APPLYING PRE-CLINICAL DATA TO CLINICAL STUDIES-I Edward A. Sausville, M.D., Ph.D. Developmental Therapeutics Program National Cancer Institute October.

Slides:



Advertisements
Similar presentations
The Drug Discovery Process
Advertisements

Regulatory Framework Leigh Shaw, Director.
Paula M. Jacobs, Ph.D. SAIC Frederick Cancer Imaging Program/DCTD/NCI/NIH September 5, 2006 Phase 0 Trials in Oncologic Drug Development DCTD Phase 0 Workshop.
Drug Research and Development (R&D) Karol Godwin DVM.
Single-Patient Use of Investigational Drugs and Biologic Products for Treating Cancer Grant Williams, M.D. Medical Team Leader DODP/CDER/FDA.
Safety and Extrapolation Steven Hirschfeld, MD PhD Office of Cellular, Tissue and Gene Therapy Center for Biologics Evaluation and Research FDA.
1 Pharmacology/Toxicology information to submit an IND for an anticancer drug.
Single Use Expanded Access IND/IDE: FDA and IRB Requirements Before and After Use IRB Webinar October 9,2014.
Mitochondrial Manipulation Technologies: Preclinical Considerations
JMV 1843 pharmacological profile
U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES NATIONAL INSTITUTES OF HEALTH Working with FDA: Biological Products and Clinical Development Critical Path.
What Do Toxicologists Do?
Title, in bold style Subtitle, in regular Max 3 lines of text totally NB! The graphic outside the slide will not show in “Slide Show” or on print WntResearch.
University of Colorado Cancer Center
REGULATORY ISSUES IN HIV CURE RESEARCH HIV Cure Research Training Curriculum Regulatory Issues Module by: Damon Deming, Ph.D. FDA Division of Antiviral.
Stefan Franzén Introduction to clinical trials.
+ Drug Development and Review Process. + Objectives Learn the processes involved in drug discovery and development Define the phases involved in FDA drug.
Cellular Therapies for Cardiac Diseases – FDA Preclinical Perspective Richard D. McFarland Ph.D., M.D. CBER/OCTGT/DCEPT BRMAC # 37 March 18-19, 2004.
Neonatal/Juvenile Animal Safety Studies Kenneth L. Hastings, Dr.P.H., D.A.B.T. Office of New Drugs, CDER.
Effectiveness Evaluation for Therapeutic Drugs for Non-Food Animals
Career Opportunities for PharmDs in the Pharmaceutical Industry: Research & Development.
NMF 3/6/03 Susan Galbraith, MB BChir PhD Vice President Clinical Discovery Oncology & Immunology Phase 0 Trials Why aren’t they more widely used by industry?
Food and Drug Administration Preclinical safety data for “first in human” (FIH) clinical trials in healthy volunteer subjects Oncology Drug Advisory Committee.
INVESTIGATIONAL DRUG SERVICES IN THE HOSPITAL Sheree Miller, Pharm.D. University of Washington Medical Center
U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES NATIONAL INSTITUTES OF HEALTH Working with FDA: Biological Products and Clinical Development IND Case Studies.
Stages of drug development
Office of Clinical Pharmacology and Biopharmaceutics IDSA/ISAP/FDA Workshop 4/16/04 1 Improvement in Dose Selection: FDA Perspective IDSA/ISAP/FDA Workshop.
Phase III Study Comparing Gemcitabine plus Cetuximab versus Gemcitabine in Patients with Locally Advanced or Metastatic Pancreatic Adenocarcinoma Southwest.
Food and Drug Administration Food and Drug Administration Center for Biologics Evaluation and Research Biological Response Modifiers Advisory Committee.
February 2008 William Petros, Pharm.D., FCCP
Obtaining and Negotiatiing a Position in Industry/Biotech Roy G. Smith Ph.D. Director, Huffington Center on Aging Professor, Department of Molecular and.
1 Safety Pharmacology for Oncology Pharmaceuticals at CDER John K. Leighton Associate Director for Pharmacology CDER/OND/OODP.
Essential Considerations for Your IND Submission: Objectives and Pitfall Avoidance in Your Preclinical Program Darren Warren.
Phase 1 Clinical Studies First-In-Human (FIH) Pharmacologically-Guided Dose Escalation Jerry M. Collins, Ph.D. Developmental Therapeutics Program Division.
1 Kepivance™ (Palifermin) Basis for Approval and Pediatric Studies Kepivance™ (Amgen) Approved 12/15/04 Joseph E. Gootenberg, M.D. Office of Oncology Drug.
Animal Models for Porcine Xenotransplantation Products Intended to Treat Type 1 Diabetes or Acute Liver Failure CTGTAC #47 May 14, 2009.
Exploratory IND Studies
Adding Safety Pharm Endopoints To General Tox Studies - II Michael J Engwall, DVM, PhD Principal Scientist Safety and Exploratory Pharmacology Toxicology.
Nonclinical Perspective on Initiating Phase 1 Studies for Small Molecular Weight Compounds John K. Leighton, PH.D., DABT Supervisory Pharmacologist Division.
CHEE DRUG PRODUCT DEVELOPMENT u Drug ä agent intended for use in the diagnosis, mitigation, treatment, cure, or prevention of disease in man or animals.
Investigational Drugs in the hospital. + What is Investigational Drug? Investigational or experimental drugs are new drugs that have not yet been approved.
ONCOLOGY Drug Development Fadi Sami Farhat, MD ONCOLOGY Drug Development Fadi Sami Farhat, MD Hematology Oncology
1 SNDA Gemzar plus Carboplatin Treatment of Late Relapsing Ovarian Cancer.
Drug - Device Combination Issues : Oncology Perspective Ramzi Dagher, M.D. DODP/CDER/FDA.
Preclinical Guidelines: Development of Radioprotective/Mitigative Agents Departments of Dermatology & Radiation Oncology University of Rochester Medical.
The New Drug Development Process (www. fda. gov/cder/handbook/develop
History of Pediatric Labeling
Initiating phase 1 clinical trials in pediatric oncology National Cancer Institute Perspective Barry Anderson, MD, PhD Pediatric Section Cancer Therapy.
Consider Incorporating Respiratory Safety Pharmacology Measurements into Your Next Repeat Dose Toxicology Study September 14, 2012 Jeff Tepper, PhD, DABT.
Enrollment and Monitoring Procedures for NCI Supported Clinical Trials Barry Anderson, MD, PhD Cancer Therapy Evaluation Program National Cancer Institute.
Chief, Gene Therapy Branch
Rationale for Developing New Drugs 25% of children with cancer will not survive 5 years25% of children with cancer will not survive 5 years The acute toxicity.
C LINICAL R ESEARCH. CONTENTS Drug Development Process Pre – Clinical Studies Clinical Trials Phase I Phase II Phase III Phase IV 2.
C-1 Pegfilgrastim (Neulasta  ) Oncologic Drugs Advisory Committee Pediatric Subcommittee October 20, 2005 Amgen Inc.
European Patients’ Academy on Therapeutic Innovation The key principles of pharmacology.
Nonclinical Perspective on Initiating Phase 1 Studies for Biological Oncology Products – Case Studies Anne M. Pilaro, Ph.D. DBOP/OODP/CDER Oncology Drugs.
PRECLINICAL PHARMACOLOGY & TOXICOLOGY OF ANTI-NEOPLASTIC AGENTS  THE NCI PERSPECTIVE Joseph E. Tomaszewski, Ph.D. Toxicology & Pharmacology Branch.
MM-005: A Phase 1, Multicenter, Open-Label, Dose-Escalation Study to Determine the Maximum Tolerated Dose for the Combination of Pomalidomide, Bortezomib,
Division of Cancer Treatment and Diagnosis Presented By: Michael Difilippantonio, Ph.D. October 1, 2015.
The process of drug development. Drug development 0,8 – 1 mld. USD.
FDA DRUG APPROVAL FDA’s Lengthy Drug Approval Process in Twelve Steps Overview of the FDA Drug Approval Process Drug Developed June 13, 2016 | Emilia Varrone.
Drug Development Process Stages involved in Regulating Drugs
for Human Pharmaceuticals Kyung-Chul Choi D.V.M., Ph.D.
The Stages of a Clinical Trial
Guidance for review of studies involving HCT/Ps and IND Basics
Regulatory– Terms & Definitions רגולציה - מונחים והגדרות
Broadening Eligibility Criteria to Make Clinical Trials More Representative Joint Recommendations of the American Society of Clinical Oncology and Friends.
Clinical Trials in STS Shreyaskumar Patel, M.D.
Speeding access to therapies
Presentation transcript:

APPLYING PRE-CLINICAL DATA TO CLINICAL STUDIES-I Edward A. Sausville, M.D., Ph.D. Developmental Therapeutics Program National Cancer Institute October 17, 2002

GOALS OF PRECLINICAL DRUG STUDIES IND = “Investigational New Drug” application = approval by FDA to conduct human studies; main criterion : SAFETY AND LIKELY REVERSIBLE TOXICITY = allows start of Phase I trials Pediatric Phase I Oncology Drugs: Special Issues *Few (thankfully) patients; many agents *Unmet medical need; ethical concerns *Prior Rx ; Concomitant meds *Unique pediatric biology (tumor and host) vs. value of adult data in study design Regulatory framework

PEDIATRIC PHASE I STUDIES: “CLASSICAL” NCI RECOMMENDATIONS Begin in pediatric patients with solid tumors and leukemias at 80% of max tol dose (MTD) in adults with solid tumors Solid tumor and leukemia pts at each level Escalate in 20% increments; distinguish myelosuppressive tox (desired in leukemia) vs non myeloid tox In absence ofnon-myelo tox, escalate beyond solid tumor MTD in leukemia patients. Marsoni et al. Cancer Treatment Reports 69, 1263, 1985

PEDIATRIC PHASE I STUDIES: BASIS FOR RECONSIDERATION OF CLASSICAL PRACTICE BIOLOGY: Pediatric tumors may have targets intrinsically different from adults; therefore adult data will never be available for drugs directd to those targets. PHARMACOLOGY: Past practice weighted toward “cytotoxics” ; ? Relevance to “targeted” agents. TIMING: Many new agents; delay in completing adult studies before application in peds neoplasms therefore exacerbate unmet need.

COMPONENTS OF AN IND “Form 1571” Table of Contents Intro Statement / Plan Investigator Brochure Clinical Protocol Chemistry, Manufacture, Control The goal of the pre-clinical process Pharmacology/ Toxicology Prior Human Experience Additional Info - Data monitoring, Quality Assurance

HOW ARE PHASE I DOSE AND SCHEDULE FIXED IN ADULTS ? Animal (usually mouse) model studies define likely active schedules in animals bearing human-derived tumors *Likelihood of human activity stochastic: more models with activity, greater likelihood of human activity *Limitations: difference between animal / human metabolism *Drug concentrations OR effect on target provide important ancillary info. Toxicology according to NCI guidelines / FDA requirements Starting dose a fraction of dose causing no or minimal reversible toxic effect; escalate dose in steps likely to capture reversible toxic effect

PROBLEMS WITH “MTD” DRIVEN ENDPOINTS Drugs regulating pathways important in oncogenesis are effective by combining with high affinity binding sites; therefore must distinguish “targeted” vs “non-targeted” toxicity related to these binding sites Whether dosing beyond effect on desired target “buys” therapeutic value not clear Therefore must define in pre-clinical studies “BIOLOGICALLY EFFECTIVE DOSE” and “MAXIMUM TOLERATED DOSE” Use BIOLOGIC rather than TOXIC endpoints in PhaseI?

REGULATORY CONSIDERATIONS FOR PRE- CLINICAL DEVELOPMENT OF ANTICANCER DRUGS [DeGeorge, et al, CCP (1998) 41: ] “… The types of preclinical studies expected for support of clinical trials and marketing of a new drug depends on both the intended use of the drug and the population of patients being studied and treated. In situations where potential benefits are greatest (Advanced, life- threatening disease), greater risks of treatment toxicity can be accepted and the required preclinical testing can be minimal. …”

FDA PRECLINICAL PHARMACOLOGY & TOXICOLOGY REQUIREMENTS DRUGS – Two Species - Rodent & Non-rodent – Clinical Route & Schedule Follow NCI Guidelines – Pharmacokinetics - Optional BIOLOGICALS – Most Relevant Species – Clinical Route & Schedule

OBJECTIVES OF PRECLINICAL TOXICOLOGY STUDIES FOR ANTI-NEOPLASTIC DRUGS DETERMINE IN APPROPRIATE ANIMAL MODELS: –The Maximum Tolerated Dose (MTD) –Dose Limiting Toxicities ( DLT ) –Schedule-Dependent Toxicity –Reversibility of Adverse Effects –A Safe Clinical Starting Dose

NCI STANDARDIZED PRECLINICAL TOXICOLOGY PROTOCOLS FOR ANTI-NEOPLASTIC AGENTS ( ) Mouse Lethality Studies Dog Toxicity Studies Rodent (Rat) Toxicity Studies Determine LD 10 on Dx1 & Dx5 Schedules Assess safety of 1/10 LD 10 ; Determine DLTs on Dx1 & Dx5 Schedules.

CURRENT NCI TOX PHILOSOPHY Agent-Directed Studies Pharmacologically- Guided Integrate With Preclinical Efficacy Data & the Proposed Clinical Protocol Rational Evaluation of Role of Schedule Dependence, Pharmacokinetics & Metabolism in the Development of Toxicity Relate Plasma Drug LeveLs and/or AUC to Safety and to Occurrence & Severity of Toxicity Extrapolate Toxic Effects Across Species

AGENT-DIRECTED versus STANDARD PROTOCOL DRUG DEVELOPMENT Greater Scientific Basis for Development Permits Greater Flexibility Data Rich IND Submission to Support Phase I Preclinical Potential … Less Expensive Permits PK-Guided Dose Escalation in Phase I Clinical Trial Potential … Shorter, Less Expensive Optimal Schedule … Greater Chance of Success? Patients … Greater Chance of Effective Therapy?

BENZOYLPHENYLUREA PRECLINICAL MTD & DLTs Bone Marrow, GI Tract Bone Marrow GI Tract DLT > 150 < 240 mg/m mg/m 2 MTD (Total Dose) DOGRAT Schedule q4Dx3, po Starting Dose: 24 mg/m 2

ADDITIONAL AGENT-DIRECTED TOXICOLOGY STUDY REQUIREMENTS Attain Efficacious Drug Levels in Plasma In Vivo Correlate Drug Plasma Levels and/or AUC with Toxicity and Safety Across Species Ameliorate Toxicity by Change in Route/Schedule Compare Toxicity with Accepted Clinical Agents as Necessary

SCHEDULE and ROUTE versus TOXICITY Pyrazoloacridine:Bolus ivNeurotoxicity 1 Hr civBone Marrow Penclomedine:Bolus ivNeurotoxicity 1 Hr Dx5BM (& Neuro) 5 Hr civNeuro & Death OralBM O 6 –Benzylguanine:BolusCNS, HR  InfusionNeutrophilia

HOW PREDICTIVE OF HUMAN EXPERIENCE ARE THESE SAFETY-TESTING ALGORITHMS? NCI data Predictive power varies with endpoint desired: - 97% safe starting dose if 2-3 species (rodent plus dog) - 83% mouse only (Smith, A.C. Proc AACR 35: 459) BUT: Human MTD accurately predicted by: -mouse in 36% -rat in 19% -dog in 44% NO in vitro or silico methodology has yet emerged to predict human toxicity or dosing scheme (?marrow)

CONCLUSIONS FROM ILSI HUMAN TOXICITY PROJECT DATA SOT.BiomarkersSymposium; Regul. Toxicol. 32: 56, % (151/214) human toxicities(HTs) associated with toxicities in animals - 63% in non-rodents - 36% in rodent plus non-rodent combinations - 43% in rodents 29% of human toxicities not predicted for by animals (8HTs insufficient animal data) Two species best predictor; for single species non-rodent (dog and/or primate) better than rodent (mainly rat)

CONSIDERATIONS IN APPLYING DATA TO PEDS POPULATIONS How closely do adult / peds MTDs correspond? *cytotoxics? *noncytotoxics Are “classical” MTDs still relevant? “Age” / maturity / nature of tox species relevant if adult human phase I data not to drive peds dosing? Importance of efficacy model PK / PD? Chronicity / reversibility / age-relatedness of target related tox’s (e.g.s): *bone growth and anti-VEGFRs? *skin (anti-EGFR)

ACKNOWLEDGEMENTS J. Covey M. Hollingshead A. Smith J. Tomaszewski S. Decker