Вы находитесь на странице: 1из 25

C H A P T E R

17
Biomarkers in Nonclinical Drug Development
A.D. Aulbach, C.J. Amuzie

O U T L I N E

Biomarkers in Nonclinical Safety Assessment 448 Clusterin457


Cystatin C 457
Biomarker Validation, Qualification, and Application
Enzymuria458
in Nonclinical Studies 449
Glomerular Injury Biomarkers 458
Biomarker Qualification 449
Total Protein, Albumin, and Microglobulins 458
Biomarker Assay Kits 449
Future Directions 458
Fit-for-Purpose (Analytical) Validation 450
Biomarker Application in Nonclinical Studies 451 Cardiac and Skeletal Muscle Injury Biomarkers 458
Lab-to-Lab and Method-to-Method Diversity 451 Biomarkers of Cardiac Structural Injury and Necrosis 458
Biomarkers of Cardiac Functional Alterations 460
Biomarkers of Liver Injury 451
Biomarkers of Skeletal Muscle Injury 460
Traditional Biomarkers 452
Biomarkers of Hepatocellular Injury 452 Vascular Injury Biomarkers 461
Alanine Transaminase 452
Biomarkers of the Immune System 461
Aspartate Transaminase 453
Indicators of Immunosuppression 462
Glutamate Dehydrogenase 453
Hematology and Clinical Chemistry 462
Sorbitol Dehydrogenase 453
Increased Incidence of Tumors 462
Total Bile Acids 454
Increased Incidence of Infections 462
Biomarkers of Hepatobiliary Injury 454 Histopathology and Lymphoid Organ Weight 463
Alkaline Phosphatase 454 Immune Function Studies 463
Total Bilirubin 454 Primary Antibody Assays 463
γ-Glutamyl Transferase and Other Hepatobiliary Natural Killer Cell Activity 464
Markers454 Macrophage Function 464
Emerging Biomarkers 454 Indicators of Adverse Immunostimulation 464
Glutamate Dehydrogenase 455 Acute-Phase Proteins 464
Malate Dehydrogenase 455 Cytokine Evaluation 464
Paraoxonase-1455 Indicators of Hypersensitivity 465
Purine Nucleoside Phosphorylase 455 Indicators of Immunogenicity 466
Other Emerging Biomarkers of Liver Injury 455 Indicators of Autoimmunity 466
Biomarkers of Toxicity in the Central Nervous System 466
Renal Injury Biomarkers 456
Biomarkers of Toxicity in the Reproductive System 467
Tubular Injury Biomarkers 456
Kidney Injury Molecule-1 Conclusion467
(Kim-1/TIM-1/HAVCR-1)456
References468
Neutrophil Gelatinase-Associated Lipocalin or
Lipocalin-2457

A Comprehensive Guide to Toxicology in Nonclinical Drug Development, Second Edition 447


http://dx.doi.org/10.1016/B978-0-12-803620-4.00017-7 © 2017 Elsevier Inc. All rights reserved.
448 17.  BIOMARKERS IN NONCLINICAL DRUG DEVELOPMENT

BIOMARKERS IN NONCLINICAL SAFETY magnitude of acetaminophen toxicity or the effective-


ASSESSMENT ness of acetaminophen antidote, N-acetyl cysteine. This
NAPQI example illustrates that a single biomarker can
The use of biomarkers in nonclinical drug develop- provide a picture of both exposure and effect of the test
ment covers a broad range of disciplines, modalities, article, highlighting that biomarker categories can over-
and development stages that extend from the identi- lap and should be contextually understood in order to
fication and assessment of drug targets to issue reso- be meaningfully applied.
lution during the clinical trial of investigative drugs. Biomarkers of susceptibility are specific genotypes in
Biomarkers became more applicable in drug devel- animals or people that make them more prone to a partic-
opment after the complete sequencing of the human ular disease or more likely to be affected by a particular
genome and the expansion of omics technologies that therapy. Warfarin (Coumadin) is an anticoagulant used
followed. The National Institutes of Health (USA) has to manage thrombotic disorders and acts by inhibiting
defined biomarkers as a characteristic that is objec- vitamin K-dependent clotting factors. Specific alleles of
tively measured as an indicator of biological processes the gene that encode the metabolizing cytochrome P-450
or pharmacological responses [1]. When adapted to enzyme (CYP2C9*2 and CYP2C9*3) are associated with
the context of nonclinical drug development, a bio- reduced metabolism of Warfarin, a higher circulating
marker may be defined as any specific parameter that drug concentration, and an increased risk for bleeding
is objectively measured and evaluated as an indicator disorders in patients that are taking Warfarin [5]. The
of normal biological processes, pathologic processes, or genotyping of patients provides information from these
pharmacologic responses to a drug or test article, which alleles, from which patients are ranked according to their
helps to diagnose or monitor a disease and/or the out- susceptibility to bleeding disorders while taking Warfa-
come of therapeutic intervention [2,3]. rin. Therefore patients with thrombotic disorders who
In safety assessment, biomarkers are most commonly also have these alleles are given lower doses of Warfarin
used for monitoring early signs of toxicity; as such they to mitigate this risk. The understanding of biomarkers of
are often called “safety” biomarkers in nonclinical and susceptibility and their application to a particular drug
clinical environments. However, scientists in drug dis- or target is useful in selecting the correct animal model
covery are aware that biomarkers are used in a broader of disease and/or safety in drug discovery and develop-
context than “safety” and can sometimes be selected as ment, understanding toxicokinetics in the appropriate
a molecular index to evaluate the efficacy of new ther- haplotype, and selecting the right patient population for
apeutic entities. For example, the human epidermal early clinical trials.
growth factor receptor (HER2) is a protein in 20% of Pharmacodynamic (efficacy) or toxicity (safety) bio-
breast cancers and its presence is associated with very markers are essentially biomarkers of effect. They indi-
aggressive tumor behavior, yet it is a target for several cate that a specific alteration or injury has occurred in a
effective drugs targeting breast cancer [4]. This example receptor, tissue compartment, or body fluid after a test
illustrates that a discussion on the utility of biomarkers article has been administered. The ideal biomarker of
should be broad to include their physiology, expose the effect is a characteristic that can be objectively measured
range of possible applications, and also keep the appli- as an index of test article-related alterations in a mini-
cable details to their most common uses. mally invasive way. Therefore biomarkers that can be
There are three broad categories of biomarkers in obtained in body fluids (saliva, urine, blood, and plasma)
nonclinical drug development based on the drug expo- or imaging are generally preferred to those that can be
sure–effect continuum. They are biomarkers of expo- obtained by biopsies or other invasive techniques. The
sure, susceptibility, and effect. Most of the discussion identification of biomarkers of effect requires a thorough
in this chapter will be focused on biomarkers of effect, understanding of the effect pathway in a specific tissue
but the other categories will be discussed briefly. A bio- or the signaling pathway and their interaction(s) with
marker of exposure is the administered test article or a other systems within the whole organism. For example,
metabolite that is measured in a specific tissue or body growth hormone is released from the anterior pituitary
fluid as evidence that the animal or human biological and binds to growth hormone receptors in the liver to
system was exposed to the test article. For example, produce an elevation of insulin-like growth factor-1
acetaminophen is an oral analgesic that is metabolized (IGF-1) and its acid-labile binding subunit (IGFALS) in
to a reactive intermediate N-acetyl-p-benzoquinone the plasma. Therefore molecules that alter growth or
imine (NAPQI). Both acetaminophen and NAPQI can growth hormone signaling, such as the toxin deoxyniva-
be measured as biomarkers of exposure to help investi- lenol, are also expected to change the levels of IGF-1 or
gators understand acetaminophen exposure and toxic- IGFALS in plasma, and these circulating proteins can be
ity. Note that in acetaminophen overdose, NAPQI levels used as biomarkers to determine the effect of a xenobi-
are used more like a biomarker of effect to monitor the otic on growth hormone signaling [6]. In general, a good

III.  CLINICAL PATHOLOGY, HISTOPATHOLOGY, AND BIOMARKERS


Biomarker Validation, Qualification, and Application in Nonclinical Studies 449
understanding of the circulating kinetics of the test arti- candidate. Submission of qualification data is largely
cle and the biomarker is absolutely necessary to properly driven by international consortiums such as the Predic-
understand and interpret any biomarker of effect data. tive Safety Testing Consortium (PSTC), the International
Life Sciences Institute—Health and Environmental Sci-
ences Institute (ILSI-HESI), whose work ultimately leads
BIOMARKER VALIDATION, to the discovery, critical evaluation, and application of
QUALIFICATION, AND APPLICATION IN promising new biomarker candidates. Once qualified,
NONCLINICAL STUDIES results from specific biomarkers are then officially “con-
sidered” by the endorsing regulatory body as part of
It is clear that biomarkers are playing an increasingly nonclinical data submission packages under a very spe-
important role in pharmaceutical development and are cific set of circumstances as outlined by the agency. That
receiving increased attention from both regulators and said, biomarkers that have not been officially qualified by
industry. Although there are many biomarkers that the FDA, but have demonstrated repeated utility in non-
would be considered “traditional,” in the sense that they clinical settings, are still routinely applied to nonclinical
have been in use for many years and utilize standardized safety data packages to provide additional perspective.
methodologies, most of the newly emerging biomark-
ers discussed in this section are quantified by relatively
novel immunoassay methodologies, often using propri- Biomarker Assay Kits
etary commercially available assay kits. Many of these Unlike traditional clinical pathology, clinical chem-
kits and their associated reagents are not consistently istry, and other well-established testing methods, com-
regulated, monitored, or standardized across indus- mercially available first-to-market biomarker kits often
try. These factors result in inconsistent and sometimes use ligand-based (immunoassay) testing methods and
improper use and application of biomarkers across dif- reagents that vary greatly in their quality, performance,
ferent organizations/laboratories and emphasize the and overall value. Despite the availability of a multitude
importance of stringent validation and assay character- of commercial kits from numerous vendors, the extent of
ization practices. In this section some of the challenges in assay characterization and critical reagent performance
biomarker validation and their application in nonclinical of these assays is often insufficient to support their use.
toxicology studies are reviewed. Because these products are distributed as “research use
only,” there is essentially no oversight over the manufac-
ture, distribution, or marketing of such products. These
Biomarker Qualification kits are often marketed for the broadest possible use and
The need for high-quality biomarkers that are robust, are often not designed for specific applications in which
predictive, and cost effective has been thoroughly dis- they are used (ie, cross-species, multiple matrices, insuf-
cussed by a variety of industry and regulatory groups for ficient stability). Additionally, the specific techniques
a number of years [7–11]. Regulatory organizations like and performance assessment procedures used by the
the Food and Drug Administration (FDA), the Organ- end-users prior to undertaking sample testing varies
isation for Economic Co-operation and Development greatly. These factors can negatively impact the quality
(OECD), and the European Medicines Agency (EMEA) of biomarker data used in making key decisions during
have engaged in a dialogue with the pharmaceutical drug development. Hence when using novel biomarkers
industry on what they deem necessary to develop a to generate nonclinical data, it is critical to obtain assay
qualified biomarker. The term “qualification” should not kits from reputable vendors, execute a robust site-spe-
be confused with the term “validation,” since the latter cific fit-for-purpose performance characterization, and
represents the performance characterization of a method maintain consistency of the materials and methods used
or assay. In a regulatory context, qualification represents throughout a nonclinical program.
the endorsement of a particular biomarker by a regula- Two main categories of commercial biomarker kits
tory body under a very specific set of conditions (eg, are used in drug development and research purposes:
“…may be used for the characterization of renal effects in vitro diagnostic assays (IVD) and research use
in rats…”). The FDA has established a Biomarker Quali- only (RUO) assays. An understanding of these cate-
fication Review Team (BRQT) that provides evaluations gories and their inherent differences is important to
of qualification data for biomarkers submitted as part of gain insight into whether or not a given assay may
IND (investigational new drug)/NDA (new drug appli- be suitable for the intended use. IVD assays make up
cation) applications to the FDA [12]. The BRQT aims to a large majority of the traditional clinical pathology
ensure that biomarkers are integrated in the review pro- assays as well as a number of the newer biomarker
cess and to encourage development of biomarkers that kits. These assays have been approved by the FDA
add value to the development program of a given drug (defined in 21 CFR 809.33) for intended use in human

III.  CLINICAL PATHOLOGY, HISTOPATHOLOGY, AND BIOMARKERS


450 17.  BIOMARKERS IN NONCLINICAL DRUG DEVELOPMENT

clinical diagnosis or patient care and undergo intense in which there was a known cardiac liability. Briefly,
screening/monitoring, and regulation as to how these the key elements included in a fit-for-purpose valida-
materials are manufactured, distributed, and used in tion include preparation of a validation plan, testing
laboratories [13]. For these reasons, traditional clinical of dynamic range, intra- and interassay precision and
pathology assays and IVD biomarker tests have been accuracy, dilutional linearity, parallelism, establish-
better characterized, standardized from lab-to-lab, and ment of quality control (QC) samples in species-specific
generally have better performance characteristics than matrix, normal baseline ranges from species of interest
novel first-to-market biomarker assays. Conversely, in matrix, and short/long-term stability in matrix. In
the large majority of commercially available novel bio- accordance with the fit-for-purpose approach, the over-
marker kits used in nonclinical laboratories to which all level of rigor (ie, number of runs, length of stability
most of this chapter is devoted fall under the RUO cat- testing, standard curve points, lot-to-lot variability test-
egory. Assays that are labeled RUO are exempt from ing, etc.) is determined by the level of confidence one
the regulatory requirements and approvals needed puts into the data being generated. For a more detailed
for clinical diagnosis or patient management. They and comprehensive discussion of an analytical fit-for-
are intended for use only in research and discovery purpose validation readers are directed to the refer-
work and since there are no rigorous requirements or ences discussed in this section.
guidelines for the RUO label, the extent of manufac- Recommendations for fit-for-purpose biomarker
turing quality compliance, assay characterization, and assay development and validation have been described
documentation varies considerably across different for immunoassays developed de novo at pharmaceuti-
vendors and from assay to assay. Hence most novel cal companies and contract research laboratories [7,8].
biomarker assays are not standardized between labo- However, specific regulatory guidance has been sparse
ratories and need to have a robust performance char- and incomplete, which does not help the pharmaceutical
acterization/validation prior to use. end user determine the appropriate practices to select,
adapt, validate, and ensure long-term supplies of reliable
commercial biomarker assay kits for drug development
Fit-for-Purpose (Analytical) Validation purposes. The most comprehensive of the nonbinding
The term validation is used in a multitude of dif- recommendations on the subject are found in the 2013
ferent contexts, sometimes inappropriately, and often Bioanalytical Method Validation, which that states the
interchangeably with the term qualification. Gener- following points regarding commercial biomarker assay
ally the term validation represents the actual ana- validation [14]:
  
lytical performance characterization of a method or
• T he performance of diagnostic kits should be
assay by executing various experiments (eg, spike
assessed in the facility conducting (site-specific) the
and recovery, dilutional linearity, stability, etc.) to
sample analysis.
gain an understanding of the ability of the assay to
• Diagnostic kit validation data provided by the
accurately quantitate the analyte of interest. For many
manufacturer may not ensure reliability of the kit for
years industry investigators went without a consistent
drug development purposes.
approach to this process, which led to disharmony and
• Specificity, accuracy, precision, and stability should
confusion between laboratories, particularly when
be demonstrated under actual conditions of use.
attempting to compare results between different labs.
• Quality control (QC) samples with known
Eventually leading ligand-binding scientists began to
concentrations should be prepared and used,
harmonize this process by promoting the fit-for-pur-
independent of the kit-supplied materials.
pose validation model.
• Standards and QCs should be prepared in the same
The fit-for-purpose paradigm has been in use for over
matrix as the subject samples.
a decade now and was initially described in 2006 with
• If multiple kit lots are used within a study, lot-to-lot
an intention to provide a framework for the validation
variability and comparability should be addressed
and performance characterization of ligand-binding
for critical reagents.
(immunoassay) biomarker kits [8]. The core principle   

of the fit-for-purpose approach surrounds the notion Although these guidelines remain somewhat vague,
of executing a performance characterization/valida- they are consistent with most published fit-for-purpose
tion equal in robustness to the intended use of the data documents as well as those inherent to bioanalytical
being generated. For example, the level of robustness method validation practices, which share many features
of a validation for a renal biomarker assay being used of immunoassay biomarker methodologies. Currently,
during an early non-GLP discovery phase would not be most laboratories conducting biomarker analysis have
as great as the validation of a cardiac biomarker being adopted some form of the fit-for-purpose validation
used during a 13-week GLP study for a compound model when establishing new methods.

III.  CLINICAL PATHOLOGY, HISTOPATHOLOGY, AND BIOMARKERS


Biomarkers of Liver Injury 451

Biomarker Application in Nonclinical Studies For example, in the case of inflammation, IL-6 (6–24 h),
CRP (1–2 days), and fibrinogen (2–3 days) are all posi-
Inherent to the use of biomarker assays in toxicology tive acute phase reactants that will increase in response
studies is an understanding of the identification and to the same inflammatory stimulus. However, each will
interpretation of a positive biomarker signal. We have have a different window in which you can capture their
discussed the relative diversity of assays, levels of perfor- increases (listed in parentheses). Attempting to draw
mance, and lack of standardized methods for biomarker blood at 3 days postdose for a cytokine like IL-6 will gen-
assays. All of these factors contribute to the difficulty in erally result in no positive signal, which can lead to mis-
identifying a positive signal, which must be distinguished characterization of the safety profile for the compound.
not only from background biologic variability, but also
from the analytical “noise” that is generally much higher
than traditional clinical pathology tests. Thus it remains Lab-to-Lab and Method-to-Method Diversity
crucial for the interpreting scientist to understand the One final important feature in the application and
inherent variation of the specific assay/reagent set based interpretation of biomarkers in nonclinical studies is
on the analytical validation data, as well as the biologic related to the lack of standardization between laborato-
variation, which can be determined by establishing his- ries and methods. For any given biomarker, the methods
torical control data sets for the intended reference pop- and reagents used for quantification will vary by labora-
ulation. Once a positive signal has been identified, a tory and by kit or platform. This results in a number of
thorough understanding of what a positive signal repre- different methods being used by different laboratories
sents is fundamental to accurate drug hazard identifica- to measure the same biomarker. Objective comparisons
tion. For example, for cardiac biomarkers, some markers between methods/kits from different laboratories have
are indicators of direct cardiac myocyte injury (troponins) been made for a number of routinely used biomarker
while others are indicators of a functional defect (natri- assays, and have generally demonstrated the lack of con-
uretic peptides). The renal biomarker NGAL (neutrophil cordance between them. A recent study comparing renal
gelatinase-associated lipocalin) is an indicator of renal biomarkers across different platforms revealed differ-
tubular injury, but can also be seen in cases of systemic ences in individual biomarkers that ranged up to 15-fold
and urinary tract inflammation [15]. and showed that these differences were probably due
A key feature to interpreting data from novel biomark- to the use of different antibodies with varying degrees
ers is to have an appreciation for the heterogeneity of pos- of affinity and specificity [17]. This phenomenon is not
itive biomarker response signals in timing, magnitude, uncommon and emphasizes the importance of establish-
and by species. Every individual biomarker will have its ing laboratory/method-specific reference ranges.
own unique signature in response to a specific compound
and pathophysiologic mechanism. For example, in the
dog, compound A may induce a 10-fold increase in car- BIOMARKERS OF LIVER INJURY
diac troponin I (cTnI) within 24 h, but with compound B
the response is twofold and isn’t seen until 72 h postdose. The liver is a critical organ because of its broad role
If this same example is used, replacing cTnI with another in nutrient homeostasis, as well as metabolism/detoxi-
commonly used cardiac biomarker like proBNP, the mag- fication and excretion of xenobiotics or endogenous
nitudes and timing would be completely different that compounds. Examples of liver function are synthesis of
those observed with cTnI. These differences are related to albumin and blood clotting factors, synthesis and storage
the specific pathology induced by the compounds both of glucose, use and recycling of bilirubin and cholesterol,
in timing and severity, and what each biomarker actually metabolism of hormones, and detoxification of various
represents pathologically. In addition, some species may drugs. In order to accomplish these functions the liver
show different magnitudes or even nonexistent responses is organized into lobular units that comprise cords of
to injury when using certain biomarkers. The best exam- hepatocytes that are separated by sinusoidal spaces. The
ples of this phenomenon are seen with the acute-phase sinusoids contain spaced-out (fenestrated) endothelial
proteins (eg, CRP, α2-macroglobulin, serum amyloid A, cells and resident macrophages (Kupffer cells) as well as
etc.). These biomarkers show a large degree of species- stellate cells that reside in small spaces (Space of Disse)
specific diversity among their individual responses to between the hepatocyte cords and sinusoidal basement
the same inflammatory stimuli [16]. All of these factors membrane. Within each lobular cord, individual hepa-
emphasize the importance of correlating biomarker data tocytes are separated by bile canaliculi, which are useful
with study data generated from more traditional end- for bile transport and recycling. Each liver lobule has a
points (eg, pathology, clinical pathology). portal (periportal) blood input that supplies the peripor-
Another important concept is to have an understand- tal hepatocytes with the most oxygenated blood within
ing of the specific timing of individual biomarker signals. the lobule, hence the periportal area has been referred to

III.  CLINICAL PATHOLOGY, HISTOPATHOLOGY, AND BIOMARKERS


452 17.  BIOMARKERS IN NONCLINICAL DRUG DEVELOPMENT

as Zone 1—the highest oxygen tension within a hepatic interpretation is outside the scope of this chapter and
lobule. A central (centrilobular) vein with least oxygen- has been addressed elsewhere [19–21]. It is important to
ated blood (Zone 3) is at the opposite end of the peripor- remember that there may be a biologically and/or sta-
tal blood supply within a liver lobule, while the midzone tistically significant alteration in markers of liver injury
(Zone 2) is an intermediate zone of oxygenation that without a corresponding effect on the indices of liver
resides between Zones 1 and 3. The gradient of metabo- function, and vice-versa.
lizing enzymes such as cytochrome p450 is opposite to There are increasing numbers of in vitro systems that
that of oxygenation, so hepatocytes around the central are useful for prioritizing chemical and biological entities
vein (Zone 3) have the most metabolizing enzymes, while in development. These in vitro systems use alterations
those around the periportal area usually have the least in several molecular markers to predict toxicity. They
metabolizing enzymes. This simplified understanding of are predominately used in non-GLP discovery settings
the liver architecture, oxygen, and metabolizing enzyme and will not be discussed specifically in this chapter. The
gradients is frequently used to understand and interpret discussion of biomarkers in this section will be largely
patterns of liver injury in toxicology. For example, the focused on liver injury that occurs in vivo in integrated
liver injury related to acetaminophen overdose is often animal systems. Currently there are two broad classes
driven by the reactive intermediate N-acetyl-p-quinone of liver injury biomarkers, called traditional and novel/
imine and is predominately centrilobular because of the emerging biomarkers. The traditional markers are most
high concentration of metabolizing enzymes that create often routine clinical chemistry markers, most of which
the reactive intermediate around the central vein. On the are part of a standard drug development program and
other hand, injury related to iron toxicity/overdose is will be described in detail below. Most of the emerging
predominately periportal because iron toxicity involves biomarkers and their associated methods have not been
redox cycling of ferrous and ferric iron, which occurs standardized or fully qualified but are currently being
more in the zone with higher oxygen tension. The zoning used on a case-by-case basis to understand or further
of injury within the liver is a useful and relatively com- characterize xenobiotic-related liver injury.
mon practice in safety assessment because it can help
identify potential mechanism(s) of injury and is useful Traditional Biomarkers
for investigative toxicologists who design experiments
to further understand toxicity or characterize the injury. The current best practice recommendation for non-
Drug-induced liver injury (DILI) is among the leading clinical safety assessment is that a minimum of four
causes of pharmaceutical withdrawals from the market serum parameters should be used to assess hepatocellu-
in the last three decades [18,19], and these withdrawals lar (minimum of two markers) and hepatobiliary (mini-
are impactful for the patients who need/rely on the drug mum of two markers) injury [19]. Any two markers from
and the drug maker who often has large economic loses alanine transaminase (ALT) activity, aspartate trans-
and liabilities. Efforts to use biomarkers for improved aminase (AST) activity, sorbitol Dehydrogenase (SDH)
prediction of DILI in nonclinical safety assessment are activity, and glutamate dehydrogenase (GLDH) will
increasing in scope and complexity. In order to appreci- assess hepatocellular injury, while any two from alkaline
ate the scope of biomarkers of liver injury, we need to phosphatase (ALP) activity, gamma-glutamyltransferase
remember that xenobiotic-related liver injury may take (GGT) activity, total bilirubin (TBILI), total bile acids
several forms and involve different cell types depending (TBA), and 5′-nucleotidase will assess hepatobiliary
on the chemical stimuli, dose, and/or duration of expo- injury. Among these serum parameters, ALT, AST, ALP,
sure. Examples of key biological events in different types and TBILI are also used clinically to identify liver injury
of toxic liver injury are cell death, alteration of canalicu- in humans, so they tend to be used relatively more fre-
lar patency and function, disruption of cytoskeleton, quently. It is important to note that these markers vary
increased accumulation of lipid molecules, alteration of in their utility among nonclinical species. Therefore the
sinusoidal patency and function, activation of Kupffer proper selection of liver injury markers for each program
cells and inflammation, hapten-mediated liver injury, is a thoughtful deliberation that often involves consulta-
and increased fibrosis/collagen deposition. tion with a qualified and experienced pathologist.
Several traditional markers indicate liver injury in
both clinical and nonclinical species. Some of these Biomarkers of Hepatocellular Injury
markers may be occasionally and often incorrectly
termed “liver function tests,” or LFTs. The concepts of Alanine Transaminase
liver injury and liver function need to be carefully delin- Alanine transaminase (ALT), which may be referred
eated in order to understand and communicate toxic to in other literature as alanine aminotransferase (ALAT)
responses of the liver to injury. The details of such delin- or serum glutamate-pyruvate transaminase (SGPT),
eation during nonclinical safety data acquisition and is found in blood and many tissues. ALT catalyzes the

III.  CLINICAL PATHOLOGY, HISTOPATHOLOGY, AND BIOMARKERS


Biomarkers of Liver Injury 453
transfer of an amino group from alanine to alpha-keto- necessary for nonclinical predictive testing, hence the
glutarate to yield glutamate and pyruvate as a part of need for emerging biomarkers, discussed below.
amino acid metabolism and gluconeogenesis. Dam-
aged hepatocytes leak their ALT into the extracellular
Aspartate Transaminase
space and ultimately plasma, so that ALT activity and/
or amount will be increased in animals with damaged Aspartate transaminase (AST), which may also be
hepatocytes when compared to those with normal referred to as aspartate aminotransferase or serum
hepatocytes. Among the traditional markers of hepato- glutamic-oxaloacetic transaminase (SGOT) in other lit-
cellular injury, ALT is considered to be a sensitive and erature, catalyzes the reversible transfer of amino group
translatable indicator of hepatocellular in the common between aspartate and glutamate. Like ALT, AST is
preclinical species. Serum ALT activity is the most fre- found in the cytoplasm of hepatocytes and other tissues,
quently relied upon indicator of hepatotoxic effects of including skeletal muscle. Injury to hepatocytes causes
drugs [22], although it does not always correlate well leakage of AST into the extracellular compartment with
with histopathology data [23]. Increases in ALT activ- subsequent elevation in serum AST activity. AST activity
ity may be observed with enzyme induction in rats and may also be elevated in times of skeletal muscle injury
dogs [24,25] or muscle injury [19]. Furthermore, there in preclinical species. The magnitude of ALT elevation
may be instances of hepatic injury where ALT activity is usually greater than AST when both are elevated due
is not elevated due to inhibitory factors such as Vitamin to hepatocellular injury because of the longer half-life
B12 deficiency [26], or interference by the presence of of ALT and the greater fraction of AST that is bound to
pyridoxal-5′-phosphate inhibitors such as isoniazid or the mitochondria [29]. The practice of considering a high
lead [23]. Due to the nonspecificity of ALT to hepatocel- ratio of AST/ALT to be more indicative of skeletal mus-
lular injury, ancillary clinical chemistry and/or histopa- cle injury [23] has been suggested and may aid in dis-
thology tests are often used to help interpret ALT values. tinguishing muscle versus liver injury. Animal restraint
In human clinical trials, it is an acceptable and recom- procedures leading to mild muscle trauma may also ele-
mended practice to interpret a greater than 3× elevation vate AST activity in nonclinical studies [30]. Given the
of ALT above the upper limit of normal (ULN) combined nuances associated with interpreting leakage enzymes in
with TBILI elevation of greater than 2× above ULN as the liver, careful integration of enzyme kinetics, enzyme
indicative of severe injury with/without any other evi- half-lives, and all other data by well-qualified personnel
dence. This practice in human clinical medicine is called is very important in the practice of safety assessment.
Hy’s Law. A related recommendation for ALT interpre-
tation in the absence of correlative histopathology in a
Glutamate Dehydrogenase
nonclinical setting is the biomarker of two rule, which
considers an ALT elevation of 3× ULN combined with Glutamate dehydrogenase (GLDH) is a mitochondrial
a significant alteration in another liver injury biomarker enzyme that reversibly converts glutamate to alpha-
as a sign of liver injury [27]. In general, caution should ketoglutarate as part of the urea cycle. GLDH activity
be exercised when applying human clinical recommen- increases with liver injury and the magnitude of eleva-
dations to preclinical species because enzymes such tion may be higher and longer lasting when compared
as TBILI that reflect biliary function in humans do not to ALT [31]. Unlike ALT and AST, compounds that inter-
accurately reflect liver function in all preclinical species fere with pyridoxal-5′-phosphate are less likely to alter
[26]. ALT may also have reduced sensitivity and specific- the GLDH activity assay [23]. Unlike ALT, serum GLDH
ity to liver injury in some species such as swine, guinea activity is neither affected by skeletal muscle damage
pigs, and in some strains of rats [19]. The interpretation nor induced by corticosteroids. GLDH is not currently
of ALT response in short-term nonclinical studies, espe- used routinely in nonclinical settings often due to the
cially in the absence of other indices of hepatic injury, limited availability of reliable reagents, but there may be
needs to consider the so-called “adapter” phenomenon instances where GLDH is an appropriate adjunct to help
where transient, nonclinically relevant ALT elevations delineate liver injury in preclinical species. Interest in
occur after the introduction of new drugs, but return to GLDH as a more specific nonclinical biomarker of hepa-
normal following continuous exposure [28]. Due to the tocellular injury in some species when compared to ALT
challenges associated with using ALT, some have sug- or AST is increasing and will be discussed further in the
gested an inclusion of immunoassay for ALT1, an ALT section “Emerging Biomarkers.”
isozyme that is more specific for the liver, as a measure
of hepatocellular injury, although these tests have not
Sorbitol Dehydrogenase
yet reached widespread acceptance on routine nonclini-
cal studies [23]. Since ALT has not always been predic- Sorbitol dehydrogenase (SDH) is present in several tis-
tive of DILI in the clinical setting another marker(s) is sues where it catalyzes the reversible oxidation reduction

III.  CLINICAL PATHOLOGY, HISTOPATHOLOGY, AND BIOMARKERS


454 17.  BIOMARKERS IN NONCLINICAL DRUG DEVELOPMENT

of sorbitol, fructose, and NADPH (nicotinamide adenine bilirubin production from erythrocyte destruction,
dinucleotide phosphate). SDH is a sensitive and specific altered bilirubin metabolism from drug-related inhibi-
indicator of acute hepatocellular injury in rodents, dogs, tion of metabolizing enzymes such as uridine diphos-
and nonhuman primates and has been reported as valu- phate glucuronosyltransferase, or faltering liver function
able in humans [23]. However, SDH has a short half- [34]. A careful and integrated assessment of all study
life (4 h in canine) and has limited stability compared data associated with hyperbilirubinemia is often needed
to most other clinical chemistry analytes requiring it to to identify a specific cause of TBILI elevations.
be analyzed as soon as possible after necropsy [27]. To
our knowledge, no large study has determined whether
γ-Glutamyl Transferase and Other Hepatobiliary
SDH adds any additional value to ALT quantitation.
Markers
γ-glutamyl transferase (GGT) is a liver canalicular
Total Bile Acids enzyme responsible for the transfer of glutamyl moi-
Total bile acids are important in the hepatic metabo- ety to other acceptors as part of gluthathione recycling,
lism of cholesterol and absorption of fat and fat-soluble and is present in other tissues. GGT is elevated through
vitamins. The levels of circulating bile acids are influ- induction in conditions of cholestasis and may be used
enced by diet and fasting but their elevation may also be to confirm ALP activity elevations that are associated
a sign of hepatocellular injury and/or functional change with cholestasis. In dogs and cats GGT is a less sensitive
in the liver [32]. TBA is a useful adjunct to assess the marker of cholestasis than ALP, but is more specific [19].
extent and consequence of liver injury because it is an Two other hepatobiliary markers, 5′-nucleotidase
index of hepatocellular function in preclinical species. (5′NT) and total bile acids (TBA), may be used to assess
biliary function in nonclinical studies. However, they do
not offer additional information when compared to ALP
BIOMARKERS OF HEPATOBILIARY and TBILI and will not be discussed further.
INJURY
Emerging Biomarkers
Alkaline Phosphatase
Due to the specificity issues with existing liver injury
Alkaline phosphatase (ALP) is a hydrolase that biomarkers such as ALT efforts are being made to iden-
removes the phosphate group from various proteins and tify additional biomarkers that are more specific and
nucleotides. ALP is a leading biomarker of hepatobiliary those that may provide additional information about
injury in common preclinical species. Serum ALP levels the zone of injury and/or affected cell types. Within the
increase when the patency of the bile duct is reduced, so last decade, proteomic approaches were used by inves-
ALP is widely used in nonclinical and human clinical set- tigators to identify a group of 19 putative liver injury
tings as a marker of cholestatic liver injury. Like ALT, ALP biomarkers from a mechanistically diverse group of hep-
is not specific for hepatobiliary injury as increased activity atototoxins based on early onset of biomarker alteration
of ALP may be seen in conditions of bone growth and dis- relative to time of liver [35]. The Hepatototoxcity Work-
ease, glucocorticoid treatment, and microsomal enzyme ing Group of the Predictive Safety Testing Consortium
induction [25,33]. Fluctuations of ALP need to be inter- (PSTC) has listed some promising classic and newly
preted cautiously in nonclinical settings because there is identified markers of injury for further qualification.
an intestinal isoform of ALP that drives a decrease in ALP The first set of biomarkers for qualification includes glu-
activity, particularly in rodents, during fasting or anorexia tamate dehydrogenase (GLDH), malate dehydrogenase
and a transient increase postprandially [19]. Specific ALP (MDH), paraoxonase-1 (PON1), and purine nucleoside
isoforms may be assessed to separate liver from bone and phosphorylase (PNP). PSTC also listed a second set of
intestinal sources, but isoform measurement does not biomarkers sorbitol dehydrogenase (SDH), arginase 1
offer any additional diagnostic information in preclinical (ARG-1), and gluthathione-s-transferase alpha (GSTα)
species compared to total ALP [19]. for qualification while a circulating microRNA marker
(miR-122) is being considered for qualification under the
third set. This decision is based, in part, on published
Total Bilirubin
evidence in specific liver injury situations indicating that
Total bilirubin is a yellowish-green breakdown prod- these biomarkers are likely to provide additional value
uct of hemoglobin from aging red blood cells. TBILI is in predicting liver injury. A robust qualification process
made of hepatic/conjugated and extrahepatic/uncon- is currently ongoing and some of these markers may
jugated sources. Elevations in TBILI are usually indica- become more routine in nonclinical testing panels after
tive of cholestasis, but may be a reflection of increased the qualification process.

III.  CLINICAL PATHOLOGY, HISTOPATHOLOGY, AND BIOMARKERS


Biomarkers of Hepatobiliary Injury 455

Glutamate Dehydrogenase because it decreases with injury. PON1 is currently under-


going qualification as a liver injury biomarker.
Glutamate dehydrogenase (GLDH) is a mitochon-
drial enzyme that is predominantly located in the
centrilobular (Zone 3) region of the liver lobule [36]. Purine Nucleoside Phosphorylase
GLDH is a classic serum enzyme like ALT and AST. Purine nucleoside phosphorylase (PNP) is an enzyme
Unlike ALT, serum GLDH activity is neither affected that catalyzes phosphorolysis of nucleosides in the
by skeletal muscle damage nor induced by cortico- purine salvage pathway. It is located in the cytoplasm
steroids, and is not sensitive to interference by pyri- of hepatocytes, Kupffer cells, and endothelial cells and
doxal phosphate inhibitors. GLDH activity increases is released into the sinusoids in times of hepatocellular
with liver injury and the magnitude of elevation may necrosis [23]. PNP has also been shown to be relevant
be higher and longer lasting when compared to ALT in clinical settings. PNP is a good biomarker candidate
[31]. However, in the past there has been limited avail- because it also has the possibility to detect nonparen-
ability of reliable GLDH reagents in the United States. chymal hepatic injury [26]. PNP is among the first set
Recently, GLDH was reported as a specific biomarker of markers that are currently being qualified for liver
of liver injury in a cohort of human clinical cases of injury.
drug-induced liver injury [37]. Furthermore, a human
reference interval has been established for GLDH from
a relatively large cohort. GLDH is emerging from a less Other Emerging Biomarkers of Liver Injury
commonly used classic biomarker to one that may be Arginase 1 (Arg-1) is a liver-specific hydrolase that
used more routinely to detect acute liver injury based catalyzes the breakdown of arginine to urea and orni-
on possible increases in sensitivity compared to ALT, thine. In a limited number of investigative studies
and transnational use in humans. ARG-1 has been shown to be an early onset marker of
liver injury [23]. Arginase is not commonly measured in
Malate Dehydrogenase routine nonclinical safety assessment.
Gluthathione-s-transferase (GST) is a phase II detoxi-
Malate dehydrogenase (MDH) is a predominately fying enzyme with four isozymes (alpha, pi, mu, and
periportal enzyme that is expressed highly in the extra- theta) expressed in mammals. The alpha isozyme, GSTα,
mitochondrial cytoplasm of the liver, although 10% of is found in high concentrations in centrilobular hepato-
MDH has been reported in the mitochondria [23]. It is an cytes; as such, it is a good biomarker candidate to iden-
enzyme in the citric acid cycle that catalyzes the revers- tify injuries that occur in the metabolic zone of the liver.
ible conversion of malate into oxaloacetate. Serum MDH The occurrence of GSTα within the metabolic zone of the
activity is correlated with liver injury, although cardiac liver makes it a good candidate for qualification.
injury can cause an elevation in activity. Like GLDH, miR-122 is a single-stranded noncoding (micro)
MDH levels are stable in healthy patients across gender RNA with about 22 nucleotides that is involved in post-
and age groups, but are elevated with liver injury [37]. transcriptional control of protein synthesis within the
Due to the predominantly periportal location of MDH eukaryotic genome. miR-122 is predominately expressed
and possible translation to human clinical setting, MDH in the liver and has been shown to be elevated in circula-
is among the first set of biomarkers undergoing qualifi- tion during conditions of drug overdose [26]. Although,
cation through the PSTC. the evidence for miR-122 association with DILI is lim-
ited, qualification of a microRNA biomarker is poten-
tially useful because of its stability and the availability
Paraoxonase-1
of more sensitive PCR-based assays compared to colori-
Paraoxonase-1 (PON1) is an esterase that protects metric and ligand-based assays for current biomarkers.
low-density lipoproteins from oxidation and detoxifies In conclusion, there are several markers currently
organophosphates. PON1 is predominantly produced available to assess liver injury, and these markers offer
in the liver, although enzyme activity has been detected a lot of useful information about liver injury when inter-
in the kidney and brain. Unlike MDH and GDH, PON1 preted properly. However, the occurrence of DILI after
decreases during liver injury, probably due to decreased drug approval indicates that more predictive biomarkers
synthesis and/or secretion. Initial baseline data suggests are necessary in nonclinical safety assessment. PSTC has
that PON1 varies among healthy people based on their taken the lead in qualifying new markers. As the new
ethnicity [37]; therefore it is not clear how PON1 will markers become qualified and adapted, further under-
translate from nonclinical to clinical settings. However, standing of their comparative relevance in different pre-
since all other liver injury biomarkers are elevated in clinical species should be an integral part of improving
times of injury, PON1 is an attractive biomarker candidate nonclinical safety assessment.

III.  CLINICAL PATHOLOGY, HISTOPATHOLOGY, AND BIOMARKERS


456 17.  BIOMARKERS IN NONCLINICAL DRUG DEVELOPMENT

RENAL INJURY BIOMARKERS of data from other biomarkers for informational pur-
poses, or to provide additional perspective.
Nephrotoxicity and acute kidney injury (AKI) is a When measuring urinary biomarkers it is important
common occurrence in nonclinical safety studies and to utilize metabolism cages to collect urine and insti-
continues to rank among the top reasons for drug attri- tute efforts to minimize urine sample contamination by
tion of promising new drug candidates. The kidneys are environmental materials such as food, bedding, feces,
uniquely susceptible to a wide array of xenobiotics due and other debris, which can introduce undesired pre-
to their receiving a large proportion of circulating blood analytical variation resulting in poor datasets. When
volume (up to 25% of cardiac output) resulting in high quantifying any urinary-based analyte it is also critical
exposure to compounds and/or their metabolites in cir- to normalize analyte concentration to urine concentra-
culation. The nephrons are also involved in the urine tion or volume by performing either a creatinine ratio
formation process, which further serves to concentrate or an analyte over timed urine volume ratio (eg, ana-
toxicants in renal tubular fluid. Some of the most com- lyte/16 h) to correct for fluctuations in urine output.
mon nephrotoxins include heavy metals (eg, chromium, Although the release kinetics of each biomarker into the
lead, mercury), nonsteroidal antiinflammatories thera- urine will be compound-dependent and vary slightly,
peutics (eg, acetaminophen), aminoglycoside antibiotics collecting urine for 12–16 h at least 2–3 days following
(eg, gentamicin), and immunosuppressive and chemo- anticipated renal injury is a standard approach in non-
therapeutic agents (eg, cyclosporine, cisplatin) [38]. clinical toxicology studies. Some biomarkers have been
The use of traditional clinical pathology endpoints shown to be released into the urine within hours fol-
such as blood urea nitrogen (BUN), serum creatinine lowing an acute insult [42].
(sCr), phosphorus, and urine-specific gravity as indi- Renal biomarkers generally are classified into sev-
cators of renal function have long been established; eral groups indicating their association with injury to a
however, these parameters are relatively insensitive specific part of the nephron (eg, proximal tubule, glom-
indicators of renal injury and will not demonstrate dis- erulus, collecting duct). However, recent work suggests
cernible alterations until a functional deficit of up to that there is a fair amount of overlap and redundancy
65–75% has occurred [39]. Clearly, the performance of among individual biomarkers in regards to their specific
these markers is not sufficient for monitoring kidney nephron segment, hence classifying biomarker effects
injury in many nonclinical settings. The need for renal to either the glomerulus or the tubules appears to be
biomarkers with improved sensitivity has led to more the most appropriate approach in most cases [41]. The
recent efforts, largely driven by collaborative groups magnitude, timing, and sensitivity of positive signals
like the Predictive Safety Testing Consortium (PSTC) among specific biomarkers will differ between test com-
and the International Life Sciences Institute—Health pounds, and between assay methodologies and labora-
and Environmental Sciences Institute (ILSI–HESI), tories, hence the selection of specific biomarkers should
resulting in the discovery of a number of promising be made with consideration of assay availability, spe-
next-generation renal injury biomarkers that provide cies, anticipated pathophysiology, and analyte stability
earlier and more specific detection of renal injury for [17,43]. Lastly, it is important to have an understanding
use in drug development. of the relationship between positive biomarker signals
Initially, a set of seven urinary biomarkers was quali- and microscopic pathology as histologic effects on the
fied by the FDA for use in the rat for monitoring drug- kidney as assessed by light microscopy continues to be
induced kidney injury in conjunction with BUN and SCr the reference standard used to assess the overall perfor-
[40]. These seven biomarkers include kidney injury mol- mance and sensitivity of the emerging renal biomarkers.
ecule-1 (Kim-1), albumin, total protein, β2-microglobulin
(B2M), cystatin C, clusterin, and trefoil factor-3. Addi- Tubular Injury Biomarkers
tional data for four more biomarkers were later submit-
ted to the FDA and EMA for nonclinical qualification Kidney Injury Molecule-1 (Kim-1/TIM-1/
and two of them, clusterin and renal papillary antigen-1 HAVCR-1)
(RPA-1), were accepted for use in detecting acute drug- Kidney injury molecule-1 (Kim-1/TIM-1/HAVCR-1)
induced renal tubule alterations in rat. In addition to this is probably the most popular and commonly utilized of
list, a fair number of other renal biomarkers have been next-generation or novel renal biomarkers for detecting
described (eg, osteopontin, NGAL, GST-α, calbindin) acute kidney injury (see Table 17.1). It is an 85-kDa type
and are being used in drug development settings [17,41]. I cell membrane glycoprotein conserved across many
It is worth noting that qualification of a biomarker by a species including, rodents, dogs, primates, and humans
regulatory body represents their endorsement of its use and is classically considered a proximal tubular marker
under very specific circumstances, in a specific species, [44]. Kim-1 is shed from tubular epithelial cells into the
and does not discount the utility or prevent the inclusion urine in response to injury (eg, toxic, ischemic, septic,

III.  CLINICAL PATHOLOGY, HISTOPATHOLOGY, AND BIOMARKERS


Renal Injury Biomarkers 457
TABLE 17.1  Summary of Renal Injury Biomarkers
Biomarker Sources Interpretation

Kidney injury molecule 1 (KIM-1, TIM-1, Proximal tubules Toxic, ischemic, septic renal tubular injury
HAVCR-1)

Neutrophil gelatinase-associated lipocalin Proximal and distal tubules, neutrophils, Renal tubular injury (urinary), inflammatory
(NGAL, lipocalin-2) bone marrow (blood)

Clusterin Proximal and distal tubules Toxic, ischemic, septic renal tubular injury

Cystatin C All nucleated cells, renal tubules Plasma levels inversely related to GFR; renal
tubular injury (urine)

Alkaline phosphatase (ALP) Renal tubules Renal tubular injury

N-acetyl-β-glucosaminidase (NAG) Renal tubules Renal tubular injury

Gamma glutamyltransferase (GGT) Renal tubules Renal tubular injury

Total protein Plasma protein Glomerular injury

Albumin (micro albumin) Plasma protein Glomerular injury

α1/β2-microglobulin Plasma protein Glomerular injury

GFR, glomerular filtration rate.

and transplant), and has been shown to be a sensitive inflammation, infections (eg, urinary tract), and neo-
and early diagnostic indicator of renal injury in a variety plasia, which should be considered when positive
of animal models and human disease states [42,45–47]. NGAL signals are seen [53,54].
In humans, increased levels have also been shown to
be predictive of adverse disease outcomes [48]. Kim-1 Clusterin
is routinely used in rat and nonhuman primate studies; Clusterin is a 76–80 kDa protein expressed on the
however, anecdotal evidence suggests Kim-1 may not be dedifferentiated tubular cells after injury and is another
a useful indicator of renal effects in dog. Kim-1 assays promising marker for the detection of acute tubular
are widely available and are often included in species- injury. In the context of kidney injury, clusterin has
specific renal injury panels on multiplex platforms [43]. been suggested to play an antiapoptotic role and to be
involved in cell protection, lipid recycling, cell aggrega-
Neutrophil Gelatinase-Associated Lipocalin or tion, and cell attachment [55]. Increased urinary clus-
Lipocalin-2 terin levels have been seen in a variety of conditions of
Neutrophil gelatinase-associated lipocalin (NGAL) both proximal and distal tubular injury, but not in those
(lipocalin-2) is a 25-kDa protein originally observed associated with glomerular injury [45,56–58].
within specific granules of the neutrophil produced
during granulocyte maturation in bone marrow [49]. Cystatin C
However, more recently it has been shown to be unregu- Cystatin C is a low molecular weight (13.3 kDa) basic
lated in the proximal and distal tubules in response to protein (cysteine protease inhibitor) produced by all
a variety of renal injury conditions including ischemia nucleated cells at a constant rate and is freely filtered
and chemical toxicity [41,50,51]. Although it has not by the kidneys. It is a good candidate as both an index
specifically been qualified for use by regulatory agen- of function as related to ​GFR as well as a tubular injury
cies, NGAL has generated much interest as a biomarker indicator. It is not secreted in normal urine and is com-
for the early detection of acute proximal tubular injury pletely reabsorbed by proximal tubule epithelia [59].
and is routinely used in nonclinical studies. In healthy Plasma/serum levels of cystatin C are inversely related
individuals NGAL is expressed at low levels in various to GFR, so that reduced GFR is reflected in increased
tissues, filtered at the glomerulus, and reabsorbed by cystatin C concentrations. Serum concentrations appear
the proximal tubule [52]. In addition to increasing renal to be independent of sex, age, and muscle mass. Cys-
production of NGAL following injury, damage to the tatin C does not appear in the urine of normal animals
nephron may enhance NGAL presence in the urine by but will be increased in the urine in conditions of renal
impairing proximal tubular reabsorption. tubular dysfunction [60,61]. The PSTC recently reported
Given the involvement of NGAL in neutrophil that urinary cystatin C can outperform BUN and sCr in
function, false-positive increases in NGAL can be detecting early impairment of tubular reabsorption due
induced in neutrophils by various stimuli including to glomerular alterations/damage in rat studies [55].

III.  CLINICAL PATHOLOGY, HISTOPATHOLOGY, AND BIOMARKERS


458 17.  BIOMARKERS IN NONCLINICAL DRUG DEVELOPMENT

Enzymuria and clinical applications for the early detection of renal


injury. We anticipate the continued emergence of new
Enzymes released from damaged renal tubular cells biomarkers and those selected for routine use should be
of the proximal and distal tubule have been used as ones with a sufficient body of evidence to support their
markers of kidney injury in animals for several decades; intended use.
however, their use in human studies has not been widely
accepted. This group of renal injury biomarkers is not
generally considered part of next generation or novel CARDIAC AND SKELETAL MUSCLE
renal biomarkers, partly because of the traditional meth- INJURY BIOMARKERS
odology in which most of them are measured (nonim-
munoassay-based methods). A variety of enzymes (see Cardiotoxicity can be caused by a variety of xenobiot-
Table 17.1) have been utilized for this purpose includ- ics and chemical therapeutics including antiarrhythmic
ing alkaline phosphatase (ALP), the lysosomal enzymes agents (eg, β-adrenergic blockers), positive inotropic
N-acetyl-β-glucosaminidase (NAG) and gamma glutam- compounds (eg, digoxin, isoproterenol), anesthetics (eg,
yltransferase (GGT), and the glutathione S-transferases isoflurane, propofol), narcotics (eg, cocaine), and anti-
(GSTs) [62]. The appearance of these enzymes in the urine neoplastic agents (eg, doxorubicin, cyclophosphamide).
is specific to proximal and distal tubule damage, and Cardiotoxicity, and the biomarkers used to identify such
they have been shown to be fairly sensitive indicators effects, should be distinguished from cardiovascular, or
of nephrotoxicity and injury even preceding increases in drug-induced vascular injury (DIVI) biomarkers, which
sCr [62–64]. These assays are available on most modern are generally aimed at identifying injury to blood vessels
clinical chemistry analyzers. and the vascular system, discussed later in this chapter.
Biomarkers used for the identification and characteriza-
Glomerular Injury Biomarkers tion of cardiac injury effects can be broadly categorized
as those that identify structural or direct cardiac myo-
Total Protein, Albumin, and Microglobulins cyte injury (eg, troponins, fatty acid binding protein)
The glomerular capillaries create a barrier for the and necrosis, and those that indicate alterations to car-
passage of proteins any larger than those about the size diac function (eg, natriuretic peptides). Although cardiac
of albumin (36 Å) into the urine. Depending on charge, injury may be detected through a variety of modalities,
smaller proteins (eg, α and β microglobulins) will also including electrocardiogram, imaging, and other func-
pass through the filtration barrier into the tubular fluid tional assessments (eg, blood pressure, cardiac output),
ultimately being reabsorbed or broken down by the this discussion will focus on the measurement of bio-
proximal tubules. For those reasons, the detection of markers in blood and other body fluids (serum/plasma),
increased concentrations of such proteins in the urine is which are more widely used in nonclinical research and
an indicator of glomerular (albumin or larger proteins) toxicology studies.
and/or proximal tubular injury (smaller proteins like
the microglobulins). In the case of glomerular damage,
injury results in large proteins leaking into the urine,
Biomarkers of Cardiac Structural Injury and
which overwhelms the modest ability of the proximal Necrosis
tubule to remove them, whereas injury to the proximal The current gold standard for the detection of acute
tubule will impair the ability to remove smaller proteins cardiac myocyte injury/necrosis includes the measure-
such as α1 or β2-microglobulin. They have also been ment of cardiac troponin I (cTnI) and/or troponin T
shown to have good sensitivity and specificity in detect- (cTnT) in serum/plasma (see Table 17.2). Both of these
ing renal injury due to a variety of causes [17,43]. molecules are involved in regulating the excitation and
coupling process of the contractile apparatus within the
cardiac myocyte, and are released into circulation fol-
Future Directions
lowing cell injury and necrosis. These biomarkers have
Given the complexity of renal structure and function, repeatedly demonstrated their high sensitivity and spec-
the variety of traditional and emerging renal biomarkers ificity in detecting acute myocardial injury in a variety
in use, and the wide array of new test compounds and of disease and experimental conditions in both humans
associated pathophysiologic observations, it is unlikely and animals [65–67]. Elevations in these biomarkers
that a single biomarker will be the lone answer for the will often precede the first histologic evidence of car-
detection of renal injury in toxicology studies in the diac injury and correlate well with effects on echocar-
future. It is more likely that we will continue to use pan- diography and overall histologic injury scores [68,69].
els of serum and/or urine biomarkers for general assess- In healthy animals, cardiac troponins circulate in low,
ment, patient monitoring, and investigative nonclinical nearly undetectable levels and can increase within hours

III.  CLINICAL PATHOLOGY, HISTOPATHOLOGY, AND BIOMARKERS


Cardiac and Skeletal Muscle Injury Biomarkers 459
TABLE 17.2  Summary of Cardiac and Skeletal Muscle Biomarkers
Leakage Markers Sources Interpretation

Cardiac troponin I (cTnI) Cardiac muscle Cell injury/necrosis

Cardiac troponin T (cTnT) Cardiac muscle Cell injury/necrosis

Heart-type fatty acid binding protein (H-FABP or FABP3) Cardiac and skeletal muscle Cell injury/necrosis

Myoglobin Cardiac and skeletal muscle Cell injury/necrosis

Myosin light chains (Mlc) Cardiac and skeletal muscle Cell injury/necrosis

Creatine kinase (CK) Cardiac and skeletal muscle, brain, GI tract Cell injury/necrosis

CK-MM Cardiac and skeletal muscle Cell injury/necrosis

CK-MB Cardiac and skeletal muscle Cell injury/necrosis

Lactate dehydrogenase (LD) All muscle types, liver, RBCs Cell injury/necrosis

Aspartate aminotransferase (AST) All muscle types, liver, RBCs Cell injury/necrosis

Skeletal muscle troponin I (sTnI) Skeletal muscle Cell injury/necrosis


FUNCTIONAL MARKERS

Atrial natriuretic peptide (ANP) Primarily cardiac atria Atrial wall stretch

Brain natriuretic peptide (BNP, proBNP, NT-proBNP) Primarily cardiac ventricles Ventricular wall stretch

following a single cardiac insult [68]. The positive sig- cardiac and skeletal muscle, brain, liver, and small intes-
nal is short-lived as clearance of cTn molecules is rapid tine [72]. FABP3 contained in heart muscle is rapidly
and occurs within 24–48 following a single insult. Given released into circulation within hours following myo-
the sensitivity of many commercial troponin assays, it is cardial injury [73,74]. It has good sensitivity for cardiac
generally considered necessary to observe a greater than injury and has very similar release (within hours) and
twofold increase in cTnI values before considering the clearance (≤48 h) kinetics compared to the cTnI; however,
signal to be biologically/toxicologically significant [70]. it may lack the specificity of the cardiac troponins as sig-
As with many of the newer biomarker assays, the lack nificant amounts of FABP3 have been demonstrated in
of standardized methods across industry makes com- skeletal muscle in rat [75]. Similarly, myosin light chains
parison of absolute values between different methods (Mlc) are a component of the myosin molecule found in
and laboratories problematic. Careful consideration of cardiac and slow twitch skeletal muscle cells that have
method and laboratory-specific historical ranges should been used as indicators of myocardial injury [76]. They
be made before interpretation of positive signals is have been incorporated into multiplex panels and used
determined. Similar troponin molecules also are present in conjunction with other markers to aid in the character-
in skeletal muscle (skeletal troponin; sTnI) but are anti- ization of myocardial damage.
genically distinct from their cardiac counterparts adding Myoglobin is found in the cytoplasm of both skeletal
to the specificity of both groups of biomarkers for the and cardiac muscle and is primarily used for the early
detection of cardiac and skeletal muscle injuries, respec- detection of acute myocardial infarction in humans.
tively [71]. Although cTnI and cTnT have similar inter- However, it lacks specificity for cardiac muscle and
pretive utility and kinetic characteristics, cTnI assays occurs in higher concentrations in skeletal muscle pre-
have been better characterized, are more widely avail- cluding its use as an independent marker of cardiac
able, and have demonstrated good cross-reactivity and injury [77]. Myoglobin has a short half-life (as short as
translatability across multiple species. Cardiac troponin 20 min in humans) and is used for postcardiac injury
I is the most consistently utilized biomarker for detect- monitoring in clinical settings [78].
ing myocardial injury in drug safety assessments [66,67]. Historically, a variety of other enzyme biomarkers
Heart-type fatty acid-binding proteins (H-FABP have been utilized for the detection of cardiac myocyte
or FABP3) have recently been investigated as an early injury in humans and animals. These include aspartate
marker for cardiac myocyte injury and associated cell aminotransferase (AST), lactate dehydrogenase (LD),
membrane disruption. FABPs are a family of low molec- creatine kinase (CK), and creatine kinase MB isoenzyme
ular weight proteins found in multiple tissues including (CK-MB), which are no longer recommended for use as

III.  CLINICAL PATHOLOGY, HISTOPATHOLOGY, AND BIOMARKERS


460 17.  BIOMARKERS IN NONCLINICAL DRUG DEVELOPMENT

cardiac biomarkers due to their low cardiac specificity, medical device studies. Unlike the extensive research
inconsistent performance across species, wide tissue dis- devoted to the development of diagnostic assays for
tribution, and lack of clinical relevance [77]. However, myocardial disease, much less effort has been expended
many of these markers are well suited as biomarkers of on the development of biomarkers for skeletal muscle
skeletal muscle injury, discussed later in this chapter. injury detection. A number of enzymes (eg, CK, AST, LD)
Additionally, a number of proinflammatory biomarkers and protein biomarkers (eg, myoglobin) have tradition-
(eg, high-sensitivity CRP, TNF-α, and myeloperoxidase) ally been used for this purpose; however, they generally
have been used for cardiac injury detection, and occa- lack the sensitivity and specificity needed to reliably dis-
sionally included in cardiac injury biomarker panels; tinguish subtle effects on skeletal muscle from effects on
however, these types of endpoints only serve to identify other tissues.
general inflammatory signals and should always be used Creatine kinase is a cytosolic enzyme found in a vari-
in conjunction with other more cardiac-specific markers. ety of tissues including skeletal and cardiac muscle,
brain with lesser amounts in the gastrointestinal tract,
uterus, bladder, and kidney [86]. CK is released into cir-
Biomarkers of Cardiac Functional Alterations culation following myocyte injury and is rapidly cleared
The natriuretic peptides (see Table 17.2) are a family with a half-life of only 2 h in dog [39]. It is the most
of related hormones produced in the myocardium that widely used biomarker to detect general muscle injury
have potent vasoactive and diuretic properties used in and has three distinct isoenzymes (CK-MM, CK-BB, and
the regulation and homeostasis of blood pressure, renal CK-MB), which can be determined through electropho-
function, and maintenance of cardiac output [79]. Sev- retic separation. Traditionally CK-MM and CK-MB have
eral variants of these peptides exist including ANP (atrial been used as markers of skeletal and cardiac muscle
natriuretic peptide), BNP (brain natriuretic peptide), injury, respectively; however, the majority of CK activ-
and their N-terminal prohormones (N-terminal proatrial ity in both cardiac and skeletal muscle is attributed to
natriuretic peptide (NT-proANP) and N-terminal pro- CK-MM [77]. There is also a high degree of variability in
brain natriuretic peptide (NT-proBNP)), all of which are CK content and isoenzyme proportions between specific
rapidly released into circulation in response to myocar- muscle locations, muscle types (fast twitch/slow twitch),
dial wall stretch [80]. BNP is secreted from the ventricles and between species, particularly in regards to myocar-
but may also be secreted from the atria in conditions of dial CK-MB activity [87–89]. CK is a good biomarker for
heart failure [81]. Of this class of peptides, NT-proBNP skeletal muscle injury but the CK-MB isoenzyme lacks
is the most commonly used biomarker in nonclinical the specificity and consistency across species to be used
toxicology studies. NT-proBNP has been shown to be a alone as a cardiac-specific biomarker.
sensitive and specific biomarker in the diagnosis of heart Lactate dehydrogenase (LD) is an enzyme found in
failure in humans and animals and has a positive correla- most nucleated and nonnucleated cell types including
tion to ventricular systolic dysfunction [80,82]. BNPs are skeletal and cardiac muscle, liver, and erythrocytes, with
also elevated in conditions of dilated and hypertrophic highly variable distribution among different tissue types
cardiomyopathy, diastolic dysfunction, and systemic and between species [39,90]. Given its wide tissue distri-
hypertension, and have been shown to be a good gen- bution, increases in LD activity are not specific to a single
eral prognostic indicator of cardiovascular disease states organ. LD activity is generally higher in skeletal muscle
[83–85]. However, natriuretic peptides are also greatly and liver compared to heart muscle, which supports its
increased in a number of noncardiac conditions related use as a marker of skeletal muscle and liver injury [86].
to increased vascular fluid volume and blood pressure Attempts to use isoenzymes of LD (eg, LD1, LD2, etc.)
including pulmonary embolism, liver cirrhosis, renal to increase cardiac specificity have been previously per-
disease, and hyperthyroidism [79]. Natriuretic peptides formed; however, much like the use of CK isoenzymes,
are particularly useful in drug safety assessments when there is considerable variability between specific muscle
used in conjunction with more specific biomarkers of groups, species, and concentrations within the heart pre-
cardiac structural injury/necrosis, such as cTnI, to pro- cluding it as a useful cardiac injury biomarker [77].
vide an overall characterization of both structural and Aspartate aminotransferase (AST) and alanine amino-
functional effects on the heart. transferase (ALT) are both enzymes that have multior-
gan tissue distributions that include skeletal muscle and
liver. Increased AST activities are generally associated
Biomarkers of Skeletal Muscle Injury with effects on liver and skeletal muscle, but are also
Skeletal muscle toxicity and/or injury is a common seen with hemolytic conditions [39]. ALT elevations are
finding in nonclinical safety studies following expo- classically associated with hepatocellular injury; how-
sure to various chemical compounds (eg, cholinester- ever, cases of significant muscle injury have also been
ase inhibitors, carbofuran) or surgical manipulation in associated with increases in ALT activity [91].

III.  CLINICAL PATHOLOGY, HISTOPATHOLOGY, AND BIOMARKERS


Biomarkers of the Immune System 461
More recently, next-generation biomarkers with vascular regeneration and repair. Though these fami-
improved specificity for skeletal muscle have been dis- lies of molecules have many members, only a select few
covered and are gaining wider acceptance. The most were deemed as useful for characterization of DIVI in
promising of these markers is skeletal troponin I (sTnI), the rat by the VIWG. Candidate biomarkers from this
which has been shown to be a sensitive and specific bio- group included angiopoietin-2, endothelin-1, E-selec-
marker for skeletal muscle injury [92]. Skeletal muscle tin, thrombospondin-1, and VEGF-α [96]. Following
troponin is similar to, but antigenically distinct from its administration of a known vascular toxicant (PDE3i—a
cardiac-specific counterpart cTnI, and has been shown to phosphodiesterase inhibitor), positive signals in these
have the power to distinguish between skeletal and car- circulating EC biomarkers were modest (2–5-fold) and
diac injury in rats using a variety of cardiac toxins and tended to be most pronounced at 24–72 h postinjury.
myotoxins [93]. This biomarker has also been included Notably, most of these biomarkers exhibited increases
in several multiplex muscle injury panels aimed at dif- following injury, while E-selectin showed a decrease
ferentiating between cardiac and skeletal muscle injury. from baseline following injury.
The second main category of biomarkers that was
found to be helpful in characterizing DIVI by the VIWG
VASCULAR INJURY BIOMARKERS were those involved in the inflammatory component.
Although these markers are extremely sensitive indica-
Drug-induced vascular injury (DIVI) refers to any of tors of inflammation, they lack the specificity to localize
a variety of pathologic processes leading to or result- injury to the vasculature and require the use of EC-
ing in injury to blood vessels. It is a significant cause of specific markers to be useful in DIVI identification. The
drug attrition as drug companies seldom proceed into markers found to be useful included Timp-1, lipocalin-2,
the clinics following identification of a DIVI liability in KC/GRO (Cxcl1), α-1 acid glycoprotein 1, and total
nonclinical studies. The diagnosis of DIVI in nonclinical nitric oxide. These markers tended to give more robust
animal species generally relies on biomarkers related to positive signals following PDE3i administration (up to
the release of endothelial cell (EC) adhesion molecules, 200-fold) and were observed earlier (as soon as 1–4 h)
EC activation markers, and/or nonspecific acute-phase relative to biomarkers of EC activation/injury.
inflammatory proteins [94]. Although a long list of
potential biomarkers can be gathered from a literature
search, more recent evidence suggests that the vast BIOMARKERS OF THE IMMUNE SYSTEM
majority of markers will not apply equally to all animal
species and/or types of injury. This list becomes even The immune system is a complex regulatory net-
shorter when one considers the lack of species-specific work of barriers, specialized effector cells, and mol-
assays for all DIVI biomarkers. ecules organized into a defense system that is capable
The most significant DIVI work in a nonclinical spe- of distinguishing “self” from “nonself.” The cells of the
cies (rat) to date was performed by the Vascular Injury immune system include lymphocytes [bone marrow-
Working Group (VIWG) of the PSTC. The VIWG aimed derived (B-cells), thymus-derived (T-cells), natural
to identify candidate biomarkers that would be useful killer (NK cells), macrophages/monocytes, neutrophils,
in translating into humans, but also were independent eosinophils, basophils, and mast cells. Functionally, the
of pathophysiologic mechanism. The latter point being immune system is broadly divided into innate and adap-
critical as the mechanisms of vascular disease as seen in tive immunity based on whether or not the specific pro-
humans (ie, immune-mediated) appear to be distinctly tection requires immunologic memory. Innate immunity
different than those involved in DIVI in most nonclini- does not require immunologic memory and includes
cal animal species (ie, altered hemodynamics, direct EC defensive barriers such as anatomic (skin and mucous
toxicity) [95,96]. These differences highlight the chal- membranes), physiologic (temperature and pH), and cel-
lenge in identifying biomarkers that have translatability lular (phagocytic cells and pathogen-associated molecu-
into man. The VIWG investigated dozens of protein and lar patterns)]. The innate immunity mediates its function
molecular biomarkers and ultimately identified several through an array of soluble (cytokines, chemokines) and
candidates that largely fell into one of two general cate- cellular (phagocytic and cytolytic) mediators. Acquired
gories: biomarkers of EC activation/injury and biomark- immunity requires previous exposure of lymphocytes
ers of inflammation. to the specific antigen, in the context of antigen present-
Biomarkers of EC injury make up a significant por- ing cells and major histocompatibility complex, which
tion of DIVI biomarkers covered in the literature and creates immunologic memory for the next encounter of
include the vascular cell adhesion molecules (VCAMs), that specific antigen. Any subsequent encounter with the
intracellular adhesion molecules (ICAMs), integrins/ same antigen may elicit humoral (antibody-mediated)
selectins, as well as signaling molecules involved in or cell-mediated immune response. The details of basic

III.  CLINICAL PATHOLOGY, HISTOPATHOLOGY, AND BIOMARKERS


462 17.  BIOMARKERS IN NONCLINICAL DRUG DEVELOPMENT

immunology are outside the scope of this chapter. Per- often requires several concordant lines of evidence
haps the most important point to recognize about safety and uses a weight-of-evidence approach.
assessment of the immune system is the complexity
and redundancy of the system, which is further compli- Hematology and Clinical Chemistry
cated by large species differences. Successful nonclinical Evaluation of the complete blood count (CBC) with
assessment of immune safety requires a thorough appre- differential continues to be a highly effective biomarker
ciation of these complexities within and across species. in identifying alterations to the immune system for both
For example, recent observation of systemic organ fail- signals of immunosuppression as well as proinflamma-
ure during a Phase I clinical trial of anti-CD28 mono- tory immune conditions. Lymphoid and/or bone-mar-
clonal antibody, which was not predicted by nonclinical row toxicities are routinely associated with decreases
safety assessment, is a reminder of both complexity and in lymphocyte, neutrophil, and total leukocyte counts
species difference within the immune system [97]. Addi- ultimately resulting in immunosuppression. Concur-
tional information on the details of immunobiology and rent decreases in platelets and reticulocytes may also be
immunotoxicity can be found in Chapter 22 of this book seen. These findings often correlate to decreased organ
and elsewhere [98–100]. weights and microscopic cellularity of lymphoid and
Xenobiotic-related injury or alteration of the immune bone-marrow tissues. The first appearance of mild cyto-
system can be organized into five broad categories: penias on the hematology panel often precedes decreases
immunosuppression, hypersensitivity, immunogenicity, in bone-marrow cellularity as seen microscopically on
autoimmunity, and adverse immunostimulation [101]. tissue sections.
These categories are similar for chemical and biological Stress is common in toxicology studies and causes
entities, although molecular weight is a large determi- mild-to-moderate reductions in lymphocytes and
nant of immunogenicity. For example, entities with a eosinophils, often with concurrent increases in neutro-
molecular weight of 1000 Da are not likely to be immu- phils and/or monocytes. These findings must be dis-
nogenic unless their metabolites are attached to endog- tinguished from primary effects on the immune system
enous molecules in the form of a haptens. by careful consideration of other related endpoints (eg,
Biomarker testing of the immune system involves histopathology, organ weights, acute-phase proteins) as
a wide range of strategies including use of cell-based specifically suggested in the FDA S8 guidance [102].
(eg, hematology, immunophenotyping), tissue-based
(eg, histopathology, IHC), and protein-based (eg, Increased Incidence of Tumors
acute-phase proteins, cytokines, complement) assays. Test article-related increase in the incidence of tumors
The foundation of immunotoxicity testing involves in 2-year rodent bioassays or other toxicity studies that
a tiered approach beginning with the evaluation of are not due to genotoxic, hormonal, or other well-under-
standard toxicology endpoints including hematology stood carcinogenic mechanism need to be further inves-
and clinical chemistry panels, with evaluation of lym- tigated for the likely role of immunosuppression [101].
phoid tissues for effects on organ weights, and gross In cases where further investigation is necessary tumor-
and microscopic morphology. As further investiga- host resistance assays with B16F10 melanoma cells or
tions are warranted, more focused assays such as host other xenograft models may be the appropriate assay
resistance, delayed-type hypersensitivity, autoimmu- to further evaluate the role of immunosuppression in
nity, and/or macrophage/natural killer cell function tumorigenesis.
are incorporated as needed.
Increased Incidence of Infections
Treatment-related increase in infections, especially
Indicators of Immunosuppression
those that involve weakly pathogenic organisms such as
FDA guidance for immunotoxicity assessment states Candida albicans, are suggestive of immunosuppression.
that all investigational new drugs should be evalu- The increased incidence of infection may not be related
ated for the potential to produce immunosuppression. to alterations in the function and/or number of lympho-
This is usually accomplished by a 28-day daily dos- cytes but the barrier systems of innate immunity. For
ing of the test article through the applicable route(s) example, ozone exposure before an aerosol challenge of
of administration in humans. Due to the complexity infectious agents results in treatment-related infections
of the immune system, the markers of immunosup- [103]; however, this increase in infection was related to
pression are often not one molecule as is common in alterations in the innate immune barriers and macro-
other systems. Clear suppression of T cell-dependent phage phagocytic system. Investigators need to remem-
antibody response (TDAR) may be considered as one ber that some cases of increased infection may show no
marker of immunosuppression but a conclusion of effect on TDAR because the test article altered the innate
immunosuppression in nonclinical safety assessment component of the immune system.

III.  CLINICAL PATHOLOGY, HISTOPATHOLOGY, AND BIOMARKERS


Biomarkers of the Immune System 463
Histopathology and Lymphoid Organ Weight assessments [108]. Immunohistochemistry of the same
Decreases in spleen and thymic weights in conjunc- markers within lymphoid tissues, when feasible, is a nec-
tion with correlative histopathology findings may essary adjunct to further integrate changes in cell popu-
indicate immunosuppression [104]. The associated lation with the histopathology findings and hematology
histopathology findings are usually decreases in lym- observations. Overall an indication of immunosuppres-
phocytes within specific compartment(s). Lymphoid sion by some of the above indices necessitates immune
organ weight is highly variable within and across function studies to further characterize/understand the
species and this variability should be carefully con- specific signal.
sidered while interpreting organ weight data. The
variability of median organ weight within thymus of Immune Function Studies
mature Sprague Dawley rats may be up to 70%, while These studies are designed to investigate functions
the variability in Beagle dogs may be up to 170% [98]. of components of the immune system when there is
The weighing of lymph nodes and Peyer’s patches is reason to be concerned about immunosuppression or
even more variable and the quality of data it gener- occasionally immunostimulation. The principles from
ates rarely justifies the effort it requires. The weight of FDA guidance indicate that these studies are relevant
the spleen is also variable across species. For example, when (1) the drug is being designed for HIV patients
the defense spleen has a large amount of lymphocytes or other immune-compromised population, (2) mol-
and is present in rats, mice, nonhuman primates, and ecule is structurally similar to known immunosup-
rabbits, while the storage spleen, which may store up pressive compounds, (3) pharmacokinetics and/or
to one-third of the circulating blood volume, is seen toxicokinetics indicate that the compounds occurs in
in dogs [98]. The swine spleen is an intermediate immune tissues or cells at relatively high concentra-
between defense and storage spleen. The methods of tions, and (4) signs suggestive of immunosuppression
euthanasia and exsanguination may affect the spleen are seen in clinical trials [101]. In immune function
weight in several species, but this influence tends studies, the dose, duration, and route of administra-
to be greater with storage spleen than with defense tion should be consistent with the study in which the
spleen. In general, spleen and thymus weights are original immune effect was observed. The functional
often useful in rodents and should always be collected studies are divided into tier I and tier II studies based
but their utility in nonrodents is limited and collection on the ease of incorporating them into standard toxic-
of lymphoid organ weights in nonrodents should be ity studies (STS). Tier I studies are generally easier to
addressed case-by-case [105]. incorporate into STS than tier II, which often require
To enhance the detection of immunosuppressive separate studies.
effects, the Society of Toxicologic Pathology and the
majority of the toxicologic pathology community rec- Primary Antibody Assays
ommend a descriptive semiquantitative approach to T-cell dependent antibody response (TDAR) is a
immune system evaluation [104,106,107]. There are tier I assay. It is the most acceptable, general-purpose
nuances to the approaches reported by these authors, functional assay because it depends on the appropriate
but they all emphasize a semiquantitative description of function of antigen-presenting cells, helper T cells, sol-
changes in compartments of the lymphoid organs, which uble cytokines, and B cells. The TDAR assay evaluates
derives from the recognition that (1) separate compart- an individual’s ability to mount a humoral immune
ments within each lymphoid organ support-specific response to a standardized antigenic challenge (eg,
functions, (2) each compartment should be evaluated for KLH, tetanus toxoid, SRBC). Sheep red blood cells
change(s), and (3) descriptive is better than interpretive (SRBC) were previously used in a plaque assay, but
terminology for characterizing changes that occur in the this is increasingly being replaced by Keyhole Lim-
lymphoid system. pet Hemocyanin (KLH) and ELISA (enzyme-linked
Immune cell phenotyping of lymphoid tissues and/ immunosorbent assay) measurement of antigen-spe-
or whole blood may be used to further characterize the cific immunoglobulins. Evaluation of the immune sys-
nature of identified changes in immune system and their tem using a TDAR assay should be performed when
effect(s) on circulating subpopulations of immune cells. assessing compounds with known or potential sup-
Flow cytometry analysis of surface markers of T cells pressive or modulatory effects on the immune system
(CD4+ and CD8+), B cells, and NK cells are often utilized [101]. Decreases in antigen-specific antibody levels
as tier I assays in immunotoxicity assessments as recom- are indicative of decreased immune function, which
mended by the FDA [102]. Immunophenotyping by flow is often associated with effects on related endpoints
cytometry is generally not considered to be an adequate including decreased lymphoid organ weights and
standalone test of drug effects on immune function and microscopic cellularity, decreases in cell counts on the
should be used in context with other immune function hemogram, and occasional clinical infections.

III.  CLINICAL PATHOLOGY, HISTOPATHOLOGY, AND BIOMARKERS


464 17.  BIOMARKERS IN NONCLINICAL DRUG DEVELOPMENT

Natural Killer Cell Activity including increases in neutrophils, lymphocytes, acute-


Natural killer (NK) cell activity assay is a tier I assay. phase protein (eg, fibrinogen, CRP), cytokines, and other
This assay measures the cytolytic function of NK cells inflammatory markers. The mechanism of adverse immu-
and has been traditionally performed by quantifying test nostimulation should be investigated/understood in other
article-related radioactivity in 51Cr-radiolabeled YAC-1 ways to contextualize potential human risk.
tumor cells. Ideally this assay should also include appro- Standard hematology panels can be used to detect
priate immunosuppression and immunostimulation con- increases in leukocytes, neutrophils, and/or lymphocytes
trols. Due to radioactivity concerns, a more-sensitive flow that are typical of immune/inflammatory stimulation,
cytometry-based NK cell assay has been developed and is either as a primary immunotoxic effect, or secondary to
increasingly being used in drug development [109]. infection or other morbidity. These increases are often
associated with bone-marrow hypercellularity, lym-
Macrophage Function phoid organ hyperplasia, extramedullary hematopoiesis
This is a tier II assay that evaluates the ability of mac- in the spleen, and/or other inflammatory lesions. Stan-
rophages to phagocytose-labeled antigens. The assay dard clinical chemistry panels include albumin (negative
can be performed in vitro with primary peritoneal mac- acute-phase protein) and globulin (positive acute-phase
rophages isolated from animals that received the test protein), which are both fairly sensitive in the detection of
article or in vivo, which involves an antigen challenge inflammatory and immune signals [39]. There are also a
to test article-exposed animals and subsequent examina- variety of other acute-phase protein assays (eg, C-reactive
tion of marker antigen within macrophages in tissues. protein, fibrinogen, serum amyloid A) available on most
Apoptosis and cytokine profiling are tier II assays that standard clinical chemistry or coagulation analyzers that
help in identifying mechanisms of immunotoxicity or are helpful in identifying proinflammatory signals.
immunosuppression. The measurement and interpreta-
tion of circulating cytokines need to consider that xeno- Acute-Phase Proteins
biotics induce different cytokines in a variable temporal Acute-phase proteins (APPs) are circulating blood
pattern within and across tissues [6,110]. Therefore the proteins primarily synthesized in the liver in response to
temporal pattern of cytokine induction is just as important upstream inflammatory signals. APPs serve as excellent
for understanding the mechanism as the induced cyto- biomarkers of immune system perturbation and are used
kine. Depending on the nature of observed effects (acute in context with other markers of inflammation to identify
vs. chronic) the time points should be chosen to help inter- subtle inflammatory/immune signals. Acute-phase pro-
pret pathogenesis of the observed findings. At least three teins can either be positive APPs, which increase with
time points of cytokine measure will help in elucidating inflammation (eg, fibrinogen, C-reactive protein (CRP),
the mechanism in a repeat-dose study. Interpretation of serum amyloid A), or negative APP (eg, albumin), which
cytokines needs to consider that cytokine quantification decrease with inflammation. APPs have strong species-
does not provide any information about the biological specific behaviors and can be classified as either a major
activity/significance of the protein being measured [99]. or minor APP in any given species (Table 17.3). It is criti-
Host resistance is a tier II assay that may be used to cal to understand which APP is most appropriate for the
detect the influence of xenobiotics on the host response species in use to avoid mischaracterization of compound
to bacterial (Listeria monocytogenes and Streptococcus liabilities. For example, in the nonhuman primate CRP
pneumonia), parasitic (Plasmodium yoelli), viral (influenza is major APP, eliciting strong increases in response to
A2), and tumor (B16F10 melanoma) pathogens. Host- inflammatory stimuli while in rodents, the CRP response
resistance assays are typically standalone studies and will be comparatively minor or nonexistent [16]. Most
can be more challenging to perform as the results are APPs will increase in 24–72 h following an inflammatory
often highly variable, thus a greater number (compared stimulus, which should be considered when choosing
to tier I) of animals may be necessary to achieve statisti- blood-draw intervals and during data interpretation.
cal significance [99]. Results from these biomarkers should always be inter-
preted in context with other inflammatory biomarkers
(eg, hematology, cytokines, etc.). APP assays are widely
Indicators of Adverse Immunostimulation available on both automated clinical chemistry analyz-
Some compounds are capable of overstimulating the ers and as single or multiplex ELISA platforms.
immune system to produce a range of unintended findings
that may include cytokine release, leucocyte infiltration Cytokine Evaluation
in multiple tissues, complement activation, and chronic Cytokines are a heterogeneous collection of peptides
inflammation. Vaccines, adjuvants, medical devices, that serve as signaling molecules between cells and elicit
and monoclonal antibody therapeutics may also initiate biological responses, including cell activation, prolifera-
responses typical of inflammation/immune stimulation tion, growth, differentiation, migration, and apoptosis.

III.  CLINICAL PATHOLOGY, HISTOPATHOLOGY, AND BIOMARKERS


Biomarkers of the Immune System 465
TABLE 17.3  Summary of Major and Minor Positive Acute-Phase Proteins by Species
Species Major Minor

Human C-reactive protein, serum amyloid A α-1 acid glycoprotein, haptoglobin

Nonhuman primate C-reactive protein α2-macroglobulin, serum amyloid A

Rat α-1 acid glycoprotein, α2-macroglobulin C-reactive protein, fibrinogen

Mouse Haptoglobin, serum amyloid A C-reactive protein, fibrinogen

Dog C-reactive protein, serum amyloid A α-1 acid glycoprotein, haptoglobin

Rabbit Haptoglobin, serum amyloid A C-reactive protein, fibrinogen

Pig Haptoglobin, serum amyloid A α-1 acid glycoprotein

The evaluation of cytokines in immunotoxicity charac- and INF-γ with more advanced panels including MCP-1,
terization, both in vitro and ex or in vivo, have been in VEGF, IL-2, IL-4, IL-8/KC/GRO, IL-10, IL-12, and IL-13.
use for some time now for the purposes of hazard iden- Although the evaluation of cytokines in animals contin-
tification, mechanistic characterization, as markers of ues to be common in nonclinical studies, recent examples
pharmacodynamic activity and/or efficacy, as well as have demonstrated that in vivo measures of cytokines in
to monitor for adverse drug reactions [111–113]. Blood animal species are not always predictive of adverse reac-
cytokine measurements are an attractive option due tions in humans [114]. This sometimes disastrous lack of
to the ability to perform serial monitoring using read- concordance between nonclinical and human cytokine
ily accessible samples on a multitude of available assay responses has led to more widespread use of ex and in vivo
platforms. However, the use of cytokines as biomarkers cytokine release assays (CRAs) in an attempt to bridge the
poses several challenges related to their short half-life gap between nonclinical species and humans. Although
and release dynamics, lack of organ specificity, undetect- various approaches to CRAs have been published, most
able baseline levels, and lack of standardized method- often, peripheral blood leukocytes or mononuclear cells
ologies. The overall complexity of the immune system (PBMCs) are isolated from blood and incubated with test
and related cytokine interactions emphasizes the need compounds either in an aqueous solution or solid phase/
for focused and deliberate strategies when evaluating dry-coat (immobilized on plate) format [112]. Small sam-
these endpoints. ples of cell-suspension media are then measured for cyto-
Measurement of serum cytokines may have the most kines at various intervals (eg, 6–72 h) following incubation.
utility in programs or studies that evaluate intended The notion of performing CRAs in solution or in solid
or unintended inflammation and immunomodulation phase or in whole blood versus PBMCs is still under debate
produced by therapeutics. When applying cytokine with 62% of respondents performing aqueous phase CRAs
measurements to nonclinical studies in animals, it is according to a recent industry survey [112]. It has been
important to take samples at intervals that are physi- argued that whole blood more closely mimics in vivo con-
ologically relevant to the specific immunomodulation ditions by involving soluble factors, platelets, and other
or immunopathology being induced. Cytokines are cells types when compared to PBMCs. In addition, whole-
released quickly and equally rapid in their clearance, blood CRAs require less technical effort to execute. Soluble
hence serial blood sampling time points should occur at or aqueous phase CRAs may use whole-blood or PBMCs
several intervals within the first 24 h of dose administra- and are attractive due to their simplicity but have been crit-
tion [111]. A standard approach is to sample 2–3 times icized for their lack of predictivity, particularly when using
within 24 h of dose administration (eg, 2–6, 12, and 24 h TGN1412, when compared to solid-phase CRAs [113].
postdose) in order to capture the positive cytokine sig-
nals, although this will need to be customized to the
particular project based on the goals of the study and Indicators of Hypersensitivity
the known pharmacology of the compound. Ongoing Type I hypersensitivity reactions are usually IgE-
immune/inflammatory stimulation may allow positive mediated, in humans and many nonclinical species,
signals to be detected at later intervals, although this is although IgG can also mediate systemic anaphylaxis in
more the exception than the rule. Increases in cytokines guinea pigs, rabbits, rats and mice [98]. Type I hyper-
(2–24 h) will usually precede increases in APPs (24–72 h). sensitivity could be systemic (anaphylaxis or urticarial)
In general, larger multiplex panels including multiple or respiratory (asthma). The systemic reaction involves
cytokines are used for these purposes. Some of the most the release of one or more vasoactive amines from hista-
common markers evaluated include IL-1, IL-6, TNF-α, mine, kinins, and serotonin. FDA requires that all drugs

III.  CLINICAL PATHOLOGY, HISTOPATHOLOGY, AND BIOMARKERS


466 17.  BIOMARKERS IN NONCLINICAL DRUG DEVELOPMENT

developed for dosing by inhalation route be evaluated Indicators of Immunogenicity


for ability to induce Type I hypersensitivity [101]. Thera-
peutic proteins may also elicit Type I reactions in mon- Molecules above 1000 Da may be immunogenic, thus
keys and other nonclinical species [115], but the relevance raising a twofold concern. The first is alteration of phar-
of such reaction to human risk assessment needs to con- macodynamics and loss of efficacy due to binding of
sider the species source of the protein. The demonstra- the test article to antidrug antibodies (ADA) in circula-
tion of drug-specific IgE should be considered a hazard tion, while the second is allergenicity. In general ADA
with or without the elevation of Th2 cytokines such IL-4, are used to determine the immunogenicity while mod-
IL-5, IL-10, and IL-13 [99,101]. The murine local lymph els of allergenicity testing have not undergone robust
node assay (LLNA) has undergone extensive validation validation.
since the first report [116] and has almost replaced other
sensitization assays in guinea pigs. It measures lympho- Indicators of Autoimmunity
cyte proliferation after topical exposure to xenobiotics.
A chemical is considered a sensitizer if, at any dose, it Xenobiotic-related autoimmunity has been associ-
induces threefold or greater cell proliferation in local ated with several drugs from different classes. They are
lymph node when compared to concurrent control. For related to the formation of hapten for molecules less
human risk assessment, the comparative thickness of than 1000 Da and often include a genetic predisposi-
human versus murine skin, which is thinner and more tion such as the HLA-DR3 allele in humans [110]. Due
permeable, needs to be considered [117]. to the complex nature of autoimmune diseases there are
Types II and III hypersensitivities are antibody-­ no generally recognized nonclinical testing approaches.
mediated and tend to cooccur in xenobiotic exposed ani- Drugs that cause autoimmune diseases tend to cause
mals, although Type III is more common in animals dosed hematological disorders such as neutropenia, thrombo-
with therapeutic proteins [101,115]. They result from IgG cytopenia, and anemia. Biotherapeutics are now known
and/or IgM mediated antibody-dependent/complement- to cause unexpected hematological findings and some
mediated cytotoxicity or lysis of somatic cells (Type II) or of these are immune-mediated and not predicted from
immune complex-related complement-mediated destruc- nonclinical studies. However, nonclinical studies have
tion of tissue (Type III). Pathology findings may include predicted clinical hematotoxicity for hemophagocytosis
one or more of vasculitis, glomerulonephritis, anemia, and (occurs with recombinant cytokines and growth factors)
thrombocytopenia. Type III hypersensitivity is increas- and thrombocytopenia, which is observed with anti-
ingly being observed in nonclinical safety assessment as CD40L monoclonal antibodies [118].
the testing of larger molecule biopharmaceuticals increase. In conclusion safety assessment of the immune sys-
Complement-related pathology is thought to arise from tem requires a weight-of-evidence approach using a
failure of the clearance system for soluble immune com- variety of modalities to evaluate lymphoid organs and
plexes. In some cases (subclinical) increases in circulat- bone marrow with careful understanding of the test
ing immune complexes and circulating IgM have been article and the immune system of the species in which
reported prior to or concurrent with the drug reaction the test is being conducted. A tiered approach starting
[115]. The kinetics of IgG/IgM and circulating complement with standard toxicology endpoints (eg, hematology,
on days preceding the drug reaction may be informative lymphoid organ, and bone-marrow pathology) with
when investigating drug reactions. The investigation of the addition of standalone immunotoxicity studies as
immune complex-mediated pathology requires a weight needed is the current paradigm for characterization of
of evidence approach using some of the following informa- the immune system.
tion (1) plasma and/or serum drug and antidrug antibody
(ADA) levels; (2) histopathology findings from animals Biomarkers of Toxicity in the Central Nervous
with drug reactions; (3) hematological indices such as
System
erythrocyte numbers and morphology, and platelet num-
bers; (4) circulating complement component levels, which The central nervous system (CNS) contains a vast
may not be available due to the manner of death, and is array of neuronal cell bodies (nuclei) that have very
rapid in some cases; and (5) immunohistochemical dem- specific functions. These cell bodies are protected from
onstration of granular deposits of drug, endogenous IgG, most xenobiotics by the blood–brain barrier. However,
IgM, and other complement components [115]. some molecules are designed to cross the blood–brain
Type IV hypersensitivity is a T-cell mediated delayed barrier or do so by nature of their physicochemical
type hypersensitivity. Drugs intended for topical use need properties. There are no routinely used molecular/
to be evaluated for their ability to sensitize [101], and the chemical validated biomarkers to assess xenobiotic-
LLNA is increasingly being accepted for the sensitization related injury to the nervous system. Functional obser-
aspect of Type IV reaction, in addition to Type I testing. vation battery (FOB) or other analogous test is required

III.  CLINICAL PATHOLOGY, HISTOPATHOLOGY, AND BIOMARKERS


Conclusion 467
for assessing the effects of test articles on indices of safety assessment there are elaborate toxicity studies
neurobehavioral function such as coordination, motor designed to assess the effects of xenobiotics on reproduc-
activity, behavioral changes, sensory/motor reflex tion. However, these studies are expensive, longer term,
responses, and body temperature [119]. The brain and and tend to occur later in the drug development process.
spinal cord are also evaluated histologically as part of Ideally, noninvasive biomarkers of reproductive toxic-
most standard nonclinical safety assessment programs. ity that can be measured in a 28-day repeat-dose toxicity
The current neurotoxicity testing paradigm relies heav- study will be good predictors to provide early warning
ily on morphological examinations, but the future will about reproductive toxicity and help contextualize the
probably include additional methods for understand- morphological findings that occur in the reproductive
ing receptor dynamics as well as cellular and biochemi- system during shorter-term studies.
cal alterations in the brain [120]. Two methods that Currently, hormones such as estrogen, progesterone,
might increase the sensitivity of detection and help elu- prolactin, testosterone, LH, and FSH are sometimes used
cidate mechanisms of CNS toxicity are cerebrospinal to provide additional context when there is a signal of
fluid (CSF) analysis and molecular imaging. reproductive effect. However, hormone endpoints can be
Cerebrospinal fluid analysis may reveal test article- difficult to assess in large toxicology studies due to the
related changes in CNS alteration through changes in difficulty in controlling variations due to stress, diurnal
the color and quantity of CSF, number and composi- variation, and menstrual/estrus cycling [123]. For female
tion of mononuclear cells in the CSF, and biochemical animals, estrus cycle evaluation provides another index
alterations of CNS [121]. Neurotoxic effects can result of reproductive health, although the variable length of
in the alteration of neurotransmitters and their metab- estrus cycle in different species makes it more useful
olites such as dopamine, glutamate, γ-aminobutyric in those with short cycles (eg, rat) and very difficult in
acid, epinephrine, serotonin, homovanillic acid, species with longer cycles (eg, dog). For testicular tox-
5-hydroxyindolacetic acid. For example, homovanillic icity, histopathology and organ weight of accessory sex
acid can be measured in the CSF of Collie dogs that organs are currently the most sensitive and reliable ways
develop neurologic signs after Ivermectin exposure of detecting test article-related effects [124]. However,
[121]. The concentrations of total protein, albumin, there is industry-wide interest in validating the utility
C-reactive protein, myelin basic protein, and S-100 can of inhibin B or other potential toxicogenomic mark-
also be indications of toxicant-related alterations in ers that will translate to human clinical trial scenario
blood–brain barrier and/or CNS. With the continued [125]. Androgens stimulate sertoli cells to secrete inhibin
refinement of metabolomics and spectrometry tech- B, which regulates spermatogenesis in seminiferous
niques, CSF analysis might become a better-utilized tubules. Clinically, serum inhibin B tends to be higher in
adjunct for CNS safety assessment. fertile men than in infertile men [126]. Therefore inhibin
Recently imaging techniques such as magnetic B appears to be a good biomarker for testicular toxicity
resonance imaging (MRI), positron-emission tomog- because it can be measured in a nonterminal longitudi-
raphy (PET), and single photon-emission computed nal fashion and is translatable.
tomography (SPECT) have been used in drug dis-
covery and development. These imaging techniques
have been used to (1) detect activated microglia in CONCLUSION
neuroinflammation by using a radiotracer to target
a translocator protein (TP-18), (2) access the quan- Several routinely and seldom used biomarkers were
tity and integrity of adhesion molecules (CD62E) and presented for different tissues and organ systems. A
P-glycoptoteins(CD243) in the blood–brain barrier, key feature of interpreting data from these biomarkers
and (3) CNS glucose metabolism by using [18F]fluo- is to have an appreciation for the heterogeneity of posi-
rodeoxyglucose uptake [122]. As these imaging tech- tive biomarker response signals in timing, magnitude,
nology capabilities improve and reduce in cost, they and by species, and to have a good understanding of
are likely to provide more comprehensive and specific the analytical performance each individual assay. Such
approaches to assess CNS safety of xenobiotics. appreciation frequently involves professionals who
are trained and experienced in the molecular and com-
parative physiological foundations of these biomarkers
Biomarkers of Toxicity in the Reproductive
and their clinical relevance. A careful case-by-case and
System integrative interpretation of data from these biomark-
The reproductive system has a complex hormonal ers often provides a better perspective on the potential
regulatory network that ranges from gonadal sex steroid risk(s) of xenobiotic-related safety issues and informs
hormones to centrally released luteinizing hormone (LH) on the appropriate risk mitigation strategies in drug
and follicle stimulating hormone (FSH). In nonclinical development.

III.  CLINICAL PATHOLOGY, HISTOPATHOLOGY, AND BIOMARKERS


468 17.  BIOMARKERS IN NONCLINICAL DRUG DEVELOPMENT

References [20] Tomlinson L, Boone LI, Ramaiah L, Penraat KA, von Beust BR,
Ameri M, et al. Best practices for veterinary toxicologic clinical
[1]  Biomarkers Definition Working Group. Biomarkers and sur- pathology, with emphasis on the pharmaceutical and biotechnol-
rogate endpoints: preferred definitions and conceptual frame- ogy industries. Vet Clin Pathol 2013;42(3):252–69.
works. Clin Pharmacol Ther 2001;69(3):89–95. [21] Weingand K, Brown G, Hall R, Davies D, Gossett K, Neptun D,
[2] Sasseville VG, Mansfield KG, Brees DJ. Safety biomarkers in et al. Harmonization of animal clinical pathology testing in tox-
preclinical development: translational potential. Vet Pathol icity and safety studies. The Joint Scientific Committee for Inter-
2014;51(1):281–91. national Harmonization of Clinical Pathology Testing. Fundam
[3] Lock EA, Bonventre JV. Biomarkers in translation; past, present Appl Toxicol 1996;29(2):198–201.
and future. Toxicology 2008;245(3):163–6. [22] Amacher DE. A toxicologist’s guide to biomarkers of hepatic
[4] Zhu X, Verma S. Targeted therapy in HER2-positive metastatic response. Hum Exp Toxicol 2002;21:253–62.
breast cancer: a review of the literature. Curr Oncol March [23] Ozer J, Ratner M, Shaw M, Bailey W, Schomaker S. The cur-
2015;22(Suppl. 1):S19–28. rent state of serum biomarkers of hepatotoxicity. Toxicology
[5] Dean L. Warfarin therapy and the genotypes CYP2C9 and 2008;245:194–205.
VKORC1. In: Medical genetics summaries. Bethesda (MD): [24] Amacher DE, Schomaker SJ, Burkhardt JE. The relationship
National Center for Biotechnology Information; 2012. among microsomal enzyme induction, liver weight, and his-
[6] Amuzie CJ, Pestka JJ. Suppression of insulin-like growth fac- tological change in rat toxicology studies. Food Chem Toxicol
tor acid-labile subunit expression – a novel mechanism for 1998;36:831–9.
deoxynivalenol-induced growth retardation. Toxicol Sci [25] Amacher DE, Schomaker SJ, Burkhardt JE. The relation-
2010;113(2):412–21. ship among enzyme induction, liver weight, and histologi-
[7] Lee J, Russ S, Weiner R, Sailstad JM, Bowsher RR, Knuth DW, cal change in beagle toxicology studies. Food Chem Toxicol
et al. Method validation and measurement of biomarkers in non- 2001;39:817–25.
clinical and clinical samples in drug development: a conference [26] Ramaiah SK. Preclinical safety assessment: current gaps, chal-
report. Pharm Res 2005;22(4):499–511. lenges, and approaches in identifying translatable biomarkers of
[8] Lee J, Devanarayan V, Barrett YC, Weiner R, Allinson J, Fountain drug-induced liver injury. Clin Lab Med 2011;31(1):161–72.
S, et al. Fit-for-purpose method development and validation [27] Ozer JS, Chetty R, Kenna G, Koppiker N, Karamjeet P, Li D, et al.
for successful biomarker measurement. Pharm Res 2006;23(2): Recommendations to qualify biomarker candidates of drug-
312–28. induced liver injury. Biomarkers Med 2010;4(3):475–83.
[9] Cummings J, Ward TH, Dive C. Fit-for-purpose biomarker [28] Regev A. How to avoid being surprised by hepatotoxicity at the
method validation in anticancer drug development. Drug Dis- final stages of drug development and approval. Clin Liver Dis
cov Today 2010;15(19/20). 2013;17:749–67.
[10] Policy issues for the development and use of biomarkers in [29] Blair PC, Thompson MB, Wilson RE, Esber HH, Maronpot RR.
health. 2011. www.Oecd.Org/Sti/Biotechnology. Correlation of changes in serum analytes and hepatic histopa-
[11] Lee JW, Pan P, O’Brien P, et al. Development and validation of thology in rats exposed to carbon tetrachloride. J Comp Pathol
ligand-binding assays for biomarkers. In: Khan MN, Findlay 1988;98:381–404.
JWA, editors. Ligand-binding assays: development, validation, [30] Landi MS, Kissinger JT, Campbell SA, Kenney CA, Jenkins Jr EL.
and implementation in the drug development arena. Hoboken The effects of four types of restraint on serum alanine amino-
(NJ): John Wiley and Sons, Inc.; 2010. p. 129–62. transferase and aspartate aminotransferase in the Macaca fascicu-
[12] US FDA. Biomarker qualification program. 2012. http://www. laris. J Am Coll Toxicol 1990;9:517–23.
fda.gov/Drugs/DevelopmentApprovalProcess/DrugDevelop- [31] O’Brien PJ, Slaughter MR, Polley SR, Kramer K. Advantages of
mentToolsQualificationProgram/ucm284076.htm. glutamate dehydrogenase as a blood biomarker of acute hepatic
[13] Khan M, Bowsher RR, Cameron M, Devanarayan V, Keller S, injury in rats. Lab Anim 2002;36:313–21.
King L, et al. Recommendations for adaptation and validation [32] Zollner G, Marschall HU, Wagner M, Trauner M. Role of nuclear
of commercial kits for biomarker quantification in drug develop- receptors in the adaptive response to bile acids and cholesta-
ment. Bioanalysis 2015;7(2):229–42. sis: pathogenetic and therapeutic considerations. Mol Pharm
[14] Bioanalytical method validation. U.S. Department of Health and 2006;3:231–51.
Human Services, Food and Drug Administration, Center for [33] Meyer DJ, Harvey JW. Hepatobiliary and skeletal muscle
Drug Evaluation and Research (CDER), Center for Veterinary enzymes and liver function tests. In: Meyer DJ, Harvey JW, edi-
Medicine (CVM); 2013. tors. Veterinary laboratory medicine: interpretation and diagno-
[15] Han M, Li Y, Liu M, Cong B. Renal neutrophil gelatinase asso- sis. 3rd ed. St. Louis (MO): Saunders; 2004. p. 169–92.
ciated lipocalin expression in lipopolysaccharide-induced acute [34] Zucker SD, Qin X, Rouster SD, Yu F, Green RM, Keshavan P,
kidney injury in the rat. BMC Nephrol 2012;13:25. et al. Mechanism of indinavir-induced hyperbilirubinemia. Proc
[16] Cray C, Zaias J, Altman N. Acute phase response in animals: a Natl Acad Sci USA 2001;98:12671–6.
review. Comp Med 2009;59(6):517–26. [35] Amacher DE, Adler R, Herath A, Townsend RR. Use of proteomic
[17] Gautier JC, Gury T, Guffroy M, Khan-Malek R, Hoffman D, methods to identify serum biomarkers associated with rat liver
Petit S, et al. Normal ranges and variability of novel urinary toxicity or hypertrophy. Clin Chem 2005;51(10):1796–803.
renal biomarkers in Sprague-Dawley rats: comparison of con- [36] Yang X, Schnackenberg L, Shi Q, Salminen WF. Hepatic toxic-
stitutive values between males and females and across assay ity biomarkers. In: Gupta R, editor. Biomarkers in toxicology.
platforms. Toxicol Pathol 2014;42:1092–104. Waltham (MA): Elsevier; 2013. p. 241–60.
[18] Weiler S, Merz M, Kullak-Ublick GA. Drug-induced liver injury: [37] Schomaker S, Warner R, Bock J, Johnson K, Potter D, Van Winkle
the dawn of biomarkers. F1000 Prime Rep 2015;7:34. J, et al. Assessment of emerging biomarkers of liver injury in
[19] Boone L, Meyer D, Cusick P, Ennulat D, Provencher Bolliger A, human subjects. Toxicol Sci 2013;132(2):276–83.
Everds N, et al. Selection and interpretation of clinical pathology [38] Schnellman R. Toxic responses of the kidney. In: Casarett, Doulls,
indicators of hepatic injury in preclinical studies. Vet Clin Path editors. Toxicology: the basic science of poisons. 7th ed. New
2005;34(3):182–8. York (NY): McGraw-Hill; 2008. p. 583–608.

III.  CLINICAL PATHOLOGY, HISTOPATHOLOGY, AND BIOMARKERS


References 469
[39] Stockham S, Scott M. Enzymes. In: Fundamentals of veterinary [56] Davis J, Goodsaid F, Bral C, Obert L, Mandakas G, Garner C,
clinical pathology. 2nd ed. Ames (IA): Blackwell Publishing; et al. Quantitative gene expression analysis in a nonhuman pri-
2008. mate model of antibiotic-induced nephrotoxicity. Toxicol Appl
[40] Dieterle F, Sistare F, Goodsaid F, Papaluca M, Ozer J, Webb C, Pharmacol 2004;200:16–26.
et al. Renal biomarker qualification submission: a dialog between [57] Rosenberg ME, Paller MS. Differential gene expression in the
the FDA-EMEA and predictive safety testing consortium. Nat recovery from ischemic renal injury. Kidney Int 1991;39:1156–61.
Biotechnol 2010b;28:455–62. [58] Witzgall R, Brown D, Schwarz C, Bonventre J. Localization of
[41] Ennulat D, Adler S. Recent successes in the identification, proliferating cell nuclear antigen, vimentin, c-Fos, and clusterin
development, and qualification of translational biomarkers: in the postischemic kidney. Evidence for a heterogenous genetic
the next generation of kidney injury biomarkers. Toxicol Pathol response among nephron segments, and a large pool of mitotically
2015;43:62–9. active and dedifferentiated cells. Clin Invest 1994;93:2175–88.
[42] Han W, Bailly V, Abichandani R, Thadhani R, Bonventre J. Kid- [59] Filler G, Bokenkamp A, Hofmann W, Le Bricon T, Martinez-Bru
ney injury molecule-1 (KIM-1): a novel biomarker for human C, Grubb A. Cystatin C as a marker of GFR: history, indications,
renal proximal tubule injury. Kidney Int 2002;62:237–44. and future research. Clin Biochem 2005;38(1):1–8.
[43] Vlasakova K, Erdos Z, Troth S, McNulty K, Chapeau-Campredon [60] Conti M, Moutereau S, Zater M, Lallali K, Durrbach A, Manivet
V, Mokrzycki N, et al. Evaluation of the relative performance of P, et al. Urinary cystatin C as a specific marker of tubular dys-
12 urinary biomarkers for renal safety across 22 rat sensitivity function. Clin Chem Lab Med 2006;44:288–91.
and specificity studies. Toxicol Sci 2014;8(1):3–20. [61] Uchida K, Gotoh A. Measurement of cystatin-C and creatinine in
[44] Ichimura T, Bonventre J, Bailly V, Wei H, Hession C, Cate R, urine. Clin Chim Acta 2002;323:121–8.
et al. Kidney injury molecule-1 (KIM-1), a putative epithelial [62] Clemo F. Urinary enzyme evaluation of nephrotoxicity in the
cell adhesion molecule containing a novel immunoglobulin dog. Toxicol Pathol 1998;26(1):29–32.
domain, is up-regulated in renal cells after injury. J Biol Chem [63] Emeigh Hart SG. Assessment of renal injury in vivo. Pharmacol
1998;273:4135–42. Toxicol Methods 2005;52:30–45.
[45] Bonventre J, Vaidya V, Schmouder R, Feig P, Dieterle F. Next- [64] D’Amico G, Bazzi C. Urinary protein and enzyme excretion
generation biomarkers for detecting kidney toxicity. Nat Biotech- as markers of tubular damage. Curr Opin Nephrol Hypertens
nol 2010;28(5). 2003;12:639–43.
[46] Perez-Rojas J, Blanco J, Cruz C, Trujillo J, Vaidya V, Uribe N, et al. [65] Antman E, Bassand J, Klein W, Ohman M, Sendon J, Rydén L,
Mineralocorticoid receptor blockade confers renoprotection in et al. Myocardial infarction redefined—a consensus document of
preexisting chronic cyclosporine nephrotoxicity. Am J Physiol the Joint European Society of Cardiology/American College of
Renal Physiol 2007;292:F131–9. Cardiology Committee for the redefinition of myocardial infarc-
[47] Vaidya V, Ramirez V, Ichimura T, Bobadilla N, Bonventre J. tion: the Joint European Society of Cardiology/American Col-
Urinary kidney injury molecule-1: a sensitive quantitative bio- lege of Cardiology Committee. J Am Coll Cardiol 2000;36:959–69.
marker for early detection of kidney tubular injury. Am J Physiol [66] Apple F, Murakami M, Ler R, Walker D, York M. Analytical char-
Renal Physiol 2006;290:F517–29. acteristics of commercial cardiac troponin I and T immunoassays
[48] Liangos O, Perianayagam M, Vaidya V, Han W, Wald R, Tighi- in serum from rats, dogs, and monkeys with induced acute myo-
ouart H, et al. Urinary N-acetyl-beta-(d)-glucosaminidase cardial injury. Clin Chem 2008;54(12):1982–9.
activity and kidney injury molecule-1 level are associated with [67] Bertinchant J, Robert E, Polge A, Marty-Double C, Fabbro-Peray P,
adverse outcomes in acute renal failure. J Am Soc Nephrol Poirey S, et al. Comparison of the diagnostic value of cardiac tropo-
2007;18:904–12. nin I and T determinations for detecting early myocardial damage
[49] Borregaard N, Sehested M, Nielsen B, Sengelov H, Kjeldsen L. and the relationship with histological findings after isoproterenol-
Biosynthesis of granule proteins in normal human bone marrow induced cardiac injury in rats. Clin Chim Acta 2000;298(1–2):13–28.
cells. Gelatinase is a marker of terminal neutrophil differentia- [68] York M, Scudamore C, Brady S, Chen C, Wilson S, Curtis M, et al.
tion. Blood 1995;85:812–7. Characterization of troponin responses in isoproterenol-induced
[50] Matthaeus T, Schulze-Lohoff E, Ichimura T, Weber M, Andreucci cardiac injury in the Hanover Wistar rat. Toxicol Pathol 2007;35.
M. Co-regulation of neutrophil gelatinase-associated lipocalin [69] Bertanchant J, Polge A, Juan J, Oliva-Lauraire M, Giuliani I,
and matrix metalloproteinase-9 in the postischemic rat kidney. J Marty-Double C, et al. Evaluation of cardiac troponin I and T
Am Soc Nephrol 2001;12:787A. levels as markers of myocardial damage in doxorubicin-induced
[51] Mishra J, Ma Q, Prada A, Mitsnefes M, Zahedi K, Yang J, et al. cardiomyopathy rats, and their relationship with echocardio-
Identification of neutrophil gelatinase-associated lipocalin as a graphic and histological findings. Clin Chim Acta 2003;329.
novel early urinary biomarker for ischemic renal injury. Am Soc [70] Schultze A, Carpenter K, Wians F, Agee S, Minyard J, Lu Q, et al.
Nephrol 2003;14:2534–43. Longitudinal studies of cardiac troponin-i concentrations in
[52] Singer E, Marko L, Paragas N, Barasch J, Dragun D, Muller DN, serum from male Sprague Dawley rats: baseline reference ranges
et al. Neutrophil gelatinase-associated lipocalin: pathophysiol- and effects of handling and placebo dosing on biological vari-
ogy and clinical applications. Acta Physiol (Oxf) 2013;207:663–72. ability. Toxicol Pathol 2009;37:754–60.
[53] Ohlsson S, Wieslander J, Segelmark M. Increased circulating lev- [71] Metzger J, Westfall M. Covalent and noncovalent modification
els of proteinase 3 in patients with antineutrophilic cytoplasmic of thin filament action. The essential role of troponins in cardiac
autoantibodies-associated systemic vasculitis in remission. Clin muscle regulation. Circ Res 2004;94:146–58.
Exp Immunol 2003;131:528–35. [72] Chmurzyńska A. The multigene family of fatty acid-binding
[54] Xu S, Pauksen K, Venge P. Serum measurements of human neu- proteins (FABPs): function, structure and polymorphism. J Appl
trophil lipocalin (HNL) discriminate between acute bacterial and Genet 2006;47(1):39–48.
viral infections. Scand J Clin Lab Invest 1995;55:125–31. [73] Clements P, Brady S, York M, Berridge B, Mikaelian I, Nicklaus R,
[55] Dieterle F, Perentes E, Cordier A, Roth D, Verdes P, Grenet O, et al. Time course characterization of serum cardiac troponins,
et al. Urinary clusterin, cystatin C, β2-microglobulin and total heart fatty acid binding protein, and morphologic findings
protein as markers to detect drug-induced kidney injury. Nat with isoproterenol-induced myocardial injury in the rat. Toxicol
Biotechnol 2010;28(5):463–8. Pathol 2010;38.

III.  CLINICAL PATHOLOGY, HISTOPATHOLOGY, AND BIOMARKERS


470 17.  BIOMARKERS IN NONCLINICAL DRUG DEVELOPMENT

[74] Ghani F, Wu A, Graff L, Petry C, Armstrong G, Prigent F, et al. [93] Tonomuraa Y, Matsushimaa S, Kashiwagi E, Fujisawaa K, Takagi
Role of heart-type fatty acid-binding protein in early detection of S, Nishimuraa Y, et al. Biomarker panel of cardiac and skeletal
acute myocardial infarction. Clin Chem 2000;46:718–9. muscle troponins, fatty acid binding protein 3 and myosin light
[75] Zhen E, Berna M, Jin Z, Pritt M, Watson D, Ackermann B, et al. chain 3 for the accurate diagnosis of cardiotoxicity and musculo-
Quantification of heart fatty acid–binding protein as a biomarker skeletal toxicity in rats. Toxicology 2012;302:179–89.
for drug-induced cardiac and musculoskeletal necroses. Pro- [94] Weaver J, Snyder R, Knapton A, Herman E, Honchel R, Miller T,
teomics 2007;1(7):661–71. et al. Biomarkers in peripheral blood associated with vascular injury
[76] Katus H, Yasuda T, Gold H, Leinbach R, Strauss H, Waksmon- in Sprague-Dawley rats treated with the phosphodiesterase IV
ski C, et al. Diagnosis of acute myocardial infarction by detec- inhibitors SCH 351591 or SCH 534385. Toxicol Pathol 2008;36:840–9.
tion of circulating cardiac myosin light chains. Am J Cardiol [95] Kerns W, Schwartz L, Blanchard K, Burchiel S, Essayan D, Fung
1984;54(8):964–97. E, et al. Drug-induced vascular injury – a quest for biomarkers.
[77] Walker D. Serum chemical biomarkers of cardiac injury for non- Toxicol Appl Pharmacol 2005;203:62–87.
clinical safety testing. Toxicol Pathol 2006;34. [96] Mikaelian I, Cameron M, Dalmas D, Enerson B, Gonzalez R, Gui-
[78] Ellis A, Little T, Zaki R, Masud A, Klocke F. Patterns of myoglo- onaud S, et al. Nonclinical safety biomarkers of drug-induced
bin release after reperfusion of injured myocardium. Circulation vascular injury: current status and blueprint for the future. Toxi-
1985;72:639–47. col Pathol 2014;42:1533–601.
[79] Clerico A, Emdin M. Diagnostic accuracy and prognostic rel- [97] Bhogal N, Combes R. TGN 1412: time to change the paradigm for
evance of the measurement of cardiac natriuretic peptides: a testing of new pharmaceuticals. Altern Lab Anim 2006;34:225–39.
review. Clin Chem 2004;50:133–50. [98] Haley P. Species differences in structure and function of the
[80] Bay M, Kirk V, Parner J, Hassager C, Nielsen H, Krogsgaard K, immune system. Toxicology 2003;188:49–71.
et al. NT-proBNP: a new diagnostic screening tool to differenti- [99] Kaminski NE, Faubert Kaplan BF, Holsapple MP. Toxic
ate between patients with normal and reduced left ventricular responses of the immune system. In: Klaassen CD, editor. Casa-
systolic function. Heart 2003;89(2):150–4. rett & Doull’s toxicology: the basic science of poisons. 7th ed.
[81] Yasue H, Yoshimura M, Sumida H, et al. Localization and mech- New York (NY): McGraw-Hill Companies; 2008. p. 485–555.
anism of secretion of B-type natriuretic peptide in comparison [100] House RV, Luster MI, Dean JH, Johnson VJ. Immunotoxicology:
with those of A-type natriuretic peptide in normal subjects and the immune system response to toxic insult. In: Hayes AW, Kru-
patients with heart failure. Circulation 1994;90:195–203. ger CL, editors. Hayes’ principles and methods of toxicology. 6th
[82] MacDonald K, Kittleson M, Munro C, Kass P. Brain natriuretic ed. Boca Raton (FL): CRC Press; 2014. p. 1793–830.
peptide concentration in dogs with heart disease and congestive [101]  FDA. Guidance for industry: immunotoxicology evaluation
heart failure. J Vet Intern Med 2003;17:172–7. of investigational new drugs. Center for Drug Evaluation and
[83] Mizuno Y, Yoshimura M, Harada E. Plasma levels of A and Research (CDER), Food and Drug Administration, U.S. Depart-
B-type natriuretic peptides in patients with hypertrophic cardio- ment of Health and Human Services; 2002.
myopathy or idiopathic dilated cardiomyopathy. Am J Cardiol [102]  FDA. Guidance for industry: S8 immunotoxicity studies for
2000;86:1036–40. human pharmaceuticals. US Department of Health and Human
[84] Lang C, Prasad N, McAlpine H. Increased plasma levels of brain Services, Food and Drug Administration, Center for Drug Evalu-
natriuretic peptide in patients with isolated diastolic dysfunc- ation and Research (CDER), Center for Biologics Evaluation and
tion. Am Heart J 1994;127:1635–6. Research; 2006.
[85] Cheung B. Plasma concentration of brain natriuretic peptide is [103] Coffin DL, Blommer EJ. Acute toxicity of irradiated auto exhaust.
related to diastolic function in hypertension. Clin Exp Pharmacol Its indication by enhancement of mortality from Streptococcal
Physiol 1997;24:966–8. pneumonia. Arch Environ Health 1967;15(1):36–8.
[86] Cardinet G. Skeletal muscle function. In: Kaneko J, Harvey J, [104] Haley P, Perry R, Ennulat D, Frame S, Johnson C, Lapointe J-
Bruss M, editors. Clinical biochemistry of domestic animals. 5th M, et al. STP position paper: best practice guideline for the rou-
ed. San Diego (CA): Academic Press; 1997. tine pathology evaluation of the immune system. Toxicol Pathol
[87] Fontanet H, Trask R, Haas R, Strauss A, Abendschein D, Bil- 2005;33:404–7.
ladello J. Regulation of expression of M, B, and mitochondrial [105] Sellers RS, Morton D, Michael B, Roome N, Johnson JK, Yano
creatine kinase mRNAs in the left ventricle after pressure over- BL, et al. Society of toxicologic pathology position paper: organ
load in rats. Circ Res 1991;68:1007–12. weight recommendations for toxicology studies. Toxicol Pathol
[88] Sharkey S, Murakami M, Smith S, Apple F. Canine myocardial 2007;35(5):751–5.
creatine kinase isoenzymes after chronic coronary artery occlu- [106] Kuper CF, Harleman JH, Richter-Reichelm HB, Vos JG. Histo-
sion. Circulation 1991;84:333–40. pathologic approaches to detect changes indicative of immuno-
[89] Hironaka E, Hongo M, Azegami M, Yanagisawa S, Owa M, toxicity. Toxicol Pathol 2000;28(3):454–66.
Hayama M. Effects of angiotensin-converting enzyme inhibition [107] Elmore SA. Enhanced histopathology of the immune system: a
on changes in left ventricular myocardial creatine kinase system review and update. Toxicol Pathol 2012;40(2):148–56.
after myocardial infarction: their relation to ventricular remodel- [108] Immunotoxicology Technical Committee, International Life Sci-
ing and function. Jpn Heart J 2003;44:537–46. ences Institute Health and Environmental Sciences Institute.
[90] Chow C, Cross C, Kaneko J. Lactate dehydrogenase activ- Application of flow cytometry to immunotoxicity testing: sum-
ity and isoenzyme pattern in lungs, erythrocytes, and plasma mary of a workshop. Toxicology 2001;163:39–48.
of ozoneexposed rats and monkeys. J Toxicol Environ Health [109] Kim GG, Donnenberg VS, Donnenberg AD, Gooding W, Whi-
1977;3:877–84. teside TL. A novel multiparametric flow cytometry-based cyto-
[91] Nathwani R, Pais S, Reynolds T, Kaplowitz N. Serum alanine toxicity assay simultaneously immunophenotypes effector cells:
aminotransferase in skeletal muscle diseases. Hepatology comparisons to a 4 h 51Cr-release assay. J Immunol Methods
2005;41(2):380–2. 2007;325(1–2):51–66.
[92] Simpson J, Labugger R, Hesketh G, D’Arsigny C, O’Donnell D, [110] Pestka JJ, Amuzie CJ. Tissue distribution and proinflamma-
Matsumoto M, et al. Differential detection of skeletal troponin I tory cytokine gene expression following acute oral exposure to
isoforms in serum of a patient with rhabdomyolysis: markers of deoxynivalenol: comparison of weanling and adult mice. Food
muscle injury. Clin Chem 2002;48(7):1112–4. Chem Toxicol 2008;46(8):2826–31.

III.  CLINICAL PATHOLOGY, HISTOPATHOLOGY, AND BIOMARKERS


References 471
[111] Tarrant J. Blood cytokines as biomarkers of in Vivo toxicity in pre- [119] EMEA. ICH topic S 7 A. Safety pharmacology studies for human
clinical safety assessment: considerations for their use. Toxicol pharmaceuticals. European Medicines Agency; 2006. p. 6.
Sci 2010;117(1):4–16. [120] Bolon B, Butt M. Toxicological neuropathology: the next two
[112] Finco D, Grimaldi C, Fort M, Walker M, Kiessling A, Wolf B, et al. decades. In: Bolon B, Butt M, editors. Fundamental neuropathol-
Cytokine release assays: current practices and future directions. ogy for pathologists and toxicologists. Hoboken (NJ): John Wiley
Cytokine 2014;66(2):143–55. and Sons; 2011. p. 537–40.
[113] Vessillier S, Eastwood D, Fox B, Sathish J, Sethu S, Dougall T, [121] Vernau W, Vernau KM, Bolon B. Cerebrospinal fluid analysis in
et al. Cytokine release assays for the prediction of therapeutic toxicological neuropathology. In: Bolon B, Butt M, editors. Fun-
mAb safety in first-in man trials – whole blood cytokine release damental neuropathology for pathologists and toxicologists.
assay are poorly predictive for TGN1412 cytokine storm. J Hoboken (NJ): John Wiley and Sons; 2011. p. 271–81.
Immunol Methods 2015;424:43–52. [122] Gabrielson K, Fletcher C, Czoty PW, Nader AM, Gluckman T,
[114] Eastwood D, Bird C, Dilger P, Hockley J, Findlay L, Poole S, et al. In vivo imaging applications for the nervous system in animal
Severity of the TGN1412 trial disaster cytokine storm correlated models. In: Bolon B, Butt M, editors. Fundamental neuropathol-
with IL-2 release. Br J Clin Pharmacol 2013;76(2):299–315. ogy for pathologists and toxicologists. Hoboken (NJ): John Wiley
[115] Rojko JL, Evans MG, Price SA, Han B, Waine G, DeWitte M, et al. and Sons, Inc.; 2011. p. 271–81.
Formation, clearance, deposition, pathogenicity, and identifica- [123] Everds N, Snyder P, Bailey K, Bolon B, Creasy D, Foley G, et al.
tion of biopharmaceutical-related immune complexes: review Interpreting stress responses during routine toxicity studies: a
and case studies. Toxicol Pathol 2014;42(4):725–64. review of the biology, impact, and assessment. Toxicol Pathol
[116] Kimber I, Weisenberger C. A murine local lymph node assay 2013;41:560–614.
for the identification of contact allergens. Assay develop- [124] Creasy DM. Pathogenesis of male reproductive toxicity. Toxicol
ment and results of an initial validation study. Arch Toxicol Pathol 2001;33:404–7.
1989;63(4):274–82. [125] Sasaki JC, Chapin RE, Hall DG, Breslin W, Moffit J, Saldutti
[117] Gwaltney-Brant S, et al. Immunotoxicity biomarkers.. In: Gupta L, et al. Incidence and nature of testicular toxicity find-
R, editor. Biomarkers in toxicology. Waltham (MA): Elsevier; ings in pharmaceutical development. Birth Defects Res B
2014. p. 373–83. 2011;92:511–25.
[118] Everds N, Tarrant J. Unexpected hematologic effects of biothera- [126] Myers GM, Lambert-Messerlian GM, Sigman M. Inhibin B ref-
peutics in nonclinical species and in humans. Toxicol Pathol erence data for fertile and infertile men in Northeast America.
2013;41(2):280–302. Fertil Steril 2009;92(6):1920–3.

III.  CLINICAL PATHOLOGY, HISTOPATHOLOGY, AND BIOMARKERS

Вам также может понравиться