Office of Hematology and Oncology Products

Slides:



Advertisements
Similar presentations
U.S. Food and Drug Administration Notice: Archived Document The content in this document is provided on the FDAs website for reference purposes only. It.
Advertisements

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.
Rare Diseases and FDASIA
U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES NATIONAL INSTITUTES OF HEALTH Working with FDA: Biological Products and Clinical Development IND Regulatory.
Single Use Expanded Access IND/IDE: FDA and IRB Requirements Before and After Use IRB Webinar October 9,2014.
Clinical Trials — A Closer Look. The Food and Drug Administration (FDA) is the main consumer watchdog for numerous products: Drugs and biologics (prescription.
Have FDA Expedited Programs Shortened Drug Development Timelines? An analysis of newly approved therapies and how FDA expedited programs impacted drug.
Wednesday, December 17, :00pm-4:30pm EST National Coalition for Cancer Survivorship Post-Training Webinar ©2014 National Coalition for Cancer Survivorship.
Introduction to Regulation
What Do Toxicologists Do?
Special Topics in IND Regulation
Meeting Agenda Presentations on endpoints –Regulatory issues –Scientific issues Pros and cons of end points –Classical end points –Non-classical end points.
The FDA Landscape AdvaMed September 2008 Judith K. Meritz
Investigational New Drug Application 21 CFR Part 312 A Review for OCRA US RAC Study Group September 2005 Ginger Clasby, MS Promedica International
Mrs. Brandi Robinson Office of New Animal Drug Evaluation Center for Veterinary Medicine Regulating Animal Drugs.
INVESTIGATIONAL DRUG SERVICES IN THE HOSPITAL Sheree Miller, Pharm.D. University of Washington Medical Center
Regulatory Background and Past FDA Approvals in Colorectal Cancer Amna Ibrahim M.D DODP, FDA.
Stages of drug development
Adverse Events, Unanticipated Problems, Protocol Deviations & other Safety Information Which Form 4 to Use?
Special Issues in FDA-Regulated Studies: The Good, the Bad, and the Ugly C. Karen Jeans, MSN, CCRN, CIP COACH Program Analyst VA Office of Research & Development.
Nonclinical Studies Subcommittee Advisory Committee for Pharmaceutical Science CMC Issues for Screening INDs Eric B. Sheinin, Ph.D. Acting Deputy Director.
FDA, PDUFA AND MEDICAL INNOVATION Spring WHAT IS THE FOOD AND DRUG ADMINISTRATION (FDA)? The FDA is an agency within the U.S. Department of Health.
ODAC SCHERING-PLOUGH RESEARCH INSTITUTE 1 Temozolomide Oncology Drug Advisory Committee March 13, 2003 Craig L. Tendler, M.D. Vice President, Oncology.
Investigational New Drug Application (IND)
Clinical Trial Review and Approval: New Regulations and their implications Siddika Mithani, Ph.D Clinical Trials & Special Access Programme Therapeutic.
Animal Models for Porcine Xenotransplantation Products Intended to Treat Type 1 Diabetes or Acute Liver Failure CTGTAC #47 May 14, 2009.
Nonclinical Perspective on Initiating Phase 1 Studies for Small Molecular Weight Compounds John K. Leighton, PH.D., DABT Supervisory Pharmacologist Division.
Investigational Drugs in the hospital. + What is Investigational Drug? Investigational or experimental drugs are new drugs that have not yet been approved.
Key Compliance Risks in Clinical Trials Kathleen Meriwether Principal, ERNST & YOUNG, LLP Fraud Investigation & Dispute Services.
Supporting Informed Formulary Decision Making: CADTH’s Common Drug Review Denis Bélanger, Director, CADTH New Brunswick Stroke Summit November 27, 2010,
Single Patient Use of Investigational Anticancer Agents: An Industry Perspective Gerard T. Kennealey, MD Vice President, Clinical Research, Oncology AstraZeneca.
Proposal for End-of-Phase 2A (EOP2A) Meetings Advisory Committee for Pharmaceutical Sciences Clinical Pharmacology Subcommittee November 17-18, 2003 Lawrence.
The FDA: Basic Facts It takes 12 to 15 years to develop a single drug Only 1 in 10,000 potential medications makes it completely through the process Only.
History of Pediatric Labeling
U.S. Food and Drug Administration Notice: Archived Document The content in this document is provided on the FDA’s website for reference purposes only.
Questions to the Committee. Question 1. The Agency has accepted durable responses in hematologic malignancies for approval for both chronic leukemias.
1 Operation of the Prescription Drug User Fee Program Janet Woodcock, M.D. Deputy Commissioner for Operations November 14, 2005.
1 Pharmacists’ Roles in Emergency Preparedness CDR Louis Flowers LT Evan Wearne.
General Regulatory Issues in the Development of Drugs Intended for Treatment of Chronic Illness Sharon Hertz, M.D. Medical Officer Division of Anesthetic,
Research in the Office of Vaccines Research and Review: Vision and Overview Jesse Goodman, M.D., M.P.H. Director, Center for Biologics Evaluation and Research.
October 28, F OOD AND DRUG ADMINISTRATION AMENDMENTS ACT OF 2007 (FDAAA) and Risk Evaluation and Mitigation Strategies (REMS) Presented to the Ninth.
Research in the Office of Cellular, Tissue and Gene Therapies: Vision and Overview Jesse Goodman, M.D., M.P.H. Director, Center for Biologics Evaluation.
Initiatives Drive Pediatric Drug Development January 30, 2002.
Regulatory Considerations for Endpoints Ann T. Farrell, M.D. FDA/CDER/DODP.
Center for Biologics Evaluation and Research, FDA Site Visit Introduction Kathryn M. Carbone, M.D. Associate Director for Research.
Agency Review of sNDA SE-006 DOXIL for Ovarian Cancer Division of Oncology Drug Products Office of Drug Evaluation 1 Center for Drug Evaluation.
FDA Risk Communication Nancy M. Ostrove, PhD Senior Advisor for Risk Communication Risk Communication Advisory Committee February 28, 2008.
The Regulation on Cell Therapy Products in Japan
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.
November 9, 2015 February 20, 2017 Using real world evidence – industry perspective Pma indication expansion Melissa hasenbank, phd Sr. Clinical Research.
My Experiences as an FDA Statistician
Regulatory– Terms & Definitions רגולציה - מונחים והגדרות
FDA’s IDE Decisions and Communications
How to Put Together an IDE Application
Clinical Trials — A Closer Look
Expedited Drug Approval Programs
FDA Perspective on Cardiovascular Device Development
Balancing Pre and Postmarket Requirements Different Scenarios
Reasonable Assurance of Safety and Effectiveness: An FDA Division of Cardiovascular Devices Perspective Bram Zuckerman, MD, FACC Director, FDA Division.
Medical Device Regulatory Essentials: An FDA Division of Cardiovascular Devices Perspective Bram Zuckerman, MD, FACC Director, FDA Division of Cardiovascular.
Bozeman Health Clinical Research
Speeding access to therapies
Streamlining IRB Procedures for Expanded Access
Suzanne M. Sensabaugh, MS, MBA
Phase 2 to phase 3 clinical trial transitions: Reasons for success and failure in immunologic diseases  Dhavalkumar D. Patel, MD, PhD, Christian Antoni,
IND Review Process Seoul National University
Patient Involvement in the Development and Safe Use of
Pediatric Therapeutics Still working to get it right for kids
Presentation transcript:

Office of Hematology and Oncology Products Ann T. Farrell, M.D. Division Director Division of Hematology Products Office of Hematology and Oncology Products Center for Drug Evaluation and Research

Disclosure I have nothing to disclose and will not discuss off-label uses.

FDA Mission protect the public health by assuring the safety, effectiveness, quality, and security of human and veterinary drugs, vaccines and other biological products, and medical devices. responsible for the safety and security of most of our nation’s food supply, all cosmetics, dietary supplements and products that give off radiation. responsible for regulating tobacco products

FDA Structure Center for Drug Evaluation and Research (CDER) Regulate Drugs and Biologics except for vaccines, blood and plasma products and cellular based therapies – 16 review divisions Center for Biologic Evaluation and Research (CBER) Cellular therapies - Office of Cellular, Tissue and Gene Therapy Vaccines – Office of Vaccines Blood and Plasma Products – Office of Blood Research and Review

Four Main Areas that Impact Public Determine whether protocols submitted under an investigational new drug (IND) application are safe to proceed Approve Drugs/Biological Products for widespread commercial marketing Work with Pharmaceutical Manufacturers to manage production and safety issues that arise Permit access to drugs or biological products in development for compassionate use

FDA and You Patient Advisors – Special Government Employees Consultation Advisory Committee Meetings -- ODAC Patient Focused Drug Development Meetings – 5th authorization of Prescription Drug User Fee Act/Food Drug Administration Safety and Innovation Act (July 9, 2012) – 5 years (lung cancer – July 2013, sickle cell disease – February 2014) CDER’s Center Director Office – Professional Affairs and Stakeholder Engagement Federal Register Notice – 11/4/14 – wants suggestions Professional Affairs and Stakeholder Engagement -a focal point for advocacy, to enhance two-way communication and collaboration with healthcare professionals, patients, patient groups, and others on CDER issues concerning drug development, drug review, and drug safety.

Division Structure Divisions organized by area of expertise Project Manager (PM) coordinates multidisciplinary teams Discipline-specific teams Primary and secondary review Within the Center for Drug Evaluation and Research, there are 16 different divisions which are organized by areas of expertise. The RHPM or CSO coordinates a multidisciplinary team. Discipline specific teams exist with a primary reviewer and a team leader who performs primary and secondary reviews respectively.

Staff with Significant Expertise Reviewers are Clinicians – Medical Officers, Physician Assistants, Nurse Practitioners Chemists, Biologists, Microbiologists Pharmacologists/Toxicologists Clinical Pharmacologists Statisticians Epidemiologists Social Scientists, Ethicists Regulatory Expertise - Lawyers, Project Managers External Advisors (Special Government Employees)

Drug Development Timeline End of Phase I Meeting End of Phase II Meeting IND Submission Clinical Start Pre-NDA Meeting NDA/BLA IND Review Annual reports IND Amendments 30 days Clinical hold Phase I Phase II Phase III

Orderly and Logical Progression of Studies Lab studies first, then animal, then human Small studies before large Short studies before long Low dose before high dose Each step builds on prior experience As outlined on the slide, drug development is an orderly process. The process starts with lab studies and then progresses into animal studies and then clinical humans trials. In general Small studies before large Short studies before long Low dose before high dose Each step builds on prior experience

Oncology Drug Development Lab studies first, then animal, then human Small studies before large ones Phase 1 (dose escalation accelerated titration 1 by 1 or 3 plus 3) Phase 2 (sometimes main trial for accelerated approval) Available therapy Unmet need Risk/benefit Phase 3 (usually one trial – pivotal trial) Each step builds on prior experience Phase 4 studies post-approval to address safety/efficacy issues

Investigational New Drug (IND) Determine whether protocols submitted under an investigational new drug (IND) application are safe to proceed Research (physician working at a university/center) Commercial (Pharmaceutical Sponsors or other) Expanded Access Intermediate protocol Single Patient Emergency

IND Formal application to the Agency to conduct research Requirements for submission FDA has 30 days to get back to the sponsor regarding whether the trial is safe to proceed or not Team reviews Deficiencies – notify the sponsor and recommend how to correct them If not corrected --- clinical hold

INDs Fast Track Designation – nonclinical data Break Through Designation – clinical data Focused attention on the development program

Basis for NDA and BLA Approval Demonstration of efficacy with acceptable safety in adequate and well-controlled studies Ability to generate product labeling that Defines an appropriate patient population for treatment with the drug Provides adequate information to enable safe and effective use of the drug So when we approve a drug or biologic product, we generally have sufficient information from adequate and well-controlled studies to generate product labeling. The product labeling must define the patient population for whom the treatment is used and adequate information to enable safe and effective use. I am going to stress a couple of points here: we have to be able to describe for whom the treatment should be prescribed and that is guided by the data submitted for review because in general the study population is what goes into the description we have to be able to describe a dosing regimen, what the expected benefits and what the expected risks are

Requirements for Drug Approval Safety (FDAC 1938) Efficacy established in adequate and well-controlled investigations (FDAC 1962) Use of data from one trial plus supportive evidence (FDAMA 1997)

Approval Accelerated or Regular Regular Approval Accelerated Approval Based on clinical benefit Accelerated Approval Affect a surrogate endpoint other than mortality or irreversible morbidity Surrogate endpoint must be reasonably likely to predict clinical benefit, based on epidemiologic, therapeutic, pathophysiologic, or other evidence Additional studies confirming clinical benefit must be completed after approval

Clinical Benefit Improvement in survival Improvement how a patient feels Improved functioning Clinical benefit must be demonstrated to receive regular approval. Clinical benefit is defined as prolonged patient survival or an improvement in how a patient feels or functions. Cancer and hematologic conditions often cause symptoms that lead to patient suffering. Improvement in symptoms of cancer, properly measured, using an instrument that is ‘fit for purpose’ in the intended patient population, could provide evidence of clinical benefit and support a regular approval. A symptom endpoint may support a claim of treatment benefit if it is used as a primary or secondary endpoint in adequate and well-controlled oncology or hematology clinical trials and if the studies also demonstrate a favorable treatment effect on the disease. The Office of Hematology and Oncology Products works with Sponsors to incorporate Clinical Outcome Assessments into their drug development plans when appropriate. 18 18

Accelerated Approval Serious and life-threatening illness New therapy must provide advantage over available therapy ability to treat patients unresponsive to or intolerant of improved patient response

Evidence for Accelerated Approval Substantial evidence from well-controlled clinical trials regarding a surrogate endpoint NOT: Borderline evidence regarding a clinical benefit endpoint

Greatest Potential for Problems Chemistry, Manufacturing and Control Efficacy Data Safety Data Clinical Data goes to an Oncologics Drug Advisory Committee (AC) Meeting or other AC Meet several times a year to discuss scientific issues

How many trials? Usually more than one trial is needed. Substantial evidence: “Adequate and well-controlled investigations” Sometimes a single trial may suffice. FDAMA (1997) single trial plus other supportive evidence 1998 FDA Effectiveness Guidance: Statistically strong evidence Multicenter trial Important clinical benefit Additional trials not ethical

Review Time Standard 10 months plus 2 months Priority 6 months plus 2 months Expedited less than 6 months (usually)

Oncologic Drugs Advisory Committee Meetings November 2014 – Panobinostat and Triferic Applications September 2013 – Perjeta in combination with trastuzumab/docetaxel demonstrating neoadjuvant pathologic CR May 2013 – Animal data for approval of agents to treat radiation-induced myelosuppression associated with a radiological/nuclear incident November 2011 – Facilitating anticoagulant drug development in pediatrics

2014 NME Recent Approvals Cyramza (ramucirumab) – gastric cancer Sylvant (siltuximab) – multicentric Castleman’s disease Zykadia – non-small cell lung cancer Belodaq (bellinostat) – peripheral t-cell lymphoma Zydelig – CLL, Follicular B-cell non-Hodgkins Lymphoma, small cell lymphocytic lymphoma Keytruda – melanoma

Permit access Permit access to drugs or biological products in development for compassionate use widespread – commercial marketing not marketed – emergency INDs, Single Patient INDs physician working with a pharmaceutical sponsor process which includes providing documentation, letters, etc.

Risk/Benefit

Ponatinib- original indications kinase inhibitor indicated for the treatment of adult patients with chronic phase, accelerated phase, or blast phase chronic myeloid leukemia (CML) that is resistant or intolerant to prior tyrosine kinase inhibitor therapy or Philadelphia chromosome positive acute lymphoblastic leukemia (Ph+ALL) that is resistant or intolerant to prior tyrosine kinase inhibitor therapy The indication on this slide is for the original approval of Iclusig. There is no test specified here because the requirement for a test was not considered necessary for the product to be used. Although much of the development was for patients whose disease had the T315I mutation, there was data showing a similar response rate for those without the mutation.

Ponatinib original approval Efficacy (MCyR) for CML Relapsed CML – 49% CML with T315I -70% Accelerated Phase CML (CHR) – 47% Blast Phase CML (CHR) – 21% Ph+ Acute Lymphoblastic Leukemia – 34% Safety Arterial thrombosis – 8% (MI, Stroke, gangrene)

Ponatinib approval October 2013 – voluntarily withdrawn from market available via expanded access protocol December 2013 – returns to market Efficacy – unchanged Safety – Changed Box Warning to reflect Increase in Vascular Occlusive Events – 27% New warning for Heart Failure

Ponatinib – latest indications Treatment of adult patients with T315I-positive chronic myeloid leukemia (chronic phase, accelerated phase, or blast phase) or T315Ipositive Philadelphia chromosome positive acute lymphoblastic leukemia (Ph+ ALL). Treatment of adult patients with chronic phase, accelerated phase, or blast phase chronic myeloid leukemia or Ph+ ALL for whom no other tyrosine kinase inhibitor (TKI) therapy is indicated. The Agency takes its post-marketing surveillance seriously, and after the percentage of adverse events appeared to triple in a short time frame, we felt it best to restrict the drug for those patients who truly needed it. Those who were T315I positive or those who had no other options. The decision on whether it was absolutely necessary for a test can be summed up this way – you can get here either by multiple trials with various approved TKIS or get mutation testing done in either instance you would be a candidate for this product. So the indication was felt to be low risk from a CDRH perspective for a companion diagnostic.

References www.fda.gov