Vous êtes sur la page 1sur 10

Scientific and Regulatory Policy Committee

Toxicologic Pathology
2016, Vol. 44(2) 163-172

STP Best Practices for Evaluating Clinical ª The Author(s) 2016


Reprints and permission:
sagepub.com/journalsPermissions.nav
Pathology in Pharmaceutical DOI: 10.1177/0192623315624165
tpx.sagepub.com
Recovery Studies

Lindsay Tomlinson1, Lila Ramaiah2, Niraj K. Tripathi3,


Valerie G. Barlow4, Allison Vitsky5, Florence M. Poitout-Belissent6,
Denise I. Bounous7, and Daniela Ennulat8

Abstract
The Society of Toxicologic Pathology formed a working group in collaboration with the American Society for Veterinary Clinical
Pathology to provide recommendations for the appropriate inclusion of clinical pathology evaluation in recovery arms of nonclinical
toxicity studies but not on when to perform recovery studies. Evaluation of the recovery of clinical pathology findings is not
required routinely but provides useful information on risk assessment in nonclinical toxicity studies and is recommended when the
ability of the organ to recover is uncertain. The study design generally requires inclusion of concurrent controls to separate
procedure-related changes from test article–related changes, but return of clinical pathology values toward baseline may be suf-
ficient in some cases. Evaluation of either a select or full panel of standard hematology, coagulation, and serum and urine chemistry
biomarkers can be scientifically justified. It is also acceptable to redesignate dosing phase animals to the recovery phase or vice versa
to optimize data interpretation. Assessment of delayed toxicity during the recovery phase is not required but may be appropriate in
development programs with unique concerns. Evaluation of the recovery of clinical pathology data for vaccine development is
required and, for efficacy markers, is recommended if it furthers pharmacologic understanding.

Keywords
clinical pathology, toxicologic pathology, preclinical research and development, preclinical safety assessment/risk management

Introduction and Background Group of STP and the Regulatory Affairs Committee of the
American Society for Veterinary Clinical Pathology (ASVCP)
Previous literature has addressed best practices in study design to provide insight on the following points regarding inclusion
and evaluation of the recovery phases for nonclinical toxicity
of clinical pathology evaluation in recovery studies: (1) appro-
studies (Perry et al. 2013; Pandher, Leach, and Burns-Naas
priate comparisons for assessment of recovery of clinical
2012; Sewell et al. 2014). While clinical pathology was
pathology, (2) considerations of tissue recovery, (3) selection
mentioned in these articles, specific considerations for the
of recovery biomarkers based on dosing phase data, (4) consid-
appropriate inclusion of clinical pathology evaluation were not
erations for exploratory or newly approved biomarkers,
explored in detail. Standard clinical pathology biomarker
(alternatively known in general as ‘‘variable’’ and as ‘‘analyte’’
for a chemistry biomarker) evaluation is critical for the moni-
toring of potential toxicity in the clinic following characteriza-
1
tion in nonclinical studies, and it is also crucial to the Pfizer Inc., Andover, Massachusetts, USA
2
understanding of recovery from toxicity. Since regulations for Envigo (formerly Huntingdon Life Sciences), East Millstone, New Jersey, USA
3
Covance Inc., Madison, Wisconsin, USA
inclusion of clinical pathology in recovery arms of toxicity 4
Valerie G. Barlow Consulting, LLC
studies are neither detailed nor stringent, an opportunity exists 5
Pfizer Inc., La Jolla, Massachusetts, USA
6
for a review of the common practices in the pharmaceutical Charles River Laboratories, Senneville, Quebec, Canada
7
industry and to make recommendations based on sound scien- Bristol-Myers Squibb Company, Princeton, New Jersey, USA
8
tific practices, with input from pathologists having diverse GlaxoSmithKline, King of Prussia, Pennsylvania, USA
backgrounds. The Scientific and Regulatory Policy Committee Corresponding Author:
of the Society of Toxicologic Pathology (STP) formed a work- Lindsay Tomlinson, Pfizer Inc., 1 Burtt Rd., Andover, MA 01810, USA.
ing group in collaboration with the Clinical Pathology Interest Email: lindsay.tomlinson@pfizer.com
164 Toxicologic Pathology 44(2)

Table 1. Common Study Designs for Clinical Pathology Recovery Evaluation.a

Recovery
Dosing phase phase N/dose/ Length of Length of
Nb/dose/sex sex (N of dosing recovery
Species (N of groups) groups) phase phase Clinical pathology time points Clinical pathology evaluation
c
Mouse 10 to 20 (4) 5–10 (2 or 4) 2–13 weeks 2–4 weeks End of dosing + end of recovery Hematology and/or chemistry
Rat 10 (4) 5 (2 or 4) 2–26 weeks 2–13 weeks +Interim, end of dosing, and Hematology, coagulation,
+end of recovery chemistry, urinalysis, and/or
biomarkers or noncore
biomarkers
Dog/monkey 3–4 (4) 2–4 (2 or 4) 2–26 weeks 2–13 weeks Pretreatment(s), +interim(s) Hematology, coagulation,
and end of dosing, +interim chemistry, urinalysis, and/or
recovery and end of recovery biomarkers or noncore
biomarkers
a
Regulated good laboratory practice studies form the basis of this experience.
b
N ¼ number.
c
Different cohorts will be evaluated at end of dosing and end of recovery.

(5) evaluation of delayed toxicity, (6) biomarker recovery in phase as recommended by Weingand et al. (1996) and then mon-
vaccine studies, and (6) recovery of efficacy markers. itored at a frequency that allows assessment of changes over time
(EMA Committee of Human Medicinal Products/Safety Working
Party 2010). If there is a need for single-dose toxicity studies,
Current Regulatory Guidelines hematology and clinical chemistry data should be evaluated
*24 hr after a single administration, with further evaluations
Regulatory guidelines from organizations and agencies responsible
conducted 2 weeks later to assess for delayed toxicity and/or
for the investigation of the safety and/or efficacy of chemicals or
recovery (EMA Committee of Human Medicinal Products 2010).
human health products, such as the International Conference of
The World Health Organization (WHO) guidelines on vac-
Harmonization (ICH), U.S. Food and Drug Administration (FDA),
cines (Guidelines on Nonclinical Evaluation of Vaccines; WHO
Environmental Protection Agency (EPA), European Medicines
2005) are an exception as they provide detailed recommendations
Agency (EMA), Organisation for Economic Cooperation and
for the evaluation of findings during the recovery phase. WHO
Development (OECD), and the Japanese Ministry of Health,
recommends that the pivotal supporting nonclinical study should
Labor, and Welfare, provide specific recommendations for the
include additional treatment groups, which will be evaluated at
inclusion of body weight, food/water consumption, hematologic
later time points after vaccination to allow evaluation of the rever-
and clinical biochemistry measurements, and morphologic pathol-
sibility of adverse effects observed during the dosing phase and to
ogy investigations in nonclinical toxicity studies. Guidelines for
screen for potential delayed adverse effects. WHO indicates that
recovery evaluation have been recently reviewed (Pandher, Leach,
hematology and serum chemistry evaluation should be consid-
and Burns-Naas 2012; Perry et al. 2013).
ered within 1 to 3 days following the first and last dose adminis-
Few guidelines for pharmaceutical development provide
tration and at the end of the recovery phase (2 weeks or more
detailed recommendations regarding clinical pathology evalua-
following the last dose; WHO 2005). At a minimum, biomarkers
tion in the recovery phase of nonclinical regulatory toxicology
to be evaluated include an evaluation of relative and absolute dif-
studies. Recommendations for the inclusion of a recovery
ferential white blood cell counts, albumin/globulin ratio, enzyme
phase state that at least one nonclinical study over the course
activities (specific enzymes not detailed in the guideline), and
of the development of a compound should incorporate a recov-
electrolyte concentrations. In some cases, assessment of coagula-
ery phase at the end of the study to determine the reversibility
tion times and fibrinogen concentration, urinalysis, and serum
or potential worsening of test article–related effects; guidelines
immunoglobulin classes are recommended.
specify that recovery groups should be monitored until reversi-
bility is demonstrated but full recovery is not necessary (FDA
2003; OECD 2008, 2009; ICH 2011; EPA 2001). However, an
alternative to including a recovery phase is using scientific jus-
Current Pharmaceutical Practices
tification as to whether a toxicologic effect with potential Common practices for the evaluation of clinical pathology dur-
adverse clinical impact is reversible. A nondosing period is ing the recovery phase of nonclinical toxicity studies were
warranted in a chronic study if there is severe toxicity at rele- compiled based on evaluation of the literature and the experi-
vant clinical exposures or if toxicity is only detectable at an ence of the working group and are presented in Table 1. The
advanced stage of the pathophysiology in humans. number of recovery groups may be similar to the number of
A few of the EMA guidelines indicate that in repeat-dose stud- dosing phase groups or limited to control and high-dose groups.
ies, clinical pathology should be evaluated during the dosing The study design relative to inclusion of recovery groups may
Tomlinson et al. 165

be influenced by many factors including the availability of data included in the recovery phase. In a few instances, selected
from previous studies, company preference, or an attempt to clinical pathology biomarkers were included if a test article–
minimize animal usage. The number of animals designated for related effect was observed in these test(s) during the dosing
the recovery phase is usually smaller than that used during the phase.
dosing phase (e.g., one-third to two-thirds of that in the dosing
phase), although this could vary depending on the anticipated
Appropriate Comparisons for Assessment
incidence of findings. Recommendations in this article are not
based on a specific study design because the number of recov-
of Recovery in Clinical Pathology
ery groups and the number of animals evaluated per recovery Nonclinical study design recommendations by the FDA to sup-
group is unique to each test article. However, it is strongly rec- port clinical trials do not specify which comparisons should be
ommended that the clinical pathology recovery assessment made for evaluation of reversibility. Reference values or poten-
strategy be determined in consultation with a clinical patholo- tial comparators in toxicology studies are often obtained from 1
gist and ideally with the one who will be interpreting the data. or more of 3 sources: a concurrent control group not exposed to
Standard clinical pathology biomarkers (Tomlinson et al. the test article, baseline (alternatively known as ‘‘pretest’’ or
2013) are generally evaluated from individual recovery ani- ‘‘prestudy’’) values compiled from all animals prior to dosing,
mals at the end of the dosing phase in most laboratory species or historical reference intervals collected from defined control
(e.g., rats, dogs, and monkeys) other than mice as this helps and/or predose animals. Importantly, utilizing historical control
with the assessment of reversibility in individual animals. In values or ‘‘reference ranges/intervals’’ can result in misinter-
some studies, changes in individual clinical pathology biomar- pretation of data, unless there is strict partitioning for animals
kers may help to determine which animals are to be retained for of the same strain; age; sex; provider; comparable routes of
evaluation during the recovery. Particularly in rodents, evalua- administration; and vehicles and exclusion of animals with
tion of different cohorts at the end of the dosing and recovery individual conditions, histories, and/or treatments (Hall 1997;
phases may be required due to blood and sample volume lim- National Committee for Clinical Laboratory Standards 1995;
itations, thus preventing evaluation of recovery in the same James 1993). The practice of strict partitioning prevents highly
individual animals. Repeated collections are rarely possible variable historical data which would be derived if control
in mice as blood collection is usually a terminal procedure in groups from nonclinical studies were given a variety of vehi-
this species. The length of the recovery phase is generally cles (Figure 1). The ASVCP has also developed guidelines for
determined by the half-life of the test article or the nature of the establishing reference intervals (http://www.asvcp.org/pubs/
test article–related findings. In most cases, all clinical pathol- pdf/RI%20Guidelines%20For%20ASVCP%20website.pdf).
ogy biomarkers that were assessed during the dosing phase are In rodent studies, a concurrent control group is considered to
evaluated during the recovery phase. However, in the recovery be necessary for the interpretation of data (Weingand et al. 1996;
phase, some investigators may opt to evaluate only the subset Leissing, Izzo, and Sargent 1985; James 1993). Using concur-
of biomarkers that were altered by target organ toxicity and/or rent controls is recommended because there is less interanimal
test article pharmacology during the dosing phase. This deci- variation than with the use of historical data, it eliminates con-
sion can be determined on a case-by-case basis as long as there cerns with collecting baseline data from the same animals, and
is scientific justification for it. For appropriate data compari- the animals are the same age at the end of the study. In accor-
sons to be made, blood sampling and analysis conditions (site dance with suggested animal numbers (ICH 2009), rodent stud-
of collection, fasting, anesthesia, and type of tube and analysis) ies commonly incorporate larger numbers of genetically similar
should be the same for the dosing and recovery phases. animals per group than nonrodent studies, resulting in lower bio-
A large global retrospective analysis detailing general prac- logical variance. Due to the small differences between individ-
tices for use of recovery groups (Sewell et al. 2014) provided ual animals, a control group of 5 animals/gender is sufficient
recommendations for minimizing recovery animals where to assess recovery for the standard biomarkers. Because of low
scientifically justified, including avoiding recovery phases in blood volumes, particularly in young rodents, baseline blood
first in human (FIH)-enabling studies. A broad review of the collection can negatively impact results and interfere with study
literature suggested that clinical pathology is most often incor- interpretation or overall animal health. Satellite groups for other
porated in the recovery groups to evaluate reversibility of purposes such as toxicokinetics are often included to avoid
organ dysfunction (such as renal, hematopoietic, immune, and impact of blood collection procedures on recovery animals. Due
endocrine systems), with fewer reports incorporating clinical to the shorter life span of rodents, the periadolescent age that
pathology to monitor tissue injury (Abraham, Gottschalk, and rodent studies typically start, and potential age-related effects
Ungemach 2005; Boorman et al. 1982; Henzen et al. 2000; on certain analytes (e.g., age-related decreases in serum phos-
Derelanko et al. 1985; Lefebvre et al. 1984; Rouse et al. phorus; Wolford et al. 1987), inclusion of a concurrent control
2011; Streck and Lockwood 1979; Terse et al. 2011). group ensures comparison to appropriate age-matched controls
A retrospective analysis of studies performed over the past 6 (Pandher, Leach, and Burns-Naas 2012).
years at a large U.S.-based contract research organization indi- Studies in nonrodent species (dogs and nonhuman primates)
cated that recovery groups were included in *25% of studies. generally use smaller numbers of more genetically diverse ani-
In most cases, the full clinical pathology profile was routinely mals with higher interanimal variation but lower intraanimal
166 Toxicologic Pathology 44(2)

Figure 1. Variable alanine aminotransferase values in control male Han-Wistar rats given a variety of vehicles.

variation; therefore, evaluation of baseline values is recom- phases (Leissing, Izzo, and Sargent 1985; Boone et al. 2005;
mended to better understand the biological variance and aid James 1993). However, the study purpose and/or complexity
in the interpretation of results during the dosing and recovery of the study design may dictate the need for concurrent controls
Tomlinson et al. 167

in the recovery phase. For short-term studies with low animal changes (e.g., alanine aminotransferase and AST activities)
numbers and limited blood collection or other evaluations dur- would not reflect a full recovery of liver structure or function.
ing the dosing phase, low intra-animal variation may make it When tissue injury is expected to fully recover and can be
preferable to monitor the return toward baseline of potential easily monitored in a clinical setting, whether associated with
test article–related changes in individual animals. However, microscopic changes or not, assessment of the recovery of bio-
as the study design becomes more complicated with additional markers associated with that tissue injury may not be required.
pharmacokinetic, pharmacodynamic, antidrug antibody, immu- This decision should be based on the information available for
nophenotyping or other evaluations, separation of procedure- the test article and/or pharmacologic mechanism. If the time
related changes from test article–related changes may not be course of damage and recovery is well understood for a test
possible without a concurrent control group. Nonhuman pri- article and its related toxicity, then monitoring associated bio-
mates are particularly susceptible to impact red cell mass markers after the recovery phase adds no additional informa-
because of the blood volume requirements for additional biomar- tion and is not required. For example, altered biomarkers
kers that can be more easily translated to the clinic than in other from labile tissues (those that are continuously dividing), such
nonrodent species. as hematopoietic cells in the bone marrow and surface epithe-
lium of the gastrointestinal tract, are expected to return to base-
line/control levels. In these cases, the timing of the return
Considerations of Tissue Recovery toward baseline/control levels may be more critical than
A crucial component of evaluating the reversibility of toxicity demonstration of full recovery. In agreement with Perry et al.
is the assessment of biomarkers indicative of structural organ (2013), when loss of architecture occurs in a labile tissue (such
damage and/or alteration of organ function. The standard as bone marrow) or there is injury to a stable tissue (such as
recommended evaluations during the dosing phase and/or liver) but normal architecture is retained, there is more uncer-
recovery phase include hematology, coagulation, clinical tainty about the ability of the tissue to recover. In those cases,
chemistry, and urinalysis panels to assess alterations in major morphologic recovery should be evaluated along with appro-
organ systems (Tomlinson et al. 2013). priate associated biomarkers, with consideration of the half-
For the majority of test article–related changes in specific life of the test article and biomarkers and length of the recovery
biomarkers, it is useful to monitor the return of those results phase. For example, monitoring liver enzyme activity can pro-
back toward baseline levels, particularly when the recovery vide information regarding ongoing liver injury, as well as
of clinical pathology findings is not well documented for a timing of recovery. This could be particularly helpful when
given test article. However, with some test article–related evaluating either small molecules or biologics with long termi-
changes, there is no realistic potential that a biomarker will nal half-lives (several days to weeks).
return to baseline, and thus measurement of the relevant bio- The concurrent recovery of clinical pathology changes and
markers would not be warranted. An example is a permanent clinical observations may help to distinguish changes that are
change in an organ such as the endocrine pancreas where the secondary to poor clinical condition (e.g., decreased body
utility of evaluation of functional markers (e.g., insulin and glu- weight or stress-related signs) from primary test article–related
cagon) during the recovery phase is questionable in the pres- effects. For example, the return toward baseline/control levels of
ence of advanced nonrecoverable damage to the islets. Perry biomarkers associated with dehydration (e.g., urea and creati-
et al. (2013) provided examples of anatomic pathology findings nine) and decreased food consumption (e.g., ALP, albumin, tri-
that are not expected to recover; one example is injury to per- glycerides, and effects on hematopoietic biomarkers such as
manent or terminally differentiated tissues, such as cardiac reticulocytes) in the recovery phase should be correlated to the
muscle. As cardiomyocytes are considered postmitotic, there clinical signs of the recovering animals. If clinical pathology
is no reasonable expectation that regeneration will occur. Thus, changes in the dosing phase are clearly identified as secondary
Perry et al. (2013) contend that recovery of the microscopic or have been previously characterized in the program as second-
lesions should not be evaluated in such cases. With cardiac ary, evaluation at the end of the recovery phase is not required.
muscle injury, common analytes of assessment may include
aspartate aminotransferase (AST) and creatine kinase (CK) Selection of Recovery Biomarkers Based on
activities and cardiac troponin I (cTnI). These analytes may
be used to monitor cessation of active degeneration, but they
Dosing Phase Data
are not indicative of restored functional capacity of the heart In general, options for clinical pathology assessment following
since irreversible cardiac injury resolves with fibrosis. Simi- a recovery phase can include evaluation of the full panel,
larly, evaluating analytes should be carefully considered when portions of the panel such as hematology but not clinical chem-
stable tissues (quiescent with the ability to regenerate with istry, or only affected biomarkers. A prescriptive recommenda-
appropriate stimuli, such as renal tubular epithelial cells or tion cannot be made because there are aspects, such as
hepatocytes) have severe enough structural injury that tissue biomarker storage stability, that affect the ability to delay
architecture is not expected to be restored. For example, with assessment and thus, decision making on the assessment. An
severe liver injury resulting in hepatic fibrosis that bridges and abbreviated panel can be considered if there are only changes
dissects liver lobules, recovery of hepatic injury marker in a portion of the clinical pathology panel, such as fibrinogen.
168 Toxicologic Pathology 44(2)

If a decrease in fibrinogen is the only change at the end of the time points to interpret the recovery phase. For example, when
dosing phase, the decision could be to evaluate the coagulation only two time points can be evaluated per animal, it is most ben-
panel (e.g., prothrombin time [PT], activated partial thrombo- eficial to collect data from end of dosing and end of recovery.
plastin time [APTT], and fibrinogen) or fibrinogen alone dur- Animals from the control and selected dose group(s) are usu-
ing the recovery phase. In the latter case, the individual ally assigned to a cohort recovery group at the onset of the study.
laboratory or sponsor should consider logistical considerations However, under certain circumstances, the assignment of ani-
such as computer software templates to determine if there is an mals may be altered to include animals that have been identified
appropriate cost–benefit scenario to run one test instead of a as having test article–related clinical pathology findings to
typical panel of biomarkers. However, each laboratory has to ensure that representative changes are present in the recovery
weigh these considerations against the risk of creating unneces- group and subsequently analyzed during the recovery phase. For
sary data versus the risk of producing a limited data set that example, if 5 of the 10 animals in the high-dose group exhibit
cannot be interpreted properly. For example, if there are marked hematologic changes, but none are assigned to the
increases in fibrinogen alone at the end of the dosing phase, recovery group, then reassignment may be considered so that
evaluation of related inflammatory parameters in hematology recovery of the finding can be assessed. However, if animals
and serum chemistry panels should also be considered. Consul- assigned to the recovery phase have critical levels of a biomar-
tation with a clinical pathologist is recommended during this ker (e.g., severe life-threatening thrombocytopenia or neutrope-
decision-making process. nia), it might be advisable to terminate them at the end of the
There is limited utility in evaluating biomarkers after a dosing phase. Similarly, if all animals with an increase in an
recovery phase when there were no test article–related altera- injury biomarker are in the recovery group, it may be impossi-
tions at the end of the dosing phase. However, if clinical pathol- ble to determine the histologic nature of the lesion present at
ogy results from the dosing phase are not available in time for the end of dosing without reassigning one or more animals to
interpretation and if waiting for evaluation compromises sam- the end of dosing phase. Such reassignments are only recom-
ple quality, then it may be preferable to routinely collect clin- mended if they will make the data set more interpretable and
ical pathology samples and/or evaluate them at the end of a are more likely to be utilized in studies with large animals
recovery phase. Also, if there are specific concerns of potential rather than in rodents due to the smaller group size. A reason-
alterations due to test article exposure during the recovery able justification of any reassignment should be well documen-
phase because of a long half-life, clinical pathology evaluation ted in a study amendment. Reassignment is not recommended
may be warranted. for animals with minor changes and may not be necessary for
Assessment of clinical pathology during the recovery phase animals with changes that are expected to fully recover. Ensur-
should also be considered when test article–related clinical ing and documenting that appropriate animals are part of the
pathology changes indicate a functional effect that is not expected recovery group to allow adequate assessment of recovery is the
to have morphologic correlates or when clinical pathology responsibility of the study director.
changes are more sensitive predictors of toxicity than morpholo-
gic ones. For example, a decreased reticulocyte count may indi-
Considerations for Exploratory or Newly
cate injury to the bone marrow that is not yet apparent
histologically or has not yet impacted red blood cell (RBC) mass
Approved Biomarkers
because of the long life span of RBCs. If damage to the marrow is Standard clinical pathology biomarkers have been used for
severe enough in a short-term study, the reticulocyte count may decades and their performance features (sensitivity, specificity,
not improve or may decrease further during the recovery phase, kinetics), assay formats, and caveats are well understood. This
and RBC mass may be decreased at the end of the recovery phase is not necessarily the case with novel safety biomarkers. Novel
even though it was unchanged at the end of the dosing phase. exploratory safety or nontraditional biomarkers should be used
Interpretation of data can be challenging if appropriate time within a clearly defined context. The goals, anticipated out-
points and animals are not evaluated. In rodent studies, suffi- comes, and reference standard or comparators such as the
cient blood volume may not be available to sample every animal associated histopathology or functional change, should be iden-
at every desired time point. Special attention must be paid to tified prior to use in a study. The interpretation of the novel bio-
how animals are bled during the dosing phase and during the markers should be based on these predefined endpoints. In
recovery phase to minimize preanalytic variation and optimize some instances, very little is known about the biological role,
interpretation. For example, multiple collections during a study sensitivity, kinetics, and species differences of novel biomar-
(interim, end of dosing, and/or end of recovery) would enable kers. The assays themselves are often at the exploratory stage
assessment at multiple time points in a single rat and correlation of development and characterized as ‘‘fit for purpose’’ (Lee
with histopathology data for animals terminated at the end of et al. 2006; Wagner 2008). Gathering data during the recovery
dosing or recovery phases. Bridging of doses between studies phase will enable a progressive understanding of the candidate
can facilitate comparisons of clinical pathology and histopathol- biomarker, as interpretation of changes in standard clinical
ogy data across studies; by utilizing data from shorter term stud- pathology biomarkers and other study data can be used to put
ies, it is possible to minimize blood collection during the dosing information on the novel marker biology in health and disease
phase of longer term studies, allowing collection of appropriate into context.
Tomlinson et al. 169

In 2011, the FDA accepted the qualification of cTnI as an and the kinetics of repopulation is essential for characterization
indicator of the presence and extent of cardiac structural dam- of the pharmacologic activity of the test article, identification
age in dogs and rats based on data from studies of drugs with of potential adverse events, and determination of the most
known cardiotoxicity or drugs that have shown evidence of car- appropriate clinical administration regimen. However, the
diac damage when previously tested (http://www.fda.gov/ addition of recovery phases in order to assess immunogenicity
downloads/Drugs/DevelopmentApprovalProcess/DrugDeve is not required as per ICH harmonized tripartite guideline, pre-
lopmentToolsQualificationProgram/UCM294644.pdf). The clinical safety evaluation of biotechnology-derived pharma-
intent of using cTnI as a biomarker of cardiac toxicity is to bet- ceuticals S6 (R1).
ter define the lowest cardiotoxic dose in nonclinical studies of
test articles known to induce structural cardiac damage and to
help choose starting doses in clinical studies of those test arti-
Evaluation of Delayed Toxicity
cles; there is no intent to use cTnI as a general screening marker With the exception of nonclinical studies supporting microdos-
to detect unexpected cardiac toxicity. CTnI often peaks 2 to 6 ing in the clinic, there is no regulatory requirement for the eva-
hr after cardiac injury in nonclinical studies, depending on the luation of delayed toxicity in nonclinical studies. However, in
duration and extent of cardiac injury (Clements et al. 2010; nonclinical drug development, the addition of recovery groups
O’Brien 2008). Therefore, cTnI should be evaluated taking the should be considered when there is a specific concern for test
short circulating half-life and the timing of the peak concentra- article–related findings that could develop after the dosing
tion of the drug into account, usually within 24 hr after drug phase. For example, for test articles with known class liabilities
administration. An increase in cTnI values, however, should caused by effects on early hematopoietic or spermatogenic pro-
be carefully evaluated. As in people, obtaining serial samples genitors, a recovery phase may be used to monitor changes that
might help to evaluate the relevance of these changes in noncli- may not manifest within the course of an FIH-enabling study.
nical studies. Given the kinetics and short circulating half-life Data from recovery groups may allow decisions on the design
of cTnI, measurement during a recovery phase may not be war- of clinical dosing as well as subsequent nonclinical studies. In
ranted, but the half-life of the test article must also be consid- the case of biopharmaceuticals, recovery groups are commonly
ered. A negative cTnI result does not indicate an absence of used to monitor for the potential of a test article to produce
structural cardiac damage. delayed toxicity during the off-dose phase due to either pro-
Beginning in 2010, FDA accepted several qualification sub- longed exposure or extended pharmacodynamic effects of the
missions for 9 novel urinary biomarkers including kidney test article, although this is not required per ICH S6 (R1).
injury molecule-1 (Vaidya et al. 2010), clusterin, cystatin C, Although most of the concerns for delayed toxicity relate to
and renal papillary antigen-1 (Dieterle et al. 2010) that demon- morphologic changes, clinical pathology data can aid in the
strated improvement in the detection of acute drug-induced evaluation of target organ toxicities through identification of
kidney injury in rats (Harpur et al. 2011). Subsequently, rat effects on organ function that might not be evident morpholo-
urinary osteopontin and neutrophil gelatinase-associated lipo- gically. For example, a compound affecting lymphoid stem
calin (NGAL) have received regulatory endorsement in the cells in bone marrow could have delayed effects on peripheral
form of a Letter of Support signed by Janet Woodcock on lymphoid subpopulations due to the progressive depletion of
August 20, 2014. Most of the data supporting the use of these circulating lymphocytes with a long half-life. Similarly, as
biomarkers were generated in 4- to 15-day studies, and recov- T-cell maturation occurs in the thymus, a test article causing
ery was not evaluated. Even though these renal biomarkers are concurrent hematopoietic and thymic toxicity might exhibit a
accepted, there is still limited information about the kinetics, delayed or more severe effect on peripheral T-cell counts than
sensitivity, specificity, recovery, or assay interferences for a compound causing thymic injury alone due to the combined
them. The widespread use of these novel urinary biomarkers effects of senescence of mature T cells and decreased bone
in rodent nonclinical studies, including their use to evaluate marrow lymphopoiesis. Finally, in the case of a nephrotoxicant
recovery of the target organ toxicity, will enable progressive with proximal tubular injury, concurrent urinalysis biomarkers,
qualification and further understanding of the biology unique including evaluation of protein or albumin excretion with his-
to these markers. topathology, could support demonstration of reversibility, par-
Immunotoxicity findings (excluding immune-related find- ticularly if there is histological evidence of tubular basophilia
ings secondary to stress) may be observed as intended pharma- in recovery animals. As tubular basophilia can reflect either a
cological effects or as unexpected (off-target) effects of test regenerative response or a toxic change (Seely and Frazier
articles. It is recommended that immunotoxicity findings 2015), recovery of urinalysis biomarkers can aid in the discrim-
undergo further evaluation either in follow-up studies, as ination between regenerative and adverse tubular changes.
described in the FDA guidance on immunotoxicology evalua-
tion of investigational new drug (FDA Center for Devices and
Radiological Health: U.S. Department of Health and Human Biomarker Recovery in Vaccine Studies
Services 1999), or during the recovery phase of toxicity studies. As described earlier, regulatory guidelines for the recovery
For example, in the case of a lymphocyte-depleting antibody, of clinical pathology biomarkers in the evaluation of vac-
thorough immunophenotypic evaluation of lymphocyte subsets cines are more detailed than for other areas of drug
170 Toxicologic Pathology 44(2)

development. Vaccine programs are unique since a dose– A primary objective for the use of efficacy markers in non-
response evaluation is not required as part of the basic toxi- clinical toxicity studies is to identify the relationship between
city assessment, and maximum tolerable doses do not need intended pharmacologic effects and off-target or adverse find-
to be determined. As vaccines are given episodically rather ings. For example, comparison of the peak exposure for desired
than daily, the dosing intervals may be based on the kinetics pharmacology with the exposure level where adverse findings
of the primary and secondary antibody responses observed occur can be used to define the relationship between on- and
in the animal studies. off-target effects. Subsequently, efficacy markers or clinical
Vaccine administration causes acute phase protein increases pathology biomarkers related to either desired pharmacology
that will usually be at their peak 24 to 48 hr postinjection. or target organ toxicity may be evaluated during a recovery
Therefore, only a mild, if any, change in acute phase proteins phase in conjunction with histopathology to define the relation-
should be observed during or at the end of the recovery phase. ship between various findings or to inform the risk assessment
While residual chronic inflammation is often described after of both intended and unintended findings. Utilizing the desired
the 2-week off-dose phase and is reflective of an expected tis- pharmacology, half-life, mode of action, and associated on- or
sue response, some recovery of the inflammation is expected off-target effects will optimize the design for evaluation of effi-
during that time. Unless the immunological and inflammatory cacy markers during the recovery phase.
response to the vaccine is severe and prolonged, it is not con- The biological time course of a pharmacologic effect often
sidered to be adverse. determines duration of recovery. For example, increased glu-
Extensive recommendations for vaccine evaluation have cose production due to treatment with a glucagon analog
recently been published and are being readily adopted by com- quickly returns to normal levels after test article exposure
panies developing vaccines (Green and Al-Humadi 2013). diminishes, whereas excessive production of RBCs caused by
These recommendations provide a larger and updated list of erythropoiesis-stimulating agents or an excessive number of
biomarkers to consider during vaccine studies. The clinical leukocytes produced as a result of bone marrow stimulation
pathology biomarkers recommended for evaluation at all time by granulocyte-colony stimulating factor reverse slowly because
points include a complete blood cell count, a serum chemistry the target cell populations may decrease slowly over time. In
panel, a coagulation panel including fibrinogen concentration, cases where cell populations are expected to change slowly or
and the measurement of acute phase proteins. The recom- where long-lasting effects are expected, demonstration of com-
mended acute phase proteins are C-reactive protein (CRP, plete recovery to baseline may not be necessary. However, fre-
which is the primary acute phase protein in rabbits, monkeys, quent monitoring of efficacy markers can help to define the
and humans) and a 2-macroglobulin and a 1-acidic glycopro- length of recovery required in some cases, such as for biologic
tein in rodents (Green and Al-Humadi 2013) or more specifi- test articles with longer half-lives. It may not be necessary or
cally in rats (Cray, Zaias, and Altman 2009). This STP/ beneficial to assess a full panel of clinical pathology biomarkers
ASVCP working group concurs with the recommendations but if only intended pharmacology is being monitored.
also recommends CRP in dogs and serum amyloid A and Besides persistence of a pharmacologic effect, the need for
haptoglobin in mice (Cray, Zaias, and Altman 2009). When recovery assessment may depend on the organ involved and the
specific acute phase proteins are measured, serum electrophor- mechanism of the effect. With some immunomodulators, pro-
esis analysis is not required. The measurement of CK activity, longed decreases in B- or T-lymphocyte populations can persist
an analyte with a short half-life, is useful for the evaluation of without clinical or other hematologic manifestations. In rare
the recovery from muscle injury, particularly in dogs (Tomlin- cases, adverse events can occur during the recovery phase due
son et al. 2013; Vassallo et al. 2009). Since vaccine administra- to immunosuppression or unintended immunostimulation. If
tion may cause local muscular degeneration, the evaluation of such events are anticipated, close monitoring of efficacy mar-
CK activity or other biomarkers of skeletal muscle injury such kers, traditional clinical pathology biomarkers, and clinical
as skeletal troponin I (Vassallo et al. 2009), dependent on the observations may be warranted throughout the dosing and
species being evaluated, is also recommended during the off- recovery phases. Also, with certain classes of test articles that
dose phase of nonclinical vaccine studies. are known to have long-lasting effects (e.g., some bisphospho-
nates or endocrine modulators), slowly progressive or delayed
pharmacologic effects can occur and monitoring may be
Recovery of Efficacy Markers desired during the recovery phase.
Efficacy markers enable monitoring of desired pharmacologic Finally, it is recommended that for the evaluation of the
effects in both nonclinical studies and clinical trials and may be recovery of efficacy markers, the pharmacologic effect should
useful to evaluate in a recovery phase to understand persistence be present in animals designated for recovery. This is of partic-
of these pharmacologic effects, particularly for biologic or ular concern in studies with biologic test articles that induce
large-molecule test articles that have long half-lives. For small immunogenicity in the test species. Depending on the level
molecules with short half-lives, efficacy is likely to decrease of development and the effects on test article exposure of
quickly following cessation of dosing, and evaluation of recov- antitest article antibodies, not all animals will have similar
ery for efficacy markers induced by these test articles is gener- pharmacologic effects that can be monitored during the recov-
ally unnecessary. ery phase.
Tomlinson et al. 171

Conclusions (10) For the evaluation of recovery of efficacy markers, the


pharmacologic effect should be present in animals
Regulatory guidelines provide the flexibility required for
intended for the recovery phase.
adapting the recovery phase to the needs of the program, but
offer limited direction for the evaluation of clinical pathology These recommendations are intended to complement rather
in the recovery phase. The mandate of this STP/ASVCP work- than replace recommendations currently available in the litera-
ing group was to consider the assessment of and to provide con- ture and to provide additional clarification for the clinical
siderations and recommendations on the appropriate inclusion pathology evaluation in the recovery phase of nonclinical toxi-
of clinical pathology biomarkers in the recovery phase of non- city studies.
clinical toxicology studies.
The following recommendations represent the ‘‘best prac- Acknowledgments
tice’’ for appropriate evaluation of clinical pathology biomar- The working group acknowledges the input from members of STP and
kers in the recovery phase of nonclinical toxicity studies. ASVCP.

(1) The clinical pathology recovery assessment strategy should Author Contributions
be determined after consultation with a clinical pathologist, Authors contributed to conception or design (LT, DE, and DB); data
ideally the one who will be interpreting the data. acquisition, analysis, or interpretation (LT, DE, LR, NT, VB, DB,
(2) Data from concurrent controls are recommended for FP, and AV); drafting the manuscript (LT, DE, LR, NT, VB, DB,
assessment of recovery data in rodent studies and com- FP, and AV); and critically revising the manuscript (LT, DE, and
plicated nonrodent studies but in simple short-term AV). All authors gave final approval and agreed to be accountable for
(<28 days) nonrodent studies, comparisons of recovery all aspects of work in ensuring that questions relating to the accuracy
data in test article–treated animals to baseline may be or integrity of any part of the work are appropriately investigated and
resolved.
adequate.
(3) Recovery of biomarkers of toxicity should be assessed
Authors’ Note
when the timing or ability of the organ to recover is of
concern (e.g., loss of architecture in a labile tissue or This article is a product of a Society of Toxicologic Pathology (STP)
Working Group involving the Scientific and Regulatory Policy Com-
injury to a stable tissue while normal architecture is
mittee (SRPC) and Clinical Pathology Interest Group (CPIG) of the
retained). STP in collaboration with the Regulatory Affairs Committee (RAC)
(4) An evaluation of biomarkers is not required after the of the American Society for Veterinary Clinical Pathology (ASVCP)
recovery phase if an associated group of biomarkers is and has been reviewed and approved by the SRPC and Executive
unchanged after the dosing phase, the recovery of test Committee of the STP.
article–related clinical pathology changes are well under-
stood, there is no expectation of recovery, the changes are Declaration of Conflicting Interests
secondary effects, and/or the changes have no relevance The author(s) declared no potential conflicts of interest with respect to
to recovery. the research, authorship, and/or publication of this article.
(5) Assessment of clinical pathology during the recovery
phase should be considered when clinical pathology Funding
changes indicate a functional effect with no morphologic The author(s) received no financial support for the research, author-
correlates or when clinical pathology changes are more ship, and/or publication of this article.
sensitive predictors of toxicity than morphologic changes.
(6) Reassignments of dosing and recovery phase animals are References
only recommended if they will make the data set more Abraham, G., Gottschalk, J., and Ungemach, F. R. (2005). Evidence for ototo-
interpretable. pical glucocorticoid-induced decrease in hypothalamic-pituitary-adrenal
(7) Evaluation of clinical pathology biomarkers of delayed axis response and liver function. Endocrinology 146, 3163–71.
Boone, L., Meyer, D., Cusick, P., Ennulat, D., Bolliger, A., Everds, N., Mea-
toxicity should be considered on a case-by-case basis in
dor, V., Elliott, G., Honor, D., Bounous, D., and Jordan, H. (2005). Selec-
development programs with specific concerns for delayed tion and interpretation of clinical pathology indicators of hepatic injury in
toxicity that can be monitored using clinical pathology. preclinical studies. Vet Clin Pathol/Am Soc Vet Clin Pathol 34, 182–88.
(8) In addition to the guidelines for clinical pathology assess- Boorman, G. A., Luster, M. I., Dean, J. H., and Campbell, M. L. (1982).
ment of recovery from vaccine administration, evaluation Assessment of myelotoxicity caused by environmental chemicals. Environ
Health Perspect 43, 129–135.
of acute phase proteins (CRP in dogs and monkeys, a 2-
Clements, P., Brady, S., York, M., Berridge, B., Mikaelian, I., Nicklaus, R.,
macroglobulin or a 1-acidic glycoprotein in rats, and Gandhi, M., Roman, I., Stamp, C., Davies, D., McGill, P., Williams, T.,
serum amyloid A or haptoglobin in mice) and biomarkers Pettit, S., Walker, D., Group, I. H. C. T. W., and Turton, J. (2010). Time
of skeletal muscle injury is recommended. course characterization of serum cardiac troponins, heart fatty acid-binding
(9) Evaluation of novel exploratory or nontraditional clinical protein, and morphologic findings with isoproterenol-induced myocardial
injury in the rat. Toxicol Pathol 38, 703–14.
pathology biomarkers during the recovery phase should
Cray, C., Zaias, J., and Altman, N. H. (2009). Acute phase response in animals:
be used within a clearly defined context. A review. Comp Med 59, 517–26.
172 Toxicologic Pathology 44(2)

Derelanko, M. J., Gad, S. C., Powers, W. J., Mulder, S., Gavigan, F., and OECD (Organisation for Economic Co-operation and Development). (2008).
Babich, P. C. (1985). Toxicity of cyclohexanone oxime. I. Hematotoxicity Guideline for the testing of chemicals in chronic toxicity studies, 40CFR
following subacute exposure in rats. Fundam Appl Toxicol 5, 117–27. Part 261. U.S. Government Printing Office, Washington, DC.
Dieterle, F., Perentes, E., Cordier, A., Roth, D. R., Verdes, P., Grenet, O., Pan- OECD (Organisation for Economic Co-operation and Development). (2009).
tano, S., Moulin, P., Wahl, D., Mahl, A., End, P., Staedtler, F., Legay, F., OECD Guidelines for the Testing of Chemicals, 40CFR Part 261. U.S.
Carl, K., Laurie, D., Chibout, S. D., Vonderscher, J., and Maurer, G. (2010). Government Printing Office, Washington, DC.
Urinary clusterin, cystatin C, beta2-microglobulin and total protein as mar- Pandher, K., Leach, M. W., and Burns-Naas, L. A. (2012). Appropriate use of
kers to detect drug-induced kidney injury. Nat Biotechnol 28, 463–69. recovery groups in nonclinical toxicity studies: Value in a science-driven
EPA (Environmental Protection Agency). (2001). OCSPP Harmonized Test case-by-case approach. Vet Pathol 49, 357–61.
Guidelines Series 870- Health Effects Test Guidelines, 40CFR Part 261. Perry, R., Farris, G., Bienvenu, J. G., Dean, C. Jr., Foley, G., Mahrt, C., and
US Government Printing Office, Washington, DC. Short, B., and Society of Toxicologic, P. (2013). Society of Toxicologic
EMA (European Medicines Agency) Committee of Human Medicinal Prod- Pathology position paper on best practices on recovery studies: The role
ucts. (2010). Questions and Answers on the Withdrawal of the ‘‘Note for of the anatomic pathologist. Toxicol Pathol 41, 1159–69.
Guidance on Single Dose Toxicity,’’ 40CFR Part 261. US Government Rouse, R. L., Zhang, J., Stewart, S. R., Rosenzweig, B. A., Espandiari, P., and
Printing Office, Washington, DC. Sadrieh, N. K. (2011). Comparative profile of commercially available urin-
EMA (European Medicines Agency) Committee of Human Medicinal Prod- ary biomarkers in preclinical drug-induced kidney injury and recovery in
ucts/Safety Working Party. (2010). Guideline on Repeated Dose Study, rats. Kidney Int 79, 1186–97.
40CFR Part 261. US Government Printing Office, Washington, DC. Seely, J. C., and Frazier, K. S. (2015). Regulatory forum opinion piece*: Dis-
FDA (Food and Drug Administration). (2003). Guidance for Industry and pelling confusing pathology terminology: Recognition and interpretation of
Other Stakeholders. Toxicological Principles for the Safety Assessment selected rodent renal tubule lesions. Toxicol Pathol 43, 457–63.
of Food Ingredients. Redbook 2000: IV.B.1 General Guidelines for Design- Sewell, F., Chapman, K., Baldrick, P., Brewster, D., Broadmeadow, A., Brown,
ing and Conducting Toxicity Studies, 40CFR Part 261. US Government P., Burns-Naas, L. A., Clarke, J., Constan, A., Couch, J., Czupalla, O., Danks,
Printing Office, Washington, DC. A., DeGeorge, J., de Haan, L., Hettinger, K., Hill, M., Festag, M., Jacobs, A.,
FDA (Food and Drug Administration) Center for Devices and Radiological Jacobson-Kram, D., Kopytek, S., Lorenz, H., Moesgaard, S. G., Moore, E.,
Health: U.S. Department of Health and Human Services. (1999). Guidance Pasanen, M., Perry, R., Ragan, I., Robinson, S., Schmitt, P. M., Short, B.,
for Industry and FDA Reviewers: Immunotoxicity Testing Guidance, Lima, B. S., Smith, D., Sparrow, S., van Bekkum, Y., and Jones, D. (2014).
40CFR Part 261. US Government Printing Office, Washington, DC. Recommendations from a global cross-company data sharing initiative on the
Green, M. D., and Al-Humadi, N. H. (2013). Preclinical toxicology of vac- incorporation of recovery phase animals in safety assessment studies to sup-
cines. In A comprehensive guide to toxicology on preclinical development, port first-in-human clinical trials. Regul Toxicol Pharmacol 70, 413–29.
edited by A.S. Faqi, 619–45. Waltham, MA: Elsevier Inc. Streck, W. F., and Lockwood, D. H. (1979). Pituitary adrenal recovery follow-
Hall, R. L. (1997). Lies, damn lies, and reference intervals (or hysterical con- ing short-term suppression with corticosteroids. Am J Med 66, 910–14.
trol values for clinical pathology data). Toxicol Pathol 25, 647–49; discus- Terse, P. S., Johnson, J. D., Hawk, M. A., Ritchie, G. D., Ryan, M. J., Vascon-
sion 650–41. celos, D. Y., Contos, D. A., Perrine, S. P., Peggins, J. O., and Tomaszewski,
Harpur, E., Ennulat, D., Hoffman, D., Betton, G., Gautier, J. C., Riefke, B., J. E. (2011). Short-term toxicity study of ST-20 (NSC-741804) by oral
Bounous, D., Schuster, K., Beushausen, S., Guffroy, M., Shaw, M., Lock, gavage in Sprague-Dawley rats. Toxicol Pathol 39, 614–22.
E., Pettit, S., and Nephrotoxicity, H. C. O. B. O. (2011). Biological quali- Tomlinson, L., Boone, L. I., Ramaiah, L., Penraat, K. A., von Beust, B. R.,
fication of biomarkers of chemical-induced renal toxicity in two strains of Ameri, M., Poitout-Belissent, F. M., Weingand, K., Workman, H. C., Aul-
male rat. Toxicol Sci 122, 235–52. bach, A. D., Meyer, D. J., Brown, D. E., MacNeill, A. L., Bolliger, A. P.,
Henzen, C., Suter, A., Lerch, E., Urbinelli, R., Schorno, X. H., and Briner, V. and Bounous, D. I. (2013). Best practices for veterinary toxicologic clinical
A. (2000). Suppression and recovery of adrenal response after short-term, pathology, with emphasis on the pharmaceutical and biotechnology indus-
high-dose glucocorticoid treatment. Lancet 355, 542–45. tries. Vet Clin Pathol 42, 252–69.
ICH (International Conference on Harmonization). (2009). ICH S9. Guidance Vaidya, V. S., Ozer, J. S., Dieterle, F., Collings, F. B., Ramirez, V., Troth, S.,
on Nonclinical Safety Studies for the Conduct of Human Clinical Trials and Muniappa, N., Thudium, D., Gerhold, D., Holder, D. J., Bobadilla, N. A.,
Marketing Authorization for Pharmaceuticals, 40CFR Part 261. US Gov- Marrer, E., Perentes, E., Cordier, A., Vonderscher, J., Maurer, G., Goering,
ernment Printing Office, Washington, DC. P. L., Sistare, F. D., and Bonventre, J. V. (2010). Kidney injury molecule-1
ICH (International Conference on Harmonization). (2011). ICH S6 (R1). Pre- outperforms traditional biomarkers of kidney injury in preclinical biomar-
clinical Safety Evaluation of Biotechnology Derived Pharmaceuticals, ker qualification studies. Nat Biotechnol 28, 478–85.
40CFR Part 261. US Government Printing Office, Washington, DC. Vassallo, J. D., Janovitz, E. B., Wescott, D. M., Chadwick, C., Lowe-Krentz, L.
James, R. (1993). The relevance of clinical pathology to toxicology studies. J., and Lehman-McKeeman, L. D. (2009). Biomarkers of drug-induced ske-
Comp Haematol Int 3, 190–95. letal muscle injury in the rat: troponin I and myoglobin. Toxicol Sci 111,
Lee, J. W., Devanarayan, V., Barrett, Y. C., Weiner, R., Allinson, J., Fountain, 402–12.
S., Keller, S., Weinryb, I., Green, M., Duan, L., Rogers, J. A., Millham, R., Wagner, J. A. (2008). Strategic approach to fit-for-purpose biomarkers in drug
O’Brien, P. J., Sailstad, J., Khan, M., Ray, C., and Wagner, J. A. (2006). development. Annu Rev Pharmacol Toxicol 48, 631–51.
Fit-for-purpose method development and validation for successful biomar- Weingand, K., Brown, G., Hall, R., Davies, D., Gossett, K., Neptun, D., Waner,
ker measurement. Pharm Res 23, 312–28. T., Matsuzawa, T., Salemink, P., Froelke, W., Provost, J. P., Dal Negro, G.,
Lefebvre, F. A., Belanger, A., Pelletier, G., and Labrie, F. (1984). Recovery of Batchelor, J., Nomura, M., Groetsch, H., Boink, A., Kimball, J., Woodman,
gonadal functions in the adult male rat following cessation of five-month D., York, M., Fabianson-Johnson, E., Lupart, M., and Melloni, E. (1996).
daily treatment with an LHRH agonist. J Androl 5, 181–92. Harmonization of animal clinical pathology testing in toxicity and safety
Leissing, N., Izzo, R., and Sargent, H. (1985). Variance estimates and indivi- studies. The Joint Scientific Committee for International Harmonization
duality ratios of 25 serum constituents in beagles. Clin Chem 31, 83–86. of Clinical Pathology Testing. Fundam Appl Toxicol 29, 198–201.
National Committee for Clinical Laboratory Standards. (1995). How to Define Wolford, S. T., Schroer, R. A., Gallo, P. P., Gohs, F. X., Brodeck, M., Falk, H.
and Determine Reference Intervals in the Clinical Laboratory: Approved B., and Ruhren, R. (1987). Age-related changes in serum chemistry and
Guideline, 40CFR Part 261. US Government Printing Office, Washington, hematology values in normal Sprague-Dawley rats. Fundam Appl Toxicol
DC. 8, 80–88.
O’Brien, P. J. (2008). Cardiac troponin is the most effective translational WHO (World Health Organization). (2005). WHO Guidelines on Nonclinical
safety biomarker for myocardial injury in cardiotoxicity. Toxicology Evaluation of Vaccines, 40CFR Part 261. US Government Printing Office,
245, 206–18. Washington, DC.

Vous aimerez peut-être aussi