systems for Inpatient and Hospital Outpatient Care
For inpatient care under Part A, payment is provided via lump sum based on the patient’s
diagnosis. Hospital diagnosis-related groups (DRGs) are designed to cover institutional costs,
excluding physicians’ services that are paid for through the Part B fee schedules. Included in the
DRGs are any laboratory tests that are bundled with other services for a given hospital admission.
Other inpatient facilities use prospectively set groupings that are similar to DRGs; some use per
diem rates.i Generally, a standardized base rate is determined according to the diagnosis
grouping and associated relative value, which may be adjusted to accommodate case-mix (i.e.,
health condition, clinical characteristics) and geographic differences in wages. (Relative value
payment methodology is discussed below.) For hospitals, additional adjustments can be made to
sites treating a disproportionate share of low-income patients, sites in rural locations, or outlier
cases of extraordinarily high cost.
Both operating and capital payment rates are updated annually for all PPSs. Operating costs are
updated according to the projected increase in CMS’ market basket index (which measures price
increases of goods and services hospitals buy to produce patient care). Capital updates are
determined by the Secretary of DHHS. In addition, payments to hospitals that fail to provide data
on specified quality indicators are reduced by 2%.59 (Reporting on quality indicators as a
condition to maintain payment level is not required of other inpatient facilities at this time.)
The change to PPSs for inpatient care had a significant effect on clinical laboratories. Until 1984,
hospital laboratories generally were recognized as profit centers. However, the institution of
prospective payment based on DRGs and similar capped payment rates transformed inpatient
laboratory testing into a cost center, creating incentives to reduce the number of tests ordered as
well as to shift inpatient care to the hospital outpatient and ambulatory care settings.4 When
DRGs were introduced, CMS assumed that the mean cost attributable to the laboratory as a
proportion of the revenue generated by DRGs would not exceed 9.5%.60
The proportion of payment attributed to laboratory tests is mediated, in part, by the patient’s
health condition. A 2000 report of a study that examined 486 DRGs drawn from a large
University HealthSystems Consortium database found that the proportion of payment associated
with laboratory tests and services averaged 6% for surgical DRGs and 9% for other DRGs
designating the management of medical conditions.61 The highest proportion of total costs
attributable to the laboratory was 18.3% for acute leukemia, 13.6% for HIV with surgical
procedure, and about 8 to 10% for various transplant procedures. Tests for kidney and urinary
signs and symptoms in children also were higher than average. Median laboratory costs were
i

Separate Medicare PPSs have been developed for other for health care facilities, including skilled nursing facilities,
long-term care, hospice care, inpatient psychiatric care, home health and inpatient rehabilitation.

May 2008

323

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

<1.0% for only 15 DRGs. Since the publication of this study in 2000, there has been little research
on the contribution of laboratory costs to total costs or the effect of variations in laboratory costs
on quality of care.
In August 2000, CMS began using an outpatient prospective payment system (OPPS) for outpatient
hospital care in order to restrain cost increases. This system provides a fixed, prospectively
determined, bundled payment for products and services provided by hospitals in outpatient
settings, excluding those services provided by physicians and other health care providers.62 Similar
to DRGs, the OPPS classified services into ambulatory payment classifications (APCs). All services
within a given APC are assumed to be clinically comparable and have comparable resource
requirements, and therefore, have the same payment rate. APCs pertain to laboratory tests,
implantable devices, items used in diagnostic radiography, and durable medical equipment (DME)
associated with hospital outpatient services.63 Physicians’ fees, including those for certain
pathologist-related consultative services, are paid through the Part B fee schedule. (Splitting of
laboratory fees into physician fees and “technical components” paid differently is discussed in the
next section.)
Along with bundling, the move to OPPS significantly affected the ways that independent
laboratories can bill for their services. Independent laboratories that provide pathology services
to registered hospital outpatients must bill the hospital directly for the technical component. The
hospital receives payment from Medicare under Part A and the independent laboratory receives
payment from Medicare for pathologist fees.64 Laboratory professional organizations have
opposed the OPPS, citing concerns about administrative and financial burdens on hospitals and
clinical laboratories.65 Stakeholder actions prompted the inclusion of an exception in the Benefits
Improvement and Protection Act of 2000.66 The legislation allows independent laboratories with
contracts in place by the date of the proposed rule-making to continue to bill Medicare directly for
technical component services.
Health care facility costs associated with blood banking and transfusion medicine are paid via the
inpatient and outpatient PPSs. Fees for services associated with end stage renal disease are paid
via the outpatient dialysis services payment system.67 This system uses a composite rate that is
intended to cover the bundle of services, laboratory tests, certain drugs, and supplies that are
routinely required for dialysis treatment. Providers must bill Medicare separately for certain
injectable medications and laboratory tests that are not included in the bundle.
Other government payers, i.e., Medicaid, TRICARE, VHA, use the Medicare DRGs, with some
modifications, for payment of inpatient and hospital outpatient services for their respective
beneficiaries.68-70 However, Medicare Part C (in which CMS contracts with health plans to
provide care to Medicare beneficiaries), TRICARE, and the Federal Employee Health Benefits
have switched from PPSs to competitive bidding contracts.71
The change to PPS by Medicare prompted a similar move by private sector. Private payers
typically use one of several PPS methodologies for inpatient and outpatient hospital settings, such
as all-inclusive case rates, per diem rates, and DRGs.10 As is the case with the Medicare PPS, allinclusive rates used by private payers do not provide separate payment for each resource used.
These PPSs rely on their ability to capture or accurately estimate how much each resource costs in
order to account for it when determining payment rates. Private payers usually use the Medicare
DRG groupings, but will assign their own relative weights to the individual DRGs. While the
May 2008

324

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

payment amounts negotiated between the hospital and the payer have been proprietary
information, at least one major private payer recently announced that it will make public the
actual payment rates negotiated with physicians.72, 7372, 73

Fee-for-Service Payment Systems for Ambulatory Care
Public and private sector payers cover ambulatory care and other services such as laboratory tests
according to predetermined, fixed fee schedules, negotiated contracts, or competitive bidding
contracts.
A fee schedule is a list of allowable fees representing the average or maximum amount that the
payer will reimburse providers for the service.6969 HCPCS codes, including CPT codes, describe
specific tests or services and link the service to the fee schedules. Medicare uses three fee
schedules under Part B to provide payment for a given service, procedure, or item. Most private
sector payers base their fee schedules on the Medicare schedules.
(1) Medicare physician fee schedule (MPFS). This fee schedule covers physician and other
licensed health care practitioner services, including office visits, surgical procedures
and certain laboratory services that require professional interpretation, such as
anatomic pathology tests and certain gene-based, molecular or similarly complex tests.
Physician services can occur in different settings. Determinations of the payment rate
are based on three aspects affecting physician services: physician work, practice
expense (all costs other than physician time required for the physician to provide the
service), and malpractice expense.
(2) Clinical laboratory fee schedule (CLFS). Medically necessary diagnostic and monitoring
laboratory tests and, with recent legislation, certain preventive and screening tests, are
reimbursed according to fee schedules differentiated by geographic region and the
national limitation amounts (NLAs). Testing and services may be furnished by
hospital, independent, or physician office laboratories, which bill Medicare directly for
tests performed. Technologies covered under the CLFS include diagnostic test kits and
reagents, devices that analyze test results, and other laboratory equipment essential to
testing.
(3) Durable medical equipment, prosthetics, orthotics, and supplies (DMEPOS). Medicare pays
for non-implantable DMEPOS through this fee schedule, although some types of
equipment are paid on a reasonable cost basis, such as dialysis supplies and
equipment. While the DMEPOS fee schedule does not directly affect payment for
laboratory services, cost containment strategies used for DMEPOS are being examined
to cut costs associated with the CLFS.

Using Fee Schedules for Laboratory Tests and Services
Use of the MPFS, CLFS, and other payment methods varies according to test type. Payment for
anatomic pathology tests and certain clinical pathology and highly complex molecular or genebased tests have two components—a professional component and a technical component. The
professional component covers the costs of interpretive consultation when the pathologist
discusses test results with the patient’s clinician. The technical component covers specimen
collection, transport, processing, preparation (e.g., centrifugation, tissue block cutting), analysis
(e.g., automated, microscopic exam), and storage.

May 2008

325

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

While all covered anatomic pathology tests are afforded payment for consultative services, only a
few clinical pathology and molecular and genetic laboratory tests can be reimbursed for
consultations. For anatomic pathology tests, both the professional and technical components are
paid according to codes on the MPFS. For clinical pathology, including molecular and gene-based
testing, both components are paid via codes on the CLFS.

Medicare Physician Fee Schedule
The MPFS is a set of physician services and payment rates tied to CPT codes. A resource-based
relative value scale is the methodology used to classify and calculate the physician payment rates.
Physician payments in the fee schedule are calculated by ranking medical services (as defined by
CPT and other HCPCS codes) according to the relative costs of resources required to provide
them. Physicians who treat Medicare patients are paid for their services through the MPFS,
including those providing inpatient, hospital outpatient, and other ambulatory care.j This
includes pathologists associated with laboratory tests and services.
Payment rate calculations involve several steps. First, for each CPT code, CMS assigns a relative
valuek that is weighted based on three factors: (1) the amount of work required to provide the
service (e.g., differences in time and intensity of work between a physician exam and surgical
procedure), (2) expenses related to maintaining a practice (e.g., supplies, staff), and (3) malpractice
liability insurance costs. Each factor’s relative value is the national average determined through
periodic national surveys sponsored by CMS.l Second, the relative values given to each factor are
adjusted for geographic differences in input prices. Separate geographic practice cost indexes are
used for this purpose. Third, the sum of the relative values (of the three factors) is multiplied by
the standard dollar amount (the fee schedule’s conversion factor) to arrive at the payment amount
for a particular service.74
Unlike the CLFS, the MPFS is updated regularly. The relative values are updated every five years
at a minimum. The conversion factor updates payments for physician’s services annually
according to a formula-based sustainable growth rate (SGR).74 The SGR sets a targetm for growth
in Medicare spending on physician services (per volume growth) that is no greater then growth in
the national economy, after accounting for percent changes in enrollment in the Medicare
program.75 The annual update to the MPFS depends on comparison to the target. If spending is
below the target, the fee update for the next year is increased. Conversely, if actual spending
exceeded the target, the fee update is reduced. If spending exceeds the target by a large enough
amount, fees will be cut for one year, up to a maximum of 7% below the underlying rate of
inflation. Since 1995, fee increases have averaged 2.5-3.0%.
For most physician services, Medicare pays 80% of the MPFS amount and the beneficiary is responsible for the
remaining 20% as coinsurance. Additional modifiers may be used to adjust the payment depending on the type of
service provided, type of clinician providing the service, and whether or not a physician participates in the Medicare
program or is working in an underserved area.
k The relative value unit is a unit of measure designed to permit comparison of the amount of resources required to
perform various provider services by assigning weights to such factors as personnel time, level of skill, and
sophistication of equipment required to render service.69
l Payments for physician assistants, nurse practitioners, and nurse midwives also are tied to relative value amounts.
m Four factors define the target: (1) long-term trend in U.S. real gross domestic produce per capita; (2) growth in the
Medicare fee-for-service population; (3) increases in Medicare fees (payment rate, conversion factor); and (4) the
impact of changes in law and regulation (e.g., additional costs due to expansion of benefits to include more
screening services).23
j

May 2008

326

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

In recent years, overall growth in Medicare spending on physician services has significantly
exceeded the SGR target, although growth in spending on laboratory tests during 2000-2004
(10.8%) was consistent with that of all SGR-covered services (11%).23 Steep physician fee cuts
were planned for 2003. However, after strong opposition by physician groups, Congress agreed
to consecutive, conservative increases of 1.5% for 2004 and 2005. More recently, the planned cuts
of 4.4% for 2006 were replaced with a one-year price freeze. Given the multitude of factors
affecting Medicare’s sustainability, continued use of the SGR in its present form could result in
physician fee cuts of 4-5% per year (the result of 2-3% inflation less the 7% penalty, according to
one analysis.23 However fee cuts of this magnitude have only occurred once before in 2002. The
matter of revising the SGR formula remains a central issue pertaining to the MPFS.
CMS’ last estimate of laboratory practice expenses was reported in 2003.76 The supplemental
expense data for this estimate was obtained from a survey conducted by CAP of independent
laboratories reporting physician hours and expenses associated with anatomic pathology.n The
survey found that the total practice expenses in anatomic pathology per physician hour at
independent laboratories averaged $160.50. Although this amount was substantially higher than
the all-physician average practice expense per hour and was at the upper end of the distribution
of practice expenses per hour across all specialties, an objective auditor found that the data
submitted for the survey met formal CMS requirements for supplemental practice expense data.
On the basis of its review, the auditor recommended that CMS use these values in its calculation
of 2004 practice expense relative value units for the 2004 MPFS. This implies that the costs to
provide laboratory services are higher than for other medical disciplines.

Clinical Laboratory Fee Schedule
Independent laboratories, POLs, and hospital laboratories when functioning as reference
laboratories (except hospital outpatient services) receive payment for their services through
prospectively set payment rates. Payment rates are tied to laboratory services listed in HCPCS.
The main factors that influence payment include: (1) prevailing charges; (2) national limitation
amounts; (3) annual updates; and (4) entry of new technologies.
Prevailing charges. Medicare pays laboratories directly for services performed in the ambulatory care
setting through the use of the 56 fee schedules that coincide with geographic areas and designated
carriers that process claims. These fee schedule amounts differ from carrier to carrier. The original
fee schedules were established by Congress in 1984. Although the original intent was to move
toward a single, national fee schedule, efforts to do so have been repealed repeatedly over the years.
Payments for each laboratory test were set separately for each carrier’s geographic market based
on a percentage of the prevailing charges for 1983. Specifically, the 75th percentile of the carrier
customary charges defined the prevailing charge for a given area. The payment rate for hospital
laboratories was set at 62% of the prevailing charge (reduced to 60% in 1987), while the rate for
independent laboratories and POLs was set at 60% of the prevailing charges. Because some
carriers’ charges were significantly lower, the rules also include a provision mandating that all
laboratories be paid the lower of submitted charges or the fee schedule rate.

n

The survey asked respondents to exclude physician hours and expenses related to clinical pathology services.

May 2008

327

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

National limitation amounts. Beginning in 1986, Congress established upper limits on laboratory
payment rates, known as NLAs, at 115% of the median of all carrier rates for each test. Since their
institution, NLAs have been reduced seven times, and have remained at 74% of the median for
the past decade.12 The actual payment paid to laboratories is the lowest of the providers’ charge,
carrier’s fee schedule amount, or NLA. Most laboratories are paid at or close to the NLA.
According to the 2000 IOM report, 85% of all pricing amounts across the 56 carriers and more
than 1,100 test codes were subject to NLAs in 2000. Further, these pricing amounts applied to a
disproportionately greater number of Medicare beneficiaries. As such, NLAs appeared to
constrain more than 98% of Medicare’s laboratory spending, making carrier fee schedules and
provider charges of minimal relevance to Medicare. Therefore, as noted by the IOM, “in practical
terms there is now a de facto single fee schedule.”4 This constrained fee schedule remains largely
stagnant, falling behind price increases applying to other elements of health care in the U.S.
An exception to the NLA determination was made in the Benefits Improvement and Protection
Act of 2000. Congress approved setting the NLA at 100% of the national median for tests for
which the cap was set on or after January 1, 2001.6 To date, CMS has applied this provision to 12
diagnostic and screening codes associated with Pap tests.
Fee updates. In 1984, the initial CLFS included a mechanism for annual inflation adjustments
consistent with the Consumer Price Index (CPI) for all urban consumers, determined by OMB.
However, after a few years, the updates were reduced to a rate less than the CPI or zero, then
eventually eliminated altogether.4 Following an update in 1997, payment rates for laboratory tests
were frozen from 1998 to 2002, and a modest 1.1% inflation update was provided in 2003. The
MMA of 2003 cancelled a scheduled 2.6% update for 2006 and enacted another five-year freezeo
for 2004 through 2008.6 A longitudinal comparison of the fee updates for laboratory and other
services are provided in Table 8.1. This comparison indicates that the 1995-2007 cumulative and
average increases for the CLFS were 6.4% and 0.48% as compared to significantly higher increases
for all other updates indexes, including 39.4% (2.6%) for CPI-U, 66.3% (4.0%) for CPI-U Medicare,
48.6% (3.1%) for the Medicare inpatient hospital basket, 28.5% (3.2%) for Medicare outpatient
hospital care, and 42.3% (2.8%) for the MPFS.
New technologies. For newly developed tests considered to be similar to existing tests, CMS
assigns a payment rate based on the rates of the similar existing tests, a process known as “crosswalking.”12 If a test is considered to be a truly novel or breakthrough technology for which there
is no existing similar test, CMS relies on carriers to independently set rates for the first year of use.
Carriers assign a new code and use data from manufacturers, other carriers, or other information
to determine appropriate payment levels for the test along the range of existing payments, a
process known as “gap-filling.” In turn, CMS sets the NLA for new technologies at 74% of the
median rate of all carriers. (This process and corresponding issues are discussed below in the
section on coding.)

o

The latest five-year freeze (2004-2008) was established under MMA of 2003 as an alternative to a 20% co-payment
requirement for Medicare beneficiaries, whose likelihood of being collected was considered to be low by many
laboratories.10

May 2008

328

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

Negotiated Contracts and Competitive Bidding
Increasingly, federal payers are contracting with private payers (local and regional plans) to offer
coverage to their beneficiaries. For example, the Medicare Part C plans (Medicare Advantage) are
private sector health plans approved by Medicare to provide Part A and Part B benefits to
enrollees who choose this option. 77, 78 Under Part C, Medicare negotiates contracts to make
monthly payments to the private plans.78 Medicare pays private plans according to the categories
under which they fall: local plans or regional plans.
However, in 2006, CMS implemented a system of competitive bidding to set payments for new
contracts with preferred provider organizations (PPOs) as required under MMA of 2003. Bidsp
made by private plans to offer Part A and Part B coverage to Medicare beneficiaries can directly
influence Medicare payment rates. That is, payment rates will be based on the relationship
between the private plan’s bid rate and the benchmark (which is the county-level payment rates
used to pay for Medicare Part C plans before 2006).q For plans whose standard bid exceeds the
benchmark, Medicare’s base payment rate is set equal to the benchmark and enrollees are
required to pay an additional premium that equals the difference between the bid and the
benchmark. For plans whose standard bid is below the benchmark, Medicare’s base payment rate
is set equal to the standard bid.r Methodology to determine benchmarks for regional plans is very
similar, although CMS uses a more complex formula to calculate the benchmark. (See discussion
of competitive bidding further along.)

The bid represents the cost to cover an average, or standard, beneficiary; it includes any plan administrative cost
and profit.
q County-level payment rates were at least as high as per capita fee-for-service Medicare spending in each county
before 2006 and were often substantially higher.
r Medicare also pays a rebate to plans that bid below the benchmark, which is defined by law as 75% of the difference
between the plan’s bid and its case mix-adjusted benchmark. The plan is required to return the rebate to
enrollees through supplemental benefits or by charging lower premiums.
p

May 2008

329

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

Table 8.1: Medicare pricing updates for certain provider types: percentage changes
1995-2006
1995

1996

1997

2006

2007

Cum.

Avg.

CPI-U (July)a

2.8%

3.0%

2.2%

1.7%

2.1%

3.7%

2.7%

1.5%

2.1%

3.0%

3.2%

3.2%

2.5%

39.4%

2.6%

CPI-U Medical
Care (July)±,b

4.4%

3.6%

2.7%

3.4%

3.5%

4.1%

4.5%

4.9%

3.8%

4.5%

4.2%

4.0%

4.3%

66.3%

4.0%

Medicare
Inpatient
Hospital Mkt.
Basketc

3.1%

2.4%

2.0%

2.9%

2.5%

3.6%

4.1%

2.9%

2.95%

3.4%

3.3%

3.7%

3.4%

48.6%

3.1%

Cost-based system – no defined updates

3.6%

4.1%

2.9%

2.95%

3.4%

3.3%

2.2%

3.0%

28.5%

3.2%

Outpatient
Hospitald

1998

1999

2000

2001

2002

2003

2004

2005

Physician
Updatee

7.5%

0.8%

0.6%

2.3%

2.3%

5.5%

4.8%

-4.8%

1.7%

1.5%

1.5%

0.2%

0.0%

42.3%

2.8%

Actual CLFS
Updatef

0.0%

2.9%

2.3%

0.0%

0.0%

0.0%

0.0%

0.0%

1.1%

0.0%

0.0%

0.0%

0.0%

6.4%

0.48%

Sources: a For data from 1995 to 2000, Medicare laboratory payment policy: Now and in the future. Washington, DC: Institute of Medicine, 2000.
2006 annual update for clinical laboratory fee schedule and laboratory services subject to reasonable charge payment. CMS manual system. Pub. 100-04 Medicare claims processing.
Baltimore, MD: Centers for Medicare and Medicaid Services, 2005.
2007 annual update for clinical laboratory fee schedule and laboratory services subject to reasonable charge payment. CMS manual system. Pub. 100-04 Medicare claims processing.
Baltimore, MD: Centers for Medicare and Medicaid Services, 2006.
b

Section 1834(h)(4)(A) of the United States Social Security Act. Payment for durable medical equipment.

c

Medicare program; Changes to the hospital outpatient prospective payment system and calendar year 2006 payment rates; Final rule. Federal Register 70, no. 217 (November 2005):
69516. Medicare program; Hospital outpatient prospective payment system and CY 2007 payment rates; CY 2007 update to the ambulatory surgical center covered procedures list; Medicare
administrative contractors; and reporting hospital quality data for FY 2008 inpatient prospective payment system annual payment update program—HCAHPS survey, SCIP, and mortality. 42
Code of Federal Regulations, parts 410, 416, 419, 421, 485, and 488.
d

2005 annual report of the Boards of Trustees of the Federal hospital Insurance and Federal Supplementary Medical Insurance trust funds. Washington, DC: Boards of Trustees of the
Federal Hospital Insurance and Federal Supplementary Medical Insurance Trust Funds, 2005. ; 2006 annual report of the Boards of Trustees of the Federal hospital Insurance and Federal
Supplementary Medical Insurance trust funds. Washington, DC: Boards of Trustees of the Federal Hospital Insurance and Federal Supplementary Medical Insurance Trust Funds, 2006.
2007 annual report of the Boards of Trustees of the Federal hospital Insurance and Federal Supplementary Medical Insurance trust funds. Washington, DC: Boards of Trustees of the Federal
Hospital Insurance and Federal Supplementary Medical Insurance Trust Funds, 2007.
Consumer price index—All urban consumers. Washington, DC: Bureau of Labor Statistics, 2007.
e

Medicare program; Hospital outpatient prospective payment system and CY 2007 payment rates; CY 2007 update to the ambulatory surgical center covered procedures list; Medicare
administrative contractors; and reporting hospital quality data for FY 2008 inpatient prospective payment system annual payment update program—HCAHPS survey, SCIP, and mortality. 42
Code of Federal Regulations, parts 410, 416, 419, 421, 485, and 488. Medicare program; Changes to the hospital outpatient prospective payment system and calendar year 2006 payment
rates. Federal Register 70, no. 217 (November 2005): 68516.
History of the Medicare clinical laboratory fee schedule. Washington, DC: Prepared by Avalere Health for the American Clinical Laboratory Association, 2005.

May 2008

330

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

Medicaid Payment Methodologies
In general, Medicaid payment rates are determined by individual states; however, payment for
laboratory services may not be set higher than the Medicare NLA. According to the Kaiser
Family Foundation’s online Medicaid benefits database, 49 states use a fee-for-service payment
methodology to pay for laboratory services and one state (Alabama) uses a reasonable charge
methodology.79 Specific data indicate that nine states use the NLA as the payment rate, two states
use a percentage of the NLA as the payment rate, and other states use some combination of these
calculations depending on whether the test is a high or low volume procedure. Eleven states
currently require a co-payment for laboratory testing and x-ray services received outside a
hospital or clinic.

TRICARE Payment Methodologies
The MHS provides direct care to beneficiaries and allows them to choose an option to receive care
from civilian providers. TRICARE Prime is a managed care option, TRICARE Standard (formerly
known as CHAMPUS) is a fee-for-service option, and TRICARE Extra is similar to TRICARE
Standard but offers discounts when beneficiaries use network providers.80, 81 TRICARE has
negotiated contracts with approved managed care providers that accept the negotiated rate as
payment in full. In 2002, TRICARE issued a request for proposals for new contracts through
competitive bid.71
For laboratory services, TRICARE pays either the rate on the Medicare CLFS or the contract rate
negotiated by HMOs and PPOs, whichever is lower.s,82 Three regional fiscal intermediaries have
been contracted to administer payments for services accrued by TRICARE beneficiaries at nonmilitary health care facilities.83
Payment and contract-related information could not be obtained for the Veterans Health
Administration.

Private Sector Payment Methodologies
Private payers often use Medicare’s MPFS and CLFS for setting their own payment rates. For
example, a payer may pay a multiple or percentage of the rate designated for a CPT or other
HCPCS code on the Medicare fee schedule (e.g., multiple of 1.2=120% or 80% of the CLFS).
Private insurance companies also may use other methods to determine payment rates, such as a
percentage of the laboratory’s actual charges (e.g., an 80% payment for a laboratory charge of $100
for a test) or predetermined rates for services negotiated via contracts with employers or
laboratories participating in their network. Private payers also may negotiate rates with
employers or participating laboratories, that become contractually binding.39 Unlike Medicare’s
fee schedule, which is publicly available, data on payment rates and methodologies used by
private payers are generally not publicly available.
In addition to Medicare business, private sector payers also negotiate contracts independently with
clinical laboratories. Two such contracts were awarded recently. In October 2006, United
s

TRICARE uses an adjustment factor equal to the ratio of the local average charge (standardized for the distribution
of clinical laboratory services) to the national average charge for all laboratory services during the base period to
establish laboratory service local maximum allowable charges.

May 2008

331

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

Healthcare awarded an exclusive ten-year managed care contract to Laboratory Corporation of
America (LabCorp) worth an estimated $3 billion.84 Along with providing laboratory services to
United’s 28 million members, LabCorp is obligated to develop a series of laboratory networkst in
selected regions of the country.6 The goal of the contract was to cut a minimum of 15% to 20% off
United’s over $2 billion per year laboratory testing expenses (including clinical and anatomic
pathology outpatient and outreach testing.) Also at that time, Aetna awarded an exclusive contract
to Quest Diagnostics. While the financial details of the contracts are not available, a news article
quoted laboratory industry professionals who speculated that these contracts provide the insurance
companies with testing services at costs that are as much as 45%–55% less than what Medicare pays
for the tests.85, 86

Outstanding Issues with the Current Payment Systems
The 2000 IOM report, the 2005 report prepared by The Lewin Group, and others have concluded
that the Medicare payment policy for clinical laboratory services is outdated and inadequate, and
could inhibit beneficiary access and stifle innovation in testing technology.4, 10, 87, 88 The IOM
Committee on the Medicare’s Payment Methodology for Clinical Laboratory Services evaluated
the current methodology against five desirable goals of a payment system:

ƒ

Beneficiary access to services on a timely basis

ƒ

Flexibility to promptly recognize and determine fair payment for new technologies

ƒ

Transparency in processes for setting payment policies and amounts that are
understandable and open to input from the public and providers

ƒ

Value that reflects efficient and appropriate use of laboratory services to support
positive health outcomes and quality of care, and eliminate fraud and abuse

ƒ

Administrative simplicity and efficiency of the system for the provider, payer, and patient4

The IOM committee found significant shortfalls toward all of these goals except beneficiary
access. However, other experts, including some in government, assert that access remains limited
for screening purposes and nonexistent for predictive purposes.35, 47 The IOM committee
developed 12 recommendations to begin redesigning the current methodology toward a more
rational approach for attaining the five goals. Only one of the recommendations has been
implemented since publication of the IOM report in 2000—improved transparency in the
participation of stakeholders in annual meetings of the CMS Council on Technology and
Innovation to assess the relationship between current CPT codes on the fee schedule and new
testing technologies.89

Multiple Fee Schedules Add to Administrative Burden and Result in Payment
Inconsistencies
The complexity and inefficiency resulting from use of 56 different fee schedules was cited as a
major problem in the 2000 IOM report. The administrative value of the original fee schedule
system has been greatly diminished since many of the individual test fees on the schedules are
now close to the NLA.4 Except for national coverage determinations, the use of different fee
t

LabCorp will reimburse United Healthcare $200 million for the first three years of the contract for transition costs
related to developing the expanded network in certain local markets.

May 2008

332

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

schedules and corresponding carrier processes for coverage determinations contributes to
administrative burden and inefficiencies for all parties involved.
Because the original allowable payments set in 1983 were not linked to laboratories’ relative costs
associated with performing tests or adjusted for inflation, some fees are most likely low relative to
costs while others may be high.10 Such payment variations are arbitrary, and payment disparities
for new tests that are integrated into the existing system often are compounded by other preexisting variations. For tests where there is not convergence with the NLA (an estimated 25-30%
of payments), wide variations in rates among carriers add to the challenges of setting appropriate
payment levels.90 Variations in 2007 fee schedule payments for the top 15 clinical laboratory
procedures (by allowable charges) are listed in Table 8.2. Payment variations are as high as 40%
for complete blood count and white blood cell count tests and 54% for a urine culture.90
Based on principles of inherent reasonableness, Congressu gave CMS the authority to modify
payment levels for Part B services it considered grossly inappropriate (excessive or deficient) by
as much as 15% annually without using public notice and comment procedures.10 To date, CMS
has not employed this authority. The authority notwithstanding, this method of payment
adjustment may be impractical, especially as the number of laboratory tests on the market
continues to increase.

u

CMS was given this authority under the Balanced Budget Act of 1997.

May 2008

333

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

Table 8.2: Variations across the Medicare Carrier Fee Schedules for the
Top 15 Laboratory Tests Ranked by Allowable Charges
2006
Ranked by
Total
Allowed
Charges in
2006

HCPCS
Code

1

Short Description

NLA

2006
MidPoint

85025

Complete CBC
w/auto diff WBC

$10.86

$14.68

$6.50

$10.86

$4.36

40%

2

84443

Assay thyroid
stimulating
hormone

$23.47

$31.72

$21.98

$23.47

$1.49

6%

3

80061

Lipid panel

$0.00

$0.00

$13.69

$18.72

$5.03

27%

4

80053

Comprehensive
metabolic panel

$14.77

$19.96

$11.74

$14.77

$3.03

21%

5

36415

Routine
venipuncture

$0.00

$3.00

$3.00

$3.00

$0.00

0%

6

83036

Glycosylated
hemoglobin test

$13.56

$18.33

$9.77

$13.56

$3.79

28%

7

83970

Assay of
parathormone

$57.67

$77.93

$57.67

$57.67

$0.00

0%

8

85610

Prothrombin time

$5.49

$7.42

$4.44

$5.49

$1.05

19%

9

80048

Basic metabolic
panel

$11.83

$15.98

$8.93

$11.83

$2.90

25%

10

84153

Assay of PSA, total

$25.70

$34.73

$20.28

$25.70

$5.42

21%

11

82728

Assay of ferritin

$19.03

$25.72

$12.22

$19.03

$6.81

36%

12

G0103

PSA screening

$25.70

$34.73

$20.28

$25.70

$5.42

21%

13

87086

Urine culture/
colony count

$11.28

$15.24

$5.24

$11.28

$6.04

54%

14

82607

Vitamin B-12

$21.06

$28.46

$14.56

$21.06

$6.50

31%

15

83550

Iron binding test

$12.21

$16.50

$7.58

$12.21

$4.63

38%

Minimum

Maximum

$
Difference

%
Difference

Source: List of top 100 lab procedures by carrier in descending order by allowed charges. From the 2006 BESS procedure data
completed year. Baltimore, MD: Centers for Medicare and Medicaid Services, 2007.
Clinical diagnostic laboratory fee schedule, 2006. Baltimore, MD: Centers for Medicare and Medicaid Services, 2007. History of the
clinical laboratory fee schedule. Washington DC: Prepared by Avalere Health for American Clinical Laboratory Association, 2005.

May 2008

334

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

Insufficient Research on Alternatives to the CLFS
In 2000, the IOM committee recommended replacement of the CLFS with a single national fee
schedule based on a methodology similar to that of the MPFS. The building blocks for this system
would be a relative value scale; adjustments for laboratory, beneficiary, or other characteristics,
including geographic location; a dollar conversion factor; and periodic updates. On an interim
basis, relative value payments could be calculated using the current NLA.4 This would provide
time for more rigorous cost-based analyses of alternative methods for gathering data to be used in
the calculation of the relative values. The committee identified four approaches worthy of further
study by CMS:

ƒ

Microcosting studies to determine costs of individual procedures in order to set both the
relative value and the conversion factor

ƒ

Competitive bidding demonstration project to set the relative value (but not the
conversion factor)

ƒ

Negotiated fee demonstration project to set both the relative value and the conversion
factor

ƒ

Analysis of charges to set the relative value (but not the conversion factor)91

Among these approaches, negotiated rulemaking has proven to be a successful model for
updating the physician and ambulance fee schedules.10 Each alternative offers certain advantages
and disadvantages and may have varied implications for the laboratory medicine sector. Since
publication of the IOM report, one study of a charge-based relative value payment system has
been completed and plans for the competitive bidding demonstration project are underway
(following a three-year negotiation of terms with stakeholders). Both of these studies are
discussed in greater detail below.
Even though some of the proposed studies have not been completed, many stakeholders contend
that an entirely new methodology is needed rather than conversion of the current system to one
based on relative value. Among potential alternative approaches are payment based on evidence,
test complexity, episodes of care, and cost-effectiveness.

Study of charge-based relative value
In 2002, CMS funded a preliminary study that examined the use of charge data to determine relative
values of laboratory tests and compared payment levels across several hypothetical fee schedules in
selected carrier markets. All charge-based relative values (CBRV) also were compared to NLAbased relative values. Although it was not a systematic effort to evaluate relative values for
laboratory tests, this analysis found that payments for many tests would change under a chargebased approach, including some that would increase or decrease dramatically. A large number of
outliers resulted from the comparison of charge-based and NLA-based relative values. Calculations
of standard deviations indicated that payment for outlier procedures would either decrease by at
least 44% or increase by at least 82% (assuming a constant conversion factor) when a CBRV
approach was used compared to the NLA approach.91 Further analysis revealed that low-volume

May 2008

335

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

procedures accounted for a disproportionate share of outliers.v In addition, the study found that
there would be relatively little redistribution across procedure classes. However, at the level of the
individual procedure, differences would be significant for certain tests.w,91
Using a CBRV methodology offers advantages. Since it is based on available claims data, it is
relatively inexpensive and administratively straightforward. Supporters of this approach contend
that the correlation between charge data and costs is valid. In addition, CBRVs provide an
automatic and timely methodology for accommodating the need to set payments for new tests
and for updating payments on a regular basis.
The CBRV approach has a few weaknesses that would have to be resolved prior to adoption. First,
CBRVs would not help to set payment rates for automated test panels, which are currently bundled.
In the current system, automated test panels are paid based on the number of tests performed, the
rationale being that the marginal cost of performing an additional test is less than the average cost of
the set of tests. The methodology used in this study assumed that the charges associated with a test
are independent of other tests. If submitted charges also reflect bundling, then the CBRV
methodology could be applied, in principle. Second, because there are limited data on relative costs
to compare CBRVs, it was not possible to determine whether the CBRVs provide a good measure of
the relative costs. The investigators suggested the use of microcosting studies for this purpose.
Third, this study did not examine how to set payment rates for new technologies. Further, use of a
CBRV methodology would require adequate provisions to control overcharging.

Competitive Bidding Demonstration Project
Another CMS study to assess payment alternatives involves a competitive bidding
demonstration. Competitive bidding refers to cost containment mechanisms whereby providers
of a service or product submit price bids to a purchaser (e.g., Medicare). Based on predetermined
criteria and the proposed bids, the purchaser selects a winner or group of winners that will
provide the services or products at a set price for a set period of time. The goal of competitive
bidding is to secure a set of prices that reflect the cost of efficient production, including a normal
profit.4 A core assumption of competitive bidding is that competitors will reveal the minimum
price at which a sale is acceptable, obviating the need for extensive data collection. The approach
of competitive bidding has provoked significant controversy among stakeholders. While
government payers generally have advocated it, providers, patient groups, and other
stakeholders have opposed it.
Since the mid-1980s, the Medicare program has attempted to implement competitive bidding
demonstrations for payment of clinical laboratory services, health plans (Part C), drugs and
biologics (Part B, e.g., for end-stage renal disease), and DMEPOS (Part B).92 Only the latter two
have been implemented thus far. Laboratory industry and provider opposition to competitive
bidding for outpatient clinical laboratory services helped to persuade Congress to halt
implementation of any of these demonstrations. Similarly, there were four attempts to conduct
competitive bidding demonstrations for Part C services; Congress and the courts intervened and
CBRVs for independent laboratories were significantly different than CBRVs calculated for POLs, and the CBRVs for
five of the ten CMS regions included in the study differed considerably from the CBRVs calculated for the nation as
a whole.
w Charge-based payment would be at least 33% lower than the NLA-based payment for 15% of procedures, and it
would be at least 33% higher than the NLA-based payment for 28% of procedures.91
v

May 2008

336

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

the demonstrations were not initiated.93 However, as stated earlier, competitive bidding was
implemented in 2006 to set payments rates for new regional PPOs under Part C. In addition,
competitive bidding is now used by the Federal Employee Health Benefits program and
TRICARE.
CMS is proceeding with the competitive bidding demonstration project for laboratory services. The
attractiveness of competitive bidding derives from its potential for substantial savings to the health
system. By limiting existing firms’ market power, competitive bidding could encourage efficiency,
innovation, and lower costs. The competitive bidding demonstration project (1999-2002) for
DMEPOS reportedly reduced Medicare payments by $7.5 million and reduced beneficiary
payments by $1.9 million without a reduction in access (aggregate savings of nearly 20%).94, 95
Supporters expect substantially higher savings for laboratory services, estimated at $1.43 billion
over the 2007 to 2011 period (if a national competitive bidding program was implemented).96
Many experts believe that overpayment for laboratory services originated in the late 1980s, when
the fee schedule was established using payment rates provided by clinicians that had marked up
the real cost of the test and kept the difference. Thus, current prices on the fee schedule have no
substantial relationship to actual costs.86 The IOM recommended a demonstration project in
competitive bidding to gather information about needs for administrative resources, management
at the local level, and impact on beneficiaries and providers.4 Congress included a provision in
MMA of 2003 authorizing a competitive bidding demonstration project for clinical laboratory
services that would otherwise be paid under Medicare Part B fee schedule.97 Congress awarded a
task order contract in 2004 to initiate the demonstration. The demonstration project may provide
a more realistic assessment that can be used to identify those tests that are relatively expensive to
produce and those that are inexpensive. In addition, the data could be used to establish an initial
set of relative values for a new payment methodology. Whether CMS will use the data to develop
a relative value-based payment methodology or as the pricing mechanism outright is unclear.
Along with these strengths, the IOM Committee examined previous assessments of competitive
bidding methodology and expert opinion and identified certain weaknesses of this approach
when initiated via exclusive contracts. Specifically, competitive bidding initiatives that rely on
exclusive or selective contracting allow only those firms submitting winning bids to participate;
losing firms are barred from receiving any payment from these contracts during the time of the
procurement.4 Such arrangements could have significant impact on the financial health of
excluded laboratories and the structure of the industry as well as disproportionately disadvantage
certain segments of the market.
CMS will include the 358 HCPCS codes that represented approximately 99% of Medicare laboratory
testing by volume and payment in the demonstration project. However, in terms of other
parameters, the CMS contractor developed a highly exclusive model as outlined in Box 8.3.

May 2008

337

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

Box 8.3: Design of the Competitive Bidding Demonstration Project for Clinical Laboratory Services
•

The demonstration project will use two metropolitan statistical areas (MSAs) to define demonstration sites, and
cover all tests paid under Medicare except Pap tests, colorectal cancer screening tests, and new tests added to
the CLFS during the demonstration.

•

Independent, hospital, and physician office laboratories with $100,000 or more in annual Medicare Part B
payments for nonpatient services will be required to participate in the demonstration, though they do not need
to bid on coverage of the entire MSA.* Those that do not submit a bid will be barred from receiving any
Medicare Part B payments for the duration of the project.

•

A laboratory’s bids for individual tests will be weighted according to expected demonstration volume and
summed to form a single composite bid. The composite bids will be organized from lowest to highest and, along
with unspecified criteria, a “pivotal” composite bid will determine the winners and losers.

•

CMS will use the pivotal bids to set and freeze payment rates for the three years of the project. Laboratories
with a composite bid equal to or lower than the pivotal bid will be winners.

•

Laboratories with a composite bid higher than the pivotal bid will be losers, and will be barred from receiving
any Medicare Part B payments for laboratory services for three years.

•

To ensure quality, CMS will rely on winning laboratories’ compliance with CLIA regulations, PT data, and
reporting of data on standardized measures of turnaround time, log-in error rates, and physician satisfaction.
TAT measures include: (1) total TAT; (2) transport TAT; (3) processing turnaround time; (4) total turnaround
time for statim (STAT) testing; (5) reporting TAT for critical values; (6) reporting TAT for public health disease
notification. CMS also will review complaints received through a toll-free hotline for the demonstration.

* POLs and hospital outpatient testing are exempt, except where they function as an independent reference laboratory
earning Medicare payments of $100,000 or more per year.
Source: Report to Congress—Initial report on the Medicare Clinical Laboratory Competitive Bidding Demonstration. Baltimore,
MD: Centers for Medicare and Medicaid Services, 2006.

According to 1999 data, laboratories derive about 29% of their income from Medicare Part B
payments.4 Laboratories that are not selected in the competitive bidding process likely would be
subject to steep, immediate cuts in revenue for three years. Industry stakeholders contend that,
without the ability to offer laboratory services to Medicare beneficiaries, many local area
laboratories may have to close their business. Stakeholders have sought to revise the exclusivity
provision, allowing all interested CLIA-certified laboratories to continue providing services for
Medicare beneficiaries during the demonstration at the amount accepted as the winning bid.98-100
As another alternative, stakeholders have suggested that CMS simply indicate the percentage of
cost reductions needed for the Medicare program. However, CMS has maintained the initial
framework that bars losing laboratories from any Medicare payments.
Several other prominent issues with the current framework have been voiced by stakeholders. In
particular, industry representatives have stated that the $100,000 threshold is insufficient to
protect many laboratories that qualify as small businesses, such as those performing low volume,
highly complex testing as a reference laboratory or those that grow their business beyond the
threshold.98-100 For example, if a laboratory earns $100,000 or less at the time of the initiation of the
competitive bid and elects not to participate, that laboratory will be penalized if, at any time
during the demonstration, Medicare payments exceed the $100,000 threshold.101 The penalty is
the forfeiture of any further Part B Medicare payments for the remainder of the demonstration.
The demonstrations likely would force approximately 90% of laboratories in the designated areas
to participate. Many small laboratories that earn $1-2 million per year have expressed that they
May 2008

338

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

cannot afford to lose Medicare payments.102 With more hospital laboratories expanding outreach
testing programs, hospitals also are concerned about how the demonstration would affect their
financial status. Laboratories that serve as subcontractors or reference laboratories to other
laboratories that win the competitive bid also would be affected by the current design.
Supporters of the competitive bidding proposal disagree as to the financial effect on local
laboratories. Several contractors (i.e., winners) will be selected in each of the metropolitan
statistical areas, but being named a contractor does not guarantee business. Physicians can choose
among several laboratories; thus, even the winning laboratories must compete for business. In
theory, competition would increase overall quality of laboratory services. Also, if physicians
choose to obtain services from multiple laboratories, this may decrease the potential for loss of
Medicare market share among the winners.103 However, it is not yet known whether these market
effects will occur, and loss of market share may be sizable.
The extent to which competitive bidding of laboratory services affects beneficiaries and providers
also is unknown. Because the project confines the provider network, beneficiaries may be required
to travel long distances to obtain even basic testing, thereby reducing access to necessary services. A
small group of winning laboratories may not be able to accommodate all Medicare beneficiaries in
every setting in which laboratory services are needed. Such restrictions could have a detrimental
impact on continuity of patient care and, perhaps, health outcomes.104, 105 Potential problems with
the current framework could be accentuated in rural areas and for specialized patient populations.
In addition, patient preferences and patient satisfaction also were not incorporated into the
competitive bidding process or in its standardized measures for reporting on quality.
A laboratory industry stakeholder group, the Clinical Laboratory Coalition, has been seeking
legislative action to address the competitive bidding framework. The House Committee on Small
Business held a hearing on this matter in July 2007. Shortly thereafter, Representative Nydia M.
Velázquez (D-NY) introduced H.R. 3453 (110th Congress), The Community Clinical Laboratory
Fairness in Competition Act of 2007. The Senate version of the bill, S.2099, the Protecting Access
to Clinical Laboratory Services Act of 2007, was introduced by Senators Ken Salazer (D-CO) and
Pat Roberts (R-KS) in September 2007.

Additional Studies Needed
The IOM proposed microcosting studies using standard accounting practices to collect data on
direct costs and develop an appropriate basis for determining indirect costs.4 Microcosting (also
called activity-based costing) attempts to allocate itemized costs by identifying the components of
each individual cost.106 In the context of the clinical laboratory, microcosting determines the total
direct labor and supply costs that are required to produce a laboratory test and can serve as the
starting point to determine the total cost and ultimately the price of a test.107 Data acquisition
through microcosting studies is critical to the development of a relative value-based payment
system, and could contribute to periodic evaluations of payment appropriateness.
For its report, the IOM committee used a very small resource-costing study to gain an
understanding of service level costs. However, the study surveyors obtained only general
information.55 The lack of standard cost accounting systems for the laboratory’s costs separate
from the hospital and physician’s office, and the general reluctance of laboratories to reveal
sensitive, proprietary cost information may be challenges to conducting these studies. New

May 2008

339

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

software programs are making such analyses significantly and, as a result, an increasing number
of health care organizations, including clinical laboratories, are incorporating microcosting into
their overall management strategies.
A major advantage of microcosting is that it avoids or minimizes distortions in product costing
resulting from subjective allocations of indirect costs and thus generates useful data on how
money is being spent and whether an organization or entity is operating cost-effectively.108
Findings derived from microcosting studies could provide insight into whether public and
private payers are paying appropriate prices for particular laboratory tests and could be used to
educate clinicians on the true cost of ordering laboratory tests.4, 109 Because of their level of detail
and specificity, microcosting studies also allow others to see how well an analysis matches their
own situation, even where patterns of care may differ.110 The investment in carefully designed
microcosting studies, along with a standardized accounting mechanism, could yield the type of
high-quality data needed to redesign the payment methodology.

Reducing Fraud and Abuse
As in other sectors of health care, laboratory medicine is subject to fraud and abuse in areas
ranging from defrauding of payers to billing methods that combine legitimate claim information
with falsified information.111 Examples of common types of fraud in laboratory medicine include
billing for services that were not performed, ordered or needed; up-coding;x unbundling;y
duplicate billing; and falsifying diagnoses.
Several laws and regulations have been applied to reduce fraud and abuse in clinical laboratories.
Under the False Claims Act Amendments of 1986 (Public Law 99-562, 100 Statute 3153), any
person who knowingly presents or causes to be presented false claims for payment of
government funds is subject to a civil penalty between $5,000 and $10,000 plus three times the
amount of damages sustained by the government because of the act of that person.z,113 This act
has been the primary means by which the DHHS OIG and Federal Bureau of Investigation have
investigated fraudulent clinical laboratory billing practices.6 The Health Care Fraud and Abuse
Control Program, established by the Health Insurance Portability and Accountability Act of 1996,
is intended to coordinate federal, state, and local law enforcement efforts to prevent and disclose
health care fraud and abuse, including in laboratory services.114
The DHHS OIG has been involved in many actions and projects intended to curb clinical laboratory
fraud and abuse. For example, Project LabScam was the first nationwide law enforcement project to
occur in the medical field and applied to all major independent clinical diagnostic laboratories in the
U.S. It arose from information revealed during the investigation that led to the 1992 guilty plea of
National Health Laboratory and its agreement to repay $111 million.111 The Hospital Outpatient
Laboratory Project has documented hospital laboratory abuses related to test unbundling, double
billing of tests, and improper billing of medically unnecessary tests. With input from medical care
industries, the OIG has developed model compliance guidelines for laboratories for assisting
Up-coding, also referred to as up-charging, is the misuse of standardized codes (i.e., increasing the bill by
exaggerating or falsely representing medical conditions and serves provided) to obtain higher payment than
legally allowed.112
y In laboratory medicine, unbundling occurs when a laboratory bills separately for some or all tests that were
analyzed simultaneously by a single piece of equipment on a single patient specimen.111
z The False Claims Act does not encompass tax fraud.
x

May 2008

340

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

development of effective internal controls to detect and prevent fraud, abuse, and waste.115 Among
other aspects, the compliance plan includes written standards of conduct, policies, and procedures
that address potential fraud, designation of a chief compliance officer, and a hotline or other means
by which complaints can be received anonymously.
Government investigations related to clinical laboratory businesses conducted from 1992 to 2006
are estimated to have resulted in penalties exceeding $1.727 billion.6 Monetary penalties imposed
as a result of laboratory fraud and abuse represent a fraction of penalties imposed throughout all
health care sectors. In 2005, the federal government won or negotiated an estimated $1.47 billion
in settlements and judgments pertaining to health care fraud cases and proceedings.116 In 2006,
state Medicaid fraud control units recovered more than $1.1 billion in court-ordered restitution,
fines, civil settlements, and penalties and obtained 1,226 convictions.117 These units recovered
$709 million in 2005.118

Pod or Condo Laboratories
Another type of fraud-and-abuse pertaining to laboratory medicine is physician self-referral, which
is addressed by the Stark laws. Enacted under Section 1877 of the Social Security Act, the laws
prohibit physicians from referring Medicare patients for certain designated health services to an
entity with which the physician or a member of the physician's immediate family has a financial
relationship, unless an exception applies.119 When first enacted in 1989, the law applied only to
clinical laboratory services; however, in 1993 and 1994, Congress expanded application of the law to
10 other designated health services and financial arrangements involving physicians. In 2003,
Congress authorized certain exceptions in which physicians receive non-monetary remuneration
that is used solely to send and receive electronic prescription information. Additional clarification
and exceptions were defined in the phase III provisions of the law published on August 27, 2007.
However, the final rule did not fully address pathology-related self referrals (discussed below).120
The potential for clinical laboratory-related fraud and abuse arises from contractual joint ventures
that enable non-pathologist physicians and other entities to profit from self-referrals of pathology
services.121 These arrangements are often cited in connection with regulatory changes affecting
billing of anatomic pathology services as well as a loophole in how Medicare assigns benefits, which
allows an independent contractor physician or non-physician to reassign Medicare billing privileges
to a health care entity.121, 122 CMS plans to address these issues in a separate rulemaking.
Two types of these referral arrangements have arisen: “pod” or “condo” laboratories and
referring physician billing arrangements.123 Pod or condo laboratories are established by a
“manager” in a single office space that subdivides each room or cubicle into a separate, fully
equipped laboratory. Each “laboratory” is subleased to a physician group practice. The manager
hires technologists/scientists and technicians for each laboratory and one pathologist to supervise
all laboratory staff. In seeking to be technically in compliance with applicable exceptions to the
federal self-referral law, the staff rotate from one laboratory to the next, reviewing each group
practice’s slides. Each physician practice group compensates the laboratory at a discounted rate
for each slide reviewed, but bill payers for the entire pathology service. In referring physician
billing arrangements, a laboratory offers to perform anatomic pathology services for referring
physicians and bills such services at a discount; the referring physician then marks up the bill
from the laboratory and bills Medicare.

May 2008

341

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

These arrangements can create incentives for physicians involved in these arrangements to order
more laboratory tests or to perform biopsies and other anatomic pathology procedures that may not
be necessary, leading to increased and inappropriate utilization.120 Medical decision-making and
the quality of patient care may also be compromised.124 The potential profits to the test-ordering
physician are far greater than for clinical laboratory services. Pod or condo laboratories also may
violate the anti-kickback statute. Until the rulemaking on pathology services is completed, CMS has
emphasized that parties involved in shared arrangements in the same building must comply with
the in-office ancillary services exception in operation of their business, not just on paper.120
The proposed 2007 Physician Fee Schedule released by CMS included a series of proposals to
prevent pod or condo laboratories and other contractual joint ventures.125 CMS chose to wait to
address the issue in its final fee schedule, stating that it needed more time to study the issues
involved. The proposed 2008 fee schedule, published in July 2007, included a series of proposals
to prevent pod or condo laboratories; however, these proposals differ substantially from those
outlined in the previous 2006 proposed rule.126 Specifically, the proposed 2007 billing reforms
address self-referral and reassignment abuses by strengthening anti-markup restrictions on the
technical component of diagnostic services and by creating new anti-markup restrictions on the
professional component. Stakeholders are advocating adoption of these proposed rules.124

Limitations Based on Volume
Section 1128(b)(6)(A) of the Social Security Act grants the Secretary of DHHS power to exclude
from any federal health care program individuals or entities that have submitted bills or requests
for payments containing charges for items or services substantially in excess of such individuals’
or entities’ usual charges for such items or services.127 As such, a supplier or provider submitting
a claim to Medicare or to a state health care program that contains charges “substantially in
excess” of its usual charges may be excluded from participating in these programs.128
In 1990, 1997, and 2003, the OIGaa proposed regulations to provide guidance on the Medicare and
Medicaid programs’ exclusion authority for submitting claims that contain excessive charges.129
The 2003 proposed rule stated that individuals and entities (including clinical laboratories) could
be excluded from participating in federal health care programs if their charges or costs are more
than 120% of their usual charges or costs.bb
Several laboratory medicine organizations opposed the rule proposed by OIG. In June 2007, the
OIG announced that it would not proceed with the 2003 proposed rule, basing its decision on
public comments and lack of sufficient information required to establish a fixed benchmark for
substantially excessive charges or costs that could be applied across the health care sector.130
Despite its decision not to issue a final rule, OIG expressed concern about disparities in the
amounts charged to Medicare and Medicaid relative to private payers.cc

The Secretary has delegated this authority to the OIG.127
The proposed 2003 rule exempted physicians on the basis that their payment is based on actual costs and is
updated annually.129
cc In its decision not to promulgate the 2003 proposed rule, the OIG also stated its continued concern about disparities
between the amounts charged to Medicare and Medicaid and private payers.130
aa

bb

May 2008

342

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

CODING
Standardized coding systems are used to categorize claims for payment for health care services
and ensure that they are processed consistently and systematically.131 Coding systems use
alphanumeric nomenclatures to identify particular health conditions, services, or products. The
code assigned to a health condition, service, or product is linked to a payment amount
reimbursed to providers. For example, under Medicare, codes are linked to fixed payment
amounts via fee schedules. The coding systems that apply to laboratory medicine include CPT
codes and HCPCS codes for laboratory tests and services and ICD-9-CM codes for diagnoses.132
The CPT-4, published by the AMA, describes the professional services performed by physicians and
is widely used as the standard for outpatient and ambulatory care procedural coding and payment.
CPT codes fall within the larger national HCPCS, which was designed initially to represent services
provided by physicians and non-physicians to Social Security beneficiaries covered by Medicare,
currently is used by physicians to report services provided to Medicare and Medicaid patients.10, 132
In 2004, there were approximately 1,000 clinical laboratory and pathology codes listed in the 8000089399 CPT-4 code series.10 The process for updating these coding systems varies by organization.
The ICD is a product of the World Health Organization; however, CDC’s National Center for
Health Statistics in collaboration with other U.S. government agencies has developed a clinical
modification (ICD-9-CM) for use in the American health system. It is used to describe illnesses,
conditions, and injuries of people seeking medical services in the inpatient departments of
hospitals.35 Box 8.4 depicts the main characteristics of these coding systems.
The CMS HCPCS Workgroup comprises representatives of CMS, the state Medicaid agencies, and
the Statistical Analysis Durable Medical Equipment Regional Carrier.133 The workgroup is
responsible for considering each request for a change to a HCPCS level II national code at
regularly scheduled monthly meetings.dd
The CPT Editorial Panel, which consists of 17 members, 11 of whom are physicians nominated by
the National Medical Specialty Societies and approved by the AMA Board of Trustees, is
responsible for revising, updating, and modifying the CPT codes.ee,134 The Editorial Panel is
supported by the CPT Advisory Committee, primarily comprising physicians nominated by the
national medical specialty societies represented in the AMA House of Delegates. Applications for
new CPT codes are reviewed quarterly by the Editorial Panel and must be submitted by a specific
date each year. Because CPT coding changes are made effective only once each year for Category
I codes and twice per year for Category II and III codes, the review process for CPT coding
applications requires at least 8-15 months.10 Additional delays of 6-28 months can occur if the
medical specialty societies and other reviewers do not reach a consensus regarding coding
decisions within the appointed timeframe.
The AMA has recently made efforts to increase the transparency of the coding process. In order
to provide public stakeholders more time to comment on coding changes, the public release of
proposed changes was moved up from the fall to the summer of each year.10 The AMA also
dd
ee

Prior to 2006, the National Panel was responsible for final coding decisions.133
The seven other members of the CPT Editorial Panel consist of four physicians nominated from the Blue Cross Blue
Shield Association, America’s Health Insurance Plans, AHA, and CMS; one performance measures representative,
and two members of the CPT Health Care Professionals Advisory Committee (including one at-large member).134

May 2008

343

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

established the Pathology Coding Caucus to allow non-physician stakeholders to play a larger
role in the development of CPT laboratory and pathology codes and to review code revision
proposals.ff,35 The group reviews proposed new codes, suggests revisions to existing codes, and
develops consensus recommendations.
Box 8.4: Description of Payment Coding Systems
HCPCS
• Developed by HCFA (now CMS)
• Originally designed to represent physician and non-physician services provided to Medicare beneficiaries
• Federal government currently requires physicians to use HCPCS codes to report services provided to
Medicare and Medicaid patients
Level I
• Comprises CPT codes
• Used to report hospital visits, surgical procedures, radiological procedures, supervisory services, and
other medical services
Level II
• Known as national codes
• Developed by CMS to report medical services that are not covered in CPT
CPT-4
• Published and maintained by the AMA
• System for describing and reporting physician services in an outpatient setting
• Also used for planning outpatient services, benchmarking, assessing quality of patient services
Category I
• Codes for evaluation and management, anesthesia, surgery, radiology, pathology and laboratory, and
medicine
Category II
• Supplementary tracking codes for use in performance assessment and QI
Category III
• Temporary codes representing emerging medical technologies, services, procedures not yet approved by
FDA and not otherwise covered by CPT codes
Modifiers
• Supplementary codes that can be reported along with Category I codes to report additional information
about unusual circumstances under which a procedure was performed
• Meant to support the medical necessity of procedures that otherwise might not qualify for payment
ICD-9-CM
• Based on classification system developed and maintained by WHO
• Clinical modification published by CDC’s National Center for Health Statistics
• CMS, private payers require physicians and other medical providers to report ICD-9-CM diagnostic codes on
almost all payment claims
Diagnostic Codes
• Represent the reason why a patient is receiving medical care
Supplementary Codes
• Additional information about the patient and/or circumstances surrounding the patient’s illness or injury
Source: Smith GI. Basic CPT/HCPCS coding. 2006 Edition. Chicago, IL: American Health Information Management
Association, 2006.

ff

The Pathology Coding Caucus consists of representatives from the AMA, Advanced Medical Technology Association,
American Association for Clinical Chemistry, American Clinical Laboratory Association, ASCP, American Society of
Cytopathology, CAP, National Association of Medical Examiners, U.S. and Canadian Academy of Pathology,
CLMA, ASM, and American Association of Bioanalysts.35

May 2008

344

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

Inadequate Mechanism for Adding New Tests
As noted above, CMS incorporates new CPT codes for laboratory tests into the CLFS and
establishes payment levels for them using two primary methods: cross-walking and gap filling.135
Cross walking is used when a new laboratory test is deemed similar to an existing test, multiple
existing test codes, or a portion of a test code that already exists. When a new laboratory test is
cross-walked, it is assigned the related existing local fee schedule amounts and resulting NLA.
Gap-filling is used when it is determined that a comparable laboratory test does not exist. Under
this method, each Medicare carrier is provided with instructions to determine a payment amount
for its geographic area(s) to be used for the first year; these carrier-specific amounts are used to
establish a NLA for subsequent years. However, gap-filling is rarely used as a payment
methodology.10 Figure 8.1 depicts the basic payment pathways for gap-filling and cross-walking.
The process for updating codes applicable to the CLFS has undergone certain significant changes
over the past several years, some of which have been prompted by concerns that the CLFS is not
sufficiently subject to stakeholder feedback and that greater opportunity for clinical laboratories
and diagnostic manufacturers to provide input would increase the accuracy and efficiency of
complex coding determinations.10 A provision of the Medicare, Medicaid, and State Children’s
Health Insurance Program Benefits Improvement and Protection Act of 2000 mandated that the
public be given an opportunity to consult on payment determinations for new clinical laboratory
tests in a manner similar to the procedures established for implementing coding modifications for
ICD-9-CM.gg,135 As a result of this and the IOM’s 2001 recommendations, CMS has held a
“Laboratory Public Meeting” each year since 2002.137 These public meetings, notification of which
is given in the Federal Register, are intended to allow experts to provide input on the nature of the
new test codes and for CMS to receive recommendations regarding cross-walking and gap-filling.
Despite these changes and attempts at greater transparency, the coding system through which
new laboratory technologies are added remains inadequate, particularly for new and emerging
laboratory tests such as genetic testing. CPT codes used for billing genetic tests identify the test
procedure performed (e.g., reverse transcription), but are not specific to the condition being
evaluated (unlike codes for most tests which is disease specific).35 Thus, a new genetic test
performed with existing procedures receives payment under existing codes, rather than under a
newly assigned CPT code and payment rate. (Only novel genetic technologies and testing
procedures are assigned a new Category III CPT code.) One of the main criticisms in the use of
CPT codes for genetic tests has been the lack of specificity in the codes that limits the ability of
payers to make informed claim determinations.

gg

The ICD-9-CM Coordination and Maintenance Committee, a federal interdepartmental committee co-chaired by CMS
and the National Center for Health Statistics, is charged with maintaining and updating the ICD-9-CM system.136
Public meetings for discussion or education and proposed coding changes are held by the committee, presenting an
opportunity for organizations involved in the coding field, e.g., American Health Information Management
Association and AHA, as well as physician specialty groups, to voice their opinions on coding matters. The
committee ultimately creates recommendations that must be approved by CMS and the National Center for Health
Statistics.

May 2008

345

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

Figure 8.1: Gap-Filling and Cross-Walking Payment Determination Pathways
New Laboratory Tests

Gap-Fill

Cross-Walk

•New and innovative technology
•1 or more years to obtain new/modified code

•2 generation technology replacing older tests
•Map to existing code(s)
nd

New Payment Level Assigned

Obtain Payment for Existing Code

•Varying state fee schedules
•CLFS NLA

•May be associated with new payment

Payment

Update

Adjustment

•CLFS-CPI Update (infrequent)

Addition of New/Modified CPT Codes

Adapted from: The Lewin Group. Outlook for medical technology innovation: Will patients get the care they need? Report 2: The
Medicare payment process and patient access to technology. Washington, DC: AdvaMed, 2000.

CPT code modifiers have been criticized for being too vague to allow health insurance companies
to make well-informed coverage determinations, leading them to deny coverage or request
additional information. The Genetic Test Coding Workgroup, a consortium of genetics and
laboratory organizations founded by CAP in 2003, proposed a “numeric-alpha” coding section to
be added to existing 5-digit CPT laboratory codes used for genetic testing.hh,138 The AMA CPT
Editorial Board adopted these modifiers, which were included in the 2005 CPT Coding Manual.
This coding modification will not change the rates at which these codes are reimbursed.35
Many providers who supply medical services to Medicare beneficiaries argue that payment rates do
not correspond to the cost of a genetic test.35 Testimony provided to SACGHS in 2004 indicated that
the cost to one academic laboratory in Virginia to perform a genetic test for Fragile X syndrome was
$255, but was reimbursed only $68 for it. According to this laboratory, major health plans,
including both Medicare and Medicaid, reimbursed 60-90% of claims filed for genetic tests.
In its 2006 report on coverage and payment of genetic tests and services, SACGHS recommended
that, by 2009, when the freeze on laboratory test payment rates is lifted, DHHS should be ready to
revise payment rates to reflect the actual cost of genetic testing.35 SACGHS also recommended
hh

This coding system includes a numeral that indicates the disease category and a letter that denotes the gene type,
thereby conveying information about the nature of the test being billed.35

May 2008

346

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

that DHHS assess the adequacy of CPT E&M (Category I evaluation and management) codes and
allow non-physician health providers deemed qualified to provide genetic counseling services
who are currently billing incident to a physician to use the complete range of CPT E&M codes
that apply to genetic counseling.

Need for a Better Coding System
Some organizations have advocated the replacement of HCPCS, CPT, and ICD-9-CM with the
ICD 10th revision, Clinical Modification (ICD-10-CM) and ICD 10th revision, Procedure Coding System
(ICD-10-PCS).ii,140, 141 In contrast to the ICD-9-CM, the ICD-10-PCS includes a unique code for
each substantially different procedure, expandability to allow new procedures to be easily
incorporated as unique codes, a multi-axial structure with each code character having the same
meaning within a specific procedure section and across procedure sections, and a standardized
terminology in which each term has a unique and specific meaning. MMA 2003 includes
language that encouraged the Secretary of DHHS to proceed with developing and promulgating
rules to adopt ICD-10-CM and ICD-10-PCS.142
The transition to ICD-10-CM and ICD-10-PCS has been supported by the federal government and
prominent national health care organizations (e.g., AHA, Federation of American Hospitals, and
American Health Information Management Association).143 A 2005 study by the RAND
Corporation estimated that the conversion would cost $425 million to $1,150 million in one-time
costs, in addition to between $5 million and $40 million per year in lost coder and physician
productivity over 10 years following conversion.144 Nevertheless, RAND concluded that the
potential benefits of conversion outweighed costs. One of the major benefits of conversion to
ICD-10 would be its ability to more finely differentiate between new and old procedures,
allowing the value and applicability of new procedures to be more fully realized. Concerns about
the high cost of implementation, the magnitude of benefits, and other issues associated with
implementation have slowed the transition from ICD-9-CM to ICD-10 CM and PCS in the U.S.139
The current coding system also is inadequate for supporting and enabling implementation of HIT.
A coding system that provides more accurate, detailed clinical information capable of supporting
quality measurement and patient safety efforts will enable a smoother, more effective transition to
EHRs and other electronic storage and transfer of health information. Widespread adoption of
EHRs and interoperable information networks depends on classification systems that can
summarize and report data.142 The capacity to integrate standardized laboratory data into nextgeneration clinical practice applications is critical to establishing a national health information
network. For example, according to some HIT experts, U.S. efforts to invest in and promote use
of SNOMED-CT, a comprehensive, multilingual clinical health care terminology developed
jointly by the National Health Service in England and CAP, will be undermined if the U.S. fails to
adopt ICD-10-CM and ICD-10-PCS.142, 145 The Health Information Technology Promotion Act of
2006 (H.R. 4157), passed by the House in July 2006 but not signed into law, called for the
implementation of the ICD-10 coding system by October 2009.146

ii

ICD-10 has been used for mortality classification in the U.S. since 1999 and by health systems in much of the world
since the mid-1990s.139

May 2008

347

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

CONCLUSIONS
The design and updating of coverage, coding, and payment systems should strive to enable
patient access to medically necessary care, support delivery of high-quality care, and sustains
innovation of new technologies. Further, they should discourage inefficiency, fraud and abuse,
and non-competitive practices. However, these systems can pose significant barriers to achieving
these ends in laboratory testing. Changing demographics and disease patterns in the population,
corresponding increases in utilization and expenditures, and attributes of emerging technologies
are intensifying the challenges to the current laboratory services payment system.
Medicare is the single largest payer in the country, accounting for 29% of all revenues for
laboratory services. All public payers and approximately 67% of private payers use Medicare’s
payment methodologies as the basis for their own and as a tool for negotiating discounts with
providers. As such, suboptimal practices and other shortcomings in the Medicare reimbursement
system pertaining to laboratory testing affect other public and private sector payers in the U.S.
health system.
Key reimbursement challenges to laboratory medicine include the following:

ƒ

Medicare’s statutory restriction of coverage for screening tests and related preventive
services remains a shortcoming in the scope of benefits for Medicare beneficiaries.
Adding preventive services to Medicare benefits on a case-by-case basis via the
legislative route is cumbersome and impedes access to certain proven, beneficial tests.
Legislation is needed that would expand Medicare benefits to include such preventive
services that are evidence-based and determined to be reasonable and necessary for
prevention and detection of illness or disability among Medicare beneficiaries.

ƒ

Continued use of 56 different fee schedules is inefficient and unnecessarily complex. For
certain commonly ordered tests, the multiple schedules result in large regional
variations, while for other tests, NLAs constrain Medicare payment rate variations.

ƒ

There is a notable lack of reliable data on the relationships among historical costs on
which the CLFS is based, current production costs, and the effects of economies of scale
and other cost-reducing effects of technological changes.

ƒ

Studies of data-derived methods for evaluating the appropriateness of payment rates
and for designing of potential new payment systems, such as resource-based relative
value, microcosting, and negotiated rulemaking, have not been completed.

ƒ

CMS is proceeding with a competitive bidding demonstration project for laboratory
services, with the expectation of substantial savings. Supporters of the project believe
that current prices on the fee schedule have no substantial relationship to actual costs;
thus competitive bidding may provide information about resources and costs. However,
the project model is highly exclusive and could have significant, detrimental effect on
clinical laboratories that loose in the bidding process since many depend on Medicare
reimbursement for a sizable portion of their revenues.

ƒ

Despite modest improvements in the transparency, processes for establishing payment
levels for new laboratory tests, including assignment of new and existing CPT codes to
tests and related methods of cross-walking and gap-filling, remain archaic and

May 2008

348

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

inadequate. The expansion of genetic testing, including new types of testing technology
and the use of PGx that links tests to therapies, are placing greater strains on flaws in
these payment processes.

ƒ

From 1992 to 2006, federal government investigations of clinical laboratory-related fraud
and abuse resulted in penalties exceeding $1.727 billion. Current government efforts aim
to control fraud and abuse arising from contractual joint ventures that enable nonpathologist physicians and other entities to profit from self-referrals of pathology
services. CMS plans to address these issues in a separate rulemaking.

ƒ

Redesign of the current Medicare payment system for laboratory services is needed in
order to meet the growing scientific, technical, clinical, and economic challenges of the
U.S. health care system.

May 2008

349

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

REFERENCE LIST
1. Medicare reform. Health Aff (Millwood) 2000;19(5).
2. Rethinking health reform. Health Aff (Millwood) 2005;24(6).
3.

Overview. In: Restoring fiscal sanity 2007: the health spending challenge. Antos JR, Rivlin
AM, ed. Washington DC: Brookings Institution Press, 2007.

4. Institute of Medicine. Medicare laboratory payment policy: now and in the future.
Washington, DC: National Academy Press, 2000.
5. CMS financial report, fiscal year 2006. Baltimore, MD: Centers for Medicare and
Medicaid, 2006.
6. Terry M. Lab industry strategic outlook: market trends and analysis 2007. New York, NY:
Washington G-2 Reports, 2007.
7. Fiscal year 2007 budget submission (section on medical programs). Washington, DC: U.S.
Department of Veterans Affairs, 2006.
http://www.va.gov/budget/summary/1514Chapter3B.pdf.
8. Evaluation of the TRICARE program: FY2006 report to Congress. Washington, DC: U.S.
Department of Defense, 2006. http://www.tricare.mil/ocfo/_docs/eval_report_fy06.pdf.
9. Trends and indicators in the changing health care market place, 2004 update. Menlo Park,
CA: Kaiser Family Foundation, 2004. (Accessed November 28, 2007, at
http://www.kff.org/insurance/7031/index.cfm.)
10. The Lewin Group. The value of diagnostics innovation, adoption and diffusion into health
care: final report. Washington, DC: Advanced Medical Technology Association, 2005.
11. Neumann PJ, Divi N, Beinfeld MT, et al. Medicare's national coverage decisions, 19992003: quality of evidence and review times. Health Aff (Millwood) 2005;24(1):243-54.
12. Payment basics: clinical laboratory services payment program. Washington, DC:
MedPAC, 2006.
http://www.medpac.gov/publications/other_reports/Sept06_MedPAC_Payment_Basics
_clinical_lab.pdf.)
13. HCFA legislative summary: Balanced Budget Act of 1997, Medicare and Medicaid
provisions. Baltimore, MD: Centers for Medicare and Medicaid, 1997.
http://www.cms.hhs.gov/demoprojectsevalrpts/downloads/cc_section4016_bba_1997.pdf.
14. Leavitt MO. Report to Congress on national coverage determinations for fiscal year 2004.
Baltimore, MD: Centers for Medicare and Medicaid Services, 2006.
http://www.cms.hhs.gov/InfoExchange/Downloads/2004reporttocongress.pdf.
15. Reducing Medicare complexity and regulatory burden. Washington, DC: MedPAC, 2001.
16. Young DS. Reforming laboratory reimbursement: issues, impact, and innovations. A
summary of the clinical chemistry forum held on November 15, 2001. Clin Chem
2002;48(5):792-5.
17. National intelligence report: what's ahead for clinical labs, pathologists in '07? A quick
guide to key Medicare policy changes. New York, NY: Washington G-2 Reports, 2006.

May 2008

350

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

(Accessed November 2, 2007, at
http://www.g2reports.com/issues/NIR/2006_24/1610939-1.html.)
18. Greenberg AL. Centers for Medicare and Medicaid Services. Personal communication.
November 22, 2007.
19. Medicare: divided authority for policies on coverage of procedures and devices results in
inequities. Washington, DC: General Accounting Office, 2003.
20. Summary of H.R. 1, Medicare Prescription Drug, Improvement, and Modernization Act of
2003, Public Law 108-173. Baltimore, MD: Centers for Medicare and Medicaid, 2004.
http://www.cms.hhs.gov/MMAUpdate/downloads/PL108-173summary.pdf.
21. Increase utilization of preventive services in Medicare and Medicaid. Washington, DC:
Partnership for Prevention, 2007. (Accessed October 5, 2007, at
http://www.prevent.org/images/stories/2007seniors/Position%20Statement%20Give%2
0CMS%20Authority.pdf.)
22. Report to the Congress: Medicare payment policy. Washington, DC: MedPAC, 2002.
http://www.medpac.gov/documents/Mar02_Entire%20report.pdf.
23. Hogan C. Trends in Medicare carrier-paid laboratory testing services. Vienna, VA: Direct
Research, LLC, 2006.
24. Fee for service: Advance Beneficiary Notice--general use and Advance Beneficiary Notice-laboratory. Baltimore, MD: Centers for Medicare and Medicaid Services, 2005. (Accessed
October 4, 2007, at http://www.cms.hhs.gov/BNI/02_ABNGABNL.asp#TopOfPage.)
25. Sedor F. AACC submits comments to IOM study panel on clinical laboratory
reimbursement. Washington, DC: American Association for Clinical Chemistry, 2007.
(Accessed October 5, 2007, at
http://www.aacc.org/AACC/gov/gov_affairs/positions/pos_stat_00/01_20_00.htm.)
26. Advance Beneficiary Notice (ABN)--laboratory (form no. CMS-R-131-L). Baltimore, MD:
Centers for Medicare and Medicare Services, 2002.
http://www.cms.hhs.gov/BNI/Downloads/CMSR131L.pdf.
27. Agency information collection activities; submission for OMB review; comment request.
Fed Regist 2007;72(101):29322-3.
28. Mertz A. Advance Beneficiary Notice of noncoverage ("ABN") (CMS-R-131). Washington,
DC: American Association for Clinical Chemistry, 2007. (Accessed October 5, 2007, at
http://www.clinical-labs.org/documents/ACLAABNcommentsApril2007.pdf.)
29. Baselski. ASM sends comments to OMB regarding CMS's draft Advanced Beneficiary
Notice. Washington, DC: American Society for Microbiology, 2007. (Accessed October 5,
2007, at http://www.asm.org/Policy/index.asp?bid=51347.)
30. Root CB. CodeMap, LLC. Personal communication. December 5, 2007.
31. Weil AR, Rossiter LF. The role of Medicaid. In: Restoring Fiscal Sanity 2007: The Health
Spending Challenge. Antos JR, Rivlin AM, ed. Washington. DC: The Brookings Institution,
2007.

May 2008

351

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

32. Brief summaries of Medicare and Medicaid. Title XVIII and Title XIX if the Social Security
Act. Hoffman ED, Klees BS, Curtis CA, ed. Baltimore, MD: Office of the Actuary, Centers
for Medicare and Medicaid, 2006.
33. The Medicaid resource book. Washington, DC: The Kaiser Commission on Medicaid and
the Uninsured, The Henry J. Kaiser Family Foundation, 2002.
http://www.kff.org/medicaid/2236-index.cfm.
34. Administration of Medicaid in New York State: key players and their roles. New York,
NY: Medicaid Institute at United Hospital Fund, 2006.
http://www.uhfnyc.org/usr_doc/Administration_of_Medicaid_in_New_York_State.pdf.
35. Coverage and reimbursement of genetic tests and services. Bethesda, MD: Secretary's
Advisory Committee on Genetics, Health, and Society, 2006.
http://www4.od.nih.gov/oba/sacghs/reports/CR_report.pdf.
36. Whitman B. 2007 rings in changes to anticoagulation management. ACP Observer 2007.
37. Key elements of reimbursement. Minneapolis, MN: Princeton Reimbursement Group,
2006. (Accessed December 3, 2007, at
http://www.prgweb.com/resources/elements.html.)
38. Ullrich CG. Focusing the private payer: education, collaboration and negotiation. Reston,
VA: American College of Radiology, 2006.
http://www.acr.org/Hidden/Economics/FeaturedCategories/ManagedCare/Informati
onabouttheACRManagedCareCommittee/UllrichAMCLC2007.aspx#260,1,FOCUSING
THE PRIVATE PAYER: Education, Collaboration and Negotiation.
39. Schoonmaker MM. Reimbursement for molecular diagnostics. IVD Technology 2007.
40. Technology Evaluation Center assessments. Chicago, IL: Blue Cross Blue Shield
Association, 2007. (Accessed November 26, 2007, at
http://www.bluecares.com/betterknowledge/tec/tec-assessments.html#topics.)
41. Melanson SE, Lindeman NJ, Jarolim P. Selecting automation for the clinical chemistry
laboratory. Arch Pathol Lab Med 2007;131:1063-9.
42. Technology Evaluation Center Medical Advisory Panel. Chicago, IL: Blue Cross Blue
Shield Association, 2007. (Accessed October 4, 2007, at
http://www.bcbs.com/betterknowledge/tec/medical-advisory-panel.html.)
43. Welcome to Hayes. Lansdale, PA: Hayes, Incorporated, 2007. (Accessed October 4, 2007, at
http://www.hayesinc.com/.)
44. Enterprise integration: defining the landscape. Chicago, IL: Healthcare Information and
Management Systems Society, 2007.
45. Athenahealth releases first "PayerView" ranking for Texas health insurers and payers.
Watertown, MA: Athenahealth, Incorporated, 2006. (Accessed October 8, 2007, at
http://www.athenahealth.com/articles/press/2006-10-06.php.)
46. Preventive services: a healthier U.S. starts here. Baltimore, MD: Centers for Medicare and
Medicaid Services, 2007. (Accessed May 16, 2007, at
http://www.medicare.gov/Health/Overview.asp.)

May 2008

352

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

47. S. 2115--110th Congress (2007): A bill to amend title XVIII of the Social Security Act to
extend for 6 months the "Welcome to Medicare" physical examination and to provide for
the coverage and waiver of cost-sharing for preventive services under the Medicare
program. GovTrack.us, 2007. (Accessed October 5, 2007, at
http://www.govtrack.us/congress/bill.xpd?bill=s110-2115.)
48. Curriculum directory: genetics. Washington, DC: American Association of Medical
Colleges, 2007. (Accessed August 27, 2007, at
http://services.aamc.org/currdir/section4/start.cfm.)
49. Realizing the promise of pharmacogenomics: opportunities and challenges. Draft report
of the Secretary's Advisory Committee on Genetics, Health, and Society. Bethesda, MD:
Secretary's Advisory Committee on Genetics, Health, and Society, 2007.
50. Oversight of genetic tests. Bethesda, MD: Secretary's Advisory Committee on Genetics,
Health, and Society, 2007.
51. The fiscal survey of states. Washington, DC: National Governors Association and
National Association of State Budget Officers, 2007.
http://www.nasbo.org/Publications/PDFs/Fiscal%20Survey%20of%20the%20States%20
June%202007.pdf.
52. Technical summary. Baltimore, MD: Centers for Medicare and Medicaid Services, 2005.
(Accessed December 3, 2007, at
http://www.cms.hhs.gov/MedicaidGenInfo/03_TechnicalSummary.asp#TopOfPage.)
53. Schoonmaker MM. Private health insurance coverage and payment policies and
decisionmaking processes for genetic technologies and services. SACGHS March 2004
Meeting. March 1, 2004. Bethesda, MD: Secretary's Advisory Committee on Genetics,
Health, and Society, 2007.
54. Ramsey SD, Veenstra DL, Garrison LP, Jr., et al. Toward evidence-based assessment for
coverage and reimbursement of laboratory-based diagnostic and genetic tests. Am J
Manag Care 2006;12(4):197-202.
55. Dyckman Z. Study of fees and payment system characteristics for clinical laboratory
services. In: Medicare laboratory payment policy: now and in the future. Washington DC:
National Academy Press, 2000.
56. Medicare at a glance. Kaiser Family Foundation, 2007. (Accessed September 18, 2007, at
http://www.kff.org/medicare/index.cfm.)
57. The budget and economic outlook: an update. Washington, DC: Congressional Budget
Office, 2007. http://www.cbo.gov/ftpdocs/77xx/doc7731/01-24-BudgetOutlook.pdf.
58. Wilensky GR. The challenge of Medicare. In: Restoring fiscal sanity 2007: the health
spending challenge. Antos JR, Rivlin AM, ed. Washington DC: Brookings Institution
Press, 2007.
59. Payment basics: hospital acute inpatient services payment system. Washington, DC:
MedPAC, 2006. http://medpac.gov/medicare_basics.cfm.)
60. Young DS. Earning your keep: succeeding in laboratory reimbursement. Clin Chem 1998;
44(8):1701-12.

May 2008

353

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

61. Young DS, Sachais BS, Jefferies LC. Laboratory costs in the context of disease. Clin Chem
2000;46(7):967-75.
62. Smith JJ, Maida A, Henderson JA. Hospital outpatient prospective payment under
Medicare: understanding the system and its implications. Radiology 2002;225(1):13-9.
63. Smith JJ, Maida TR, Agraz JA. Medicare coverage for new medical technology: the basics
of Health Care Financing Administration policy. Am J Roentgenol 2001;176(2):313-6.
64. Outpatient PPS at a glance. Northfield, IL: College of American Pathologists, 2000.
(Accessed October 5, 2007, at
http://www.cap.org/apps/cap.portal?_nfpb=true&cntvwrPtlt_actionOverride=%2Fportl
ets%2FcontentViewer%2Fshow&_windowLabel=cntvwrPtlt&cntvwrPtlt%7BactionForm.c
ontentReference%7D=cap_today%2Ffeature_stories%2F700sidebar5.html&_state=maximi
zed&_pageLabel=cntvwr.)
65. Graziano C. Independent labs on the outs with new Medicare TC billing. CAP Today 2000.
66. H.R. 5661--106th Congress (2000): Medicare, Medicaid, and SCHIP Benefits Improvement
and Protection Act of 2000. GovTrack.us, 2000. (Accessed October 5, 2007, at
http://www.govtrack.us/congress/bill.xpd?bill=h106-5661.)
67. Outpatient dialysis services payment system. Washington, DC: MedPac, 2006.
http://www.medpac.gov/publications/other_reports/Sept06_MedPAC_Payment_Basics
_dialysis.pdf.
68. Carter GM. Use of diagnosis-related groups by non-Medicare payers. Medicare payment
systems: Moving toward the future. Health Care Financing Review, 1994.
http://findarticles.com/p/articles/mi_m0795/is_n2_v16/ai_16863011/print.)
69. Casto AB, Layman E. Principles of healthcare reimbursement. Chicago, IL: American
Health Information Management Association, 2006.
70. TRICARE/CHAMPUS DRG-Based Payment System. TRICARE, 2007.
http://www.tricare.mil/drgrates/.)
71. Cawley JH, Whitford AB. Improving the design of competitive bidding in Medicare
Advantage. Journal of Health Politics, Policy, and Law 2007;32(2):317-47.
72. Price and quality transparency in the health care sector. Testimony of Robin Downey, Vice
President and Head of Product Development, Aetna, before the House Committee on
Ways and Means Subcommittee on Health, 109th Congress, 2nd session. 2006.
http://www.aetna.com/about/aoti/business_solutions/price_quality_testimony.html.
73. Reinhardt UE. The pricing of U.S. hospital services: chaos behind a veil of secrecy. Health
Aff (Millwood) 2006;25(1):57-69.
74. Payment basics: physician services payment system. Washington, DC: MedPAC, 2006.
http://medpac.gov/medicare_basics.cfm.)
75. Medicare's physician payment rates and the sustainable growth rate system. Washington,
DC: Congressional Budget Office, 2006.
76. Dobson A, Koenig L, Siegel J. Recommendations regarding supplemental practice expense
data submitted for 2004. Evaluation of survey data for: independent laboratory. Prepared

May 2008

354

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

for: Centers for Medicare and Medicaid Services #500-95-0059/TO#6. Falls Church, VA:
The Lewin Group, 2003.
77. How Medicare plans work. Baltimore, MD: Centers for Medicare and Medicaid Services,
2007. (Accessed December 5, 2007, at
http://www.medicare.gov/MPPF/Include/DataSection/Questions/Welcome.asp?versio
n=default&browser=IE%7C6%7CWinXP&language=English&year=2008&PDPYear=2008
&MAPDYear=2008&defaultstatus=1&pagelist=MPPFHome&MPDPF%5Fzip=&type=ZIP
COUNTY&ExternalSourceID=&MPPF%5FPDP%5FIntegrate=N.)
78. Medicare Advantage program payment system. Washington, DC: MedPAC, 2007.
http://www.medpac.gov/documents/MedPAC_Payment_Basics_07_MA.pdf.
79. Medicaid benefits: online database. Benefits by service: laboratory and x-ray services,
outside hospital or clinic (October 2006). Menlo Park, CA: Kaiser Family Foundation, 2007.
http://www.kff.org/medicaid/benefits/service.jsp?gr=off&nt=on&so=0&tg=0&yr=3&ca
t=7&sv=18.
80. TRICARE Prime. Falls Church, VA: TRICARE Management Activity, 2007. (Accessed
December 6, 2007, at http://www.tricare.mil/mybenefit/home/overview/Plans/Prime?)
81. TRICARE Standard and Extra. Falls Church, VA: TRICARE Management Activity, 2007.
(Accessed December 6, 2007, at
http://www.tricare.mil/mybenefit/home/overview/Plans/StandardExtra?)
82. Civilian Health and Medical Program of the Uniformed Services (CHAMPUS);
reimbursement of providers, claims filing, and participating provider program. Fed Regist
1993;58(189):51227-41.
83. TRICARE regions. Falls Church, VA: TRICARE Management Activity, 2007. (Accessed
December 5, 2007, at
http://www.tricare.mil/mybenefit/ProfileFilter.do;jsessionid=HXpZ0SWMpbvrK47hVH
mhTzk3QLbMjJsclGw9GLYj4Q2x0NBJnXxc!546270539?puri=%2Fhome%2Foverview%2F
Regions.)
84. LabCorp lands 10-year contract to provide services to United Healthcare. 2006. WRAL
Local Tech Wire.
85. McDowell J. Managed care contract shakeups for lab testing. Is a price war underway?
Clinical Laboratory News 2007;33(5).
86. Plandowski J. Counterpoint: time is right for competitive bidding demo. Washington G-2
Reports 2007;11(12).
87. The Lewin Group. Outlook for medical technology innovation: will patients get the care
they need? Report 2: the Medicare payment process and patient access to technology.
Washington, DC: The Advanced Medical Technology Association, 2000.
88. Raab GG, Logue LJ. Medicare coverage of new clinical diagnostic laboratory tests: the
need for coding and payment reforms. Clinical Leadership and Management Review
2001;15(6):376-87.

May 2008

355

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

89. 2005 annual update for the clinical laboratory fee schedule and laboratory services subject
to reasonable charge payment. Baltimore, MD: Centers for Medicare and Medicaid, 2004.
http://www.cms.hhs.gov/transmittals/downloads/R363CP.pdf.
90. History of the Medicare clinical laboratory fee schedule. Washington DC: American
Clinical Laboratory Association, 2005.
91. Calculation of charge-based relative values for laboratory procedures. Baltimore, MD:
Centers for Medicare and Medicaid Services, 2002.
http://www.aacc.org/NR/rdonlyres/1EAFFFEC-0976-46BE-824F016F49B20DED/0/CMSrvsone.pdf.
92. Medicare. Baltimore, MD: Centers for Medicare and Medicaid Services, 2007. (Accessed
October 1, 2007, at http://www.cms.hhs.gov/home/medicare.asp.)
93. Dowd B, Coulam R, Feldman R. A tale of four cities: Medicare reform and competitive
pricing. Health Aff (Millwood) 2000;19(5):9-29.
94. Thompson T. Final report to Congress: evaluation of Medicare's competitive bidding
demonstration for durable medical equipment, prosthetics, orthotics, and supplies.
Baltimore, MD: Centers for Medicare and Medicaid Services, 2004.
http://www.cms.hhs.gov/DemoProjectsEvalRpts/downloads/CMS_rtc.pdf.
95. DeParle NM, Berensen RA. The need for demonstrations to test new ideas. Health Aff
(Millwood) 2000;19(5):57-9.
96. Hansen K, Lavanty D. Reimbursement concerns. Clin Lab Sci 2006;19(2):66-7.
97. Demonstration project for competitive acquisition of clinical laboratory service. Baltimore,
MD: Centers for Medicare and Medicaid Services, 2005.
http://www.cms.hhs.gov/DemoProjectsEvalRpts/downloads/MMA302b_Summary.pdf.
98. Mensh S. Competitive bidding demonstration project for clinical laboratory tests.
Washington, DC: Advanced Medical Technology Association, 2004.
99. CMS open forum on the clinical laboratory services demonstration project. Washington,
DC: American Association for Clinical Chemistry, 2004.
http://www.aacc.org/govt/2004/comp_bidding.doc.
100. The Medicare competitive bidding demonstration project must be repealed. Washington,
DC: American Clinical Laboratory Association, 2007. http://www.clinicallabs.org/documents/TalkingPointsCompetitiveBiddingFeb07.pdf.
101. Report to Congress--initial report on the Medical Clinical Laboratory Competitive Bidding
Demonstration. Baltimore, MD: Centers for Medicare and Medicaid Services, 2006.
http://www.cms.hhs.gov/DemoProjectsEvalRpts/downloads/MMA302b_NewCongress.pdf.
102. McDowell J. Competitive bid repeal bill introduced as CMS moves on: will demo drive
smaller labs out of the market? Clinical Laboratory News 2007;33(9):1-4.
103. Hoerger TJ, Lindrooth RC, Eggleston JL. Medicare's demonstration of competitive bidding
for clinical laboratory services: what it means for clinical laboratories. Clin Chem 1998;44(8
Pt 1):1728-34.
104. Chambers A. Washington's forecast for 2005. MLO Med Lab Obs 2005.

May 2008

356

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

105. Waters RJ. The outlook for CMS competitive bidding: part II. MLO Med Lab Obs 2006.
106. Arenson RL, van der Voorde F, Stevens JF. Improved financial management of the
radiology department with a microcosting system. Radiology 1988;166(1 Pt 1):255-9.
107. McPherson RA, Pincus MR. Henry's clinical diagnosis and management by laboratory
methods. St. Louis, MO: Elsevier, 2007.
108. Activity-based costing. Washington, DC: Office of the Under Secretary of Defense
(Comptroller) iCenter, 2007. (Accessed December 17, 2007, at
http://www.defenselink.mil/comptroller/icenter/learn/abcosting.htm.)
109. Paxton A. Bean counting basics for laboratories. CAP Today 2001.
110. Luce, Manning WG, Siegel JE, Lipscomb J. Estimating costs in cost-effectiveness analysis.
In: Cost-effectiveness in health and medicine. Gold MR, Siegel JE, Russell LB, Weinstein
MC, ed. New York, NY: Oxford University Press, 1996.
111. Grob GF. Medicare payments for clinical laboratory services: vulnerabilities and controls.
Washington DC: Office of the Inspector General, 2000.
112. Marcum S. What is "upcoding" or "upcharging?". Little Rock, AR: DHS Division of Aging
and Adult Services, Arkansas Department of Human Services, 2007.
http://www.arkansas.gov/dhs/aging/A3-UpcodingDefinition.pdf.
113. False Claims Act. 31 U.S. Code 3729.
114. Health Care Fraud and Abuse Control Program: annual report for fiscal year 2000.
Washington, DC: Department of Health and Human Services and Department of Justice,
2001. http://www.usdoj.gov/dag/pubdoc/hipaa00ar21.htm.
115. Model compliance plan for clinical laboratories. Washington, DC: Office of Inspector
General, 1998. http://oig.hhs.gov/fraud/docs/complianceguidance/cpcl.html.
116. Health Care Fraud and Abuse Control Program: annual report for fiscal year 2005.
Washington, DC: Department of Health and Human Services and Department of Justice,
2006.
117. Levinson DR. State Medicaid Fraud Control Units annual report; fiscal year 2006.
Washington, DC: Office of Inspector General, 2006.
118. Levinson DR. State Medicaid Fraud Control Units annual report; fiscal years 2004 and
2005. Washington, DC: Office of Inspector General, 2005.
119. Physician self referral, overview. Baltimore, MD: Centers for Medicare and Medicaid,
2007. (Accessed November 27, 2007, at
http://www.cms.hhs.gov/PhysicianSelfReferral/01_overview.asp#TopOfPage.)
120. Physician self referral, phase III (1810-F). Baltimore, MD: Centers for Medicare and
Medicaid Services, 2007. (Accessed November 27, 2007, at
http://www.cms.hhs.gov/PhysicianSelfReferral/04a_regphase3.asp#TopOfPage.)
121. 2007 Physician Fee Schedule released: good and bad news for pathology. LabMedicine
2006;37(11):649.

May 2008

357

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

122. About the CPOE initiative. Westborough, MA: Massachusetts Technology Collaborative,
2007. (Accessed August 15, 2007, at http://www.mtpc.org/ehealth/about_1_26_07.html.)
123. "Condo" labs. Washington, DC: American Clinical Laboratory Association, 2007.
(Accessed December 17, 2007, at http://www.clinicallabs.org/issues/condo/index.shtml.)
124. CMS recommits to stopping potential pod lab abuses--ASCP launches phase II of the Stop
Pods Campaign. ASCP e-Policy News 2007;4(8).
125. Medicare program; revisions to payment policies under the Physician Fee Schedule for
calendar year 2007 and other changes to payment under Part B; proposed rule. Fed Regist
2006;71(162):48982-9252.
126. Medicare program; proposed revisions to payment policies under the Physician Fee
Schedule, and other Part B payment policies for CY 2008; proposed revisions to the
payment policies of ambulance services under the ambulance fee schedule for CY 2007;
and the proposed elimination of the e-prescribing exemption for computer-generated
facsimile transmissions; proposed rule. Fed Regist 2007;72(133):38122-395.
127. Exclusion of certain individuals and entities from participation in Medicare and state health care
programs. 42 U.S. Code 1320a-7. Section 1128.
128. Grinstead DJ. OIG proposes new regulation limiting excessive charges to Medicare.
Ambulance Service Journal 2004:1,5.
129. Medicare and federal health care programs; fraud and abuse; clarification of terms and
application of program exclusion authority for submitting claims containing excessive
charges. Fed Regist 2003;68(178):53939-45.
130. Medicare and state health care programs: fraud and abuse; clarification of terms and
application of program exclusion authority for submitting claims containing excessive
charges. Fed Regist 2007;72(116):33430-2.
131. HCPCS background information. Baltimore, MD: Centers for Medicare and Medicaid
Services, 2007. (Accessed October 2, 2007, at
http://www.cms.hhs.gov/MedHCPCSGenInfo/01_Overview.asp#TopOfPage.)
132. Smith GI. Basic CPT/HCPCS coding: 2006 edition. Chicago, IL: American Health
Information Management Association, 2006.
133. Healthcare Common Procedure Coding System (HCPCS) level II coding procedures.
Baltimore, MD: Centers for Medicare and Medicaid Services, 2005. (Accessed October 2,
2007, at
http://www.cms.hhs.gov/MedHCPCSGenInfo/Downloads/LevelIICodingProcedures11
3005.pdf.)
134. CPT process--how a code becomes a code. Chicago, IL: American Medical Association,
2007. (Accessed October 2, 2007, at http://www.amaassn.org/ama/pub/category/3882.html.)
135. Medicare program; public meeting in calendar year 2007 for new clinical laboratory tests
payment determinations. Fed Regist 2007;72(101):29330-1.

May 2008

358

Laboratory Medicine: A National Status Report

Chapter VIII – Reimbursement for Laboratory Medicine

136. ICD-9-CM Coordination and Maintenance Committee. Hyattsville, MD: National Center
for Health Statistics, Centers for Disease Control and Prevention, 2008. (Accessed February
21, 2008, at http://www.cdc.gov/nchs/about/otheract/icd9/maint/maint.htm.)
137. Baselski VS. ASM submits comments on CMS proposed revisions to the Physician Fee
Schedule for calendar year 2007. Washington, DC: American Society for Microbiology,
2006. (Accessed October 3, 2007, at http://www.asm.org/Policy/index.asp?bid=46001.)
138. Coding. Washington, DC: American Clinical Laboratory Association, 2007. (Accessed
October 2, 2007, at http://www.clinical-labs.org/issues/coding/index.shtml.)
139. Lumpkin J. Letter to the Secretary--ICD-10 recommendations. Washington, DC: National
Committee on Vital and Health Statistics, 2003. (Accessed October 3, 2007, at
http://ncvhs.hhs.gov/031105lt.htm.)
140. Mullin R. A brief history of ICD-10-PCS. Journal of American Health Information
Management Association 1999;70(9):97-8.
141. Averill RF, Mullin R, Steinbeck BA, Goldfield NI, Grant TM. Development of the ICD-10
Procedure Coding System (ICD-10-PCS). St. Paul, MN: 3-M Health Information Systems,
1998.
142. Kloss L. AHIMA testimony on health information technology, quality data and the need
for ICD-10-CM and ICD-10-PCS before the Subcommittee on Health of the House Ways
and Means Committee, 109th Congress, 1st session. 2005.
http://www.ahima.org/dc/testimony-icd100705.asp.
143. ICD-10: capturing the complexities of health care. St. Paul, MN: 3M Health Information
Systems and The Healthcare Financial Management Association, 2004.
144. Libicki M, Brahmakulam I. The costs and benefits of moving to the ICD-10 code sets.
Santa Monica, CA: RAND Corporation, 2004.
145. SNOMED CT. Copenhagen, Denmark: International Health Terminology Standards
Development Organisation, 2007. (Accessed October 3, 2007, at
http://www.ihtsdo.org/our-standards/.)
146. H.R. 4157--109th Congress (2005): Health Information Technology Promotion Act of 2006.
GovTrack.us, 2007. (Accessed October 3, 2007, at
http://www.govtrack.us/congress/bill.xpd?bill=h109-4157.)

May 2008

359

Laboratory Medicine: A National Status Report

Appendix A – Desirable Characteristics for Performance Measures

APPENDIX A
DESIRABLE CHARACTERISTICS FOR PERFORMANCE MEASURE SELECTION
CONSIDERED BY IOM, AHRQ, AND OTHER SELECTED GROUPS
To be selected, the measure should rate highly for:
Importance: Is the measure important in a clinical sense, important to the general population,
or important to improve the quality of health care delivery.

ƒ

The health problem addressed by a measure should be a leading cause of death or
disability or associated with high resource use.

ƒ

A measure must have an impact on health, be tied to national goals, and be
susceptible to being influenced by the health care delivery system.

ƒ

A measure should be stratified by race, sex, and age.

Scientifically Sound: This criterion concerns properties of the measure that give it credibility
in terms of reliability, validity, and explicitness of the evidence base.

ƒ

Reliability means a measure consistently produces the same result when repeated
within the same population and setting.

ƒ

Validity addresses the question of whether a measure reflects what it is intended to
measure.

ƒ

The evidence base from which a measure is derived must be explicit—for example,
randomized controlled trials, case control studies, observational studies, or formal
consensus processes.

Usability: The measure should have been effectively used in the past and have high
potential for working well with other measures currently in use. This criterion assesses
whether the measure provides a workable solution for the needs of the health care
organization.
Feasibility: This criterion refers to the feasibility of implementing the selected measures by
examining the existence of measure prototypes, availability of required data across the
system, cost or burden of measurement on providers, and capacity of data and measures to
support subgroup analyses.

May 2008

ƒ

Existence of prototypes means that the measure has already been precisely defines,
field tested, and applied in a variety of settings, such that it can be used by others in
a national data set.

ƒ

Data required for the measure should be available across the health system for the
nation as a whole. The data can be readily collected in the scale and time frame
required.

ƒ

Cost of measurement should be justified and should not impose an excessive
burden on the health system or national collection systems.

A-1

Laboratory Medicine: A National Status Report

ƒ

Appendix A – Desirable Characteristics for Performance Measures

The measure should support meaningful comparisons across subgroups based on
population and by health condition.

Alignment: Optimally, measures should be selected from existing leading measure sets that
are calculated with the same technical specifications for both the numerator and
denominator to reduce redundancy and the burden of reporting.
Comprehensiveness: Measures selected should be part of a set to reflect quality in a
particular area of care or bundled services of necessary care for a given condition.

May 2008

ƒ

Each measure in the set should meet the criterion of importance to warrant inclusion.

ƒ

To demonstrate comprehensiveness, the set of measures must address the way the
care is delivered and the nature of the quality problem involved—underuse,
misuse, or overuse.1-5

A-2

Laboratory Medicine: A National Status Report

Appendix A – Desirable Characteristics for Performance Measures

REFERENCE LIST
1. Approach to performance measurement. Rockville, MD: Agency for Healthcare Research
and Quality, 2001. (Accessed November 16, 2007, at
http://www.ahrq.gov/about/gpra2001/gpra01app1.htm.)
2. Principles of performance measurement in health care. Washington, DC: National
Committee on Quality Assurance, 2001. (Accessed November 16, 2007, at
http://www.ncqa.org/communications/news/prinpls.htm.)
3. Guidance for using the AHRQ quality indicators for hospital-level public reporting or
payment. Rockville, MD: Agency for Healthcare Research and Quality, 2004.
4. Medicare Advantage PPO HEDIS measurement feasibility assessment report. Baltimore,
MD: Centers for Medicare and Medicaid, 2004.
5. Institute of Medicine. Performance measurement: accelerating improvement. Washington,
DC: National Academy Press, 2006.

May 2008

A-3

Laboratory Medicine: A National Status Report

Appendix B – Summary of Selected Performance Indicators Used by Stakeholders

APPENDIX B
COLLEGE OF AMERICAN PATHOLOGISTS QUALITY INDICATORS FOR
PERFORMANCE MEASUREMENT STUDIED IN THE Q-TRACKS PROGRAM
AND POSSIBLE ADDITIONAL INDICATORS STUDIES IN THE Q-PROBES
STUDIES AND OTHER LITERATURE
Step of Total
Testing Process

Q-Tracks Performance
Indicators

Examples of Performance Indicators
From Q-Probes and Other Studies

Structural Measures
Laboratories with written guidelines for changing solution
in tissue processors and water baths
Use or nonuse of preprinted “check off” test order forms
Laboratory policies on double checking test orders
Implementation of strategies for reducing identification
errors
•

Reorganization of phlebotomy

•

Introduction of electronic event reporting system

•

Activation of automated processing system

Implementation of strategies for reducing errors in surgical
pathology
Policies, Procedures,
and Practices

Not studied or developed.

•

Use of checklists

•

Information access

•

Use of computerized forms

•

Use of specific QC processes at each step

•

Use of standardized tasks and language in reports

•

Techniques to simplify processes and reduce handoffs

•

Use of secondary checks

•

Adjustments in work schedule and environment

•

Adequacy of staff training

•

Correct staff for correct job.

How often test directory is updated
Percentage of tests performed on site (could be POCT)
Percentage of tests that must be referred to another
laboratory
Staffing benchmarks
•

Output (workload) per technical staff

•

Management span of control ratios

ƒ Measurement of benchmarks in 4 areas: anatomic

Staff

Not studied or developed.

pathology; chemistry/hematology/ immunology;
microbiology; transfusion medicine
Percentage of employees complying with universal
precautions requirements
Number or percentage of employee exposures to
bloodborne pathogens

May 2008

B-1

Laboratory Medicine: A National Status Report

Step of Total
Testing Process

Access

Appendix B – Summary of Selected Performance Indicators Used by Stakeholders

Q-Tracks Performance
Indicators

Examples of Performance Indicators
From Q-Probes and Other Studies
Percentage of time laboratory hours of operation meet or
exceed those of the managed care organization

Not studied or developed.

Percentage of time patient waits less than 10 minutes for
specimen collection
Use of Web-based systems for reporting, analyzing, and
storing errors
Implementation of specifications and strategies to prevent
errors in point-of-care testing and measurement of
compliance rate

Technology

Not studied or developed.

•

Operator certification and validation in POCT (%)

•

Implement security, validation, performance, and
emergency systems existing and new devices

•

Require flexible user-defined error-prevention system
options on instruments as prerequisite for federal
licensing

•

Integrate connectivity standards for data exchange

•

Preserve fast TATs

•

Monitor invalid use, operator competence, quality
compliance, and other indicators

Percentage of laboratory reports reported by each of the
following:
Fax, phone, computer

Process Measures
Preanalytic Phase- Clinical and Anatomic Pathology
Physician Test
Knowledge

Appropriateness of
Test Selection

Not studied or developed.
Not studied or developed.

Measures not developed to evaluate appropriateness of test
orders.
Not studied or developed.

Not studied or developed.

Patient Preparation

Not studied or developed.

May 2008

Current substitution: Testing rates from National
Healthcare Quality Report and HEDIS measures, other
condition-related measures.
•

Physician Test
Ordering

Patient
Identification

Current substitution: Scope of care within knowledge base (%)

ƒ Breakdown of deficiencies by type
•

•

Wristband error rate (%)

•

Breakdown of wristband error
types (%)

Requisition slip accuracy and completeness
Duplicate orders (%)

Not studied or developed.
•

Patient identification errors caught before reporting (%)

•

Patient identification errors caught after reporting (%)

B-2

Laboratory Medicine: A National Status Report

Step of Total
Testing Process

Appendix B – Summary of Selected Performance Indicators Used by Stakeholders

Q-Tracks Performance
Indicators

Examples of Performance Indicators
From Q-Probes and Other Studies
Blood bank safety
•

Rate of ABO specimen labeling errors

•

Rate of ABO typing result discrepancies

Specimen labeling
Specimen Labeling/
Identification

•

Specimens with a labeling error for one or more
reasons (%)

•

Relabeled specimens due to label misalignment (%)

•

Breakdown of labeling errors by type (%)

•

Specimen/requisition identification mismatch (%)

•

Unlabeled specimen (%)

•

Mislabeled specimen (%)

Not studied or developed.

Blood culture contamination rate

Chemistry specimen quality and acceptability

•

Total contamination rate (%)

•

Median rates of acceptance

•

Neonatal contamination
rate (%)

•

Median rates of rejection due to:

•

Other contamination rate (%)

ƒ
ƒ
ƒ
ƒ
ƒ
ƒ
ƒ
ƒ

General specimen acceptability
•

Specimen rejection rate (%)

•

Breakdown of rejection
reasons (%)

Clotted specimen
Container leaking
Specimen contamination
Hemolyzed specimen
Insufficient volume
Tube over/underfilled
Specimen lost/not received
Improper container

Hematology specimen quality and acceptability
•

Percentage of submitted specimens rejected for
testing

•

Rate of rejection due to:

ƒ
ƒ
ƒ
ƒ
ƒ
ƒ

Specimen Collection

Specimen damaged in transit
Hemolyzed specimen
Clotted specimen
Specimen delayed in delivery/too old
Insufficient specimen quantity
Specimen contaminated by intravenous solution

Phlebotomy
•

Percentage of successful encounters

•

Percentage of unsuitable specimens

•

Reasons for unsuccessful encounters (%)

Compliance rate (%) with phlebotomy safety practices:

May 2008

•

Preventing recapping of needles

•

Discarding tourniquets when contaminated with blood

•

Glove replacement for each patients

•

Handwashing between patients

B-3

Laboratory Medicine: A National Status Report

Step of Total
Testing Process

Appendix B – Summary of Selected Performance Indicators Used by Stakeholders

Q-Tracks Performance
Indicators

Examples of Performance Indicators
From Q-Probes and Other Studies
Extraneous tissue in surgical pathology
•

Overall extraneous tissues contamination rate (%)

•

Location of contamination on slides (% of total)

•

Origin of contamination (% of total)

Specimen adequacy for atypical epithelian cells
•

Median rate of unsatisfactory specimen (%)

•

Median rate of satisfactory specimen but limited use (%)

•

Median rate of 3 most common reasons for inadequacy
or limited use (%)

Not studied or developed.

Specimen Delivery

Not studied or developed.

Current substitute: Delivery within specified time and
under specified conditions according to specimen type
Courier service
•

Percentage of time courier service picks up specimens
on time

Outpatient order entry

Clinical Pathology
Specimen Processing
and Preparation

•

Outpatient order entry rate
(%)

•

Order entry error rates by
category (%)

Transfusion blood product wastage
•

Overall wastage rate (%)

•

Other blood components
wastage rates (%)

•

Breakdown of wastage
reasons (%)

Order entry/transcription error rate
Not studied or developed.

Cutting errors
Anatomic Pathology
Specimen
Accessioning and
Preparation
(Gross Room)

Not studied or developed.

•

Error rates at tissue, block, slide level (%)

•

Lack of or incomplete sampling (%)

•

Injuries to laboratorians (%)

Clinical information for surgical pathology
•

Percentage of cases requiring additional information

Type of additional information needed (%)
Analytic Phase- Clinical Pathology
PT specifications and indicators
Specimen Analysis

Not studied or developed.

•

Laboratory tests reported in or out of range (reported
by analyte, diagnosis, age group) (%)

•

QC reported in or out of range (%)

Percentage of PT results reported accurately
Report Review or
Verification

May 2008

Not studied or developed.

Percentage of test repeats due to abnormal values or errors
identified through autoverification
•

Internal consistency checks

•

Delta checks

B-4

Laboratory Medicine: A National Status Report

Step of Total
Testing Process

Appendix B – Summary of Selected Performance Indicators Used by Stakeholders

Q-Tracks Performance
Indicators

Examples of Performance Indicators
From Q-Probes and Other Studies

Analytic Phase- Anatomic Pathology
Cognitive and interpretive skills
Microscopic
Specimen
Examination

Not studied or developed.

Gynecologic cytology outcomesbiopsy correlation

Results Review

•

Predictive value of a
positive cytology (%)

•

Sensitivity (%)

•

Screening/interpretation
sensitivity (%)

•

Sampling sensitivity (%)

•

Percent positive for atypical
squamous cells of
undetermined significance
interpretations

•

Percent positive for atypical
squamous cells – cannot
exclude high grade
squamous intraepithelial
Lesions Interpretations

•

Percent positive for atypical
glandular cells
interpretations

•

Visual pattern recognition of cells and structures

•

Development of hypotheses and differential diagnoses

•

Skill at clinical and histologic grading

Mammographically directed biopsies
•

Correlation of mammographic abnormality with
microscopic findings

False-negative rate
False-positive rate
Gynecologic cytology specimen adequacy
•

Mean and median rates of specimen accessioning
(e.g., percentage with at least 300 squamous cells and
a cluster of endocervical or metaplastic cells)

•

Mean and median rates of specimen rejection

•

Mean and median rates of reasons for specimen
rejection

Postanalytic Phase- Clinical and Anatomic Pathology
Stat TAT (outliers)

Physician notification of critical values

•

Stat TAT outlier rate (%)

•

Calls with read-backs (%)

•

Breakdown of outliers by
shift (%)

•

Occurrences where only one call was needed to notify
a caregiver of the critical result (%)

•

Breakdown of outliers by
day of week (%)

•

Measures of TAT for the following:

ƒ From result verification to non-physician caregiver
notification

TAT for troponin
•
Turnaround Time
and Notification of
Critical Values

May 2008

•

Median TAT of troponin
from order to ED availability

ƒ From non-physician caregiver notification to

Results reported by
deadline (%)

ƒ From result verification to physician notification

physician notification

TAT for surgical pathology reports
•

Median TATs for routine processing

•

Median TATs for complex cases

•

Median TATs for processing special-handling cases

•

Percentage of outliers

•

Percentage of reports completed within 2 working days

•

Comparison of surgeon satisfaction and outlier rate

•

Percentage of presurgical TAT met

B-5

Laboratory Medicine: A National Status Report

Step of Total
Testing Process

Appendix B – Summary of Selected Performance Indicators Used by Stakeholders

Q-Tracks Performance
Indicators

Examples of Performance Indicators
From Q-Probes and Other Studies
Surgical pathology report accuracy
•

Accuracy/completeness of descriptor analysis for
carcinomas that includes

ƒ
ƒ
ƒ
ƒ
ƒ
ƒ
ƒ
•
Report Accuracy and
Completeness

Gross measurement of primary tumor
Gross tumor configuration
Histologic type
Histologic grade
Depth of invasion
Margin status
Total number of positive/negative lymph nodes

Documentation errors

ƒ Transcription error (%)
ƒ Documentation error (%)
ƒ Poor or incomplete descriptions

Test result correction rate

Practices associated with surgical pathology report
completeness
•

Use of standard report form or check list

•

Amended report rates (%) (clinical pathology)

•

Aggregate mean rate

•

Rates according to change in diagnosis, clinically
significant information, or patient identification

Discrepancies in anatomic pathology reports

Report Delivery

Morning rounds results
availability
•

Morning rounds reporting
compliance rate (%)

•

Frequency of discrepancies by type (e.g., margin
status, diagnosis)

•

Frequency of discrepancies by patient outcome (e.g.,
no harm, near miss, harm)

Errors in report delivery (%)
•

Report delivered to wrong physician

•

Report delivered to wrong location

•

Report delivered about wrong patient

Percentage of abnormal reports not documented in patient
medical record by test type
Physician Follow-up

Not studied or developed.

Percentage of follow-up tests needed but not ordered
Percentage of reports appropriately interpreted by clinician
Laboratory tests that confirm, alter, or add nothing to
patient care
Consultation rate

Interpretive
Consultation

May 2008

Not studied or developed.

•

Aggregate consultation rate

•

Consultation rate by specimen type

•

Median TAT

•

Consultation satisfaction rates (%)

B-6

Laboratory Medicine: A National Status Report

Step of Total
Testing Process

Appendix B – Summary of Selected Performance Indicators Used by Stakeholders

Q-Tracks Performance
Indicators

Examples of Performance Indicators
From Q-Probes and Other Studies

Outcomes Measures
Patient satisfaction with
outpatient specimen collection
• Patient satisfaction score
• Percentage of patients “more
than satisfied”

Customer satisfaction in anatomic pathology
•

Overall satisfaction score

•

Aggregate satisfaction score

•

Percentage of excellent/good ratings

•

Percentage of below average/poor ratings

•

Satisfaction scores for 10 aspects of laboratory
servicea

Percentage of physician complaints per managed care
organization
Hospital nursing satisfaction with laboratory services
•

Nursing overall satisfaction score

•

Aggregate satisfaction score

•

Percentage of very satisfied/usually satisfied ratings

•

Percentage of rarely/not satisfied ratings

•

Productivity ratios:

ƒ laboratory tests per full-time employee and

number of telephone calls per full-time employee

Customer
Satisfaction

ƒ Number of complaints per million laboratory tests
•

Satisfaction scores for 13 aspects of laboratory
services (rated 1-5)b

Calls
•

Percentage of laboratories having written guidelines
for handling telephone inquiries and dealing with
security (e.g., procedures for caller identification or
access to results)

•

Median time to complete a call

•

Percentage of calls requiring transfer to another
person or laboratory section

•

Percentage of phone calls answered within 2 minutes
of ringing

•

Percentage of requests for information successfully
completed in the original phone call

Patient satisfaction
•

Percentage of patient complaints per managed car
organization

The 10 aspects of laboratory services include (1) quality of professional interaction; (2) pathologist responsiveness to
problems; (3) diagnostic accuracy; (4) courtesy of secretarial and technical staff; (5) communication of relevant
information; (6) notification of significant abnormal results; (7) pathologists’ accessibility for frozen sections; (8)
tumor board presentations; (9) teaching conferences and courses; (10) timeliness of reporting.
b The 13 aspects of laboratory services include: (1) accuracy of test results; (2) stat TAT; (3) accessibility of laboratory
management; (4) promptly answered phone calls; (5) abnormal results notification; (6) routine test TAT; (7) ability to
answer telephone questions; (8) laboratory management responsiveness; (9) telephone courtesy; (10) laboratory
point of care testing support; (11) phlebotomy courtesy toward nursing; (12) phlebotomy courtesy toward patients;
(13) phlebotomy responsiveness to service requests
a

May 2008

B-7

Laboratory Medicine: A National Status Report

Step of Total
Testing Process

Appendix B – Summary of Selected Performance Indicators Used by Stakeholders

Q-Tracks Performance
Indicators

Examples of Performance Indicators
From Q-Probes and Other Studies

Other Measures
Cost-related
outcomes
Reimbursementrelated

May 2008

•

Cost per test

•

Cost per unit of health outcome

•

Cost per QALY

•

Percentage of laboratory tests not reimbursed

B-8

Laboratory Medicine: A National Status Report

Appendix C – Development of the Medicare Payment System

APPENDIX C
DEVELOPMENT OF THE MEDICARE PAYMENT SYSTEM
The Medicare program was enacted in 1965 to ensure access to medically necessary care for the
elderly and disabled so as to diminish their financial liability, especially with regard to
catastrophic illness or disability. While the program’s initial structure that provides coverage and
payment for hospital care (Part A) and ambulatory care (Part B) remains intact, recent
developments include options allowing beneficiaries to purchase private insurance (Part C) and
an outpatient prescription drug benefit (Part D). Except for catastrophic coverage, there is no
“stop-loss” provision limiting a person’s financial liability. Therefore, about 90% of seniors also
have supplementary coverage (in addition to Medicare) obtained from various sources, including
employers, Medigap, Medicare Advantage (Part C), or via dual eligibility for Medicaid.1
From its inception until the mid-1980s, Medicare paid for inpatient and ambulatory care,
including laboratory tests and services in either setting, using a fee-for-service system based on
what providers considered to be customary and reasonable charges. Physicians billed Medicare
for laboratory services that they performed in their office and for laboratory services that they
purchased at a discount from hospital and independent laboratories.2 Many physicians routinely
marked up the cost of their purchased laboratory services when billing Medicare and other
insurers. The rule was changed in 1980 to eliminate mark ups, but enforcement was difficult.
Steep increases in health care spending prompted further attempts to contain costs. The
foundation of the current Medicare payment system was established in the Omnibus Deficit
Reduction Act of 1984. Methodologies for calculating costs were further modified through major
legislative acts, including the Consolidated Omnibus Budget Reconciliation Acts of 1985 and 1986,
Balanced Budget Act of 1997, and MMA in 2003. Each of these acts directly affected payment for
laboratory tests and services.

ƒ

May 2008

Omnibus Deficit Reduction Act of 1984
•

Eliminated reasonable charge as a basis for payment

•

Allowed physicians to bill only for laboratory tests performed in their offices

•

Established a PPS for inpatient care under Part A that provides specific lump
sum payments corresponding to particular patient diagnoses (based on
diagnosis-related groups)

•

Established regional fee schedules for physician services, laboratory services,
and durable medical equipment under Part B

•

Designated 56 geographic regions for price determinations

•

Set fee schedule payments for clinical laboratory services at 60% of prevailing
charges for ambulatory care and 62% for outpatient hospital services

•

Required annual adjustment for all fee schedules according to the CPI and
wage rates for each geographic area3

C-1

Laboratory Medicine: A National Status Report

ƒ

Appendix C – Development of the Medicare Payment System

Consolidated Omnibus Budget Reconciliation Act of 1985
•

Established payment caps (deemed NLAs) at 115% of the median of all local
fee schedule amounts for each service4
Note: Congress eliminated payment rate increases in 1988, reduced hospital
outpatient rates from 62% to 60%, restricted updates to 2% of CPI each year
from 1991 to 1993, and eliminated updates for 1994 and 1995.

ƒ

ƒ

a

Balanced Budget Act of 1997
•

Established more extensive payment provisions for inpatient care

•

Established PPS for hospital outpatient care (e.g., outpatient surgery)

•

Reduced Medicare payment caps for Part B laboratory services to the lowest
of the actual charge by the carrier, 74% of the NLA, or 100% for new test
without NLA

•

Required reduction of the number of regional carriers processing laboratory
claims from 56 to 5, with one carrier designated as a central statistical resource
for such claims

•

Required use of negotiated rulemaking to establish national coverage and
administrative policies for Part B laboratory services

•

Required independent laboratories to bill hospitals for their services when
they serve as reference laboratories

•

Required DHHS to fund an IOM study on Medicare Part B payments for
laboratory services

•

Eliminated coinsurance for Part B laboratory services

•

Established Medicare+Choice (Part C) for managed care

•

Expanded coverage of selected laboratory tests for screening and preventiona

•

Expanded anti-fraud and abuse provisions

•

Ordered competitive bidding demonstration projects for durable medical
equipment

•

Eliminated payment rate increases from 1998 through 2003 for laboratory
services5

Medicare Prescription Drug, Improvement, and Modernization Act of 2003
•

Established the Medicare prescription drug benefit program (Part D)

•

Provided process for recognition and payment of new medical technology
under Part A and Part B

Subtitle B of the Balanced Budget Act of 1997 cites coverage for certain screening and prevention tests including
mammography, Pap smear and pelvic exams, colorectal screening, diabetes self management, bone mass
measurements, and vaccines.

May 2008

C-2

Laboratory Medicine: A National Status Report

May 2008

Appendix C – Development of the Medicare Payment System

•

Improved payment for certain screening tests (e.g., for diabetes,
cardiovascular disease, mammography)

•

Increased Part B deductible to $110 in 2005; with subsequent updates by the
annual percentage increase in Medicare expenditures

•

Mandated use of competitive bidding process for durable medical equipment

•

Required competitive bidding demonstration projects for clinical laboratory
services

•

Eliminated Part B laboratory services payment increases for 5 years (2004-2008)6

C-3

Laboratory Medicine: A National Status Report

Appendix C – Development of the Medicare Payment System

REFERENCE LIST
1. Medicare chartbook. Menlo Park, CA: Kaiser Family Foundation and Health Research and
Educational Trust, 2005. http://www.kff.org/medicare/7284.cfm.
2. Grob GF. Medicare payments for clinical laboratory services: vulnerabilities and controls.
Washington DC: Office of the Inspector General, 2000.
3. Omnibus Deficit Reduction Act of 1984, Public Law 98-369. Baltimore, MD: Centers for
Medicare and Medicaid, 1984.
http://www.cms.hhs.gov/RelevantLaws/LS/ItemDetail.asp?ItemID=CMS029523.
4. HCFA legislative summary: Consolidated Omnibus Budget Reconciliation Act of 1985,
Public Law 99-272. Baltimore, MD: Centers for Medicare and Medicaid, 1986.
http://www.cms.hhs.gov/relevantlaws/downloads/LegislativeSummaryforOBRA1985.pdf.
5. HCFA legislative summary: Balanced Budget Act of 1997, Medicare and Medicaid
provisions. Baltimore, MD: Centers for Medicare and Medicaid, 1997.
http://www.cms.hhs.gov/demoprojectsevalrpts/downloads/cc_section4016_bba_1997.pdf.
6. Summary of H.R. 1, Medicare Prescription Drug, Improvement, and Modernization Act of
2003, Public Law 108-173. Baltimore, MD: Centers for Medicare and Medicaid, 2004.
http://www.cms.hhs.gov/MMAUpdate/downloads/PL108-173summary.pdf.

May 2008

C-4



Source Exif Data:
File Type                       : PDF
File Type Extension             : pdf
MIME Type                       : application/pdf
PDF Version                     : 1.6
Linearized                      : Yes
Author                          : debbie.faulk
Create Date                     : 2008:05:12 13:09:07-04:00
Modify Date                     : 2013:02:15 14:12:24-08:00
Has XFA                         : No
XMP Toolkit                     : Adobe XMP Core 5.4-c005 78.147326, 2012/08/23-13:03:03
Producer                        : Acrobat Distiller 7.0.5 (Windows)
Creator Tool                    : PScript5.dll Version 5.2.2
Metadata Date                   : 2013:02:15 14:12:24-08:00
Format                          : application/pdf
Creator                         : debbie.faulk
Title                           : Microsoft Word - 9%x%01!.DOC
Document ID                     : uuid:8d1c5afe-8a31-4ef9-87f8-9087ac49259e
Instance ID                     : uuid:d4259267-64ae-8840-8b8f-5845c01da437
Page Count                      : 385
EXIF Metadata provided by EXIF.tools

Navigation menu