27.09.2014 Views

The Toxicologist - Society of Toxicology

The Toxicologist - Society of Toxicology

The Toxicologist - Society of Toxicology

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

than genetic mutations which have long been the hallmark <strong>of</strong> cancer. Since cancer<br />

is considered a disease <strong>of</strong> clonally expanded dedifferentiated cells with stem cell-like<br />

properties, the genomic analysis <strong>of</strong> signaling pathways involved in stem cell maintenance<br />

and renewal in vivo might provide a useful alternative for estimating cancer<br />

risk following short term environmental chemical exposure. We have identified alterations<br />

in the Wnt signaling pathway in target tissues as early as two weeks following<br />

chemical carcinogen exposure while little or no change was observed in this<br />

pathway following non-carcinogen exposure. <strong>The</strong> observed alterations in Wnt signaling<br />

gene transcripts were also dose-dependent and correlated well with the relative<br />

potency <strong>of</strong> the chemical carcinogen. Bioinformatic analysis <strong>of</strong> public gene expression<br />

data from cancer studies reveals clustering <strong>of</strong> Wnt signaling genes into<br />

receptor/ligand and second messenger gene families during stem cell growth. In this<br />

talk we describe research that facilitates the genomic characterization <strong>of</strong> alterations<br />

in these signaling pathways as predictors <strong>of</strong> the carcinogenic potential <strong>of</strong> environmental<br />

chemicals.<br />

fetal exposure during this developmental window). For compounds which are pharmacologically<br />

active in non-human primates only a DART study in a single, i.e.<br />

this species is sufficient. Fertility assessment may be covered in the repeated dose<br />

toxicity test if functional assessment is not feasible (e.g. monkey). <strong>The</strong> revision <strong>of</strong><br />

the S6 will focus on the redundancy <strong>of</strong> a classical embryo-fetal developmental toxicity<br />

study for monoclonal antibodies recommending a rather integral approach e.g.<br />

with an enhanced peri-postnatal development study covering nearly the complete<br />

period <strong>of</strong> gestation.<br />

1436 PRECLINICAL STRATEGY CONSIDERATIONS FOR<br />

ASSESSING THE REPRODUCTIVE AND<br />

DEVELOPMENTAL TOXICITY POTENTIAL OF<br />

BIOPHARMACEUTICALS.<br />

J. Cavagnaro. Access BIO, Boyce, VA.<br />

1434 CURRENT THINKING AND EXPERIENCES ON<br />

DEVELOPMENTAL AND REPRODUCTIVE SAFETY<br />

ASSESSMENT OF BIOTHERAPEUTICS.<br />

C. J. Bowman 1 and T. E. White 2 . 1 Pfizer, Groton, CT and 2 GlaxoSmithKline, King<br />

<strong>of</strong> Prussia, PA.<br />

As scientific thinking and regulatory expectations around highly target-specific biotherapeutics<br />

have evolved, it has become increasingly difficult to design meaningful<br />

nonclinical strategies that reduce uncertainty around the risk <strong>of</strong> effects on human<br />

reproduction and development. Importantly, these nonclinical studies are likely the<br />

most reliable method available to prevent drug-induced birth defects and infertility<br />

since clinical evaluation <strong>of</strong> these endpoints is unethical or rare. <strong>The</strong>se studies should<br />

generally be in compliance with ICH S5, which is designed primarily to detect toxicity<br />

to reproduction and development (hazard identification). From ICH S5 relatively<br />

standard nonclinical strategies for small molecules have evolved, but for practical,<br />

technical, and sometimes ethical reasons may have limited value for large<br />

molecules or vaccines (issues ranging from placental transfer to limited <strong>of</strong>f-target<br />

toxicity). Although most biological effects <strong>of</strong> biotherapeutics have an origin in<br />

modification <strong>of</strong> a target or target signaling, it is not uncommon to have unexpected<br />

effects on reproduction and/or development since regulation/function <strong>of</strong> the target<br />

during these lifestages is <strong>of</strong>ten not well understood, particularly for novel drug targets.<br />

As described in ICH S6, for biotherapeutics careful scrutiny <strong>of</strong> the nonclinical<br />

strategy and conduct <strong>of</strong> specific studies is necessary to appropriately account for<br />

many issues, particularly species specificity, immunogenicity, biological activity<br />

and/or elimination half-life. In order to adhere to ever-changing regulatory expectations,<br />

minimize the use <strong>of</strong> animals; and improve the performance <strong>of</strong> safety assessment/toxicology<br />

around potential treatment-related effects on reproduction and<br />

development; innovative strategies using a combination <strong>of</strong> animal models (e.g.,<br />

transgenic) and study designs (e.g., use <strong>of</strong> homologues or combined pre/postnatal<br />

development in nonhuman primate) are currently being developed and applied by<br />

many companies.<br />

1435 CURRENT REGULATORY EXPERIENCE AND<br />

PROPOSED MODIFICATIONS TO ICH S6.<br />

J. van der Laan. Centre for Biological Medicines and Medical Technology, National<br />

Institute for Public Health and the Environment, Bilthoven, Netherlands. Sponsor: C.<br />

Bowman.<br />

<strong>The</strong> regulatory approach in the preclinical assessment <strong>of</strong> new biotechnology-derived<br />

human pharmaceuticals in the ICH S6 Guideline is explicitly different from<br />

the common approach for conventional small molecule, chemically-synthesized<br />

pharmaceuticals. <strong>The</strong> development <strong>of</strong> a huge number <strong>of</strong> monoclonal antibodies for<br />

a broad population including women <strong>of</strong> child-bearing potential revealed that the<br />

minimal attention to this group <strong>of</strong> products in the present S6 Guideline merits an<br />

update in the description <strong>of</strong> the regulatory approach. In preparing the revision <strong>of</strong><br />

the guideline we have evaluated the experience with respect to developmental and<br />

reproductive toxicity (DART) testing for the products with a European marketing<br />

authorization, with a further emphasis on the monoclonal antibodies and related<br />

products. This evaluation has revealed that for protein hormones and other proteins<br />

the classical DART approach <strong>of</strong> two species (mainly rats and rabbits) has been used<br />

to test the reproductive safety. For 15/20 approved monoclonal antibodies DART<br />

has been tested. <strong>The</strong> Cynomolgus monkey showed to be the species used in the majority<br />

<strong>of</strong> the cases (10 out <strong>of</strong> 15). <strong>The</strong> human fetal risk <strong>of</strong> the exposure to monoclonal<br />

antibodies is more associated with functional effects derived from influencing<br />

development in the latter part in pregnancy rather than with malformations<br />

resulting from interference with organogenesis in the first trimester (due to limited<br />

<strong>The</strong> “principles” <strong>of</strong> DART testing for biopharmaceuticals are similar to those for<br />

small molecule pharmaceuticals and in general follow the guidance outlined in<br />

ICH S5 (R2). However, because many biopharmaceuticals are species-specific, alternate<br />

approaches or “practices” are generally needed to evaluate DART potential<br />

as outlined in the initial ICH S6 guidance. <strong>The</strong> need for this “case-by-case approach”<br />

is dictated by differences in product attributes. This presentation provides<br />

both a framework for developing DART testing strategies for biopharmaceuticals,<br />

in the context <strong>of</strong> the overall clinical development strategy, as well as an overview <strong>of</strong><br />

the state <strong>of</strong> DART testing <strong>of</strong> biopharmaceuticals. <strong>The</strong> various strategies that have<br />

been successfully implemented over the past two decades will be summarized based<br />

upon regulatory reviews and the recently published BioSafe White Paper (Birth<br />

Defects Res (Part B), 33:176-203, 2009). Lessons learned will be applied to future<br />

strategies in the context <strong>of</strong> novel compounds now entering development. Current<br />

challenges will be highlighted as well as the importance <strong>of</strong> communicating and<br />

managing potential DART risks <strong>of</strong> biopharmaceuticals.<br />

1437 CHALLENGES AND SOLUTIONS FOR EVALUATING<br />

THE DEVELOPMENTAL TOXICITY POTENTIAL OF<br />

BIOTHERAPEUTICS.<br />

L. Andrews. Genzyme Corporation, Framingham, MA.<br />

As the development <strong>of</strong> biotherapeutics becomes a more advanced science based<br />

challenge, the selection <strong>of</strong> relevant animal models, utility <strong>of</strong> traditional species and<br />

alternatives to traditional safety approaches are becoming more accepted and in<br />

fact, necessary. <strong>The</strong> challenges <strong>of</strong> biotherapeutics become especially evident when<br />

considering the most appropriate science based approach for the conduct <strong>of</strong> developmental<br />

and reproductive studies.<br />

Designing an informative developmental and reproductive toxicity study for a biotherapeutic<br />

can be a challenge in light <strong>of</strong> the need to utilize alternative approaches<br />

that may be divergent from a more traditional Segment I, II, III approach and include<br />

utilization <strong>of</strong> two species. Alternatives to the traditional safety approach include<br />

the use <strong>of</strong> homologous proteins, transgenic animals, animal models <strong>of</strong> disease<br />

as well as state <strong>of</strong> the art non-invasive, non-terminal technologies such as high resolution<br />

imaging and scanning methods. In addition, a science based approach to rationale<br />

study design has allowed for a better use <strong>of</strong> animals through the development<br />

process. Study design considerations must be addressed in order to most<br />

effectively utilize animals and wherever possible reduce the need for large numbers<br />

and multiple studies. <strong>The</strong> opportunities and challenges for these approaches with<br />

respect to developmental and reproductive studies as well as the approach to implementing<br />

these areas to help reduce animal use and advance the science <strong>of</strong> biotechnology<br />

drugs will be discussed. A specific example <strong>of</strong> a monoclonal antibody in development<br />

will be illustrated including a discussion <strong>of</strong> regulatory feedback and<br />

alternatives to a traditional approach.<br />

1438 CASE STUDIES: DEVELOPMENTAL AND<br />

REPRODUCTIVE TOXICITY (DART) STRATEGIES<br />

EMPLOYED TO SUPPORT THE REGISTRATION OF<br />

GOLIMUMAB AND USTEKINUMAB.<br />

C. Sachs, G. Treacy and P. Martin. Centocor R&D, Inc., Radnor, PA.<br />

Golimumab and ustekinumab are fully human monoclonal antibodies to soluble<br />

cytokines (tumor necrosis factor α and IL-12/23, respectively). Cynomolgus monkeys<br />

were identified as biologically relevant species for toxicity studies for both antibodies.<br />

Common goals <strong>of</strong> the developmental toxicity studies for both antibodies<br />

304 SOT 2010 ANNUAL MEETING

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!