420 likes | 502 Views
Nonclinical Safety Testing for Biological Therapeutics for Cancer Treatment. Anne M. Pilaro, Ph.D. Center for Drug Evaluation and Research FDA . Disclaimer. The opinions expressed by Dr. Pilaro in this presentation are the presenter’s, and do not reflect official policy of the FDA
E N D
Nonclinical Safety Testing for Biological Therapeutics for Cancer Treatment Anne M. Pilaro, Ph.D. Center for Drug Evaluation and Research FDA
Disclaimer • The opinions expressed by Dr. Pilaro in this presentation are the presenter’s, and do not reflect official policy of the FDA • Information presented was obtained from publicly available sources, including published literature, labeling, and/or SBA documents • No official support or endorsement by FDA is intended or should be inferred
Objectives for Today’s Presentation • Discuss review responsibilities for biological therapies for cancer • Review approaches to obtain IND-enabling, nonclinical safety data for protein therapeutic agents for cancer • Introduce current guidance for toxicology testing of biological therapeutic products
What is a Biological Product? • Substances derived either from living organisms – including humans, animals, plants, and microorganisms – or produced by biotechnology methods Photo from www.gene.com
Who is Responsible for Regulating Biologic Cancer Therapies? • CBER, OCTGT • adoptive cell transfers (i.e., stem cells, embryonic stem cells [ESC], leukocytes with or without gene modification) • gene therapies • tumor vaccines (e.g., pDNA-based, viral vector-based, or autologous/allogeneic tumor with or without gene modification) • siRNAs delivered using gene transfer technology • combination therapies
Who is Responsible for Regulating Biologic Cancer Therapies? • CDER/OODP/DDOP • hormones and metabolic factors • synthetic peptides • oligonucleotides and siRNA • CDER/OODP/DBOP • monoclonal antibodies • recombinant proteins • e.g., cytokines, growth factors, fusion proteins • extracted proteins
Who is Responsible for Regulating Biologic Cancer Therapies? • For the purposes of this presentation, focus today will be on products regulated in CDER/OODP/DBOP • monoclonal antibodies for cancer treatment • cytokines and/or growth factors • e.g., interleukin-2 for cancer treatment • recombinant hematopoietic factors for treatment and/or prophylaxis of chemotherapy side effects • fusion proteins for cancer treatment • combination antibody~ or protein~small molecule drug conjugates
What Do I Need to Start an IND for a New Anti-Cancer Agent? • Product characterization • manufacturing and quality control issues • Nonclinical testing in animals • biologic or pharmacologic activity • pharmacokinetic profile • toxicology testing in animals • Phase 1 clinical trial
What are Pharmacology Studies for Anti-Cancer Biologics? • Evaluation of ability of a new agent to induce the desired therapeutic effect • in vitro studies of product binding, tumor cell killing, and other effects • in vivo studies of anti-tumor activity • e.g., human tumor xenograft models • Demonstration of pharmacologic and/or biologic activity is the first step in the development of ANY new drug or biologic
Why are Pharmacology Studies Important? • Establish basis for conducting trial • interaction with target receptor, tissue, or organ • degree of pharmacodynamic (P/D) effect • correlation of P/D effect with exposure • Optimize dosing, regimen for the clinic • Optimize route of administration • Selection of species for further testing
Where do Pharmacology Data Come From? • Most biologic activity/pharmacology data come from studies conducted in the sponsor’s own laboratory • need to include appropriate control groups • should include sufficient “n” in treatment groups to attain statistical relevance • should be described in the IND in adequate detail that FDA can judge appropriateness of model, findings to proposed human study • Published literature may also serve as data source • data in article must be of sufficient detail, as above • For previously tested agents, access to data may be obtained by permission to cross-reference existing IND or DMF
How are Pharmacology Data Usedto Support a New IND? • Data from nonclinical bioactivity and/or pharmacology studies are used to establish the rationale for conducting the first clinical trials in humans • for anti-cancer biologic agents, most early trials are done in the index (patient) population, NOT healthy volunteer subjects
What are Pharmacokinetic Studies for Anti-Cancer Biologics? • Measurement of in vivo disposition of a new biologic agent • studies of product concentration, exposure, clearance, and half-life • determined from blood, plasma, or serum levels of agent at various times after dosing • differences following different routes of dosing
Why are Pharmacokinetic Studies Important? • Demonstration of the pharmacokinetics of a new biologic allows estimation of: • exposure to agent after any given dose • correlation with pharmacologic/therapeutic effect • duration of exposure (half-life) • dosing interval for the clinical study • time to reversal of any biologic or toxic effects • development of anti-product antibodies • both total and neutralizing activity
Where do Pharmacokinetic Data Come From? • Serum levels of anti-cancer biologic measured during in vivo pharmacology studies • Stand-alone pharmacokinetic studies in responsive species • Adjunct to single- and repeat-dose toxicology studies in animals • toxicokinetics
What are Toxicology Studies for Anti-Cancer Biologics? • Dedicated in vivo studies that provide data describing the safety profile of the agent, at doses greater than those that produce the therapeutic effect • single- and repeat-dose paradigms • sub-chronic and chronic duration • include measurements of exposure, antibody development, and reversibility
Why is Toxicology Testing for Any New Biologic Important? • Provides information regarding the safety of single and/or repeated exposure to the biologic anti-cancer agent • toxicities related to the dose of product given • toxicities related to the duration of product administration • identification of target organs • reversibility of toxicities • after clearance of the biologic product • after development of neutralizing antibodies
What Does a Nonclinical Toxicology Study Tell Me? Single dose (acute) studies • designed to maximize exposure to agent (mimic overdose) • high doses, frequently i/v administration • limited endpoints – 14 days • clinical observations, body weights • gross necropsy with no histology • clinical pathology not usually done
What Does a Nonclinical Toxicology Study Tell Me? Repeat-dose administration • Designed to support the intended clinical usage of the agent • doses “bracket” those planned for clinic • dosing for at least the same number of planned administrations • duration may vary from 14 days to 6+ months • should include recovery group(s)
Where do Toxicology Data Come From? • Data in support of a clinical trial may come from: • GLP-compliant toxicology studies conducted by a contract laboratory • well-controlled studies conducted in house (“GLP-like”) • published data in peer-reviewed journals • cross-reference to previously submitted IND or DMF
What Do I Need to Measure in Toxicology Testing? • Monitoring in-life • clinical observations, behavior • signs/symptoms of toxicity • body weights, weight changes • feed and water consumption • clinical pathology • serum enzyme, electrolyte, glucose and lipid profiles as evidence of organ toxicity • hematology • urinalysis • these can all also be monitored in man!
What Do I Need to Measure in Toxicology Testing? • Pharmacokinetics/toxicokinetics • correlation of serum levels of biologic with toxicity, pharmacodynamic activity • identification of elimination half-life and antibody development (reversibility) • Pathology • gross and microscopic evaluation of tissue damage • most sensitive indicator, but final! • confirmation of target organ toxicity detected biochemically
What Else Do I Need to Include in My Toxicology Studies? • Administration/dose selection • route & dosing regimen should mimic proposed clinical use • alternative routes/regimens acceptable in some cases • attainment of toxic dose, no observable adverse effect level (NOAEL) desirable • multiples of human dose needed to determine adequate safety margins • can vary with product class and/or clinical indication
Are There Any Specific Safety Concerns for my Biologic Agent? • Monoclonal antibodies (naked or conjugated) • cross-reactivity with normal tissue • exaggerated pharmacodynamic effects • chronicity of exposure and toxicity • immunogenicity/antibody production • “bystander” toxicity of radiolabel or small molecule drug in conjugates
Are There Any Specific Safety Concerns for my Biologic Agent? • Cytokines and/or growth factors • demonstration of species-specificity • evaluation of interaction with host endogenous cascade • tumor-promoting potential • immunogenicity/antibody production • effects on neutralization of endogenous counterpart to test agent
How Are Toxicology Studies Used to Support a New IND? • Nonclinical toxicology data are used to make recommendations regarding the conduct of the clinical trial, based on the safety data obtained • initial safe starting dose, dose-escalation scheme • target organ(s), reversibility of toxicity • appropriate parameters for clinical monitoring • identify “at risk” patient populations (inclusion/exclusion criteria)
What Else Do I Need to Know for Safety Testing of Biologics? • Data from preclinical pharm/tox studies must meet the regulatory requirements set forth in 21 CFR • data must be of unquestionable integrity, quality to support safety of agent for entry into clinical trials • animal studies may be done in house as long as meet the requirements of 21 CFR 312.23(8) • full compliance with GLP (21 CFR 58) expected for any pivotal toxicity studies
What Else Do I Need to Know for Safety Testing of Biologics? • Product development allowed during preclinical testing phase • use of non-GMP protein products allowed • need to demonstrate comparability with clinical grade material(s) as development progresses • Protein therapeutics are not expected to differ from small molecule cancer drugs in the requirements for demonstration of safety, just in the approach used to obtain the data
Where Can I Find Guidance for Safety Testing of Biologics? The ICH S6 Document • Title: Preclinical Testing of Biotechnology-Derived Pharmaceuticals • Scope: Covers protein and other biological therapeutics produced by biotechnology methods • Does not address: cellular, tissue or gene therapies, blood or plasma products, traditional vaccines
ICH-S6 – What it Doesn’t Do • Does NOT provide a “one size fits all” or “cookbook” approach to toxicology study design • provides a framework for design of animal studies to address safety of biotechnology derived products, based on characteristics of the product and intended clinical use
ICH-S6 – What it Does Do • Recognizes that conventional approaches to toxicity studies used for small molecule drugs are often NOT appropriate for protein therapeutics • unique issues of species restriction, dose and formulation limitations, antibody formation limit utility of standard toxicity paradigms • offers alternative approaches to obtain adequate safety data to support human use
ICH-S6 – What it Does Do • Provides guidance to sponsors regarding the design of toxicology studies, including: • usage of pharmacologically relevant vs. nonrelevant animal species • usage of animal models of disease • immunogenicity testing and its implications • genotoxicity, chronic toxicity, carcinogenicity, and reproductive and developmental toxicity testing • GLP compliance
Additional Guidance from FDA Coming soon… • Guidance for Industry and Reviewers: Nonclinical Safety Evaluation of Biotechnology-Derived Pharmaceuticals • FDA general guidance on necessary studies to support development of protein therapies • expands upon the framework of ICH S6 • adds FDA experience with unique issues • includes Q&A section to address common questions encountered by sponsors in field
Additional Guidance from FDA Coming soon… • Guidance for Industry, Investigators and Reviewers: Nonclinical Studies for Anticancer Drugs and Biologics • OODP-specific guidance on necessary studies to support development of anti-cancer therapies • identifies differences in nonclinical study approaches for small molecule versus protein cancer therapeutics • addresses issues of timing of studies relative to duration of clinical trial, indication • incorporates principles of both ICH S6 and FDA Guidance for protein therapeutics
Summary • What do I need to start an IND for a new anti-cancer biologic agent? • The “Three R’s” of nonclinical testing: • pharmacology = RATIONALE • toxicology = RECOMMENDATIONS • demonstration of safety = REGULATORY EXPECTATIONS
Take-Home Messages • Animal testing determines safety…but caveats exist for biological products • Toxicology programs for anti-cancer biological therapeutics require novel approaches to obtain data • no “one size fits all” paradigm for biologics • traditional animal toxicology models may not be appropriate or feasible • studies may have to be “individualized” to address specific safety concerns
Take-Home Messages • Toxicology studies for anti-cancer biological therapeutics should: • follow guidance set forth by ICH S6, and additional guidances as available • be rational, scientifically designed, and problem solving • employ careful design, judicious use of animals
Take-Home Messages • But... animal studies really only give you a “best guess” of what to expect • no animal model, including humans is 100% predictive of response in man • sometimes animal studies can give a false sense of security • however, these models may be useful in evaluation of mechanism, other toxicities
Some Further Resources • ICH Guidances • ICH S6: Safety Studies for Biotechnological Products • ICH M3: Timing of Pre-clinical Studies in Relation to Clinical Trials • ICH S5a: Detection of Toxicity to Reproduction for Medicinal Products • ICH S2b: Standard Battery of Genotoxicity Testing also available at: www.ich.org/cache/compo/276-254-1.html
Some Further Resources • Points to Consider • Points to Consider in the Manufacture and Testing of Monoclonal Antibody Products for Human Use - 2/28/97 • www.fda.gov/cber/gdlns/ptc_mab.pdf • Points to Consider in the Manufacture and Testing of Therapeutic Products for Human Use Derived from Transgenic Animals – 1995 • www.fda.gov/cber/gdlns/ptc_tga.txt
Please Contact the DBOP Toxicologists Anne M. Pilaro, Ph.D. Supervisory Toxicologist DBOP Staff Andrew McDougal, Ph.D. Brenda Seidman, Ph.D. (301) 796-2320 Further Questions?