You are on page 1of 14

The Journal of Clinical

Pharmacology
http://www.jclinpharm.org

Biomarkers in Drug Discovery and Development: From Target Identification through Drug Marketing
Wayne A. Colburn
J. Clin. Pharmacol. 2003; 43; 329
DOI: 10.1177/0091270003252480

The online version of this article can be found at:


http://www.jclinpharm.org/cgi/content/abstract/43/4/329

Published by:

http://www.sagepublications.com

On behalf of:
American College of Clinical Pharmacology

Additional services and information for The Journal of Clinical Pharmacology can be found at:

Email Alerts: http://www.jclinpharm.org/cgi/alerts

Subscriptions: http://www.jclinpharm.org/subscriptions

Reprints: http://www.sagepub.com/journalsReprints.nav

Permissions: http://www.sagepub.com/journalsPermissions.nav

Downloaded from http://www.jclinpharm.org by Luciana Oliveira on April 21, 2009


DRUG DEVELOPMENT

ARTICLE
BIOMARKERS
COLBURN
10.1177/0091270003252480
DRUG DEVELOPMENT
IN DRUG DISCOVERY AND DEVELOPMENT

Biomarkers in Drug Discovery


and Development: From Target
Identification through Drug Marketing
Wayne A. Colburn, PhD, FCP

Biomarkers of disease play an important role in medicine and method development and validation; disease process and
have begun to assume a greater role in drug discovery and de- therapeutic intervention assessments; and pharmacokinetic/
velopment. The challenge for biomarkers is to allow earlier, pharmacodynamic modeling and simulation to improve and
more robust drug safety and efficacy measurements. Their refine drug development. The potential for biomarkers in
role in drug development will continue to grow for the fore- medicine and drug development will be limited by the least
seeable future. For biomarkers to assume their rightful role, effective component of the processes. The team approach
greater understanding of the mechanism of disease progres- will minimize the potential for the least effective component
sion and therapeutic intervention is needed. In addition, to be fatal to the rest of the process. As scientific/regulatory
greater understanding of the requirements for biomarker se- foundations for biomarkers in medicine and drug develop-
lection and validation, biomarker assay method validation ment begin to be established, successes and applications
and application, and clinical endpoint validation and appli- will need to be effectively communicated with all of the
cation is needed. Biomarkers need to be taken into account stakeholders, including not only internal and external
while the therapeutic target is still being identified and the drug developers and regulators but also the medical com-
concept is being formulated. Biomarkers need to be incorpo- munity, to ensure that biomarkers are totally integrated into
rated into a continuous cycle that takes what is learned from drug discovery and development as well as the practice of
the discovery and development of one series of biomarkers medicine.
and translates it into the next series of biomarkers. Optimum
biomarker development and application will require a team Keywords: Biomarkers; drug discovery and development;
approach because of the multifaceted nature of biomarker se- pharmacokinetic/pharmacodynamic modeling;
lection, validation, and application, using such techniques as surrogate and clinical endpoints
pharmacoepidemiology, pharmacogenetics, pharma- Journal of Clinical Pharmacology, 2003;43:329-341
cogenomics, and functional proteomics; bioanalytical ©2003 the American College of Clinical Pharmacology

T he current medical revolution began just prior to


World War II with the advent of antibiotics. New
approaches to treating infection brought about a move
Biochemical and molecular markers have been used
in medicine for disease characterization and diagnosis
for centuries. Together, biomarkers from the past and
to culture infections to improve the cure rate. This present provide greater possibilities to get safer and
could have been the beginning of the use of biomarkers. more effective drugs to market faster. Biomarkers are
This medical revolution continues today. Biomarkers factors that are objectively measured and evaluated as
of disease play an important role in medicine and have indicators of normal biological processes or pathogenic
begun to assume a greater role in drug discovery and processes and/or as indicators of pharmacological re-
development. The challenge for biomarkers is to allow sponses to therapeutic intervention. Clinical endpoints
earlier, more robust measures of drug safety and are variables that can be used to measure how patients
efficacy. feel, function, or survive. Surrogate endpoints are
biomarkers that are intended to substitute for a clinical
From MDS Pharma Services, Phoenix, AZ. Submitted for publication No-
endpoint. In a few select cases, biomarkers have
vember 10, 2002. Revised version accepted January 24, 2003. Address
for reprints: Deborah L. Keefe, MD, MPH, FCP, P.O. Box 500, New Ro- evolved to become partial surrogate endpoints.1 No sin-
chelle, NY 10804-0500. gle biomarker has predicted an adequate portion of the
DOI: 10.1177/0091270003252480 observed clinical safety and efficacy outcome to war-

J Clin Pharmacol 2003;43:329-341 329


COLBURN

Table I Biomarkers, Surrogate Endpoints, and Clinical Endpoints/Outcomes


Clinical Endpoint/Outcome

Biomarkers
Leukotrienes, cytokines, and chemokines Asthma, chronic obstructive pulmonary disease, and
rheumatoid arthritis
Pulmonary function tests Asthma and chronic obstructive pulmonary disease
Glucose, fructosamine, glycosylated albumin, and HbA1c Type 1 and 2 diabetes mellitus
Retinal evaluations, nephropathy measures, and peripheral Type 1 and 2 diabetes mellitus
neuropathy assessments
Cytokines Type 1 diabetes mellitus
Angiotensin-I, angiotensin-II, plasma renin, aldosterone, Hypertension
and angiotensin-converting enzyme (ACE) activity
Blood pressure and heart rate measures Hypertension
Surrogate endpoints
HbA1c Retinopathy, cardiomyopathy, neuropathy, and nephropathy
HIV/CD4 Survival time
Blood pressure Myocardial infarct/stroke—survival time
Tumor size Survival time
Cholesterol (HDL, LDL, VLDL) Myocardial infarct/stroke—survival time

rant ideal surrogate endpoint status,2 so biomarkers dation for a continually evolving biomarker team (Fig-
that have successfully predicted partial clinical out- ure 1). A continuous cycle, as well as improvements in
comes have been called surrogate endpoints. As more drug development and medicine, can occur by apply-
than 100,000 proteins are in the human body and most, ing biomarkers. Because of the complexities with and
if not all, act on a variety of biological processes, why the multifaceted nature of biomarker selection and val-
should a single protein biomarker provide that kind of idation, a multifunctional team is needed to integrate
insight? At best, a single biomarker will predict a por- biomarker concepts across discovery and develop-
tion of the overall effect. For example, CD4 cell counts ment. These teams should include the areas of
account for about 30% of the increased survival time, pharmacogenetics, pharmacogenomics, and functional
whereas CD4 plus HIV viral load accounts for approxi- proteomics; bioanalytical method development and
mately 70%a (Table I). It will take more than one validation; disease process and therapeutic interven-
biomarker to describe disease and treatment, while it tion assessments; and use of biomarkers for
will take more than one additional biomarker to de- pharmacokinetic/pharmacodynamic (PK/PD) model-
scribe treatment-related adverse events. Although sev- ing and simulation. The potential for biomarkers in
eral biomarkers may be needed to create an ideal surro- medicine and drug development is limited by the least
gate endpoint cluster to truly characterize clinical effective component of these activities. The team mini-
endpoints, individual biochemical/molecular markers mizes the potential for the least effective component to
can be used as biomarkers to evaluate disease progres- be fatal to the rest of the process. Once the scientific and
sion and to evaluate the effects of therapeutic interven- regulatory basis for biomarkers in medicine and drug
tion early in development. Mechanism-based development has been established, successes and ap-
biomarkers should be used in drug discovery and de- plications need to be effectively communicated with
velopment to support early and correct decisions. They all of the stakeholders, including, but not limited to, us-
will also be useful in later development to optimize ers of the data, corporate management, regulators, in-
dose selection, stratify patients to better understand ternal and external experts, and the medical commu-
drug effects/select treatments, and ensure prognosis. nity, to ensure that biomarkers are totally integrated
Throughout the drug discovery and development into medicine and drug development.
pipeline, biomarkers are identified that create the foun- This article is divided into five sections, including
(1) mechanism-based biomarkers, (2) PK/PD modeling
a. Personal communications with FDA biostatisticians involved
concepts, (3) biomarker assay methods designed to
in the decision to use CD4 cell counts plus HIV viral burdens as sur- meet drug development objectives, (4) clinical end-
rogates for AIDS outcomes. point issues, and (5) issues and opportunities. Each of

330 • J Clin Pharmacol 2003;43:329-341


BIOMARKERS IN DRUG DISCOVERY AND DEVELOPMENT

Other Providers
For biomarkers to be useful in drug development,
Stakeholders they should reflect biochemical/molecular changes
Clinical Laboratories
Hospitals Concept
Diagnostics that occur due to the disease process and are altered by
HMOs/PPOs
therapeutic intervention prior to a downstream impact
Insurers Marketing Discovery on the clinical endpoint.3-5 The changes are generally
Physicians Chemistry
Phase IIIb > Phase IV/V HTS
mediated through receptor or enzyme induction or in-
Pharmacists
hibition. In some cases, the effects are mediated
Patients
through direct interaction with the deleterious bio-
Etc. Preclinical chemical agent, such as when antibodies or receptors
Regulators Development are used to bind cytokines. Ultimately, some of these
Pharmacology
Toxicology biomarkers may become surrogate endpoint clusters
PK/DM that predict observed outcomes. For example, develop-
Clinical ing the first angiotensin-II (A-II) antagonist with the be-
Development lief that it would reduce blood pressure and result in re-
Phase I > Phase Ib > Phase II a >
duced incidence of strokes and myocardial infarctions
Phase II b > Phase III
represented a sequential process. First, it was impor-
tant to show that the drug bound to the A-II receptor
Figure 1. The team takes center stage. Stakeholders and other func-
and altered the renin-angiotensin system, as was pre-
tions that can produce and receive information and knowledge from
the team occupy the surrounding area. The schematic emphasizes dicted from its intended mechanism of action. Changes
the critical team concept as well as the critical nature of continuous, in biomarkers such as angiotensin-I, angiotensin-II,
effective, and efficient communication. plasma renin, and angiotensin-converting enzyme
(ACE) inhibition were used for this purpose.6 This,
however, did not ensure that blood pressure would be
reduced or that this mechanism of blood pressure re-
duction would result in reduced stroke and heart at-
these topics contributes to the impact that biomarkers tacks. Each sequential step that contributed to linking
are having and can have on drug discovery and devel- the biomarker to the clinical outcome was important to
opment. In the final analysis, the goal is to identify a se- the total understanding of the process and to establish-
ries of biomarkers that can characterize the disease pro- ing surrogate endpoints to predict clinical outcomes.
cess, be measured robustly, and assist in early and Biomarkers can provide great value in early drug devel-
correct decisions for compound selection and efficient opment if they reflect mechanism-based intervention,
drug discovery and development. even if they do not ultimately become part of surrogate
endpoint clusters.7,8 An increase in our understanding
MECHANISM-BASED of disease and drug mechanisms of action at the bio-
BIOMARKERS chemical and molecular level is producing greater num-
bers of biomarkers (see Table I for a few examples).6
Clearly, all biomarkers should represent mechanism- Useful biomarkers provide value because they occur
based processes. Early in discovery and development, earlier than clinical endpoints/outcomes and/or are
the biomarker should at least reflect activity that is me- measured by more robust methods.6 Biomarkers that
diated through the theoretical disease mechanism of are proximal to the clinical endpoint but occur earlier
action. Later in development, the biomarker should in time make drug development and approval more ef-
represent mechanism-based processes that are critical ficient. Proximal, rather than distal, biomarkers mini-
to disease progression and that are appropriately al- mize the potential influence of disparity between the
tered by effective therapeutic interventions. However, biomarker and effect that can result from other inter-
some biomarkers are simply associated with the dis- vening variables, but the biomarkers still must occur
ease, do not drive the disease process, or are not altered earlier in time and/or be measured by a more robust
by therapeutic intervention that acts on the disease method than the clinical endpoint. As earlier drug ap-
mechanism. False-positive results occur when there is provals occur using biomarkers of efficacy, biomarkers
an assumption that the biomarker is a critical part of the of drug safety will also be needed to evaluate safety and
disease process when in fact it is only loosely associ- efficacy in parallel. For example, observing seizures
ated with the disease process or a random event that that result from drug-induced epileptiform activity in
has inadvertently coincided with disease diagnosis an EEG will no longer suffice. A biochemical/molecular
and progression. marker that predicts the EEG changes and resulting sei-

DRUG DEVELOPMENT 331


COLBURN

zure early enough to anticipate and avoid the seizure is


Disease Process
needed.
Biomarker 1a Biomarker 1 Biomarker 2 Biomarker 3
Biomarkers have been used for diagnosis and, in
some cases, for prognosis for centuries, for example,
when physicians tasted urine to determine whether a Indirect Direct Direct Not Linked
person had diabetes. Biomarkers are used when look- Reliable Unreliable Reliable Reliable
ing for hypersecretion of glucocorticoids to help in the
diagnosis of Cushing’s syndrome. When elevated HER2 Surrogate Surrogate
concentrations are used to indicate the appropriate
women for Herceptin breast cancer treatment, a
biomarker is being used. Biomarkers, such as CD4 cell Clinical Endpoint/Outcome Clinical Endpoint/Outcome
counts, and viral loads have been instrumental to fast-
track drug development and approval, as well as pre- Figure 2. Biomarkers can contribute to or detract from drug devel-
opment. Biomarkers 1a and 2 can contribute positively to develop-
dict the need for treatment changes to optimize ther- ment, whereas biomarkers 1 and 3 will detract from the development
apy. In contrast, there have been examples of process. Biomarker 1 is a potentially useful marker but cannot be as-
biomarkers such as those that reduce premature ven- sayed effectively and therefore will detract from team progress.
tricular contractions, which were expected to reduce Biomarker 3 appears to be, but is not, part of the therapeutic interven-
tion process.
the incidence of sudden death but actually increased
sudden death.9
Differences that exist between biochemical/molecular
biomarker concentrations or clinical biomarker mea-
sures in disease and health may or may not be related to
the disease mechanism. Differences in biomarker val-
ues may occur because of the disease process if the ceptors that are linked to downstream protein
biomarkers are mechanism based or may occur because biomarkers can then be used as part of high-throughput
of circadian rhythms or other physiological, pathologi- screening for discovery. These screening tools are then
cal, or pharmacological processes that are not based in used to set the course for biomarker selection during
the disease of interest. In addition, apparent differ- preclinical and clinical drug development. Animal
ences can occur because of lack of reproducibility in models that incorporate relevant biomarkers can be es-
the analytical method. Or, differences between disease tablished to reduce the number of drug candidates that
and health may not be robust enough to guide future will be considered as potential lead compounds by
drug development or prognosis (Figure 2).6-8,10-12 For looking at preclinical drug effects on biomarkers of hu-
biomarkers and PK/PD modeling to be efficient and ef- man disease.12,18
fective, there must be more than just an association be- Endogenous ligands compete with drugs for recep-
tween drug concentrations and response—there must tor occupancy and can thereby influence therapeutic
be a correlation. drug intervention.19 This competition is consistent
Identifying proteins and their functions using with certain protein biomarker changes observed be-
genomic information—proteomics—allows for more tween health and disease. The observation is also con-
rapid identification of new biomarkers.13-17 Comparing sistent with dosing of receptor agonists/antagonists
a homogeneous subpopulation along with familial causing a transient increase in the circulating concen-
tracking and comparing genes within a heterogeneous tration of a naturally occurring agonist/antagonist. In
population database that encompasses the target dis- addition, this observation is consistent with dosing of
ease are two ways to identify disease-related genes. In an enzyme inducing or inhibiting agent-altering meta-
both approaches, bioinformatics software is used to de- bolic processes that result in increased or decreased
tect differences in gene expression from healthy as well concentrations of a substrate. For example, it has been
as mild and severely diseased tissues. Gene and protein shown that antibodies or biotechnology-derived recep-
sequencing and synthesis can be used to find, evaluate, tors (etanercept) that bind endogenous ligands such as
and confirm the appropriate genes and biomarker gene TNF-α reduce ligand concentrations that may be found
products for target diseases. in excess in certain disease states and thereby cause the
Differing protein profiles from diseased and normal disease to revert to a more healthy state.19 These exam-
tissues can also lead to biomarker discovery and identi- ples support using changes in biochemical/molecular
fication. Knowledge of the protein will lead to gene marker concentrations as one way to monitor the ef-
identification. Protein targets such as enzymes and re- fects of disease treatment (Figure 3).20

332 • J Clin Pharmacol 2003;43:329-341


BIOMARKERS IN DRUG DISCOVERY AND DEVELOPMENT

PK/PD MODELING

Biomarkers create value throughout the drug devel-


opment process by providing early decision-making
information and input for PK/PD modeling.20-24
Pharmacokinetics is concerned with what the body
does to the drug. Drug concentration-time profiles are
generated. Pharmacodynamics is concerned with what
the drug does to the body. Response-time profiles are
generated. PK/PD modeling is a mathematical treat-
ment of the PK and PD data in an effort to gain greater
insight into how drug concentrations and effects relate
to each other, as well as how the models can be used to
predict study outputs under a variety of different con-
ditions. More modeling of disease processes without
therapeutic intervention is needed so the disease com-
ponent of the PK/PD model can be better understood
and applied. Several recent review articles discuss the
potential for PK/PD modeling in drug discovery and
development.18,25-32 This section focuses on important
issues and concepts that apply to PK/PD modeling and
biomarkers.
A series of questions need to be answered when
identifying PK inputs for PK/PD modeling and
simulation.
Question 1: What needs to be measured? A series of ques-
tions that relate to providing sufficient input for infor-
mative PK/PD modeling need to be asked and an-
swered at this time, such as the following:
Will drug or drug plus active metabolites be measured?
If active metabolites are present, do they contribute to
the activity profile following single and/or repeated
doses of the drug?
Will unbound drug concentrations or total (unbound
plus bound) drug concentrations be assayed? Is bind-
ing linear or nonlinear? If binding is nonlinear, un-
bound concentrations need to be measured to provide
appropriate input for PK/PD modeling.
Does the drug compete with endogenous ligands for re-
ceptor binding? The answer is most likely yes. Can this
competition be built into the PK/PD models?
Question 2: Do biomarker responses parallel drug and/or
active metabolite concentrations? Generally, they do
not because there is a temporal delay between concen-
trations and effects. PK/PD models are used to better
understand this relationship and how it might change
as a function of drug input and other variables.

Figure 3. Enzyme/receptor binding of endogenous ligands in Any decision on which approach to take is based on lo-
health and disease and the influence of binding to an exogenous gistics as well as experience and receptivity of the
antibody/receptor on the unbound amount of endogenous ligand study sites and investigators to each approach.
during therapy of the disease. In segment a, endogenous receptors Having only one active moiety rather than several or
are half-filled in health. In segment b, endogenous receptors are to-
tally saturated in disease. In segment c, administration of exogenous
one that dominates the beneficial and toxic effects of
antibody or receptor reestablishes the healthy half-filled balance. therapeutic intervention makes PK/PD studies simpler.
Adapted from Colburn.19 In addition, determining whether to measure unbound

DRUG DEVELOPMENT 333


COLBURN

or total (unbound plus bound) drug concentrations can 100


be another variable. If drug-plasma binding is linear in

Unbound/Total Plasma Dry Concentration


90
the therapeutic and toxic range, unbound and total 80
concentrations are simple ratios. For example, if bind- 70
ing is 75% and linear, unbound concentrations are 60
25% of the total drug concentration across the entire 50
concentration range: 2.5 unbound at 10 total, 25 un- 40
bound at 100 total, and 250 unbound at 1000 total. In 30
contrast, nonlinear plasma binding requires unbound 20
drug concentrations to be measured because unbound 10
concentrations increase more than proportionately 0
with increasing total drug concentrations, as shown in

0
10
20
30
40
50
60
70
80
90
0
0
0
0
0
0
0
0
0
0
0
10
11
12
13
14
15
16
17
18
19
20
Figure 4. Concentration
Competitive endogenous ligands complicate the PK
Figure 4. Impact of linear and nonlinear binding. This graph shows
and PD inputs33 for PK/PD models. Environmental, di- the percentage of unbound/total drug concentration as a function of
etary, and endogenous substances compete with drugs total plasma drug concentrations under linear and nonlinear (satu-
for receptor occupancy in vivo.19 Should drug develop- rable) binding conditions. In a linear situation, percent unbound re-
mains constant as plasma drug concentrations increase. In contrast,
ers identify these competitive substances and use them during saturable binding, the unbound/total drug concentration ra-
as biomarkers to track the effects of their drug develop- tio increases as total drug concentrations increase. In this example,
ment programs? Or, are these competing substances almost no drug is unbound at concentrations less then 10 units, but
just obstacles that complicate assay development and about 90% is unbound at concentrations in excess of 100 units.
When plasma drug binding is saturable, drug assays should measure
validation as well as modeling efforts? As these sub- unbound concentrations because they vary as a function of total drug
stances compete with the drug for receptor binding, concentrations, and only unbound drug elicits drug effects.
they must be acting to stimulate or inhibit physiologi-
cal activity that could be a predecessor to pharmacolog-
ical activity at greater concentrations.34-36 It can be an-
ticipated that changes in endogenous ligand
concentrations following drug dosing may be similar to
changes observed in angiotensin-I, angiotensin-II, and
plasma renin following doses of an angiotensin-II an-
tagonist, as shown in Figure 5.6 It is likely that baseline
activity is associated with endogenous competing sub-
stances, even in the absence of drug. For comprehen-
sive PK/PD modeling, it is important to determine con-
centrations of the competing ligand(s) after both active
and placebo treatment so that competition can be built
into the models.33
PK/PD models should never be more complicated
than required to fit existing data and predict future
data. Models should only include rate-limiting,
mechanism-based components of the system that are
required to describe the data.25,37,38 For example, if the
rate-limiting PD step is receptor mediated, then the
model should reflect receptor binding. In contrast, if
Figure 5. Biomarker changes as a function of angiotensin-II inhibi-
the rate-limiting step is hormone secretion or protein
tor dosing. Angiotensin-I (A-I), angiotensin-II (A-II), and plasma
production, then the model should reflect the secretion renin activity increase from placebo/baseline in response to the ad-
process. If PK/PD models are not simple and unique, ministration of an A-II receptor antagonist. There is a direct effect of
model-predicted endpoints may provide inappropriate the antagonist on A-II when the antagonist inhibits the binding of A-II
outputs during simulation under different conditions. to the receptor. The resulting increase in A-II concentrations (䊏) then
works through a feedback loop in the renin-angiotensin system to
Sensitivity analysis has been used to identify unique
also increase A-I (starts) and plasma renin activity (䊉). This process
model solutions when transitioning from fitting a represents the types of changes that occur when a therapeutic inter-
model to data to using the model to simulate several po- vention affects biomarkers of disease to elicit a clinical endpoint.
tential outputs.39 Adapted from McShane et al.41

334 • J Clin Pharmacol 2003;43:329-341


BIOMARKERS IN DRUG DISCOVERY AND DEVELOPMENT

BIOMARKER ASSAY METHODS Reference Standards

Whether chemical assay methods or binding assays are Another challenge for biomarker assays is obtaining
used, biomarker assay development and method vali- biomarker/analyte-free matrices to prepare calibrator
dation tend to be more complicated than those for most standards. Several approaches have been used such as
drug assays.27,40,41 Several factors that can contribute to nonspecific or affinity-based removal of desired
this complexity will be discussed below. For example, biomarkers, use of matrix from a different species, or
biomarkers have diverse molecular structures ranging use of a similar protein-buffer solution. However, ma-
from simple electrolytes and amino acids to large pro- trices produced by any of these methods are not the
teins to complete microorganisms such as viruses and same as an authentic sample matrix. Therefore, assay
bacteria. As a result, it is generally not possible to vali- performance must be tested during prestudy validation
date biomarker analytical methods according to strict to show that assay performance is similar between au-
good laboratory practices (GLPs). Recent efforts have thentic and nonauthentic matrices.46 In some cases,
looked at providing GLP-like validation by evaluating pure reference standards are not available.
all aspects of the method, including precision,
reproducibility, and reliability according to GLP crite- Heterogeneity and Quantification
ria, but without attempting to determine accuracy.42-45
GLP-like validation provides scientifically valid infor- Macromolecular biomarkers exist in heterogeneous
mation about the biomarker that can be used to deter- forms. For example, vascular endothelial growth factors
mine changes in biomarkers associated with disease (VEGF) are important biomarkers for carcinogenesis.52
progression as well as therapeutic intervention provid- VEGF is expressed in at least four forms with 121, 165,
ing reliable data for critical decision making.11,28,43,45,46 189, and 206 amino acid residues. These forms are dis-
tributed differently into vascular space and
Choice of Matrix extracellular fluids.53-55 It might be possible to obtain
one pure form and use it as a reference standard to
The first challenge is to identify and select an accessi- quantify VEGF samples as standard equivalents with-
ble, meaningful sample matrix. Readily accessible ma- out specifying exact composition. This is analogous to
trices such as whole blood, plasma, serum, urine, sa- using nonspecific radioactivity measurements of drug
liva, or sweat are preferred over more difficult to obtain plus breakdown products to evaluate mass balance fol-
sample matrices such as biopsy or bronchoalveolar la- lowing a dose of radiolabeled drug.
vage samples. For example, plasma or urine drug con- Biomarkers can also exist in various bound states,
centrations are often used as surrogates for drug con- as is the case for prostate-specific antigen (PSA),
centrations in the target tissue during pharmacokinetic which occurs in serum both free and bound to α1-
studies. It is convenient to measure biomarkers in these antichymotrypsin, α1-antitrypsin, protein C, and α1-
same matrices. However, the site of biomarker produc- macroglobulin. The World Health Organization is
tion as well as the physiology and distribution of the attempting to standardize reference PSA material as
biomarkers must be taken into account before an ac- a characterized mixture.56,57 Similarly, standardiza-
ceptable matrix can be selected. Biomarker concentra- tion of biomarker calibrators is an ongoing effort for
tions are generally lower in the systemic circulation clinical chemists and bioanalytical chemists work-
than they are at the site of production. For example, ing to improve biomarker assay methodology for
cytokines can be assayed in bronchoalveolar lavage drug development.25,26
fluid, sputum, plasma, peripheral mononuclear cells, In contrast to the term definitive quantification,
or whole blood from asthma patients.47-51 The first three which is used to describe assay results for well-
matrices can be assayed directly for cytokines, whereas characterized small drug molecules that can be accu-
the latter two matrices must be stimulated to evaluate rately measured with a single standard calibrator, rela-
individualized cytokine production. If the assay tive quantification is a term that is used when endoge-
method has sufficient sensitivity, plasma or sputum is nous macromolecular biomarkers are measured and
preferred for ease of collection and analysis. If analyti- accuracy cannot be determined.45 An example is when
cal sensitivity is not sufficient to measure the VEGF-165 is used as the standard calibrator to quantify
biomarker in these matrices, bronchoalveolar lavage all VEGF isoforms. For relative quantification,
can become the preferred matrix because of greater postdose sample values are generally compared to
concentrations, but variability and complexity of sam- predose sample values from the same subject to deter-
ple collection and preparation are also greater. mine the relative effect of drug intervention. This ap-

DRUG DEVELOPMENT 335


COLBURN

proach minimizes the need for accuracy but still de- ploratory clinical research and development. The prac-
pends on precision, reproducibility, and an otherwise ticality of multiple biomarker assays, sample volume
robust assay method. Data are normalized and ex- limitations, requirements for adequate sample collec-
pressed as percent change from predose baseline tion and handling, and multiple assay methods must be
biomarker concentration. More definitive data can be considered. The number of biomarkers may be nar-
obtained by comparing biomarker concentrations over rowed to just a few that are able to show differential,
the same sampling schedule following placebo and ac- mechanism-based effects during early drug develop-
tive treatment. ment. A final cluster of biomarkers can be selected for
Semiquantitative assays are used for antidrug anti- larger Phase III proof-of-safety and efficacy trials. Out-
body, enzymatic, or receptor or cell-based assay meth- put data must be reliable and robust to pass Food and
ods when well-characterized reference standards are Drug Administration scrutiny during the end of Phase
not available.58-60 Categorical or classification results II meetings and, ultimately, new drug application re-
are often used when assay methods do not exhibit con- view for market approval. Using information and
tinuous concentration-response relationships. Dilu- knowledge gained during early development, method-
tion assays are those in which concentrations are based ology and assay dynamic ranges should be optimized,
on all or none activity at dilutions of 1:2, 1:4, 1:8, 1:16, and proper method validation should be carried out be-
1:32, and so forth. Biomarker assays based on biologi- fore designing later phase clinical protocols.70-72
cal systems can exhibit discontinuous categorical
changes such as none, low, medium, and high or di- What Is Reasonable?
chotomous changes such as yes/no or 0/1. The term
classification assay has been used to describe categori- It is not practical or necessary to perform full validation
cal assays that are used to provide descriptive order of biomarkers during early exploratory phases of drug
such as +/– or 0, 1, 2, 3, 4, 5 to biological reactions.61-63 development, as long as the methods provide reliable
Categorical quantification is more akin to clinical end- data, information, and knowledge.28 As drug develop-
point measures than to typical bioanalytical results.64-66 ment progresses, validation should keep pace with the
Therefore, to provide added value, less robust and less required precision and reliability needed to achieve
quantitative categorical biomarker assays should be re- program objectives.6,43 Prestudy validation should be
served for intermediate biomarkers that occur much completed before clinical studies are begun and should
earlier than the clinical endpoint. set the foundation for establishing method acceptance
Immunoassay methods are generally more compli- criteria. Prestudy validation should include a pilot
cated than chemical methods for biomarker measures. study to provide insight into the differences that exist
For example, tight-binding soluble receptors such as Il- between disease and health as well as differences in
2R may bind biomarkers differently between healthy biomarker variability between health and disease.
and diseased subjects, within patient groups, or in Bioanalytical results will be used to assess whether the
the same patient at different times. 45,46 Since method can provide the sensitivity, precision, and ro-
immunoassays are based on binding, each form of a bustness to distinguish the differences between disease
heterogeneous group of biomarkers can react differ- and health as well as to quantitatively evaluate the pro-
ently and thereby affect assay results in an antibody- gression from one state to the other.73,74 Assay perfor-
based assay system. Biological activity of various forms mance acceptance criteria should be established a pri-
may or may not parallel antibody-binding response ori based on the objectives of the study and the known
curves. Although functional biological assays such as assay variability.74
enzyme, cell-based, or tissue perfusion assays have In-study sample analyses and validation must use
greater variability and are not as sensitive as immuno- quality control samples (QCs) to document analytical
chemical or chemical methods, they may complement performance during clinical studies.42 In addition, QCs
quantitative analyses.67-69 In general, functional assays are used for the acceptance or rejection of an analytical
are labor intensive and time-consuming, so they would run during sample analysis. Similar to drug assay vali-
not be practical for use as a primary measurement tool dation, QCs are generally prepared to evaluate the
for large clinical trials. lower, middle, and upper limits of standard curve
ranges. QCs are made in the same matrix as the study
Multiple Biomarkers samples, so assay performance determined from the
QCs reflects that of authentic samples. Therefore, even
Several biomarkers from a multitude of discovery/ if assay standards are prepared in a nonauthentic ma-
preclinical markers can be selected to test during ex- trix, QCs must be prepared in an authentic matrix. Au-

336 • J Clin Pharmacol 2003;43:329-341


BIOMARKERS IN DRUG DISCOVERY AND DEVELOPMENT

thentic matrix samples are screened to determine basal methods as well as for biochemical marker methods
biomarker concentrations. Low basal concentrations since relative outputs are used for decision making
are selected, and then various middle and upper QC during drug development. Accuracy becomes impor-
concentrations can be prepared by adding additional tant to ensure continuity throughout the development
known biomarker amounts. For semiquantitative process when results from one method need to be com-
methods, QCs can be prepared by combining samples pared to another or one method replaces another dur-
with high, middle, and low observed concentrations.74 ing drug development. Clinical endpoint measure-
Validation using these approaches provides precise ments need to generate outputs that are as useful and
and stable data but does not provide accurate data. informative as drug assays and biochemical/molecular
marker assays.6,20 Clinical endpoint validation criteria
CLINICAL ENDPOINT ISSUES are outlined in Table III.
It should be easier to obtain reproducible GLP-like
Intuitively, it can be assumed that clinical endpoints clinical endpoint and biomarker output from a single
that measure how a patient feels, functions, or survives site than from several independent clinical trial sites.
are the best measures of disease progression and effec- Therefore, a single site or a small number of common
tiveness of therapeutic intervention. This is true for the
practice of medicine but not necessarily in drug devel-
opment and regulatory review. Except for survival or Table II Evolution of Biomarkers
cure, clinical endpoints have questionable value for during Drug Discovery and Development
determining whether a biomarker can predict reason-
able clinical endpoints in the current clinical develop- Theoretical foundations for disease processes and drug
ment environment. From a drug development perspec- mechanisms
tive, clinical endpoints and clinical endpoint measures Experimental foundations for disease processes and drug
are inextricably linked. If the clinical endpoint cannot mechanisms
be measured or cannot be measured reliably across sev- Preclinical In vitro binding to enzyme or receptor
eral patients, several sites, or several studies, it has no In vivo in animal models (transgenic?)
value within the context of drug development and the Clinical Healthy human subjects in Phase I
regulatory approval process. Clinical endpoint ratio- Healthy human disease models in Phase Ib
Human patients in Phase IIa
nalizations are often found deep in the history of dis-
Human patients in Phase IIb/III
ease, diagnosis, and prognosis. In many cases, results
Epidemiological evidence
from one clinical investigative site cannot be repro-
Previous clinical experience with therapeutic class
duced at another site because measurement methods
Previous clinical experience with biomarker
are somewhat subjective and dependent on operator-
Simulated biological systems
specific performance. Generalizing a clinical endpoint
to another patient population because it was useful in
one patient population can be misleading. It is likely
that false-positive and false-negative clinical endpoint
measures have contributed to incorrect understanding Table III Clinical Endpoint
and inappropriate application of clinical endpoints. Method Validation Requirements
Biomarker assay results tend to be more robust than
clinical endpoint measures, but they must be based on Create uniform instrument specifications
both disease and therapeutic mechanisms.23,24 Document maintenance and calibration
From a drug discovery and development perspec- Require study participants to use same instrument
tive, clinical endpoint measurements such as visual Create consistent/controlled environment for testing
analogue scales, pulmonary function tests, and cogni- Implement uniform staff and study participant training
tive function tests that assess how a patient feels or procedures
functions should meet the same GLP-like acceptance Document training procedures (standard operating
criteria that are required for biochemical/molecular procedures)
Document that training and proficiency testing is complete
biomarker assay methods. Clinical endpoints must be
Implement quality control procedures
based on known mechanisms of action or the disease
Use replicate (n ≥2) measurements if possible
process that is being treated. As stated earlier, preci-
Use baseline and placebo-controlled measurements for
sion, reproducibility, and reliability are more impor-
reference
tant than accuracy for clinical endpoint measurement

DRUG DEVELOPMENT 337


COLBURN

Table IV Types of Validity to Evaluate Biomarkers

Content validity—measuring the right thing


Construct validity—the empirical relationship is consistent with the theoretical relationship
Criterion-based validity such as (1) concurrent—consistent measures, (2) predictive—later evidence supports,
and (3) prescriptive—used to select treatment

standard operating procedure sites are preferred during acids, peptides, proteins, or electrolytes that can be
early clinical development to optimize data that can used for preclinical and early clinical assessments.
provide insight into the disease process and the influ- Although preclinical models will probably not be
ences of therapeutic intervention.75-77 At the same time, directly applicable to the clinical setting, they establish
it will be important to be able to generalize the proce- a preliminary foundation for future clinical models.78
dures that have been used and the results that have For example, transgenic animals or other animal mod-
been obtained during the early phases to later els or in vitro tests that closely reflect human disease
multicenter trials if biomarkers are to continue to pro- should be used to start the creation of PK and/or PD
vide insight during later clinical phases of develop- models that will ultimately be tested in patients to
ment.6 Following specific validation guidelines can build clinical information and knowledge. This foun-
minimize site-to-site variability (Table IV). It is critical dation can then be expanded and modified to incorpo-
to ensure that biomarkers and clinical endpoints meet rate PK/PD model components that reflect human
both content and construct validity requirements. If pharmacokinetics, biology, physiology, and pharma-
biomarkers meet predictive validity criteria, they be- cology as development moves from Phase I through
come surrogate endpoints, and it is now becoming Phase III and finally into postmarketing. These PK/PD
more common to use prescriptive biomarkers to select modeling tools are also being built into regulatory re-
appropriate treatment. Appropriate selection of both view and approval processes.79-81
predictive and prescriptive biomarkers is dependent Biomarkers and PK/PD models will guide com-
on clinical endpoints that, themselves, are appropriate pound selection and retention. Even if the selected
and measured reliably. For example, it would be appro- biomarkers are never linked to clinical endpoints,
priate to select a treatment that reduces IL-4 or TNF-α mechanism-based biomarkers will improve early drug
concentrations if IL-4 or TNF-α concentrations are ele- discovery and development by simplifying decision-
vated in the patient and are mechanistically based in making processes. Only compounds with a high proba-
the disease process. In contrast, it is pointless to take bility of medical and commercial success will enter
this approach if IL-4 or TNF-α concentrations are in the Phase III. The cost of drug development will decrease
normal range or are not involved in the disease process in response to this more efficient and effective process.
(Figure 3). Although our current PK/PD models are not suffi-
ciently mechanism based for effective prospective pre-
ISSUES AND OPPORTUNITIES dictions,82-84 biomarkers, together with PK/PD model-
ing and simulation, will provide a continuous process
Comprehensive use of biomarkers or endpoints for PK/ to link what has been learned from today’s drug devel-
PD modeling has typically been initiated during clini- opment cycle to the next generation of biomarkers, as-
cal development. Starting to think about modeling dur- says, and models. Biomarkers, together with PK/PD
ing clinical development is too late. Plans to incorpo- modeling and simulation, will provide a foundation to
rate biomarkers, clinical endpoints, and PK/PD bridge effective, multidirectional communication.
modeling in drug discovery and development need to
occur when a new therapeutic target is being identified REFERENCES
and the discovery process is starting. High-throughput
screening targets can serve as initial sources of poten- 1. Biomarkers Definitions Working Group: Biomarkers and surrogate
tial biomarkers. For example, if the discovery process is endpoints in clinical trials: proposed definitions and conceptual
going to target a specific pathogen, enzyme, or receptor, framework. Clin Pharmacol Ther 2001;69:89-95.
these targets should serve as potential biomarkers or as 2. Prentice RL: Surrogate endpoints in clinical trials: definition and
producers of downstream biomarkers such as amino operational criteria. Stat Med 1989;8:431-440.

338 • J Clin Pharmacol 2003;43:329-341


BIOMARKERS IN DRUG DISCOVERY AND DEVELOPMENT

3. Levy G: Mechanism-based pharmacodynamic modeling. Clin 24. Meibohm B, Derendorf H, Mollmann H, Frohlich P, Tromm A,
Pharmacol Ther 1994;56:356-358. Wagner M, et al: Mechanism-based PK/PD model for the
4. Keegan P, Loughman BE: Early clinical trials of chemopreventive lymphocytopenia induced by endogenous and exogenous
and biologic agents: designs, populations and endpoints. Urology corticosteroids. Int J Clin Pharmacol Ther 1999;37:367-376.
2001;57(4, Suppl. 1):216-219. 25. Derendorf H, Lesko LJ, Chaikin P, Colburn WA, Lee P, Miller R,
5. Antoine JM, Diplock AT: Validation of markers for their relevance et al: Pharmacokinetic/pharmacodynamic modeling in drug research
to health and disease. Free Radic Res 2000;33(Suppl.):S117-S120. and development. J Clin Pharmacol 2000;40:1399-1418.
6. Colburn WA: Selecting and validating biologic markers for drug 26. Meibohm B, Derendorf H: Basic concepts of pharmacokinetic/
development. J Clin Pharmacol 1997;37:355-362. pharmacodynamic (PK/PD) modeling. Int J Clin Pharmacol Ther
1997;35(10):401-413.
7. Colburn WA: Surrogate markers and clinical pharmacology. J Clin
Pharmacol 1995;35:441-442. 27. Colburn WA, Lee JW: Biomarkers in PK/PD modeling and simula-
tion. Clin Pharmacokinet; requested lead article in review.
8. Rolan P: The contribution of clinical pharmacology surrogates and
models to drug development: a critical appraisal. Br J Clin Pharmacol 28. Sim J, Arnell P: Measurement validity in physical therapy re-
1997;44:219-225. search. Phys Ther 1993;73:102-115.
9. CAST Investigators (Cardiac Arrhythmia Suppression Trial Inves- 29. Danhof M: Applications of pharmacokinetic/pharmacodynamic
tigators): Preliminary report: effect of encainide and flecainide on research in rational drug development. Meth Find Exp Clin
mortality in a randomized trial of arrhythmia suppression after myo- Pharmacol 1996;18:53-54.
cardial infarction. N Engl J Med 1989;321:406-412. 30. Balant LP, Balant-Gorgia AE: Advantages and synergism of com-
10. Mildvan D, Landay A, DeGruttola V, Machado SG, Kagan J: An ap- bined pharmacokinetic and pharmacodynamic studies during drug
proach to the validation of markers for use in AIDS clinical trials. Clin development. Boll Chim Farm 1993;132:212-213.
Infect Dis 1997;24:764-774. 31. Sheiner LB, Steimer J-L: Pharmacokinetic/pharmacodynamic
11. Lee JW, Hulse JD, Colburn WA: Biochemical markers: maximizing modeling in drug development. Annu Rev Pharmacol Toxicol 2000;
their use in new drug development. Appl Clin Trials 1995;4(3):24-32. 40:67-95.
12. Colburn WA: Early decision making during new molecular entity 32. Gieschke R, Steimer J-L: Pharmacometrics: modelling and simu-
development. Appl Clin Trials 1997;5(10):44-55. lation tools to improve decision making in clinical drug develop-
ment. Eur J Drug Metab Pharmacokinet 2000;25:49-58.
13. Ledley FD: Can pharmacogenomics make a difference in drug de-
velopment? Nature Biotech 1999;17:731. 33. Colburn WA, Gibson DM: Endogenous agonists and
pharmacokinetic/pharmacodynamic modeling of baseline effects,
14. Bandara LR, Kennedy S: Toxicoproteomics: a new preclinical
in: Kroboth PD, Smith RB, Juhl RP (eds.), Pharmacodynamic Re-
tool. Drug Disc Today 2002;7:411-418.
search: Current Problems and Potential Solutions. Cincinnati, OH:
15. Weinstein JN: Searching for pharmacogenomic markers: the syn- Harvey Whitney Books, 1988;167-184.
ergy between omic and hypothesis-driven research. Dis Markers
34. Fermnandez-Ruiz J, Berrendero F, Hernandez ML, Ramos JA: The
2001;17:77-88.
endogenous cannabinoid system and brain development. Trends
16. Li J, Zhang Z, Rosenzweig J, Wang YY, Chan DW: Proteomics and Neurosci 2000;23:14-20.
bioinformatics approaches for identification of serum biomarkers to
35. Reis DJ, Regunathan S: Agmatine: an endogenous ligand at
detect breast cancer. Clin Chem 2002;48:1296-1304.
imidazole receptor is a novel neurotransmitter. Ann NY Acad Sci
17. Petricoin EF III, Ardekani AM, Hitt BA, Levine PJ, Fusaro VA, 1999;881:65-80.
Steinberg SM, et al: Use of proteomic patterns in serum to identify
36. Teuther L, Watjens F, Jensen LH: Ligands for the benzodiazepine
ovarian cancer. Lancet 2002;359:572-577.
binding site: a survey. Curr Pharm Des 1999;5:317-343.
18. Aarons L, Karlsson MO, Mentre F, Rombout F, Steimer JL, Van
37. Bouw MR, Gardmark M, Hammarlund-Udenaes M:
Peer A, Invited COST B15 Experts: Role of modelling and simulation
Pharmacokinetic-pharmacodynamic modelling of morphine trans-
in Phase I drug development. Eur J Pharm Sci 2001;13:115-122.
port across the blood-brain barrier as a cause of the antinociceptive
19. Colburn WA: Drugs and endogenous ligands compete for receptor effect delay in rats—a microdialysis study. Pharm Res 2000;17:1220-
occupancy. J Clin Pharmacol 1994;34:1148-1152. 1227.
20. Colburn WA: Optimizing the use of biomarkers, surrogate end- 38. Harashima H, Tsuchihashi M, Iidia S, Doi H, Kiwada H:
points and clinical endpoints for more efficient drug development. J Pharmacokinetic/pharmacodynamic modeling of antitumor agents
Clin Pharmacol 2000;40:1419-1427. encapsulated in liposomes. Adv Drug Deliv Rev 1999;40:39-61.
21. Parker TJ, Della Pasqua OE, Loizillon E, Chezaubernard C, 39. Nestorov IA: Sensitivity analysis of pharmacokinetic and
Jochensen R, Danhof M: Pharmacokinetic-pharmacodynamic mod- pharmacodynamic systems: a structural approach to sensitivity anal-
elling in the early development phase of anti-psychotics: a compari- ysis of physiologically based pharmacokinetic models. J
son of the effects of clozapine, S16924, S18327 in the EEG model in Pharmacokinet Biopharm 1999;27:577-597.
rats. Br J Pharmacol 2001;132:151-158.
40. Proudfoot J, Barden A, Mori TA, Burke V, Croth KD, Beilin LJ,
22. Food and Drug Administration: Draft Guidance for Industry: Ex- et al: Measurement of urinary F(2)-isoprostanes as markers of in vivo
posure-Response Relationships: Study Design, Data Analysis, and lipid peroxidation: a comparison of enzyme immunoassay with gas
Regulatory Applications [Online]. Available: http://www.fda.gov/ chromatography/mass spectrometry. Anal Biochem 1999;272:209-
cder/guidance/4614dft.pdf 215.
23. Puchalski TA, Krzyanski W, Blum RA, Jusko WJ: Pharma- 41. McShane LM, Dorgan JF, Greenhut S, Damato JJ: Reliability and
codynamic modeling of lansoprazole using an indirect irreversible validity of serum hormone measurements. Cancer Epidemiol
response model. J Clin Pharmacol 2001;41:251-258. Biomarkers Prev 1996;5:923-929.

DRUG DEVELOPMENT 339


COLBURN

42. Miller KJ, Bowsher RR, Celniker A, Gibbons J, Gupta S, Lee JW, 58. Harris EN, Pierangeli SS: Revisiting the anticardiolipin test and
et al: Workshop on bioanalytical methods validation for its standardization. Lupus 2002;11:269-275.
macromolecules: summary report. Pharm Res 2001;18:1373-1383. 59. Hocquette JF, Brandstetter AM: Common practice in molecular
43. Lee JW, Hulse JD, Colburn WA: Surrogate biochemical markers: biology may introduce statistical bias and misleading biological in-
precise measurement for strategic drug and biologics development. J terpretation. J Nutr Biochem 2002;13:370-377.
Clin Pharmacol 1995;35:464-470. 60. Charturvedi S, Jain N, Bhatia A: Evaluation of semi-quantitative
44. Findlay JWA, Smith WC, Lee JW, Nordblom GD, Das I, DeSilva methods for protein and sugar estimation in urine. Indian J Pathol
BS, et al: Validation of immunoassays for bioanalysis: a pharmaceuti- Microbiol 2001;44:399-401.
cal industry perspective. J Pharm Biomed Anal 2000;21:1249-1273. 61. Kumpel BM, Beliard R, Brossard Y, Edelmqan L, de Haas M, Jackson
45. Lee JW, Nordblom GD, Smith WC, Bowsher RR: Validation of DJ, et al: Section IC: assessment of the functional activity and IgG Fc
bioanalytical assays for novel biomarkers: practical recommenda- receptor utilisation of 64 IgG Rh monoclonal antibodies. Coordina-
tions for clinical investigation of new drug entities, in: Bloom J (ed.), tor’s report. Transfus Clin Biol 2002;9:45-53.
Biomarkers in Clinical Drug Development. New York: Marcel Dekker, 62. Rao S, Xia L: Strategies for genetic mapping of categorical traits.
2002. Genetica 2000;109:183-197.
46. Lee JW, Colburn WA: Immunoassay techniques, in: Ohannesian 63. Kumar-Singh S, Vermeulen PB, Weyler J, Segers K, Weyn B, Van
L, Streeter AJ (eds.), Handbook of Pharmaceutical Analysis. New Daele A, et al: Evolution of tumour angiogenesis as a prognostic
York: Marcel Dekker, 2002;225-312. marker in malignant mesothelioma. J Pathol 1997;182:211-216.
47. Ho LP, Davis M, Denison A, Wood FT, Greening AP: Reduced 64. Aguirre-Hernandez R, Farewell VT: A Pearson-type goodness-of-
interleukin-18 levels in BAL specimens from patients with asthma fit test for stationary and time-continuous Markov regression models.
compared to patients with sarcoidosis and health control subjects. Stat Med 2002;21:1899-1911.
Chest 2002;121:1421-1426.
65. Sheftel FD, Fox AW: Acute migraine treatment outcome mea-
48. Out TA, Jansen HM, Lutter R: Methodological aspects in the anal- sures: a clinician’s view. Cephalalgia 2000;20(Suppl. 2):14-24.
ysis of spontaneously produced sputum. Monaldi Arch Chest Dis
66. Bateman ED: Measuring asthma control. Curr Opin Allergy Clin
2001;56:493-499.
Immunol 2001;1:211-216.
49. Stelmach I, Jerzynska J, Kuna P: A randomised double-blind trial
67. Zagon IS, Verderame MF, McLaughlin PJ: The biology of the
of the effect of glucocorticoid, antileukotriene and beta-agonist treat-
opioid growth factor receptor (OGFr). Brain Res Brain Res Rev 2002;
ment on IL-10 serum levels in children with asthma. Clin Exp Allergy
38:351-376.
2002;32:264-269.
68. Cheng L, Wang LE, Spitz MR, Wei Q: Cryopreserving whole blood
50. Biller H, Bade B, Matthys H, Lutterman W, Virchow JC: Interferon- for functional assays using viable lymphocytes in molecular epide-
gamma secretion peripheral blood CD8+ T lymphocytes in patients miology studies. Cancer Lett 2001;166:155-163.
with bronchial asthma: in vitro stimulus determines cytokine pro-
duction. Clin Exp Immunol 2001;126:199-205. 69. Crowe AJ, Barton MC: Functional analysis of chromatin assem-
bled in synthetic nuclei. Methods 1999;17:173-187.
51. Camilla C, Mely L, Magnan A, Casano B, Prato S, Debono S, et al:
Flow cytometric microsphere-based immunoassay: analysis of se- 70. Sabatine MS, Morrow DA, de Lemos JA, Gibson CM, Murphy SA,
creted cytokines in whole-blood samples from asthmatics. Clin Rifai N, et al: Multimarker approach to risk stratification in non-ST
Diagn Lab Immunol 2001;8:776-784. elevation acute coronary syndrome: simultaneous assessment of
troponin I, C-reactive protein, and B-type natriuretic peptide. Circu-
52. Braybrooke JP, O’Bryne KJ, Propper DJ, et al: A phase II study of lation 2002;105:1760-1763.
Razoxane, an antiangiogenic topoisomerase II inhibitor, in renal cell
cancer with assessment of potential surrogate markers of 71. Buccheri G, Ferrigno D: Serum biomarkers of non-neuronendocrine
angiogenesis. Clin Cancer Res 2000;6:4697-4704. origin in small-cell lung cancer: a 16-year study on carcinoembryonic
antigen, tissue polypeptide antigen and lactate dehydrogenase. Lung
53. Toi M, Takayanagi S, Tominaga T: Association of vascular endo- Cancer 2000;30:37-49.
thelial growth factor expression with tumor angiogenesis and with
early relapse in primary breast cancer. Jpn J Cancer Res 1994;85: 72. Rubin MA, Dunn R, Strawderman M, Pienta KJ: Tissue
1045-1049. microarray sampling strategy for prostate cancer biomarker analysis.
Am J Surg Pathol 2002;26:312-319.
54. Toi M, Inada K, Suzuki H, Tominaga T: Tumor angiogenesis in
breast cancer: its importance as a prognostic indicator and the associ- 73. Bonassi S, Neri M, Puntoni R: Validation of biomarkers as early
ation with vascular endothelial growth factor expression. Breast predictors of disease. Mut Res 2001;480-481:349-358.
Cancer Res Treat 1995;36:193-204. 74. Veltri RW, Miller MC, An G: Standardization, analytical valida-
55. Rodriguez CR, Fei DT, Keyt B, Baly DL: A sensitive fluorometric tion, and quality control of intermediate endpoint biomarkers. Urol-
enzyme-linked immunosorbent assay that measures vascular endo- ogy 2001;57(4, Suppl. 1):164-170.
thelial growth factor165 in human plasma. J Immunol Methods 75. Holmvang L, Hawsbak P, Clemmensen P, Wagner G, Grnade P:
1998;219:45-55. Differences between local investigator and core laboratory interpre-
56. Blijenberg BG, Storm BN, Kruger Boeken AE, Schröder FH: On tation of the admission electrocardiograph in patients with unstable
the standardisation of total prostate-specific antigen: an exercise angina pectoris or non-W-wave myocardial infarction (a thrombin in-
with two reference preparations. Clin Chem Lab Med 1999;37:545- hibition in myocardial ischemia [TRIM] substudy). Am J Cardiol
552. 1998;82:54-60.
57. Jung K, Brux B, Lein M, Rudolph B, Kristiansen G, Hauptmann S, 76. Tuinenburg JC, Koning G, Hekking E, Desjardin C, Harel F,
et al: Molecular forms of prostate-specific antigen in malignant and Bilodean L, et al: One core laboratory at two international sites: is that
benign prostatic tissue: biochemical and diagnostic implications. feasible? An inter-core laboratory and intra-observer variability
Clin Chem 2000;46:47-54. study. Catheter Cardiovasc Interv 2002;56:333-340.

340 • J Clin Pharmacol 2003;43:329-341


BIOMARKERS IN DRUG DISCOVERY AND DEVELOPMENT

77. Salani D, Saini S, Fatuga GA, Halpern EF, Lanser ME, 81. Gueyffier F, Dib M, Boissel JP: Biological markers: utilization in
Zimmerman JB, et al: Quantitative measurements of medical images drug development and approval (French). Therapie 2001;56:355-
for pharmaceutical clinical trials: comparison between on-site and 361.
off-site assessments. Am J Rheumatol 2000;174:1159-1162. 82. Kimbo HC, Reele SSB, Holford NHG, Peck CC: Prediction of the
78. Obach RS, Baxter JC, Liston TC, Silber BM, Jones BC, MacIntyre F, outcome of a phase 3 clinical trial of an antischizophrenic agent
et al: The prediction of human pharmacokinetic data from preclinical (quetiapine fumarate) by simulation with a population pharma-
and in vitro metabolism data. J Pharmacol Exp Ther 1997;283:46-58. cokinetic and pharmacodynamic model. Clin Pharmacol Ther 2000;
79. Machado SG, Miller R, Hu C: A regulatory perspective on 68:568-577.
pharmacokinetic/pharmacodynamic modelling. Stat Meth Med Res 83. Veyat-Follet C, Bruno R, Olivares R, Rhodes GR, Chaikin P: Clini-
1999;8:217-245. cal trial simulation of docetaxel in patients with cancer as a tool for
80. Lesko LJ, Atkinson AJ Jr: Use of biomarkers and surrogate end- dosage optimization. Clin Pharmacol Ther 2000;68:677-687.
points in drug development and regulatory decision making: criteria, 84. Holford NHG, Kimko HC, Monteleone JPR, Peck CC: Simulation
validation, strategies. Ann Rev Pharmacol Toxicol 2001;41:347-366. of clinical trials. Ann Rev Pharmacol Toxicol 2000;40:209-234.

DRUG DEVELOPMENT 341

You might also like