HHS Public Access Author manuscript Author Manuscript

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01. Published in final edited form as: Toxicol Pathol. 2016 August ; 44(6): 789–809. doi:10.1177/0192623316650052.

Scientific and Regulatory Policy Committee (SRPC) Points to Consider Review*1: Inclusion of Reproductive and Pathology Endpoints for Assessment of Reproductive and Developmental Toxicity in Pharmaceutical Drug Development

Author Manuscript

Wendy G. Halpern1, Mehrdad Ameri2, Christopher J. Bowman3, Michael R. Elwell4, Michael L. Mirsky3, Julian Oliver5, Karen S. Regan6, Amera K. Remick7, Vicki L. Sutherland8, Kary E. Thompson9, Claudine Tremblay10, Midori Yoshida11, and Lindsay Tomlinson12

Author Manuscript

1

Genetech, South San Francisco, California USA

2

GlaxoSmithKline, King of Prussia, Pennsylvania USA

3

Pfizer, Groton, Connecticut USA

4

Covance, Chantilly, Virginia USA

5

Incyte, Wilmington, Delaware USA

6

Regan Path/Tox Services, Ashland, Ohio USA

7

WIL Research, Hillsborough, North Carolina USA

8

NIEHS/NTP, Research Triangle Park, North Carolina USA

9

Bristol-Myers Squibb, New Brunswick, New Jersey USA

10

Charles River Laboratories, Senneville, Quebec CANADA

11

Food Safety Commission of Japan, Minato-ku, Tokyo JAPAN

12

Pfizer, Cambridge, Massachusetts USA

Abstract

Author Manuscript

Standard components of nonclinical toxicity testing for novel pharmaceuticals include clinical and anatomic pathology, as well as separate evaluation of effects on reproduction and development to inform clinical development and labeling. General study designs in regulatory guidances do not specifically mandate use of pathology or reproductive endpoints across all study types; thus, inclusion and use of these endpoints are variable. The Scientific and Regulatory Policy Committee (SRPC) of the Society of Toxicologic Pathology (STP) formed a Working Group to assess the current guidelines and practices on the use of reproductive, anatomic pathology and clinical

*This review is a product of a Society of Toxicologic Pathology (STP) Working Group commissioned by the Scientific and Regulatory Policy Committee (SRPC) of the STP. It has been reviewed and approved by the SRPC and Executive Committee (EC) of the STP. The article does not represent a formal best practice recommendation of the Society but provides expert guidance on key principles to consider in conducting regulated toxicity studies. The views expressed in this article are those of the authors and do not necessarily represent the policies, positions or opinions of their respective agencies and organizations. Readers of Toxicologic Pathology are encouraged to send their thoughts on these articles or ideas for new topics to the editor. Address Correspondence: Wendy Halpern, Genentech, 1 DNA Way, South San Francisco, CA 94080, [email protected].

Halpern et al.

Page 2

Author Manuscript

pathology endpoints in General, Reproductive, and Developmental toxicology studies. The Working Group constructed a survey sent to pathologists and reproductive toxicologists, and responses from participating organizations were collected through the STP for evaluation by the Working Group. The regulatory context, relevant survey results, and collective experience of the Working Group are discussed and provide the basis of each assessment by study type. Overall, the current practice of including specific endpoints on a case-by-case basis is considered appropriate. Points to consider are summarized for inclusion of reproductive endpoints in general toxicity studies, and for the informed use of pathology endpoints in reproductive and developmental toxicity studies.

Keywords

Author Manuscript

Preclinical research & development; Reproductive system; Safety assessment; Toxicologic pathology; Teratology; Clinical pathology; Developmental pathology

1. Background and Introduction

Author Manuscript Author Manuscript

The International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use (ICH) has developed several nonclinical safety guidelines, including those for development of drugs [ICH M3(R2), 2010], drugs for use in advanced cancer (ICH S9, 2010), and biopharmaceuticals [ICH S6(R1), 2011], all of which acknowledge assessment of the reproductive tract and/or reproductive function as an expected component of the nonclinical safety assessment. The ICH S5(R2), which is currently under revision, provides further specific guidance for assessment of reproductive function, as well as detection of maternal and developmental toxicity with exposures during pregnancy (ICH, 2005). However, the role of pathology endpoints in these assessments is less clear; specific guidance for inclusion of histologic endpoints in reproductive toxicity studies is limited, and there are no definitive requirements for the inclusion of clinical pathology endpoints in reproductive or juvenile toxicity studies, or for the addition of reproductive toxicity endpoints to general toxicity studies in pharmaceutical development. For the agrochemical and food industry, there are more detailed guidelines with regard to collecting organ weights and tissue evaluation in general toxicity studies (USEPA, 1998; CFSAN, 2000); however, since these guidelines do not address pharmaceutical development, the available best practices will be described and referenced. The combined experience of pathologists and reproductive toxicologists working in nonclinical safety assessment supports the assertion that, on a case-by-case basis, additional histologic and clinical pathology endpoints may be useful to include in reproductive toxicity studies, and likewise reproductive endpoints in general toxicity testing. As with inclusion of behavioral and functional assessments in repeat dose toxicity studies (Redfern et al 2013), inclusion of reproductive toxicity or pathology endpoints in studies that otherwise do not require them can be viewed as part of an overall strategy to potentially reduce animal use and candidate drug attrition, and increase understanding of preclinical-clinical translation. Additionally, major health authorities require consideration of nonclinical juvenile toxicity evaluation when appropriate in support of pediatric use. Currently, region-specific guidance documents from the United States Food and Drug Administration (FDA; 2006), European Medicines

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Halpern et al.

Page 3

Author Manuscript

Agency (EMA; 2008), and the Japanese Ministry of Health Labor and Welfare (MHLW; 2012) leave the juvenile toxicity strategy flexible, including pathology endpoints, to allow rationale for case-by-case approaches. The Organisation for Economic Co-operation and Development also describes an extended 1-generation test (OECD Test 443; 2011). In addition to these regional guidance documents, development of an international harmonized guidance for nonclinical studies supporting pediatrics, the ICH S11, has been initiated.

Author Manuscript

In an effort to understand the current regulatory context and practices regarding the use of reproductive and pathology endpoints for the assessment of reproductive, developmental and juvenile toxicity in pharmaceutical drug development, the Scientific and Regulatory Policy Committee (SRPC) of the Society of Toxicologic Pathology (STP) formed a Working Group. The purpose of this Working Group was to gain a broadly informed view of these activities, and thus the group included participants who are diplomates or members of the STP, American Board of Toxicology (ABT), American College of Veterinary Pathologists (ACVP), American Society of Veterinary Clinical Pathologists (ASVCP), Society of Toxicology (SOT), Teratology Society (TS) and the International Life Sciences Institute Health and Environmental Safety Institute (ILSI HESI) Developmental and Reproductive Toxicity (DART) Technical Committee.

Author Manuscript

The Working Group reviewed current practices for the inclusion of reproductive endpoints in general toxicity studies and pathology endpoints in reproductive and juvenile toxicity studies, in part through development of a survey of current practices used by pathologists and reproductive toxicologists. This survey was conducted through the STP and responses for each participating organization were collected and tabulated for analysis by the Working Group. One consequence of requesting combined responses from each organization is that there was no direct enumeration of the total number of individuals who contributed to the results overall, or the absolute number of studies reported. Some organizations gathered input from several individuals for their responses, whereas a single individual may have represented other organizations. However, useful information was evident in the numerical responses for defining areas of a) common practice, b) variable or inconsistent practices, and c) lack of cross-industry experience or consensus.

Author Manuscript

There were approximately 100 responding organizations or individuals overall, representing contributions from industry, contract research, consulting, government, and academia. Regions represented were global, with the majority of responses coming from North America and Europe, but also from Japan, Korea, and Africa. Approximately 80% included input from their pathology function, and 65% included input from their reproductive toxicology function. Not all respondents answered all questions, and in some instances the comments reflected a lack of direct experience in the subject area queried or responses were inadequately informative to draw conclusions. Overall, most questions were answered by about half of the participating organizations or individuals. The results were compiled centrally by the STP; responding organizations or individuals were anonymous to the Working Group. Both the numerical results and comments provided by respondents were comprehensively reviewed, and results were summarized in tabular form according to study type (DART, juvenile, or general toxicity study; see Tables 1-6). In many instances the

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Halpern et al.

Page 4

Author Manuscript

interpretation of results was strengthened by the comments from contributors, which are also discussed. Survey results were used as a basis for understanding current industry practices. The Working Group then developed an integrated assessment based on these practices, as well as current regulatory guidelines, review of the scientific literature, and personal experience. A summary of points to consider is provided, acknowledging that inclusion of endpoints is often contingent on program context, such as mechanism of action, target distribution, intended patient population, and expected or established toxicity. Where best practices are already established, these specific recommendations are referenced. The integrated assessment by the Working Group is summarized by study type in Tables 7 and 8.

Author Manuscript Author Manuscript

The current regional and harmonized guidelines are not particularly specific with regard to clinical or anatomic pathology endpoints in reproductive or juvenile toxicity studies, or reproductive endpoints in general toxicity studies. Rather, these guidance documents provide a fair amount of flexibility in approach with an emphasis on justifying the scientific rationale for each program. This flexibility was echoed in many survey responses, which favored the addition of ‘non-standard’ assessments on a ‘for cause’ basis for both general and reproductive/developmental studies. Specific considerations are outlined below for General Toxicity Studies, Developmental and Reproductive Toxicity (DART) Studies, and Juvenile Toxicity Studies. The scope of the survey was limited to species most frequently and broadly used for general toxicity testing (rat, dog and nonhuman primate [NHP]), although it is acknowledged that the rabbit, not the dog, is more commonly used in DART testing. Many principles for rodents may also apply to rabbits for study designs in which they are used; therefore, they will not be discussed separately. Finally, there is a brief discussion of considerations and challenges when the NHP is the only pharmacologically relevant species, such as with many highly specific biopharmaceuticals.

2. Inclusion of Reproductive Endpoints in General Toxicity Studies 2.1. Evaluation of Antemortem Reproductive Endpoints in General Toxicity Studies In the survey of toxicologic pathologists and reproductive toxicologists conducted by this Working Group, the majority of respondents agreed that they do not routinely include antemortem reproductive assessments in their general toxicity studies. Survey results for antemortem endpoints are summarized in Table 1. For the male reproductive system, endpoints queried included semen (dog/NHP only) or terminal sperm analysis (rat), testicular volume, hormone analysis, and/or mating trials. For the female reproductive system, hormone analysis, in-life cyclicity, and/or mating trials were queried.

Author Manuscript

Hormone evaluation was the most frequent antemortem assessment added to general toxicity studies for both males and females across species; this was followed by semen/sperm evaluation in males (dogs/NHP only) and in-life cyclicity evaluation in females. Survey respondents included hormone assessments most often (60-63%) when pharmacology or existing data of the test item suggested a potential issue. In nonrodent species, proper assessment is not typically possible without an altered study design due to

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Halpern et al.

Page 5

Author Manuscript

long and variable cycle lengths requiring frequent sampling in females, and due to hormone pulsatility in males. For all species, the evaluation of hormones should take into consideration the necessary blood volume, sampling frequency and circadian rhythm for the timing of blood collections, the inherent variability of hormones, the ovarian cycle in nonsynchronized females, and the number of animals required to detect a difference related to the test substance. The number of animals needed to detect a difference in hormones is often severely under-estimated. This topic has recently been addressed in several publications (Stanislaus et al., 2012, Chapin and Creasy, 2012, Andersson et al., 2013).

Author Manuscript

In dogs and NHP, serial assessment of semen may be added to a chronic or subchronic study design, and testicular volume may be added as a longitudinal measure of potential effects on testis size. In rodent studies, serial assessment is not possible since the approach is typically limited to sperm collected from the cauda epididymis or vas deferens at the time of necropsy. Based on Working Group experience, mating trials are infrequently added, but can obviate the need for a stand-alone study of fertility when included for males in chronic rodent studies (Mitchard et al., 2012). As an example, although unusual, a mating trial added to a 6-month rat study detected a functional effect on fertility in the absence of testicular histopathology, and demonstrated increased sensitivity relative to the standard 2-4 week premating period used in fertility studies (Powles-Glover et al., 2015). Finally, longitudinal monitoring of cycling is feasible by vaginal swab examination for evidence of menses in NHP and lavage for vaginal cytology in rodents, but is infrequently applied in the general toxicity study setting.

Author Manuscript

Thus, overall it is recognized that, on a case-by-case basis, directed assessments of the male or female reproductive systems can be included in general toxicity studies. These may be prompted by the specific pharmacology or pharmacologic targets, or based on previous study findings. However, careful consideration of potential impact to other study endpoints, or the limitations of the general toxicity study environment, should be considered for successful implementation. 2.2 Evaluation of Reproductive Organ Weights in General Toxicity Studies

Author Manuscript

Organ weight evaluations are routinely included in general toxicity studies. In this survey, almost all (93-98%) respondents routinely weighed the testes in general toxicity studies conducted in rodents, dogs, and NHP; ovaries (83-91%), prostate (80-87%), and pituitary gland (71-73%) were also commonly weighed in all species. The epididymides were weighed commonly in rats, but less consistently in dogs or NHP. The seminal vesicles were infrequently and inconsistently weighed across species, and in the rodent they were sometimes collected with the prostate. The uterus was weighed by approximately 50% of respondents across species. Survey responses for organ weight data in general toxicity studies are summarized in Table 2. For tissues related to the reproductive system, the STP has recommended weighing the pituitary gland in both sexes for all species except mice, testes in all species, and epididymides and prostate in rats and in other rodent and non-rodent species on a case-bycase basis (Sellers et al., 2007). Seminal vesicle weights in rodents generally provides information similar to that afforded by the prostate weight assessment (Sellers et al., 2007). Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Halpern et al.

Page 6

Author Manuscript

The STP has recommended that female reproductive organ weights including ovary and uterus weights should be performed on a case-by-case basis in rodents and non-rodent species and interpreted cautiously due to high inter-individual variability with normal cycling (Sellers et al., 2007). The Working Group is in general agreement with this position, while also acknowledging that ovarian weights from rodent studies with animals of comparable age can be useful in detecting test item effects, especially when interpreted with other data such as histopathology or hormone levels. This is consistent with the survey responses indicating that ovarian weights are typically recorded, and uterine weights are less consistently determined.

Author Manuscript

Organ weight changes in accessory sex glands should be interpreted in light of corroborating data. Fluid loss from the seminal vesicles can alter organ weights significantly, so necropsy approaches to avoid this may help to prevent leakage and improve consistency of results across individuals if this endpoint is considered necessary. When seminal vesicle weights or ventral prostate weights are collected carefully, either individually or as part of the accessory sex gland unit (prostate and seminal vesicles), they can offer valuable information because they are sensitive to detecting a low testosterone signal (O'Connor, Davis et al. 2000). Although these accessory reproductive organ weights vary with body weight, others such as testis, are not consistently affected (Marty et al., 2003, O'Connor et al., 2000). While lower accessory sex gland weights may occur as a result of test item-related changes such as hormonal perturbations (e.g., low testosterone), these changes may also occur due to nonspecific factors such as stress or decreased food consumption/body weight (Everds et al., 2013). Thus, caution should be taken to avoid over-interpretation of accessory sex gland organ weight data in isolation.

Author Manuscript

It should also be noted that sexual maturity, especially in dogs and NHP, has a substantial impact upon male and female reproductive system organ weights. Because of inherent variability between individuals, whether mature or immature, reproductive organ weights in large animals are most valuable when correlated with histologic findings and evaluation of maturity. 2.3 Evaluation of Reproductive Histopathology in General Toxicity Studies

Author Manuscript

Male and female reproductive system tissues are routinely included in the standard (core) protocol-required tissues for histopathologic examination in general toxicity and carcinogenicity studies, as per regulatory expectations. The STP has recommended that this core tissue list should include the pituitary, testes, epididymides, prostate, seminal vesicles, ovaries, uterus, and vagina (Bregman et al., 2003). This recommendation was made for all repeat-dose toxicity and carcinogenicity studies involving all drug classes, routes of administration, study durations, and laboratory animal species (Bregman et al., 2003). Routine histopathologic examination of the testes should be performed in a stage-aware manner without quantitative staging of spermatogenesis (Creasy, 1997, Lanning et al., 2002). Histopathologic examination of the testes should always be accompanied by evaluation of the epididymides. Organ weight changes in either the testes or epididymides, or histopathologic changes in the epididymides (such as reduced sperm or differences in the

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Halpern et al.

Page 7

Author Manuscript

amount/morphology of luminal cellular debris), should be correlated to histologic findings in the testis. Likewise, routine histopathologic examination of both ovaries for potential ovarian toxicity should be performed using a 2-tier approach (Regan et al., 2005). This approach uses qualitative examination of both ovaries in conjunction with microscopic examination of other female reproductive tissues, organ weight evaluation, and awareness of estrous/ menstrual cycle stage as a first tier. If warranted based on the qualitative assessment, this may be followed by additional second-tier assessments or studies to include monitoring of estrous/menstrual cyclicity, hormones, and/or ovarian follicle counting, although follicle counts are not a routine assessment for pharmaceutical development (Regan et al., 2005). 2.4 Sexual Maturation

Author Manuscript

The evaluation of sexually immature animals does not provide an accurate assessment of the potential for toxic effects on the adult reproductive system. While most general toxicity studies in rodent species use animals that are sexually mature at termination, it is not uncommon for dogs and nonhuman primates used in general toxicity studies to be sexually immature. Thus, caution must be used in assessing the risk for effects on spermatogenesis, cyclicity, or other aspects of reproductive morphology and function in large animal studies. Documentation of sexual maturity provides additional context for review and interpretation of the study data. In particular, studies intended to evaluate potential effects on fertility require mature animals, while immature animals may support pediatric use; these are discussed further in subsequent sections.

Author Manuscript Author Manuscript

Sexual immaturity is often recorded in the histopathology data for dogs and NHP when present, but histologic evidence of maturity is not routinely recorded. The survey responses highlight these current practices (Table 3), and confirm the more frequent documentation of immaturity for males than females. Survey comments also indicated that recording sexual maturity, while not standard, can be important for interpretation of findings and hazard identification, or in the development of pediatric indications (see discussion on pediatrics below). Of the 43 comments provided by respondents, about half clearly supported documenting immaturity, maturity or both, with the remainder mostly neutral. There were 4 comments that specifically noted that recording immaturity had been helpful in avoiding additional studies in juvenile animals, but there were also 5 comments that indicated that inclusion of sexual maturity status had contributed to a program delay or request for additional nonclinical studies. Although further context was not provided for these responses, the current regulatory environment supports inclusion of this information because it enables appropriate risk assessment for the target population. Based on the survey responses, most pathologists are able to discern sexual maturity (Table 4), acknowledging that it can be difficult to definitively categorize some adolescents. This is especially true for pubertal females where ovarian and uterine morphology can be similar to that of young adult females in the earliest preovulatory phases of their cycle. This is further complicated in NHP by the wide variability in age at onset of regular menses.

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Halpern et al.

Page 8

Author Manuscript

When maturation status was documented, most respondents preferred the use of the terms “immature” and “mature” with infrequent use of alternate terms such as “prepubertal,” “peripubertal,” “adolescent,” or “adult” in all species. The histologic features used most frequently to confirm sexual maturity in males of all species were the presence of spermatogenesis/spermiogenesis in the testis and sperm content in the cauda epididymis. Accessory sex organ epithelium and/or secretions were not considered good indicators of sexual maturation. The histologic features used most frequently to confirm sexual maturity in females of all species were the presence of corpora lutea and evidence of cyclicity. Mammary gland formation occurs early during puberty and therefore was generally not considered a good indicator of full sexual maturity. Age, body weight, and organ weights in both males and females, as well as cyclicity in females, were commonly used as nonhistological criteria in the assessment of sexual maturity.

Author Manuscript

While the Working Group is in general agreement with survey respondents regarding the practices of assessing sexual maturity based on histology of reproductive tissues in dogs and NHP, age and body weight are not considered robust indicators of maturity in these species. Both Working Group experience and survey responses indicate that the omission of information documenting maturity status in the study record may undermine interpretation of other data, such as testicular and ovarian weights, and can lead to inconsistent assumptions regarding potential effects on spermatogenesis or cyclicity. For these reasons, documentation of sexual maturity (whether immature or mature) should be considered. Although not routinely included in test system characterization for toxicity studies, dentition, physeal closure, or other objective measures to confirm age and/or maturity could also be considered (Bowen and Koch, 1970, Smith et al., 1994, Kilborn et al., 2002).

Author Manuscript

Recognizing and distinguishing test item-related findings from spontaneous or age-related changes in immature or maturing animals, especially dogs and NHP, is an important skill for pathologists reviewing general toxicity studies. This can be a challenging exercise, even for pathologists experienced in the evaluation of the reproductive tract, due to the small number of animals used and the high variability among individuals. Publications describing testicular development (Picut et al., 2015b, Haruyama et al., 2012, Campion et al., 2013), common spontaneous histologic features of the testes during the process of sexual maturation (Goedken et al., 2008, Rehm, 2000, Sato et al., 2012a, Sato et al., 2012b, Thuilliez et al., 2014), and ovarian development (Picut et al., 2015a, Picut et al., 2014) represent excellent references for this topic. 2.5 Specific Considerations for Evaluation of the Nonhuman Primate Reproductive System in General Toxicity Studies

Author Manuscript

Nonhuman primates are often the only pharmacologically relevant species for nonclinical safety testing of highly targeted biotherapeutics, and the cynomolgus monkey is the species most often evaluated. While the reproductive physiology and endocrinology of NHP and humans are similar (Weinbauer et al., 2008), the use of NHP imposes some practical limitations on fertility testing. For example, NHP usually have single births, relatively low conception rates, and high rates of fetal loss, which would necessitate the use of unacceptably large numbers of animals (Martin and Weinbauer, 2010). In recognition of

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Halpern et al.

Page 9

Author Manuscript Author Manuscript

these issues, the addendum to ICH S6(R1) formalized the acceptance of the use of organ weights and histopathologic examination of the reproductive tract in at least one study of at least 3 months duration using mature NHP to support the evaluation of potential effects on fertility (ICH S6(R1), 2012). This has led to a need for assessment of maturity as a component of prestudy screening, usually via assessment of sperm or cyclicity, and for toxicologic pathologists to recognize morphologic features enabling them to verify that animals exposed to a pharmaceutical candidate are sexually mature at the end of the study. As indicated in Table 3, a majority (71%) of respondents indicated that maturity status is recorded for male NHP, though fewer (43%) reported routine recording of maturity status in females. In males, criteria for evaluation included histologic assessment of spermatogenesis, spermiogenesis, and/or presence of sperm in the epididymides. Survey respondents most commonly used presence of a corpus luteum, indicating that ovulation had occurred, as evidence of sexual maturity in females. Some caution is needed when a single corpus luteum or corpus albicans is identified in a young female NHP, as isolated instances of ovulation may occur prior to the onset of regular ovulation and menses (Beckman and Feuston, 2003, Cline et al., 2008, Buse et al., 2003). When necessary, a weight of evidence approach using observations of serial menses prior to study initiation may be warranted. One approach is to require at least 2 cycles of at least 2 days duration and at least 20 days apart when selecting mature females (Weinbauer et al., 2015).

Author Manuscript

Histologic evaluation can be useful to demonstrate an absence of effects on the structural integrity of the reproductive tract in both sexes, and the synchronicity of cyclical features across reproductive tissues in individual females. However, these endpoints do not directly assess reproductive function as compared to a number of endpoints that more directly evaluate potential impact on fertility, such as sperm parameters, fertilization, or implantation. In recognition of this, the addendum to ICH S6(R1) also directs that, when pharmacology or prior findings with the candidate dictate, the inclusion of additional specialized antemortem assessments such as reproductive hormones, cycling patterns and sperm evaluations may be desirable in repeat dose toxicity studies (ICH, 2012). Although these additional evaluations may be useful to more fully characterize an effect, changes in the male antemortem endpoints in isolation are not expected to be more sensitive indicators of toxicity than histopathology (Cappon et al., 2013). Likewise, inherent variability in cycle stage and progression, along with small group sizes, limits the utility of monitoring cycling in female NHP (Bussiere et al., 2013).

Author Manuscript

While microscopic evaluation is an important criterion for verification of maturity, importance should also be given to pre-study selection of NHP for inclusion in general toxicity studies of ≥ 3 months duration. In females, the occurrence of regular menses is good evidence of sexual maturity (Weinbauer et al., 2008) and survey respondents reported using age and evidence of cyclicity most frequently to select mature females. However, many mature female NHP do not cycle regularly, or have irregular or anovulatory cycles due to stress and hierarchical status. For males, respondents reported using age most often for selecting sexually mature animals (data not shown). However, age is not entirely dependable as a marker of sexual maturity (Luetjens and Weinbauer, 2012, Vogel, 2000). When NHP are the only pharmacologically-relevant species for a highly targeted biotherapeutic, and there is a need to ensure that sexually mature animals are selected for the study, evaluation of an Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Halpern et al.

Page 10

Author Manuscript

ejaculate for the presence of sperm prior to treatment is a highly reliable functional endpoint (Luetjens and Weinbauer, 2012). Consideration should also be given to the many social and management factors that have been shown to impact the reproductive system in both sexes, and thus may complicate interpretation (Weinbauer et al., 2008, Niehoff et al., 2010, Cline et al., 2008, Bussiere et al., 2013). 2.6 Histologic Staging of the Female Reproductive Cycle

Author Manuscript

There is no regulatory guidance or requirement for histologic staging of the reproductive cycle in mature female animals in general toxicity studies. The survey did not query details of staging, but cyclicity is not routinely recorded by most respondents, although it is considered ‘for cause’ in the rodent and NHP (Table 1). As with evaluation of any organ or organ system, the pathologist must understand the underlying species-specific physiology to properly interpret apparent differences in morphology. This is particularly important for the female reproductive tract, since the morphology is highly synchronized and extremely sensitive to hormonal perturbation. Normal cyclicity can be disrupted by environmental factors, such as lighting, social hierarchy or secondary factors such as stress and body weight changes, and these in turn will affect the morphologic appearance of the various portions of the reproductive tract (Keane et al., 2015, Bussiere et al., 2013, Everds et al., 2013, Weinbauer et al., 2008).

Author Manuscript Author Manuscript

For rodent general toxicity studies, the morphology of each of the reproductive tract organs (ovaries, uterus, cervix, and vagina) as well as mammary gland and endocrine tissues should be assessed to identify possible changes due to a potential test item effect or hormonal perturbations. The microscopic evaluation of all reproductive tissues should include an awareness of the normal variation in morphology associated with stages of the estrous cycle (Dixon et al 2014). Recognition of the morphologies in these organs that are associated with synchronous progression through the estrous cycle is important, and discrepancies within animals may indicate an abnormality. The rapid estrous cycle (4-5 days) in the typical rodent used in general toxicity studies may enable detection of alterations even in studies as short as 2 weeks duration (Sanbuissho et al., 2009), although reproductive senescence may confound assessment in studies longer than 3 months duration (Ishii et al., 2012, LeFevre and McClintock, 1988, Sone et al., 2007). When alterations are suspected based on morphologic examination of the reproductive tract, documenting stages of the estrous cycle may suggest potential mechanisms (central vs. peripheral) underlying the observed changes. However, differences in the incidence of cycle stages across groups based only on vaginal histology may represent normal variation and should not be used in isolation as evidence of a test item-related effect. Evaluation may include consideration of other data from the current or previous studies, such as the morphology of other reproductive tissues, changes in organ weights, effects on reproduction or fertility, values of hormones, and/or alterations in cyclicity based on vaginal cytology, when these additional data are available. The Working Group supports the INHAND recommendation that routine documentation of stage of the estrous cycle in rodents is not necessary in general toxicity studies (Dixon et al 2014); however, evaluating the cycle stage is a routine part of the microscopic evaluation, and has been described in detail for the rat (Westwood, 2008), dog (Rehm et al., 2007b) and

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Halpern et al.

Page 11

Author Manuscript

NHP (Attia, 1998, van Esch et al., 2008). If the pathologist chooses to record the cycle stage in a standard toxicity study, the information should not be considered in isolation as evidence of a test effect unless accompanied by other correlative or supporting data. Notably, there are circumstances, such as evidence of effects on fertility, differences in organ weights, or apparent changes in reproductive morphology, in which estrous cyclicity data would provide additional weight of evidence for assessments or clarification of findings observed. In such circumstances, the pathologist should consider documentation of cycle stage.

Author Manuscript Author Manuscript

In dogs and NHP, detecting effects on estrous/menstrual cyclicity from the microscopic examination is more problematic because of their long cycle duration and low numbers of animals per group. The laboratory Beagle under controlled environmental conditions is monoestrous, with long periods of diestrus/anestrus (Rehm et al., 2007a), so often the females will be in diestrus/anestrus and will not have ovulated during the dosing period of short-term toxicity studies. Although adult cynomolgus monkeys ideally have regular menstrual cycles lasting approximately 28 days (Weinbauer et al., 2008), cycle irregularities are frequent. Social hierarchy plays an important part in menstrual cyclicity, and both adults and adolescents often have anovulatory cycles, further complicating the assessment in NHP (Cline et al., 2008). Assessment of cyclicity during the dosing phase using vaginal cytology (dog) or vaginal swabs for evidence of menses (NHP), and potentially hormone evaluation, may be important in helping to characterize the onset and progression of variations in morphology detected histologically. However, attempting to use stage alone may present interpretive difficulties in determining test item-related effects (Bussiere et al., 2013). Therefore, the considerations for rodents apply to dogs and NHP as well: evaluate female reproductive tissues with cycle stage awareness, but routine recording may not be warranted in the absence of apparent changes in the reproductive tract, expected pharmacology or by other findings in the current or previous studies. Likewise, if recorded, the information should not be interpreted as an endpoint in isolation. 2.7 Evaluation of the Mammary Gland in General Toxicity Studies

Author Manuscript

The female mammary gland is one of the core tissues recommended for examination by the STP in nonclinical toxicity studies (Bregman et al., 2003). In both males and females, it can serve as a sensitive endpoint for test item-related changes in hormone levels or receptor signaling in the hypothalamic-pituitary-gonadal axis (Lucas et al., 2007). In female rodents, the mammary gland offers little utility in assessing sexual maturity as the histology of the gland is similar in prepubertal and sexually-mature virgin animals (Russo et al., 1989). Cycle-dependent changes in glandular histology have been reported in the rat (Schedin et al., 2000); however, the differences in appearance are subtle and of little practical aid in differentiating phases of the estrous cycle in standard nonclinical safety studies. In comparison, the histology of the mammary gland in female dogs and NHP may provide corroborative information to differentiate mature and immature animals (Cline and Wood, 2008, Rehm et al., 2007a, Harleman and Foley, 2001); in fact, the mammary gland is one of the first tissues to undergo peripubertal developmental changes in NHP, including transient pubertal gynecomastia in males (Cline and Wood, 2008). In mature dogs, the appearance of the mammary gland changes predictably throughout the estrous cycle (Chandra et al., 2010), but its morphology in NHP changes little during the menstrual cycle (Stute et al., 2004). In

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Halpern et al.

Page 12

Author Manuscript

males, the mammary gland has limited development in most species; the rat is an exception, with lobuloalveolar development occurring under the influence of androgens, and tubuloalveolar development occurring under the influence of anti-androgens or prolactin (Lucas et al., 2007, Cardy, 1991). While not always included in the protocol specified tissue list for male rats, mammary gland is often present in sections of skin, and findings should be recorded if present.

Author Manuscript

Although mammary gland morphology does reflect endocrine influence, the majority of respondents in the current survey indicated that they generally do not use this tissue to document or confirm sexual maturity (data not shown). In females, corpus luteum formation and evidence of estrous/menstrual cyclicity were generally given greater importance in determination of sexual maturity. For assessment of cyclicity, only about half of the respondents indicated that mammary gland is useful, mostly in dogs, and it was not cited as a criterion for evaluation as often as were the ovaries, uterus, and vagina. In summary, review of the mammary gland in general toxicity studies provides some limited information for assessment of maturity, and may be helpful in assessing synchrony with other reproductive tissues in individual animals. A major value in assessing this tissue lies in its sensitivity to effects on reproductive hormonal balance. Thus, evaluation of the mammary gland, especially in the context of findings in the reproductive tract, may provide corroborative or mechanistic information in the evaluation of disruption of reproductive hormone balance. In addition, it may augment the assessment of sexual maturity in the dog. Overall, the common industry practices are consistent with the literature and assessment by the Working Group.

Author Manuscript

2.8 Summary of Working Group Considerations for Incorporation of Reproductive Endpoints in General Toxicity Studies

Author Manuscript

Consistent with general practice and published best practices, most general toxicity studies include a gross and histologic assessment of the male and female reproductive tract. The survey results largely confirmed, and the Working Group does not suggest changing, these established practices. In general, survey respondents indicated that specialized reproductive assessments are infrequently included routinely in general toxicity studies, but may be added ‘for cause’. The Working Group agrees with the STP recommendation of ‘stage aware’ assessment of both male and female reproductive tissues in mature animals (Creasy, 1997, Dixon et al., 2014, Lanning et al., 2002). Additional reproductive endpoints in mature animals, including sperm assessments, estrous or menstrual cycle stage, hormone evaluations and mating trials may be considered ‘for cause’, but are not routinely included. Routine evaluation of mammary gland morphology in both sexes may also add to the interpretation of findings in reproductive tissues, particularly in females. Based on the survey results and Working Group experience, documentation of the stage of male and female sexual maturity should be considered for nonrodents (dog and NHP). Finally, interpretation of findings in the reproductive tract should be cautiously evaluated in the context of potentially confounding factors including stress or body weight loss. These integrated considerations are summarized in Table 7 for General Toxicity Studies.

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Halpern et al.

Page 13

Author Manuscript

3. Inclusion of Pathology Endpoints in Reproductive Toxicity Studies 3.1. Pathology Endpoints in Developmental and Reproductive Toxicity Studies

Author Manuscript Author Manuscript Author Manuscript

Routine developmental and reproductive toxicity studies are not intended to support intentional pharmaceutical use during pregnancy, but rather to identify effects on development and reproduction in animals for informing human risk assessment. There are few regulatory guidelines governing inclusion of clinical and anatomic pathology endpoints in reproductive toxicity studies. The current ICH S5(R2) guideline on Detection of Toxicity to Reproduction for Medicinal Products and Toxicity to Male Fertility, indicates that observations should include the preservation and possible histologic evaluation of organs from adults with macroscopic findings and appropriately matched tissue from control animals in reproductive toxicity studies, including fertility and early embryonic development studies, embryo-fetal development studies, and pre- and postnatal development studies. For embryo-fetal development studies, macroscopic observations should also include evaluation of fetuses for birth defects and evaluation of the placenta. For fertility studies, testes, epididymides, prostate gland, ovaries and uteri should be preserved from all animals for possible histologic evaluation on a case-by-case basis but can then be discarded after completion and reporting of the study. In ICH S5(R2), organ weights are not specifically required unless there were reproductive organ weight effects in general toxicity studies, or evaluation of reproductive organ weights at the dose levels tested was not done previously. ICH S5(R2) states that some minimal maternal toxicity is expected to be induced in high dose group of embryo-fetal developmental (EFD) toxicity studies unless at a limit dose of 1 g/kg or saturation of exposure. Although it is not clear whether it is necessary to reproduce expected toxicities in the high dose dams on an EFD study, clinical and/or anatomic pathology findings from the general toxicity program can contribute to the justification of high dose selection of these studies. Additional considerations for high dose justification of EFD toxicity studies that are under discussion as part of the forthcoming revision of the ICH S5 guideline include adequate exposure margin relative to clinical use and/or maximum pharmacodynamic effect. For DART studies with biotherapeutics, the ICH S6(R1) emphasizes the need to limit studies to pharmacologically relevant species, and further acknowledges that reproductive studies may not be useful or necessary in cases where there are no relevant nonclinical species for toxicity testing. Overall, the ICH S6(R1) outlines differences from the ICH S5(R2) expectations for reproductive assessments when NHP are a single relevant species. For example, in most cases, fertility assessments rely on histopathologic assessment of reproductive tissues rather than a breeding endpoint. The ICH S6(R1) guideline does not include detailed recommendations for specific endpoints to include when studies are conducted, other than to state that specialized assessments of menstrual cyclicity, sperm analysis, and hormone evaluations be considered if there is a specific cause for concern. Thus, it is common practice to derive NHP fertility information from the general toxicity program with inclusion of mature monkeys in at least one study of at least 3 months duration. The survey results regarding pathology endpoints on Developmental and Reproductive Toxicity (DART) studies included responses to both general questions and study typespecific questions, and results are summarized in Table 5. Overall, when pathology

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Halpern et al.

Page 14

Author Manuscript Author Manuscript

endpoints (clinical pathology, organ weights and/or anatomic pathology) were added to DART studies either as routine or case-by-case, approximately half of the survey respondents indicated that the addition of these specific endpoints impacted the risk assessment (survey responses were split 53% yes, 47% no). Some additions may have been at the request of regulators, but some respondents indicated that they were helpful in clarifying maternal versus fetal toxicity, evaluating differences in susceptibility (i.e. adult versus fetus/offspring), justifying dose selections, and/or providing additional weight of evidence for assessments or clarification of findings observed in general toxicity studies. Specific endpoints added on a case-by-case basis were viewed as generally beneficial, and pharmacodynamic endpoints in studies with biotherapeutics were considered essential by some respondents to help define a persistence of effect or clarify if findings were a primary or secondary effect of maternal exposure. Concerns with adding pathology endpoints included difficulty in interpreting the relationship to the test item due to limited appropriate historical control data based on reproductive phase (e.g., gestation, lactation), and identification of findings not seen in general toxicity studies. However, the stated benefits gained (ability to characterize findings by identifying potential mechanisms, clarifying adult versus infant toxicity or toxicity vs pharmacology, and determining maternal toxicity versus direct pharmacology effect on offspring) were considered to be a justification to include additional targeted endpoints in these studies on a case-by-case basis. For example, histology and immunohistochemistry of lymphoid tissues of offspring are often assessed in studies with immunomodulatory drugs (Auyeung-Kim et al., 2009, Vaidyanathan et al., 2011, Martin et al., 2007). 3.2 Pathology Endpoints in Male and Female Fertility Studies

Author Manuscript

The speculative nature of the survey responses and comments confirmed that there is very limited experience with functional dog fertility studies (Daurio et al., 1987, Youssef et al., 1999), therefore, the survey data are not presented here. There is also limited experience across respondents for functional NHP fertility studies; survey data for NHP fertility studies should be interpreted acknowledging that few organizations have first-hand experience with this study type. Although experience with functional fertility studies in dogs and NHPs is limited, evaluation of the male and female reproductive systems in sexually mature animals may offer valuable insights into potential effects on fertility and in some cases may be the best available nonclinical model for evaluation.

Author Manuscript

Survey data comments indicated that clinical pathology is not typically added to fertility studies without scientific justification (e.g. previous findings, pharmacology/target). Although not the default approach, the rationale for adding clinical pathology appears similar across species since 48-57% of respondents would trigger this in all species, although 27-30% of the respondents do not ever perform these evaluations (Table 5). Of all the pathology endpoints considered across the study types, addition of organ weights to dedicated fertility studies was the only one done by default by the majority of survey respondents, 58-74% in rodents and 50-58% in NHP (Table 5). Survey data and comments indicated that reproductive tissue organ weights, especially testis weights in males, are often collected in fertility studies despite the absence of a regulatory requirement to include them.

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Halpern et al.

Page 15

Author Manuscript

However, if a fertility evaluation is performed as part of a chronic toxicity study in mature animals (e.g., when NHP is the only relevant species), then all standard organs for evaluation of general toxicity, including reproductive tissues, would be weighed as part of the necropsy assessment.

Author Manuscript

No clear trends in the survey data were observed for adding histopathology to fertility studies. For rodents, some respondents indicated that it was included de facto (35-42%) or conducted ‘for cause’ (48%) based on findings (such as fertility and/or organ weight effects) (Table 5). In the experience of the Working Group, a gross necropsy including reproductive organ weights is often performed, with preservation of reproductive tissues for possible future histologic evaluation. Thus additional characterization of potential effects on reproduction can be evaluated from specific animals if necessary, but is not included routinely, especially if reproductive tissue histopathology data is already available from previously conducted repeat dose toxicity studies at the same dose range. For NHPs, the survey responses were mixed, but the most common response was to include histopathology (44-48%); this is consistent with histopathology being standard on repeat dose studies and often used as a primary endpoint in NHP for nonclinical assessment of potential effects on reproductive tissues that could impact fertility. In summary, the practice is generally consistent with current guidance documents. Inclusion of organ weights and histology is relatively common even though not currently required for stand-alone fertility studies. 3.3 Pathology Endpoints in Embryo-Fetal Developmental (EFD) Toxicity Studies

Author Manuscript

Survey results supported the perception that there is very limited experience with dog EFD studies (Holson et al., 2015) as this is not a standard species for this study type; thus, the survey data for this species are not presented here. There is also limited experience across respondents with dedicated NHP EFD studies, although these have been more common in the biotechnology industry, especially prior to the most recent guideline (ICH S6[R1]).

Author Manuscript

Of responding organizations, 30% routinely added pathology endpoints (such as clinical pathology, organ weights, and/or histopathology) to define maternal toxicity in EFD studies whereas 48% stated that they would add them if no other signs of maternal toxicity were anticipated (data not shown). The survey did not distinguish between dose range-finding studies, where additional endpoints to characterize maternal toxicity may be more common, versus pivotal studies where endpoints would typically only be added for cause. One respondent indicated that collecting these or specific endpoints (both maternal and fetal) could help differentiate whether fetal effects were secondary to maternal toxicity or direct (primary) fetal effects. Most NHP EFD studies are likely performed to support biopharmaceutical development, and studies to support those programs may not use maternal toxicity to justify the high dose, but rather exposure and/or pharmacodynamic effects. In this situation, there is limited utility in adding maternal pathology endpoints on these studies other than to characterize pregnancy-specific pharmacologic effects on the dam. Beyond dose rationale, 47% of respondents indicated that they use general toxicity endpoints on a case-by-case basis to understand potential pharmacodynamic effects on development.

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Halpern et al.

Page 16

Author Manuscript Author Manuscript

Survey data confirm that clinical pathology is not typically performed in EFD studies (maternal or fetal) without scientific justification, but when it is triggered it is primarily for maternal evaluation. In dams, clinical pathology is rarely (14-16% respondents) done by default, and often (27-32% respondents) is not included at all (Table 5). When included if triggered by pharmacology/target or previous findings, the high response in the “maybe” category (52-59%) is consistent with the previously discussed 30% of respondents indicating that they would add clinical pathology endpoints to help characterize maternal toxicity. Only one organization appears to routinely perform clinical pathology assessments on fetuses (rodent only) from EFD studies, and the majority of respondents (64-71%) do not collect fetal clinical pathology for any species. The high response rate for not collecting these data from fetuses may appear inconsistent with the 47% of respondents that indicated they would add general toxicity endpoints on a case-by-case basis to understand potential effects on development. However, this may reflect the difficulty of interrogating the mechanism in these complex studies, as well as a lack of direct application of these data to inform clinical use or monitoring. There can also be technical/practical limitations on the amount of information that can be obtained from fetal specimens as part of an embryo/fetal developmental toxicity study. Based on Working Group experience, if clinical pathology endpoints are deemed useful from rodent fetuses it is advisable to collect blood from satellite animals to avoid damaging the specimens for fetal evaluations, and pooling samples across the litter if necessary to obtain sufficient blood volumes. Another approach is to collect these samples from a pilot study rather than the definitive study where the fetal exam is the primary outcome. Sampling earlier gestation age specimens is challenging, as blood volumes are more limited and use of tissue or whole fetal homogenates can be hard to interpret relative to maternal clinical pathology.

Author Manuscript Author Manuscript

Many (76%) respondents indicated that the apparent challenges of interpreting clinical pathology endpoints in gestating and lactating females (lack of historical data/familiarity) did not impede the interpretation of these endpoints. Some respondents indicated that if clinical pathology endpoints were added to a study it was done in a deliberate fashion and the study design was modified appropriately. In general, the survey comments indicated that opinions on this were split with some respondents stating that concurrent controls have been sufficient, but for others the lack of gestation age-appropriate historical control data was problematic. At least one respondent stated that maternal clinical pathology is not helpful in EFD studies as it can confuse and complicate regulatory applications, because the no observed effect level (NOEL) may be different when compared to general toxicity studies. Clearly, some maternal clinical pathology endpoints (such as serum proteins, lipids and some leukocyte subsets) change over the course of pregnancy (Honda et al., 2008); understanding these changes and their normal variability in specific species is important to appropriately interpret test item-related changes in these endpoints. In addition, close attention must be paid to the physiologic requirements of the dam so to avoid undue stress on the animal by added clinical pathology endpoints and interference with the interpretation of the standard embryofetal development endpoints. In the majority of cases, maternal organ weights are either not routinely collected in rodent EFD studies (40%) or are triggered on a case-by-case basis (40%) (Table 5). Although the survey question did not explicitly exclude gravid uterine weights, in Working Group Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Halpern et al.

Page 17

Author Manuscript

experience they are typically collected as a reproductive endpoint for these studies, as are individual fetal weights. Sixty-three percent (63%) of respondents do not routinely collect rodent fetal organ weights. In NHP, maternal organ weights are either typically not collected (36%) or they are triggered on a case-by-case basis (48%). In comparison with the infrequent collection of organ weights from rodent fetuses, only 36% responded that they do not collect NHP fetal organ weights (Table 5).

Author Manuscript

The survey indicates that greater than 50% of respondents would potentially include maternal or fetal histopathology, but it was unclear how often that might actually happen across studies or programs within an organization. Targeted review was considered useful to some respondents, but some (22-47%) indicated that they do not perform maternal or fetal histopathology analyses (Table 5). A low percentage of respondents routinely include histopathology for dams (12-19%) or fetuses (5-12%) in EFD studies (Table 5). Seventyeight percent (78%) of respondents indicated that the challenges of interpreting anatomic pathology in fetuses/pup/infants (such as lack of historical data/familiarity with conduct of these studies) did not impede interpretation. Survey comments consistently indicated that interpretation of microscopic evaluation can be difficult due to low incidence findings, limited/no historical data on specific (developmental) age of death, differentiating direct or indirect (maternal) fetal changes, differentiating developmental morphology from pathological changes, and confounding of histopathology evaluation by test item effects on growth (developmental delay). 3.4 Pathology Endpoints in Pre- and Postnatal Developmental (PPND) Toxicity Studies

Author Manuscript

The results of the survey on inclusion of pathology endpoints in rodent and NHP PPND studies is much like that for EFD studies (Table 5); although not considered routine, clinical pathology, organ weight and histopathology data are incorporated into the maternal and/or fetal aspects of these studies when deemed appropriate based on pharmacology and/or results of previous studies. There are not many recently published examples of rodent PPND studies in the literature, but a recently published compilation of NHP developmental toxicity studies provides examples of PPND studies that included pathology endpoints (Weinbauer et al., 2015). The survey data indicated that there is very limited to no experience with dog PPND studies as this is not a standard species for this study type; thus, survey data for this species are not presented here.

Author Manuscript

Forty-seven percent (47%) of respondents indicated that they add pathology endpoints (such as clinical pathology, organ weights, and/or histopathology) on a case-by-case basis to understand potential pharmacodynamic effects on development, but 37% of the respondents stated that they do not utilize these endpoints in this manner. Survey data confirm that clinical pathology is not typically conducted in rodent PPND studies without scientific justification, although for NHP PPND studies, responses were fairly evenly split between organizations that include clinical pathology as a default, only for-cause, or not at all. For rodents, it appears that 33% of organizations do not include clinical pathology of the dams, while 56% of respondents would include it for-cause. Most

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Halpern et al.

Page 18

Author Manuscript

organizations either do not perform clinical pathology on rodent pups (56%) or would trigger inclusion of clinical pathology based on previous findings or based on pharmacology/ target (38%) (Table 5). For NHP, 54% of respondents would include clinical pathology of the dams ‘for cause’, and another 27% of respondents routinely conduct clinical pathology evaluations on dams, whereas for NHP infants the responses were fairly evenly divided across respondents that routinely include, only for-cause, or not at all (Table 5).

Author Manuscript

As described for EFD studies, the majority of respondents stated that the challenges of interpreting clinical pathology endpoints in gestating and lactating females (such as lack of historical data/familiarity with conduct of these studies) did not impede the interpretation of these endpoints when controlled appropriately in the context of normal pregnancy and lactation-related changes. Most (82%) respondents also indicated that the apparent challenges of interpreting clinical pathology endpoints in infants/pups (lack of historical data/familiarity) did not impact the interpretation, and 77% responded that there was no negative impact of including clinical pathology endpoints in their studies (data not shown). It appears that concurrent controls can be sufficient, but respondent comments also indicated that a comprehensive review of rodent offspring hematology historical control values would be useful. As with other situations with limited historical control data and knowledge, it is not uncommon to default to a conservative interpretation that an equivocal effect may be test item related. As discussed with fetal endpoints in EFD studies, evaluating these endpoints in offspring, particularly if utilizing a pharmacodynamic biomarker, can facilitate correlation to maternal effects and help determine whether effects are direct or indirect (maternal).

Author Manuscript

Maternal organ weights are not a standard endpoint for PPND studies, but may be included for cause. Specifically, in rodents and NHP, organ weights are not typically recorded for dams by 42-46% of respondents, but 31-32% indicated that they would include these if indicated based on prior findings or mechanism of action (Table 5). In offspring, respondents generally do not record organ weights in rodents (42%), but in NHP they are either routinely collected (37% of respondents) or included for cause (37% of respondents; Table 5).

Author Manuscript

The survey indicated that approximately 50% of respondents would trigger maternal or fetal histopathology for individual programs, but it was unclear from the survey how often that might occur. For dams, histopathology is rarely performed (8-17%) or not done (36-39%); however, it may be included ‘for cause’ by 47-54% of respondents (Table 5). For rodent offspring, histopathology is not routinely performed (28% of respondents listed ‘never’), but 53% of respondents perform histopathology on a case-by-case basis (Table 5). For instance, for a drug with concerns about renal development, evaluation of kidneys from the PPND study from culled neonatal and weanling rat pups could provide information about a potential effect on nephrogenesis as, unlike humans, significant renal development occurs during the first week of postnatal life in rats (Zoetis and Hurtt, 2003a). Histopathology for NHP offspring is a relatively common (32%) default practice, and an additional 48% of respondents would include it on a case-by-case basis (Table 5). As mentioned earlier for fetal histopathology in EFD studies, most (78%) of the respondents indicated that, although there were challenges with interpreting histopathology in pups/infants (low incidence findings, limited/no historical data on specific age of death, differentiation of direct or indirect changes, differentiating developmental dysmorphology from pathological changes,

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Halpern et al.

Page 19

Author Manuscript

and confounding effects on growth), this rarely resulted in a negative impact on the study interpretation. 3.5 Summary of Working Group Considerations for Incorporation of Pathology Endpoints in Reproductive Toxicity Studies

Author Manuscript Author Manuscript

In the Working Group experience, the routine addition of clinical pathology, organ weights and/or anatomic pathology endpoints are not expected to change the human risk assessment for effects on development and reproduction. Experiences from the authors, and confirmed by the survey results, however, indicate that addition of selected endpoints to confirm anticipated pharmacodynamic effects, maternal and/or target organ toxicity may be useful to the interpretation of the study. In some cases periodic blood samples and/or tissues collected at necropsy could be stored for potential future analysis. However, if unexpected toxicities occur in a DART study, it may be more appropriate to conduct a separate study specifically designed to investigate the unexpected finding. While the guidelines do not require recording of reproductive organ weights (testes, epididymides, and accessory sex glands appropriate to the species) from males in fertility studies, it is a common practice that the Working Group supports since they are accurate, easily recorded, cannot be collected retrospectively and can offer unique insight into the androgen status of animals in the study. Male reproductive tissues should be evaluated histologically especially if not already assessed in mature males in completed general toxicity studies, or if there is antemortem evidence of an effect. It is not deemed useful to perform standard clinical pathology as a default analysis for rodent fertility studies, relying instead on data from repeat dose toxicity studies. For rodent EFD and PPND studies, it is not typical to include maternal or fetal clinical pathology, organ weights or histopathology as a default practice, but this could be considered on a case-by-case basis for the reasons described above. One general exception might be the investigation of target-specific changes in clinical pathology, organ weights or histopathology in dams, fetuses or offspring to confirm dose or pharmacodynamic effects. When NHP studies are terminal, collection of anatomic pathology data should be considered in the study design. Another case-by-case exception might be the addition of maternal clinical pathology in NHP if maternal toxicity is observed or expected, or to confirm relevant pharmacodynamic effects if no toxicity is expected or observed. These integrated considerations are summarized in Table 8 for DART Studies.

4. Pathology Endpoints in Nonclinical Studies Supporting Pediatrics 4.1 Pathology Endpoints in Juvenile Toxicity Studies

Author Manuscript

The FDA Guidance for Industry, Nonclinical Safety Evaluation of Pediatric Drug Products (FDA, 2006) indicates that tissues/organs in the test species should be macroscopically and microscopically evaluated, with particular regard to those undergoing development at the intended time of treatment. Organs to be given emphasis due to ongoing postnatal development include the brain, kidneys, and lungs, and the immune, reproductive, endocrine, skeletal and gastrointestinal systems. This guidance also includes several tables with estimated timing for completion of developmental processes, such as completion of nephrogenesis, or events, such as puberty (FDA, 2006). The EMA Committee for Human Medicinal Products (CHMP) Guideline on the Need for Non-Clinical Testing in Juvenile

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Halpern et al.

Page 20

Author Manuscript Author Manuscript

Animals on Human Pharmaceuticals for Pediatric Indications (EMA, 2008) further acknowledges that clinical pathology determinations can also be useful, but they may be limited by the technical feasibility of obtaining adequate samples for analysis, particularly in the case of juvenile rodents. The Japanese MHLW guideline for nonclinical safety studies in juvenile animals for pediatric drugs stipulates that study endpoints should be selected with reference to toxicity target organs in mature animals, and with the objective of adequate evaluation of effects on the development of organs/functions, although no specific endpoints are described (MHLW, 2012). All of these guidance documents suggest that studies in juvenile animals should be conducted infrequently, and a recent review suggests that the percent of pediatric investigation plans (PIP) supported by a juvenile toxicity study has remained fairly stable (Hurtt and Engel, 2015). However, requests for and conduct of these studies is increasing with the increasing number of pediatric programs overall, and, at present, there is no consistent message regarding the value of the data generated in these studies for improving the design, conduct and interpretation of safety data from subsequent clinical studies in children (Rose, 2011, Rose, 2014, Leconte et al., 2011, Anderson et al., 2009, Bailey and Marien, 2009, Bailey and Marien, 2011). In response to this, the ICH has endorsed the development of a new harmonized guideline for nonclinical safety assessment to support pediatric drug development (ICH S11).

Author Manuscript Author Manuscript

Studies in juvenile animals of any species can be quite complex and logistically challenging (Barrow et al., 2011, Cappon et al., 2009). Each endpoint may require a separate cohort of animals, so these studies may involve a large number of animals. Furthermore, each species grows and develops at a different rate, and it may be difficult to discriminate between a cumulative toxicity and one due to exposure during a particular developmental window of susceptibility. Although there are general metrics for age multiples in the development of the central nervous system (CNS) and reproductive systems, these do not necessarily hold up against other organ systems such as the gastrointestinal tract, renal system and pulmonary system (Morford et al., 2004). Toxicity may reflect organ system interdependencies, especially those contributing to absorption, distribution, metabolism and excretion (ADME) of pharmaceuticals (Hines and McCarver, 2002, McCarver and Hines, 2002, Blake et al., 2005). Finally, although historically pre- and peripubertal macaques and dogs have been used for general toxicity testing, the background pathology and normal tissue histology from very young animals in these species are not well described as compared to those from the older animals typically used in toxicity testing. With regard to clinical pathology endpoints, appropriateness for the age of the animal, the timing of collection, collection intervals, and need for fasting should also be considered, as well as the potential need for adapting assays or methods for neonatal and juvenile animals. For example, in a juvenile rat study starting with pups prior to weaning, sufficient blood volume may not be available to include a standard clinical pathology evaluation. In such a case, a limited directed terminal clinical pathology evaluation may be sufficient for appropriate risk assessment. If longitudinal clinical pathology assessment is desired, the use of satellite animals may be warranted to avoid compromising the normal growth physiology of the main study animals. Furthermore, there have been few publications and little direct experience to drive specific recommendations regarding the routine inclusion of clinical pathology endpoints in juvenile animals with the objective of consistently providing interpretable results.

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Halpern et al.

Page 21

Author Manuscript

A review of survey responses for inclusion of general toxicity pathology endpoints in juvenile toxicity studies revealed, to some extent, a lack of experience in these studies and/or willingness to comment specifically. The number of survey responses for adding general toxicity endpoints to juvenile toxicity studies ranged from 23 to 38 of the approximately 100 total participating organizations, with several comments indicating a lack of specific experience with these studies. However, for the organizations that did respond, the responses were fairly consistent. For all species, when dedicated juvenile toxicity studies are conducted, there is a general willingness to consider inclusion of pathology endpoints as either a routine process or based on pharmacology/target or findings. The specific distribution of responses is indicated in Table 6.

Author Manuscript Author Manuscript

For both histopathology and clinical pathology endpoints, there was some concern by respondents regarding a relative lack of historical control data in juvenile animals. For anatomic pathology, of 40 responses, 31 (78%) indicated no negative impact, while 9 (23%) responded that, yes, the challenges of interpreting anatomic pathology in fetuses/pups/ infants had a negative impact. For clinical pathology endpoints, with 34 responses, most indicated that a lack of historical data did not prevent interpretation of clinical pathology endpoints (82%), and had no negative impact on studies (77%). However, the remaining responses indicated that interpretation was compromised (18%), or studies negatively impacted (23%); therefore this remains an area where additional experience or education could be helpful. However, as with results from the DART study queries, the comments reflected a general willingness to accept this uncertainty, at least when there are available data from concurrent control animals. In addition, for rodent studies the first scheduled necropsy often occurs when the rats and mice are at least 6-8 weeks of age. Although they will not have all reached full sexual maturity at this time, it is an age range for which relevant historical data for both anatomic and clinical pathology exists and can provide additional context for interpretation (Picut et al., 2015a, Picut et al., 2014, Picut et al., 2015b, Campion et al., 2013).

Author Manuscript

In the Working Group's experience, one important ‘exception’ occurs when pathology endpoints are collected from individual animals at unscheduled intervals. In these situations, the lack of concurrent and historical control data may well preclude a confident interpretation in any species. Reference data for juvenile animals are available, and can help provide context for more common excursions from normal adult profiles, but may not cover the full spectrum of variability seen during development (Jordan et al., 2014). This is particularly challenging for NHP and dogs where small group sizes and high variability for many endpoints can make interpretation of specific test item-related effects difficult. In rodents, one study design option is to include an additional small group of pups to be kept in the same environment and euthanized only if needed as an age-matched tissue control for unscheduled necropsies during the study. Survey respondents also indicated that some general toxicity study endpoints (such as calcium, glucose and cholesterol) are being used to understand potential pharmacodynamic effects, especially for metabolically active compounds. Although this question was not only specific to studies in juvenile animals, such studies would be included in the question. The majority (63%) of respondents would include these endpoints routinely or ‘for cause’, with

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Halpern et al.

Page 22

Author Manuscript

the remaining 37% indicating that they do not add such endpoints. There were 20 comments for this question, likely directly related to the survey request to explain case-by-case use. Of the 20 comments, 18 indicated a positive experience, and 2 indicated a lack of experience but willingness to consider if appropriate. No comments described a negative experience due to inclusion of pharmacodynamic endpoints. 4.2 Pediatric Relevant Pathology Endpoints in General Toxicity Studies Most general toxicity studies use animals intended to support clinical studies with adult human healthy volunteers or patients. However, these animals are often relatively young and still in an active growth phase. When studies include skeletally or sexually immature animals, pharmacologic effects on general growth and bone elongation may be detected (Stahlmann, 2003, Ryan et al., 1999, Hall and Vallera, 2006).

Author Manuscript

Based on the age of the animals at study start for rodent general toxicity studies, most are expected to be developmentally mature prior to initiation of the dosing phase. It is recognized that it can be difficult or impossible to determine test item-related effects on reproductive tissues, especially in males, prior to maturity. By 10 weeks of age, all rats should be sexually mature with complete spermatogenesis in males and regular estrous cycles in females. However, the long bone physes remain open and general growth continues for several months (Zoetis et al., 2003). Thus, assessments of body weight gain during the in-life phase, and histologic assessment of long bones can be very useful in determining potential general effects on growth and development.

Author Manuscript

If histologic data from general toxicity studies in dogs or NHP will be used to support pediatric use, an assessment of both sexual and skeletal maturity should be considered for all animals on the study. This would be intended as a part of characterization of the test system, but not as representing a pathology ‘finding’. Although assessment of skeletal maturity was not specifically queried with this survey, it is generally feasible, requiring only standard sections of long bone (Kilborn et al., 2002). The Working Group also refers readers to the discussion of sexual maturity presented previously in this document. 4.3 Pediatric Relevant Pathology Endpoints in Developmental and Reproductive Toxicity Studies

Author Manuscript

For the most part, data from fertility and embryofetal development studies are not intended or used to directly support pre-pubertal pediatric patients, but these studies may generally contribute to the identification of developmentally sensitive targets. In contrast, the late gestation or early lactation exposures of PPND studies may provide a ‘worst case’ scenario for potential effects in the youngest patients. These studies may be particularly useful in supporting pediatrics with molecules that have a long half-life. It is also critical to understand placental and lactation transport, and exposure in the offspring following dose administration to pregnant dams. The survey results for PPND studies are presented in Table 5 and discussed in detail in the DART section. Responses and comments indicated no dominant position for inclusion of pathology endpoints for offspring, and acknowledged a general lack of experience with dog PPND studies. In rodents, substantial development, including functional maturation of the Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Halpern et al.

Page 23

Author Manuscript

heart (Hew and Keller, 2003), lungs (Zoetis and Hurtt, 2003b), digestive tract (Walthall et al., 2005), kidneys (Zoetis and Hurtt, 2003a), and nervous system (Watson et al., 2006, Wood et al., 2003) occurs in the immediate postnatal/preweaning period. Therefore, exposing the rodent fetus during gestation is not truly reflective of perinatal dosing in humans. In addition to gestational exposure, drugs that are present in milk can result in postnatal exposure of offspring. In fact, due to limited proteolytic digestion for the first 2 weeks postnatally in rats, oral delivery through milk leading to systemic exposure is possible even for some parenteral drugs such as protein therapeutics (Cooper et al., 2014, Halliday, 1955, Mackenzie et al., 1983). Thus, although inclusion of pathology of rodent pups in a PPND study has not been a standard endpoint, it has been used under limited circumstances (Spence et al., 1995) and could be considered where appropriate, possibly including testing of drugs in support of potential use in premature or neonatal infants.

Author Manuscript

Compared to rodents, dogs, and humans, NHPs are developmentally and behaviorally precocious at birth, and some prenatal exposure may be appropriate to mimic the peri/ neonatal period in humans. For example, the lungs (Zoetis and Hurtt, 2003b), digestive tract (Walthall et al., 2005), kidneys (Zoetis and Hurtt, 2003a), and bone (Zoetis et al., 2003, Hew and Keller, 2003) are relatively well-developed at birth, and the functional abilities of the CNS are more advanced than humans at birth (Wood et al., 2003). For products such as Fccontaining biotherapeutics, which are actively transported across the placenta in late gestation and have a long half-life, exposure in the offspring may be substantial and sustained well into the postnatal period (Moffat et al., 2014). Although there is some risk of biological adaptation in utero, a general understanding of pharmacologic and toxicologic effects on offspring after dosing dams during pregnancy can be useful for pediatric risk assessment.

Author Manuscript

Although both standard DART studies and dedicated juvenile toxicity studies evaluate potential effects on development, the former is based on maternal administration and the latter utilizes direct administration postnatally. Because juvenile toxicity studies are intended to support pharmaceutical use, standard pathology endpoints, typical of a general toxicity study, are commonly included. In studies with maternal administration, the focus is more on pregnancy outcomes and general growth and development to inform the human reproductive risk assessment. Depending on the scope of the nonclinical testing program for a molecule, the inclusion of anatomic pathology endpoints for offspring in a PPND study to address potential developmental impact can be critical if data from the study are also intended to support pediatric use (Morford et al., 2011).

Author Manuscript

4.4 Summary of Working Group Considerations for Incorporation of Pathology Endpoints in Nonclinical Studies Supporting Pediatrics In summary, information from the general toxicology program and/or from the DART testing can avoid unnecessarily duplicative studies in juvenile animals to support pediatrics. There are also technical and feasibility challenges in designing studies to support drugs intended for use in neonates and infants. As with many aspects of drug development, understanding the species-specific underlying developmental physiology is critical to accomplish this aim. The current developmental toxicity literature often focuses on

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Halpern et al.

Page 24

Author Manuscript

functional processes, but these may not result in developmentally critical stages recognized by histologic endpoints from animals of different ages. The nonclinical assessment of juvenile toxicity may consider information from many sources: the clinical toxicity program in adults (if available), the mechanism of action, the nonclinical toxicity profile, and developmental signals from the combined human and animal experience in juveniles. The features of each individual program will dictate areas of specific concern, and may require additional investigations or endpoints. Examples might be immunologic function assessments or expanded neuropathology, with the caveat that there will still be individual animal variability and potentially limited historical control data. In this setting, caution should be used in interpreting individual endpoints in isolation. These integrated Working Group considerations are summarized in Table 7 for Juvenile Toxicity Studies, and in Table 8 for DART Studies.

Author Manuscript

5. Conclusions

Author Manuscript

The SRPC of the STP established a Working Group to review the regulatory context and practices for pathology endpoints in reproductive and juvenile toxicity studies, and reproductive endpoints in general toxicity studies. The Working Group performed a survey of pathologists and reproductive toxicologists to understand the range of current practices. The results of the survey, collective expertise and available literature and published guidance documents were used to provide considerations regarding reproductive toxicology and pathology. These are described in the respective sections, and are summarized in Table 7 for reproductive endpoints added to general and juvenile toxicity studies, and in Table 8 for pathology endpoints added to DART studies. In summary, the standard endpoints in general, reproductive, and developmental toxicity studies are aligned with regulatory expectations for the evaluation of reproductive or juvenile toxicity, while additional nonstandard endpoints can be included when scientifically warranted and may serve to enable earlier identification or clarification of reproductive toxicities.

Acknowledgements The authors thank the contributors to the survey responses received by the STP to understand current practices in utilizing reproductive endpoints in toxicity studies, and pathology endpoints in developmental and reproductive toxicity studies. The authors also thank the SRPC, the Reproductive Pathology Interest Group of the STP, the Regulatory Affairs Committee of the American College of Veterinary Clinical Pathologists, and Drs. Darlene Dixon, Justin Vidal, Mark Cline and Robert Chapin for their detailed technical review of this manuscript.

References Author Manuscript

Anderson T, Khan NK, Tassinari MS, Hurtt ME. Comparative juvenile safety testing of new therapeutic candidates: relevance of laboratory animal data to children. J Toxicol Sci. 2009; 34(Suppl 2):SP209–215. [PubMed: 19571471] Andersson H, Rehm S, Stanislaus D, Wood CE. Scientific and regulatory policy committee (SRPC) paper: assessment of circulating hormones in nonclinical toxicity studies III. female reproductive hormones. Toxicologic pathology. 2013; 41:921–934. [PubMed: 23334695] Attia MA. Cyclic changes in genital organs and vaginal cytology in cynomolgus monkeys (Macaca fascicularis). Dtsch Tierarztl Wochenschr. 1998; 105:399–404. [PubMed: 9857561] Auyeung-Kim DJ, Devalaraja MN, Migone TS, Cai W, Chellman GJ. Developmental and peripostnatal study in cynomolgus monkeys with belimumab, a monoclonal antibody directed against B-lymphocyte stimulator. Reproductive toxicology. 2009; 28:443–455. [PubMed: 19631735] Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Halpern et al.

Page 25

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Bailey GP, Marien D. What have we learned from pre-clinical juvenile toxicity studies? Reproductive toxicology. 2009; 28:226–229. [PubMed: 19446432] Bailey GP, Marien D. The value of juvenile animal studies “What have we learned from preclinical juvenile toxicity studies? II”. Birth defects research. Part B, Developmental and reproductive toxicology. 2011; 92:273–291. Barrow PC, Barbellion S, Stadler J. Preclinical evaluation of juvenile toxicity. Methods in molecular biology. 2011; 691:17–35. [PubMed: 20972745] Beckman DA, Feuston M. Landmarks in the development of the female reproductive system. Birth defects research. Part B, Developmental and reproductive toxicology. 2003; 68:137–143. Blake MJ, Castro L, Leeder JS, Kearns GL. Ontogeny of drug metabolizing enzymes in the neonate. Seminars in fetal & neonatal medicine. 2005; 10:123–138. [PubMed: 15701578] Bowen WH, Koch G. Determination of age in monkeys (Macaca irus) on the basis of dental development. Lab Anim. 1970; 4:113–124. [PubMed: 5005918] Bregman CL, Adler RR, Morton DG, Regan KS, Yano BL, Society of Toxicologic P. Recommended tissue list for histopathologic examination in repeat-dose toxicity and carcinogenicity studies: a proposal of the Society of Toxicologic Pathology (STP). Toxicologic pathology. 2003; 31:252– 253. [PubMed: 12696587] Buse E, Habermann G, Osterburg I, Korte R, Weinbauer GF. Reproductive/developmental toxicity and immunotoxicity assessment in the nonhuman primate model. Toxicology. 2003; 185:221–227. [PubMed: 12581697] Bussiere JL, Moffat G, Zhou L, Tarlo KS. Assessment of menstrual cycle length in cynomolgus monkeys as a female fertility endpoint of a biopharmaceutical in a 6 month toxicity study. Regulatory toxicology and pharmacology : RTP. 2013; 66:269–278. [PubMed: 23680404] Campion SN, Carvallo FR, Chapin RE, Nowland WS, Beauchamp D, Jamon R, Koitz R, Winton TR, Cappon GD, Hurtt ME. Comparative assessment of the timing of sexual maturation in male Wistar Han and Sprague-Dawley rats. Reproductive toxicology. 2013; 38:16–24. [PubMed: 23434729] Cappon GD, Bailey GP, Buschmann J, Feuston MH, Fisher JE, Hew KW, Hoberman AM, Ooshima Y, Stump DG, Hurtt ME. Juvenile animal toxicity study designs to support pediatric drug development. Birth defects research. Part B, Developmental and reproductive toxicology. 2009; 86:463–469. Cappon GD, Potter D, Hurtt ME, Weinbauer GF, Luetjens CM, Bowman CJ. Sensitivity of male reproductive endpoints in nonhuman primate toxicity studies: a statistical power analysis. Reproductive toxicology. 2013; 41:67–72. [PubMed: 23792114] Cardy RH. Sexual dimorphism of the normal rat mammary gland. Veterinary pathology. 1991; 28:139– 145. [PubMed: 1676552] Center for Food Safety and Applied Nutrition (CFSAN), U.S. Food and Drug Administration, Redbook 2000. [12 Feb 2015] Guidance for Industry and Other Stakeholders.. Toxicologic Principles for the Safety of Food Ingredients. Jul. 2007 Revised July 2007. http://www.fda.gov/ downloads/Food/GuidanceRegulation/UCM222779.pdf Chandra SA, Cline JM, Adler RR. Cyclic morphological changes in the beagle mammary gland. Toxicologic pathology. 2010; 38:969–983. [PubMed: 20651098] Chapin RE, Creasy DM. Assessment of circulating hormones in regulatory toxicity studies II. Male reproductive hormones. Toxicologic pathology. 2012; 40:1063–1078. [PubMed: 22552397] Cline JM, Wood CE. The Mammary Glands of Macaques. Toxicologic pathology. 2008; 36:134s–141s. [PubMed: 21475638] Cline JM, Wood CE, Vidal JD, Tarara RP, Buse E, Weinbauer GF, de Rijk EP, van Esch E. Selected Background Findings and Interpretation of Common Lesions in the Female Reproductive System in Macaques. Toxicologic pathology. 2008; 36:142s–163s. [PubMed: 21475639] Cooper PR, Kliwinski CM, Perkinson RA, Ragwan E, Mabus JR, Powers GD, Dorai H, Giles- Komar J, Hornby PJ. The contribution of cell surface FcRn in monoclonal antibody serum uptake from the intestine in suckling rat pups. Frontiers in pharmacology. 2014; 5:225. [PubMed: 25339905] Creasy DM. Evaluation of testicular toxicity in safety evaluation studies: the appropriate use of spermatogenic staging. Toxicologic pathology. 1997; 25:119–131. [PubMed: 9125770]

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Halpern et al.

Page 26

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Daurio CP, Gilman MR, Pulliam JD, Seward RL. Reproductive evaluation of male beagles and the safety of ivermectin. American journal of veterinary research. 1987; 48:1755–1760. [PubMed: 3434921] Dixon D, Alison R, Bach U, Colman K, Foley GL, Harleman JH, Haworth R, Herbert R, Heuser A, Long G, Mirsky M, Regan K, Van Esch E, Westwood FR, Vidal J, Yoshida M. Nonproliferative and proliferative lesions of the rat and mouse female reproductive system. Journal of toxicologic pathology. 2014; 27:1s–107s. [PubMed: 25516636] European Medicines Agency, Committee for Medicinal Products for Human Use. [09 July 2015] Guideline on the need for non-clinical testing in juvenile animals of pharmaceuticals for paediatric indications. 2008. http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/ 2009/09/WC50000330 5.pdf Everds NE, Snyder PW, Bailey KL, Bolon B, Creasy DM, Foley GL, Rosol TJ, Sellers T. Interpreting stress responses during routine toxicity studies: a review of the biology, impact, and assessment. Toxicologic pathology. 2013; 41:560–614. [PubMed: 23475558] Goedken MJ, Kerlin RL, Morton D. Spontaneous and age-related testicular findings in beagle dogs. Toxicologic pathology. 2008; 36:465–471. [PubMed: 18467685] Hall WA, Vallera DA. Efficacy of antiangiogenic targeted toxins against glioblastoma multiforme. Neurosurgical focus. 2006; 20:E23. [PubMed: 16709029] Halliday R. The Absorption of Antibodies from Immune Sera by the Gut of the Young Rat. Proceedings of the Royal Society B: Biological Sciences. 1955; 143:408–413. Harleman, JH.; Foley, GL. Reproductive system In Pathobiology of the aging dog. Mohr, U., et al., editors. Vol. 1. Iowa State University Press; Ames IA.: 2001. p. 293-309. Haruyama E, Suda M, Ayukawa Y, Kamura K, Mizutamari M, Ooshima Y, Tanimoto A. Testicular development in cynomolgus monkeys. Toxicologic pathology. 2012; 40:935–942. [PubMed: 22609951] Hew KW, Keller KA. Postnatal anatomical and functional development of the heart: a species comparison. Birth defects research. Part B, Developmental and reproductive toxicology. 2003; 68:309–320. Hines RN, McCarver DG. The ontogeny of human drug-metabolizing enzymes: phase I oxidative enzymes. The Journal of pharmacology and experimental therapeutics. 2002; 300:355–360. [PubMed: 11805191] Holson JF, Stump DG, Pearce LB, Watson RE, DeSesso JM. Absence of developmental toxicity in a canine model after infusion of a hemoglobin-based oxygen carrier: Implications for risk assessment. Reproductive toxicology. 2015; 52:101–107. [PubMed: 25697570] Honda T, Honda K, Kokubun C, Nishimura T, Hasegawa M, Nishida A, Inui T, Kitamura K. Timecourse changes of hematology and clinical chemistry values in pregnant rats. J Toxicol Sci. 2008; 33:375–380. [PubMed: 18670170] Hurtt ME, Engel S. An update of juvenile animal studies in the European Union: What do the numbers say? Reproductive toxicology. 2015; 56:105–108. [PubMed: 25937597] International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use (ICH; 2005). [09 July 2015] Detection of toxicity to reproduction for medicinal products & toxicity to male fertility S5(R2). http://www.ich.org/fileadmin/ Public_Web_Site/ICH_Products/Guidelines/Safety/S5/Step4/S5_R2__Guideline.pdf ICH.. [09 July 2015] Guidance on nonclinical safety studies for the conduct of human clinical trials and marketing authorization for pharmaceuticals M3(R2). 2009. http://www.ich.org/fileadmin/ Public_Web_Site/ICH_Products/Guidelines/Multidisciplinary/M3_R2/Step4/ M3_R2__Guideline.pdf ICH.. [09 July 2015] Nonclinical evaluation for anticancer pharmaceuticals S9. 2009. http:// www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guidelines/Safety/S9/Step4/ S9_Step4_Guideline.pdf ICH.. [29 Oct 2014] Preclinical safety evaluation of biotechnology-derived pharmaceuticals S6(R1). 2011. http://www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guidelines/Safety/S6_R1/ Step4/S6_R1_Guideline.pdf

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Halpern et al.

Page 27

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Ishii M, Yamauchi T, Matsumoto K, Watanabe G, Taya K, Chatani F. Maternal age and reproductive function in female Sprague-Dawley rats. J Toxicol Sci. 2012; 37:631–638. [PubMed: 22688002] Japanese Ministry of Health Labor and Welfare (MHLW). Guideline on the Nonclinical Safety Study in Juvenile Animals for Pediatric Drugs. 2012 Jordan HL, Register TC, Tripathi NK, Bolliger AP, Everds N, Zelmanovic D, Poitout F, Bounous DI, Wescott D, Ramaiah SK. Nontraditional applications in clinical pathology. Toxicologic pathology. 2014; 42:1058–1068. [PubMed: 24705882] Keane KA, Parker GA, Regan KS, Picut C, Dixon D, Creasy D, Giri D, Hukkanen RR. Scientific and Regulatory Policy Committee (SRPC) Points to Consider : Histopathology Evaluation of the Pubertal Development and Thyroid Function Assay (OPPTS 890.1450, OPPTS 890.1500) in Rats to Screen for Endocrine Disruptors. Toxicologic pathology. 2015; 43:1047–1063. [PubMed: 25948506] Kilborn SH, Trudel G, Uhthoff H. Review of growth plate closure compared with age at sexual maturity and lifespan in laboratory animals. Contemp Top Lab Anim Sci. 2002; 41:21–26. [PubMed: 12213043] Lanning LL, Creasy DM, Chapin RE, Mann PC, Barlow NJ, Regan KS, Goodman DG. Recommended approaches for the evaluation of testicular and epididymal toxicity. Toxicologic pathology. 2002; 30:507–520. [PubMed: 12187942] Leconte I, Bailey G, Davis-Bruno K, Hew KW, Kim J, Silva Lima B, Liminga U, Moffit J, De Schaepdrijver L, Schmitt G, Tassinari M, Thompson K, Hurtt M. Value of juvenile animal studies. Birth defects research. Part B, Developmental and reproductive toxicology. 2011; 92:292–303. LeFevre J, McClintock MK. Reproductive senescence in female rats: a longitudinal study of individual differences in estrous cycles and behavior. Biology of reproduction. 1988; 38:780–789. [PubMed: 3401536] Lucas JN, Rudmann DG, Credille KM, Irizarry AR, Peter A, Snyder PW. The rat mammary gland: morphologic changes as an indicator of systemic hormonal perturbations induced by xenobiotics. Toxicologic pathology. 2007; 35:199–207. [PubMed: 17366314] Luetjens CM, Weinbauer GF. Functional assessment of sexual maturity in male macaques (Macaca fascicularis). Regulatory toxicology and pharmacology : RTP. 2012; 63:391–400. [PubMed: 22579626] Mackenzie NM, Morris B, Morris R. The binding of proteins to isolated enterocytes from the small intestine of the neonatal rat. Immunology. 1983; 48:489–496. [PubMed: 6219063] Martin PL, Oneda S, Treacy G. Effects of an anti-TNF-alpha monoclonal antibody, administered throughout pregnancy and lactation, on the development of the macaque immune system. American journal of reproductive immunology. 2007; 58:138–149. [PubMed: 17631007] Martin PL, Weinbauer GF. Developmental toxicity testing of biopharmaceuticals in nonhuman primates: previous experience and future directions. International journal of toxicology. 2010; 29:552–568. [PubMed: 20926830] Marty MS, Johnson KA, Carney EW. Effect of feed restriction on Hershberger and pubertal male assay endpoints. Birth defects research. Part B, Developmental and reproductive toxicology. 2003; 68:363–374. McCarver DG, Hines RN. The ontogeny of human drug-metabolizing enzymes: phase II conjugation enzymes and regulatory mechanisms. The Journal of pharmacology and experimental therapeutics. 2002; 300:361–366. [PubMed: 11805192] Mitchard T, Jarvis P, Stewart J. Assessment of male rodent fertility in general toxicology 6- month studies. Birth defects research. Part B, Developmental and reproductive toxicology. 2012; 95:410– 420. Moffat GJ, Retter MW, Kwon G, Loomis M, Hock MB, Hall C, Bussiere J, Lewis EM, Chellman GJ. Placental transfer of a fully human IgG2 monoclonal antibody in the cynomolgus monkey, rat, and rabbit: a comparative assessment from during organogenesis to late gestation. Birth defects research. Part B, Developmental and reproductive toxicology. 2014; 101:178–188. Morford LL, Bowman CJ, Blanset DL, Bogh IB, Chellman GJ, Halpern WG, Weinbauer GF, Coogan TP. Preclinical safety evaluations supporting pediatric drug development with biopharmaceuticals:

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Halpern et al.

Page 28

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

strategy, challenges, current practices. Birth defects research. Part B, Developmental and reproductive toxicology. 2011; 92:359–380. Morford LL, Henck JW, Breslin WJ, DeSesso JM. Hazard identification and predictability of children's health risk from animal data. Environmental health perspectives. 2004; 112:266–271. [PubMed: 14754582] Niehoff MO, Bergmann M, Weinbauer GF. Effects of social housing of sexually mature male cynomolgus monkeys during general and reproductive toxicity evaluation. Reproductive toxicology. 2010; 29:57–67. [PubMed: 19808089] O'Connor JC, Davis LG, Frame SR, Cook JC. Evaluation of a Tier I screening battery for detecting endocrine-active compounds (EACs) using the positive controls testosterone, coumestrol, progesterone, and RU486. Toxicological sciences : an official journal of the Society of Toxicology. 2000; 54:338–354. [PubMed: 10774816] Organisation for Economic Co-operation and Development (OECD). [21 Dec 2015] Guidelines for the Testing of Chemicals, Section 4, Health Effects. Test No. 443: Extended one-generation reproductive toxicity study. 2011. http://www.oecd-ilibrary.org/environment/test-no-443-extendedone-generation-reproductive-toxicity-study_9789264122550-en Picut CA, Dixon D, Simons ML, Stump DG, Parker GA, Remick AK. Postnatal ovary development in the rat: morphologic study and correlation of morphology to neuroendocrine parameters. Toxicologic pathology. 2015a; 43:343–353. [PubMed: 25107574] Picut CA, Remick AK, Asakawa MG, Simons ML, Parker GA. Histologic features of prepubertal and pubertal reproductive development in female Sprague-Dawley rats. Toxicologic pathology. 2014; 42:403–413. [PubMed: 23599412] Picut CA, Remick AK, de Rijk EP, Simons ML, Stump DG, Parker GA. Postnatal development of the testis in the rat: morphologic study and correlation of morphology to neuroendocrine parameters. Toxicologic pathology. 2015b; 43:326–342. [PubMed: 25217330] Powles-Glover N, Mitchard T, Stewart J. Time Course for Onset and Recovery from Effects of a Novel Male Reproductive Toxicant: Implications for Apical Preclinical Study Designs. Birth defects research. Part B, Developmental and reproductive toxicology. 2015; 104:91–99. Regan KS, Cline JM, Creasy D, Davis B, Foley GL, Lanning L, Latendresse JR, Makris S, Morton D, Rehm S, Stebbins K, Group, S.T.P.O.E.W. STP position paper: ovarian follicular counting in the assessment of rodent reproductive toxicity. Toxicologic pathology. 2005; 33:409–412. [PubMed: 15805082] Rehm S. Spontaneous testicular lesions in purpose-bred beagle dogs. Toxicologic pathology. 2000; 28:782–787. [PubMed: 11127291] Rehm S, Solleveld HA, Portelli ST, Wier PJ. Histologic changes in ovary, uterus, vagina, and mammary gland of mature beagle dogs treated with the SERM idoxifene. Birth defects research. Part B, Developmental and reproductive toxicology. 2007a; 80:225–232. [PubMed: 17570126] Rehm S, Stanislaus DJ, Williams AM. Estrous cycle-dependent histology and review of sex steroid receptor expression in dog reproductive tissues and mammary gland and associated hormone levels. Birth defects research. Part B, Developmental and reproductive toxicology. 2007b; 80:233– 245. Rose K. The value of juvenile animal studies: a pediatric clinical perspective. Birth defects research. Part B, Developmental and reproductive toxicology. 2011; 92:252–253. Rose K. European Union pediatric legislation jeopardizes worldwide, timely future advances in the care of children with cancer. Clinical therapeutics. 2014; 36:163–177. [PubMed: 24529288] Russo, IH.; Twari, M.; Russo, J. Morphology and development of the rat mammary gland In Monographs on pathology of laboratory animals: Integument and mammary glands. Jones, TC., et al., editors. Springer Verlag; Berlin: 1989. Ryan AM, Eppler DB, Hagler KE, Bruner RH, Thomford PJ, Hall RL, Shopp GM, O'Neill CA. Preclinical safety evaluation of rhuMAbVEGF, an antiangiogenic humanized monoclonal antibody. Toxicologic pathology. 1999; 27:78–86. [PubMed: 10367678] Sanbuissho A, Yoshida M, Hisada S, Sagami F, Kudo S, Kumazawa T, Ube M, Komatsu S, Ohno Y. Collaborative work on evaluation of ovarian toxicity by repeated-dose and fertility studies in female rats. J Toxicol Sci. 2009; 34(Suppl 1):SP1–22. [PubMed: 19265275]

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Halpern et al.

Page 29

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Sato J, Doi T, Kanno T, Wako Y, Tsuchitani M, Narama I. Histopathology of incidental findings in cynomolgus monkeys ( macaca fascicularis ) used in toxicity studies. Journal of toxicologic pathology. 2012a; 25:63–101. [PubMed: 22481861] Sato J, Doi T, Wako Y, Hamamura M, Kanno T, Tsuchitani M, Narama I. Histopathology of incidental findings in beagles used in toxicity studies. Journal of toxicologic pathology. 2012b; 25:103–134. [PubMed: 22481862] Schedin P, Mitrenga T, Kaeck M. Estrous cycle regulation of mammary epithelial cell proliferation, differentiation, and death in the Sprague-Dawley rat: a model for investigating the role of estrous cycling in mammary carcinogenesis. Journal of mammary gland biology and neoplasia. 2000; 5:211–225. [PubMed: 11149574] Sellers RS, Morton D, Michael B, Roome N, Johnson JK, Yano BL, Perry R, Schafer K. Society of Toxicologic Pathology position paper: organ weight recommendations for toxicology studies. Toxicologic pathology. 2007; 35:751–755. [PubMed: 17849358] Smith BH, Crummett TL, Brandt KL. Ages of eruption of primate teeth: A compendium for aging individuals and comparing life histories. Yearbook of Physical Anthropology. 1994; 37:177–231. Sone K, Yamamoto-Sawamura T, Kuwahara S, Nishijima K, Ohno T, Aoyama H, Tanaka S. Changes of estrous cycles with aging in female F344/n rats. Experimental animals / Japanese Association for Laboratory Animal Science. 2007; 56:139–148. [PubMed: 17460359] Spence SG, Allen HL, Cukierski MA, Manson JM, Robertson RT, Eydelloth RS. Defining the susceptible period of developmental toxicity for the AT1-selective angiotensin II receptor antagonist losartan in rats. Teratology. 1995; 51:367–382. [PubMed: 7502237] Stahlmann R. Children as a special population at risk--quinolones as an example for xenobiotics exhibiting skeletal toxicity. Archives of toxicology. 2003; 77:7–11. [PubMed: 12491034] Stanislaus D, Andersson H, Chapin R, Creasy D, Ferguson D, Gilbert M, Rosol TJ, Boyce RW, Wood CE. Society of toxicologic pathology position paper: review series: assessment of circulating hormones in nonclinical toxicity studies: general concepts and considerations. Toxicologic pathology. 2012; 40:943–950. [PubMed: 22569585] Stute P, Wood CE, Kaplan JR, Cline JM. Cyclic changes in the mammary gland of cynomolgus macaques. Fertility and sterility. 2004; 82(Suppl 3):1160–1170. [PubMed: 15474090] Thuilliez C, Tortereau A, Perron-Lepage MF, Howroyd P, Gauthier B. Spontaneous testicular tubular hypoplasia/atrophy in the Gottingen minipig: a retrospective study. Toxicologic pathology. 2014; 42:1024–1031. [PubMed: 24292387] U.S. Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research (CDER). [09 July 2015] Guidance for industry: Nonclinical safety evaluation of pediatric drug products. 2006. http://www.fda.gov/downloads/drugs/ guidancecomplianceregulatoryinformation/guidances/ucm079247.pdf U.S. Environmental Protection Agency, OPPTS (Office of Prevention, Pesticides and Toxic Substances). [12 Feb 2015] Health effects test guidelines: Nos. 870.3100 (90-Day Oral Toxicity in Rodents), 870.3150 (90-Day Oral Toxicity in Nonrodents), 870.4100 (Chronic Toxicity), 870.4200 (Carcinogenicity), 870.4300 (Combined Chronic Toxicity/Carcinogenicity). 1998. http:// www.epa.gov/ocspp/pubs/frs/publications/Test_Guidelines/series870.html Vaidyanathan A, McKeever K, Anand B, Eppler S, Weinbauer GF, Beyer JC. Developmental immunotoxicology assessment of rituximab in cynomolgus monkeys. Toxicological sciences : an official journal of the Society of Toxicology. 2011; 119:116–125. [PubMed: 20937725] van Esch E, Cline JM, Buse E, Weinbauer GF. The Macaque Endometrium, with Special Reference to the Cynomolgus Monkey (Macaca fascicularis). Toxicologic pathology. 2008; 36:67S–100S. Vogel, F. How to design male fertility investigations in the cynomolgus monkey In Towards New Horizons in Primate Toxicology. Korte, R.; Weinbauer, G., editors. Waxmann Verlag GmbH; Munster: 2000. p. 43-51. Walthall K, Cappon GD, Hurtt ME, Zoetis T. Postnatal development of the gastrointestinal system: a species comparison. Birth defects research. Part B, Developmental and reproductive toxicology. 2005; 74:132–156.

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Halpern et al.

Page 30

Author Manuscript Author Manuscript

Watson RE, Desesso JM, Hurtt ME, Cappon GD. Postnatal growth and morphological development of the brain: a species comparison. Birth defects research. Part B, Developmental and reproductive toxicology. 2006; 77:471–484. [PubMed: 17066419] Weinbauer, G.; Bowman, CJ.; Halpern, WG.; Chellman, GJ. Developmental and Reproductive Toxicity Testing In The Nonhuman Primate in Nonclinical Drug Development and Safety Assessment. Bluemel, J., et al., editors. Elsevier, Academic Press; San Diego: 2015. p. 471-499. Weinbauer GF, Niehoff M, Niehaus M, Srivastav S, Fuchs A, Van Esch E, Cline JM. Physiology and Endocrinology of the Ovarian Cycle in Macaques. Toxicologic pathology. 2008; 36:7s–23s. [PubMed: 20852722] Westwood FR. The female rat reproductive cycle: a practical histological guide to staging. Toxicologic pathology. 2008; 36:375–384. [PubMed: 18441260] Wood SL, Beyer BK, Cappon GD. Species comparison of postnatal CNS development: functional measures. Birth defects research. Part B, Developmental and reproductive toxicology. 2003; 68:391–407. Youssef AF, Fort FL, Ronsen B, Schroeder R, Williams K, Auletta C. Evaluation of apomorphine HCl effects on reproductive endpoints: studies in male rats and dogs. Toxicological sciences : an official journal of the Society of Toxicology. 1999; 51:273–279. [PubMed: 10543029] Zoetis T, Hurtt ME. Species comparison of anatomical and functional renal development. Birth defects research. Part B, Developmental and reproductive toxicology. 2003a; 68:111–120. Zoetis T, Hurtt ME. Species comparison of lung development. Birth defects research. Part B, Developmental and reproductive toxicology. 2003b; 68:121–124. Zoetis T, Tassinari MS, Bagi C, Walthall K, Hurtt ME. Species comparison of postnatal bone growth and development. Birth defects research. Part B, Developmental and reproductive toxicology. 2003; 68:86–110.

Author Manuscript Author Manuscript Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Author Manuscript

Author Manuscript

0

0

#

5

%

2

#

12

%

%

6

#

48

22

42

19

46

24

52

24

53

24

42

22

100

46

100

45

100

52

Total

2

1

4

2

4

2

Yes

No

No

a

Maybe

60

28

63

29

62

33

Maybe

38

18

33

15

34

18

100

47

100

46

100

53

Total

6

3

15

7

8

4

Yes

19

9

24

11

17

9

Maybe

75

35

61

28

75

39

No

100

47

100

46

100

52

Total

Testicular volume

6

3

11

5

13

7

Yes

28

13

46

21

60

26

Maybe

66

31

43

20

37

19

No

100

47

100

46

100

52

Total

0

0

0

0

6

3

Yes

6

3

7

3

25

13

Maybe

94

44

93

43

69

36

No

Mating Trial

100

47

100

46

100

52

Total

c Both males and females

Maybe represents both “triggered by previous observations” & “potential consequences of pharmacological target modulation” (combining these was based on feedback from the respondents’ comments)

a

bBold denotes the ‘bucket’ with the greatest percent of responses for the parameter

Dog

NHP

Rodent

Yes

c

Hormones

Sperm Assessment

Cyclicity

Author Manuscript

Survey Responses on the Incorporation of DART Endpoints in General Toxicity Studies

Author Manuscript

Table 1 Halpern et al. Page 31

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Author Manuscript

Author Manuscript

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

96

%

34

72

#

73

%

33

#

71

%

%

37

#

Yes

46

94

#

43

#

%

c

6

3

7

3

8

4

Maybe

21

10

20

9

21

11

No

Pituitary

0

0

2

4

2

4

2

2

1

100

48

100

48

100

53

100

47

100

45

100

52

83

39

83

38

91

48

60

29

54

25

70

37

4

2

9

4

2

1

13

6

9

4

8

4

No

Ovaries

8

4

11

5

8

4

Maybe

Maybe

Yes

Yes

Total

Total

No

31

15

35

16

23

12

100

48

100

46

100

53

100

47

100

46

100

53

50

24

46

21

51

27

81

39

80

36

87

45

4

2

9

4

4

2

46

22

46

21

45

24

No

Uterus

4

2

7

3

4

2

Maybe

15

7

13

6

10

5

100

48

100

45

100

52

Total

100

48

100

46

100

53

Total

No

Prostate

Maybe

Yes

Yes

Total

Total

No

Both males and females

Bold denotes the ‘bucket’ with the greatest percent of responses for the parameter

b

c

0

b

% 1

0

52

#

98

a

Maybe

Epididymides

39

18

49

26

Yes

13

6

13

7

Maybe

48

22

38

20

No

100

46

100

53

Total

Maybe represents both “triggered by previous observations” & “potential consequences of pharmacological target modulation” (combining these was based on feedback by the respondent comments)

a

Dog

NHP

Rodent

Dog

NHP

Rodent

Yes

Testes

Seminal Vesicles

Author Manuscript

Inclusion of Reproductive/Endocrine Organ Weights in General Toxicity Studies

Author Manuscript

Table 2 Halpern et al. Page 32

Halpern et al.

Page 33

Table 3

Author Manuscript

Recording of Sexual Maturity in Microscopic Observations Data for General Toxicity Studies Males

Rodent

Dog

NHP

Females

All Studies

a Some Studies

No Studies

Total

All Studies

Some Studies

No Studies

Total

#

15

12

26

53

14

12

27

53

%

28

23

49

26

23

51

#

28

8

12

18

9

21

%

58

17

25

38

19

44

#

32

6

6

19

10

15

%

71

13

13

43

23

34

48

45

a

48

44

Author Manuscript

‘Some’ includes a composite of categories including ‘specific study types’, ‘based on pharmacology/target’, ‘triggered by previous findings’, and ‘only if the NHP is the only toxicology species used’.

Author Manuscript Author Manuscript Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Halpern et al.

Page 34

Table 4

Author Manuscript

Confidence of Pathology Group in Histologic Evaluation of Sexual Maturity

a

High (majority )

Not high (majority)

Total Responses

a

Sexual Maturity in Males

Sexual Maturity in Females

General Stage of Estrous/Menstrual Cycle in Females

#

44

42

33

%

88

84

67

#

6

8

16

%

12

16

33

#

50

50

49

‘Majority’ indicated most pathologists of the responding organization

Author Manuscript Author Manuscript Author Manuscript Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Author Manuscript

Author Manuscript

Author Manuscript

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

NHP

Rodent

Fetus

NHP

Rodent

Dam

6

0

0

%

3

#

1

%

%

#

4

16

#

5

14

%

Yes

22

% 48

11

57

21

50

11

57

b

21

Maybe

a

30

7

27

10

27

6

27

10

No

Clinical Pathology

36

9

26

9

52

13

60

22

Maybe

64

16

71

25

32

8

27

10

No

23

37

22

37

25

35

25

37

28

7

11

4

16

4

21

8

Yes

Total

Total

Clinical Pathology

5

%

#

6

16

#

5

23

%

16

#

%

#

Yes

#

EFD study

NHP

Rodent

Female

NHP

Rodent

Male

Fertility study

50

12

58

22

58

14

74

29

29

7

21

8

25

6

18

7

Maybe

36

9

26

10

48

12

40

15

Maybe

36

9

63

24

36

9

40

15

No

24

38

24

39

25

38

25

38

12

3

6

2

12

3

19

7

Yes

Total

Total

21

5

21

8

17

4

8

3

No

Organ weights

Yes

Organ weights

26

6

49

18

26

6

47

18

Maybe

52

13

47

17

52

13

60

22

Maybe

36

9

47

17

36

9

22

8

No

23

37

23

38

Total

25

36

25

37

Total

30

7

16

6

26

6

11

4

No

Histopathology

44

10

35

13

48

11

42

16

Yes

Histopathology

Survey Responses about Incorporation of General Toxicity Endpoints on DART Studies

Author Manuscript

Table 5 Halpern et al. Page 35

NHP

Rodent

Pup

NHP

Rodent

Dam

8

32

%

6

%

#

2

27

%

#

7

11

%

#

4

#

32

8

38

13

54

14

56

20

Maybe

36

9

56

19

19

5

33

12

No

25

34

26

36

Total

37

10

18

7

23

6

26

10

Yes

37

10

40

15

31

8

32

12

Maybe

26

7

42

16

46

12

42

16

No

27

38

26

38

Total

32

8

19

7

8

2

17

6

Yes

48

12

53

19

54

14

47

17

Maybe

20

5

28

10

39

10

36

13

No

Histopathology

25

36

26

36

Total

Bold denotes largest bucket for that category

b

Maybe represents both “triggered by previous observations” & “potential consequences of pharmacological target modulation” (combining these was based on feedback from the respondents’ comments)

a

Author Manuscript

Yes

Author Manuscript Organ weights

Author Manuscript

Clinical Pathology

Author Manuscript

PPND study

Halpern et al. Page 36

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Author Manuscript

Author Manuscript

Author Manuscript 15 58

%

63

#

15

%

67

%

#

24

#

Yes

27

7

21

5

25

9

Maybe

a

15

4

17

4

8

3

No

Clinical Pathology

26

24

36

Total

77

20

74

17

82

31

Yes

12

3

13

3

16

6

Maybe

12

3

13

3

3

1

No

Organ weights

26

23

38

Total

73

19

67

16

84

31

Yes

15

4

21

5

14

5

Maybe

12

3

13

3

3

1

No

Histopathology

26

24

37

Total

‘Maybe’ represents both “triggered by previous observations” & “potential consequences of pharmacological target modulation” (combining these was based on feedback by the respondent comment

a

NHP

Dog

Rodent

Juvenile Toxicity Study

Survey Responses about Incorporation of General Toxicity Endpoints in Juvenile Toxicity Studies

Author Manuscript

Table 6 Halpern et al. Page 37

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Author Manuscript

Author Manuscript

Author Manuscript Immature

NHP

Not applicable

Not applicable

Sperm at necropsy and/or hormones for cause once mature; mating trials possible

Postweaning to mature

Immature

Not applicable

Cycling (vaginal smears) for cause; hormones rare

Female

Preweaning

Semen and/or hormones for cause

Male

Rarely assessed

Dog

Rodent

NHP

Semen and/or hormones for cause

Female

Cycling (vaginal smears) and/or hormones for cause

cause

c

Sperm at necropsy and/or hormones for

Fertility Assessments

Male

Female

Male

Subset

Complete; typically after 6-12 months

Complete, typically after 3-6 months

Complete, typically after PND70

Organ weights and clinical pathology rare; targeted histopathology (i.e. immune or nervous systems), typically after 6-8 weeks of age

Complete; record menstrual cycle stage if needed to clarify or interpret results

Complete

Complete; record estrous cycle stage if needed to clarify or interpret results

Complete

Complete; record estrous cycle stage as needed to clarify or interpret results

Complete

Clinical Pathology, Organ Weights, a Macroscopic and Histopathology

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01.

Recording may be limited to reproductive immaturity, with reproductive maturity being the default as long as this is stated in the report

c ‘For cause’ indicates that these would only be added when prior data or pharmacology and mechanism suggest a potential effect

b

Complete refers to clinical pathology, organ weight and anatomic pathology endpoints commonly included in general toxicity studies

a

Juvenile

Rodent

General Toxicity (GenTox)

Dog

Species

Study Type

Yes, both sexes

Yes, both sexes

Yes, both sexes

Yes, both sexes

Yes

Yes

Yes

Yes

Yes

Yes

Record Reproductive Maturity

Working Group integrated assessment of current practices and considerations for the inclusion of reproductive endpoints in general and juvenile toxicity studies for pharmaceutical drug development

Author Manuscript

Table 7

b

Halpern et al. Page 38

Author Manuscript

Author Manuscript

Author Manuscript

Toxicol Pathol. Author manuscript; available in PMC 2017 August 01. NHP

Rodent

NHP

Rodent

a

Targeted Standard/targeted (depending on age and available blood volume)

Offspring

Targeted (not typically done)

Pup Dam

Targeted

Not typically done

Fetus Dam

Targeted (i.e. to help define maternal toxicity)

Not typically done

Fetus Dam

Targeted (i.e. to help define maternal toxicity)

Targeted

Female

Dam

Targeted

Targeted

Targeted

Clinical Pathology

Male

Female

Male

Subset

Targeted

Targeted (not typically done)

Targeted (not typically done)

Targeted (not typically done)

Yes, typically collected

Targeted (gravid uterine and placental weights routinely collected)

Targeted (not typically done)

Targeted (gravid uterine weights routinely collected)

organs

c

Typically limited to reproductive

organs

c

Typically limited to reproductive

Typically limited to reproductive organs

Typically limited to reproductive organs

Organ Weights

Complete evaluation is standardly performed but not driven by guidance

Targeted

Targeted

Targeted (rarely)

No

Targeted (i.e. no other signs of maternal toxicity anticipated)

No

Targeted (i.e. no other signs of maternal toxicity anticipated)

Complete evaluation is standardly performed but not driven by guidance

Complete evaluation is standardly performed but not driven by guidance

Yes, if reproductive organs are not well characterized in GenTox studies. Otherwise preserve reproductive organs for possible future evaluation

Yes, if reproductive organs are not well characterized in GenTox studies. Otherwise preserve reproductive organs for possible future evaluation

b Macroscopic and Histopathology

When fertility endpoints in NHP are assessed as part of a general toxicity study, a standard panel of organ weights, including reproductive tissues, is typically included

c

Histopathology: Complete - tissues commonly evaluated in general toxicology studies Targeted histopathology - limited tissues to evaluate efficacy markers, pharmacologic endpoints, or specific toxicity concerns

b

“Clinical Pathology: Standard clinical pathology - endpoints commonly tested in general toxicology studies Targeted clinical pathology - limited endpoints to evaluate efficacy markers, pharmacologic endpoints, or specific toxicity concerns

a

Dogs are generally not utilized for fertility, embryofetal, or pre- and postnatal evaluations

General Notes: Controls should be utilized when evaluating any endpoints

Pre- and postnatal developmental toxicity (PPND)

Embryofetal Development (EFD)

Rodent

Fertility

NHP

Species

Study Type

Working Group integrated assessment of current practices and considerations for the inclusion of pathology endpoints in DART studies for pharmaceutical drug development

Author Manuscript

Table 8 Halpern et al. Page 39

Scientific and Regulatory Policy Committee Points to Consider Review: Inclusion of Reproductive and Pathology End Points for Assessment of Reproductive and Developmental Toxicity in Pharmaceutical Drug Development.

Standard components of nonclinical toxicity testing for novel pharmaceuticals include clinical and anatomic pathology, as well as separate evaluation ...
221KB Sizes 0 Downloads 8 Views