Protocol Development, Design and Analysis of Phase 1 Clinical Trials in Cancer Therapy

Similar documents
Overview of Phase 1 Oncology Trials of Biologic Therapeutics

Session 6 Clinical Trial Assessment Phase I Clinical Trial

Newsletter. WntResearch AB, Medeon Science Park, Per Albin Hanssons väg 41, Malmö, Sweden. Primary Objective:

Clinical Trial Design. Sponsored by Center for Cancer Research National Cancer Institute

CLINICAL TRIALS SHOULD YOU PARTICIPATE? by Gwen L. Nichols, MD

CLINICAL TRIALS: Part 2 of 2

NONCLINICAL EVALUATION FOR ANTICANCER PHARMACEUTICALS

The Clinical Trials Process an educated patient s guide

ICH M3 (R2) Guideline on Nonclinical Safety Studies for the Conduct of Human Clinical Trials and Marketing Authorization for Pharmaceuticals

Guidance for Industry

The Promise and Challenge of Adaptive Design in Oncology Trials

Guidance for Industry

Not All Clinical Trials Are Created Equal Understanding the Different Phases

The Trials and Tribulations of the CRM: the DFO Experience

COMMITTEE FOR PROPRIETARY MEDICINAL PRODUCTS (CPMP) NOTE FOR GUIDANCE ON THE PRE-CLINICAL EVALUATION OF ANTICANCER MEDICINAL PRODUCTS

GENERAL CONSIDERATIONS FOR CLINICAL TRIALS E8

Clinical Trial Results Database Page 1

Guidance for Industry

Clinical Trial Designs for Incorporating Multiple Biomarkers in Combination Studies with Targeted Agents

GT-020 Phase 1 Clinical Trial: Results of Second Cohort

Clinical Trial Protocol Development. Developed by Center for Cancer Research, National Cancer Institute Endorsed by the CTN SIG Leadership Group

ICH Topic E 8 General Considerations for Clinical Trials. Step 5 NOTE FOR GUIDANCE ON GENERAL CONSIDERATIONS FOR CLINICAL TRIALS (CPMP/ICH/291/95)

Pharmacology skills for drug discovery. Why is pharmacology important?

Challenges in the Regulation of Pediatric Clinical Trials

Guidance for Industry

Proof-of-Concept Studies and the End of Phase IIa Meeting with the FDA

INTERNATIONAL CONFERENCE ON HARMONISATION OF TECHNICAL REQUIREMENTS FOR REGISTRATION OF PHARMACEUTICALS FOR HUMAN USE S1A. Current Step 4 version

Bayesian Phase I/II clinical trials in Oncology

ICH Topic S 1 A The Need for Carcinogenicity Studies of Pharmaceuticals. Step 5

Anticancer Drug Clinical Trial Guideline. (version 2.0)

Achieving Regulatory Success: Areas of focus for biotechnology companies. Michael J. Schlosser, PhD, DABT April 21, 2013

Lung Pathway Group Nintedanib (Vargatef) in advanced Non-Small Cell Lung Cancer (NSCLC)

Lead optimization services

TGN 1412 Welche Änderungen haben sich für die Erstanwendung am Menschen aus Sicht des BfArM ergeben?

William B. Smith, MD President. Patrick R. Ayd, RN, MBA Chief Operating Officer. SNBL-CPC Baltimore, Maryland

Guideline for Industry

INTERNATIONAL CONFERENCE ON HARMONISATION OF TECHNICAL REQUIREMENTS FOR REGISTRATION OF PHARMACEUTICALS FOR HUMAN USE. Current Step 4 version

Goals & Objectives. Drug Development & the FDA Pharmacy 309. Outline. An History of Disasters. Be able to describe

Ovarian Cancer and Modern Immunotherapy: Regulatory Strategies for Drug Development

Early Drug Discovery and Development Guidelines: For Academic Researchers, Collaborators, and Start-up Companies

Definition of dose-limiting toxicity in phase I cancer clinical trials of molecularly targeted agents

Prior Authorization Guideline

INTERNATIONAL CONFERENCE ON HARMONISATION OF TECHNICAL REQUIREMENTS FOR REGISTRATION OF PHARMACEUTICALS FOR HUMAN USE M3(R2)

Phase 0 Clinical Trials in Cancer Drug Development: From Concept to Practice

Basic Overview of Preclinical Toxicology Animal Models

Subject: No. Page PROTOCOL AND CASE REPORT FORM DEVELOPMENT AND REVIEW Standard Operating Procedure

CTC Technology Readiness Levels

University of Hawai i Human Studies Program. Guidelines for Developing a Clinical Research Protocol

Guidance for Industry Determining the Extent of Safety Data Collection Needed in Late Stage Premarket and Postapproval Clinical Investigations

U.S. Food and Drug Administration

New anticoagulants: Monitoring or not Monitoring? Not Monitoring

Data Management in Clinical Trials

Effective Outsourcing of Clinical Pharmacology Studies in Europe. John Horkulak Executive Director, Eurasian External Clinical Study Operations

4.1 Objectives of Clinical Trial Assessment

New Advances in Cancer Treatments. March 2015

Guidance for Industry

BIOAVAILABILITY & BIOEQUIVALENCE TRIALS

An introduction to Phase I dual-agent dose escalation trials

Drug Development Process

Non-clinical development of biologics

Biological importance of metabolites. Safety and efficacy aspects

Guidance for Industry FDA Approval of New Cancer Treatment Uses for Marketed Drug and Biological Products

Brigham and Women s Hospital, Boston, MA, USA; 2 Verastem, Inc., Boston, MA, USA

Targeting Specific Cell Signaling Pathways for the Treatment of Malignant Peritoneal Mesothelioma

Post-operative intrapleural chemotherapy for mesothelioma

Overview of Drug Development: the Regulatory Process

Current version dated 5 March 2012

RADIOPHARMACEUTICALS BASED ON MONOCLONAL ANTIBODIES

Guidance for Industry

ADDPLAN DF: Designing More Accurate Dose Escalation Studies in Exploratory Drug Development. An ICON White Paper

Future Directions in Clinical Research. Karen Kelly, MD Associate Director for Clinical Research UC Davis Cancer Center

2 Year ( ) CLL/SLL Research Initiative CLL/SLL Collaborative Grant

This clinical study synopsis is provided in line with Boehringer Ingelheim s Policy on Transparency and Publication of Clinical Study Data.

Longitudinal Modeling of Lung Function in Respiratory Drug Development

NATIONAL HEALTH COUNCIL RESOLUTION Nº 251, DATED 7 AUGUST 1997

Docetaxel + Carboplatin + Trastuzumab (TCH) Adjuvant Breast Cancer

Clinical Study Synopsis

SAFETY PHARMACOLOGY STUDIES FOR HUMAN PHARMACEUTICALS S7A

Statistics and Pharmacokinetics in Clinical Pharmacology Studies

Van Cutsem E et al. Proc ASCO 2009;Abstract LBA4509.

Cancer Treatments Subcommittee of PTAC Meeting held 18 September (minutes for web publishing)

Regulatory Pathways for Licensure and Use of Ebola Virus Vaccines During the Current Outbreak FDA Perspective

NEW CLINICAL RESEARCH OPTIONS IN PANCREATIC CANCER IMMUNOTHERAPY. Alan Melcher Professor of Clinical Oncology and Biotherapy Leeds

Clinical Study Synopsis for Public Disclosure

Sheffield Kidney Institute. Planning a Clinical Trial

Alterações empresariais sustentadas pelo conceito de engenharia do Produto Patrício Soares da Silva, MD, PhD

The CRM for ordinal and multivariate outcomes. Elizabeth Garrett-Mayer, PhD Emily Van Meter

The NCPE has issued a recommendation regarding the use of pertuzumab for this indication. The NCPE does not recommend reimbursement of pertuzumab.

BCCA Protocol Summary for Palliative Therapy for Metastatic Breast Cancer using Trastuzumab Emtansine (KADCYLA)

Expanded Access Programs. Richard Klein Office of Special Health issues Food and Drug Administration

Edward Odenkirchen, Ph.D. Office of Pesticide Programs US Environmental Protection Agency

Guidance for Industry Safety Testing of Drug Metabolites

POLICY AND PROCEDURES OFFICE OF NEW DRUGS. Good Review Practice: Management of Breakthrough Therapy-Designated Drugs and Biologics.

What is the Optimal Front-Line Treatment for mrcc? Michael B. Atkins, MD Deputy Director, Georgetown-Lombardi Comprehensive Cancer Center

Management of low grade glioma s: update on recent trials

Roles & Responsibilities of the Sponsor

Bios 6648: Design & conduct of clinical research

1.0 Abstract. Title: Real Life Evaluation of Rheumatoid Arthritis in Canadians taking HUMIRA. Keywords. Rationale and Background:

Clinical trials for medical devices: FDA and the IDE process

Transcription:

Protocol Development, Design and Analysis of Phase 1 Clinical Trials in Cancer Therapy November 2016 S. Gail Eckhardt, M.D. Professor of Medicine/Oncology Director, Phase I Program University of Colorado Cancer Center

Conflicts I have no relevant conflicts to disclose

Types of Phase 1 Trials Dose finding, first-in-human Food effect study QTc prolongation study Bioequivalence study Approved or investigational agent with pharmacokinetic focus (adding of CYP inhibitor) Typically considered drug-drug interaction study Investigational agent + investigational agent Investigational agent + approved agent(s) Approved agent + approved agent(s) Approved or investigational agent with pharmacodynamic focus (e.g. evaluation using functional imaging) Approved or investigational agent with radiotherapy

What Is Needed To Do A Phase I Study? An Investigational New Drug Application (IND) Provides a means of advancing from pre-clinical to clinical testing (first-in-human) Required for products that have not received marketing approval May be required for already marketed products A formal application to study an intervention in patients Sponsor: usually a pharmaceutical company, but can be an academic institution Investigator

IND Application Should include: Chemistry, manufacturing, and control information Animal pharmacology and toxicology Genotoxicity Toxicology Histopathology Non-clinical justification for proposed dose/schedule Justification for duration of treatment Prior use in humans if applicable Clinical protocol and investigator information

Protocol Development: What are the Primary Endpoints of a Phase I Trial? To characterize and quantify the toxicities of a new agent. To determine the dose limiting toxicities (DLTs) and maximum tolerated dose (MTD). To characterize the pharmacology of the agent. To assess overall tolerability and feasibility. Note that in oncology, cancer patients traditionally participate in Phase I testing, not normal volunteers (although some drugs are tested in both).

What are Some Examples of Secondary Endpoints in Phase I Trials? To assess the extent of preliminary anti tumor activity. To determine if there are pharmacokinetic/ pharmacodynamic relationships (PK/PD). For targeted agents, to assess the feasibility and utility of a biological correlative assay. Increasingly phase I trials have numerous secondary endpoints that sometimes are used as go/no go decision points for moving forward with a drug

What are the Key Protocol Elements Required to Accomplish these Objectives? Justification of the starting dose and dose escalation plan. Appropriate patient eligibility/ exclusion criteria. Definition of DLT, MTD, and recommended phase II dose. Pharmacokinetic sampling schedule. A study calendar for safety assessments. Tumor assessment criteria and schedule. Reporting requirements for adverse events and serious adverse events. Consent form and other regulatory items.

Definitions of Key Concepts in Phase 1 Trials Dose limiting toxicity (DLT): Toxicity that is considered unacceptable (due to severity and/or irreversibility) and limits further dose escalation Defined with standard criteria CTCAE 4.1 (criteria that standardize language and severity of adverse events) DLT defined in advance prior to beginning the trial is protocol specific Typically defined based on toxicity seen in the first cycle Limits dose escalation (dose limiting) With select agents that have a more delayed toxicity (eg 2 nd /3 rd cycle), time allowed for DLT definition being reevaluated

Typical Definitions of DLT Dose escalation proceeds, as planned until DLT occurs, as typically defined by: Grade 4 neutropenia for > 5 days, or complicated by fever requiring antibiotics. Platelets < 25, 000/uL. Non hematologic toxicity > grade 3 (tailored for agent). Chronic > grade 2 toxicity. Once DLT occurs, more patients are treated to assess the frequency.

Definitions of Key Concepts in Phase 1 Trials Examples of chronic DLTs can be associated with daily dosing: Threshold for DLTs is lower Some Grade 2 toxicities may be unacceptable and intolerable due to their persistence and lack of time period for recovery Examples: Grade 2 intolerable or worse non hematologic toxicity despite supportive measures: Nausea, vomiting Grade 3 or worse hematologic toxicity Inability to complete a pre specified percentage of treatment during the cycle due to toxicity (e.g. missing 10 15% of doses)

Definitions of Key Concepts in Phase 1 Trials Maximum Administered Dose (MAD), Maximum Tolerated Dose (MTD): confusing Usage of these 2 phrases varies with country Typically, MTD is used and generally defined as the highest dose where less than 30% of patients have DLT in first cycle Should treat enough patients at MTD to provide confidence that it is tolerable Recommended Phase 2 Dose (RPTD or RD): Dose associated with DLT in a pre specified proportion of patients (e.g. < 33%) dose that will be used in subsequent phase 2 trials

Why the MTD is Not Always Equivalent to the RP2D!!! DLT and MTD are used for the purposes of making decisions about dose escalation. The occurrence of DLT in the first course of therapy is what limits dose escalation. The phase II dose should incorporate multiple cycle toxicity (including DLT), tolerability, dose reductions, and dose delays. This distinction should be clarified in the protocol.

Definitions of Key Concepts in Phase 1 Trials Optimal biological dose (OBD): Dose associated with a pre specified desired effect on a biomarker Examples: Dose at which > X% of patients have inhibition of a key target in tumor/surrogate tissues Dose at which > X% of patients achieve a pre specified immunologic parameter Dose at which > X% of patients achieve a specific drug exposure that correlates with preclinical activity Challenge with defining OBD is that the desired effect on a biomarker may not be known or validated before initiation of the phase 1 trial

Definitions of Key Concepts in Phase 1 Trials Expanded Cohort: Enrollment of patients at the end of a Phase I trial of a particular tumor type and/or genetic subtype. Increasingly utilized in Phase I trials and results have been submitted to the FDA for NDAs (crizotinib). At times can be as large as mini Phase II and these designs are criticized for not having statistical parameters Often used to justify launch of Phase III following Phase II or for accelerated approval

Phase 1 Trials: Fundamental Questions At what dose do you start? What type of patients? How many patients per cohort? How quickly do you escalate?

Preclinical Toxicology Typically a rodent (mouse or rat) and non rodent (dog or non human primate) species Reality of animal organ specific toxicities very few predict for human toxicity Myelosuppression and GI toxicity more predictable Hepatic and renal toxicities large false positive Toxicologic parameters: LD 10 lethal dose in 10% of animals TDL (toxic dose low) lowest dose that causes any toxicity in animals NOAEL no observed adverse effect level

Justification of the Starting Dose 1/10th of the dose that leads to deaths in 10% (LD 10 ) of rodents is used in man. 1/6 1/3rd of the lowest toxic dose (TDL) is used in larger animals This dose is usually in mg/kg, so it must be converted to mg/m 2 for man and there are conversion tables for this. If toxicology is performed in several species, then the starting dose is used from the most sensitive species. This is very rudimentary and traditional, a lot of work is going into refining these rules, particularly with targeted agents

Phase 1 Trials: Fundamental Questions At what dose do you start? What type of patients? How many patients per cohort? How quickly do you escalate? What are the endpoints?

Patient Eligibility Criteria: Inclusion The purpose of inclusion criteria are to define the minimum requirements of the phase I population under study: Regulatory: Signed consent, age > 18. Disease: Failed standard therapy (or no standard therapy exists), histologically confirmed. Constitutional: Performance status 0 1/2 (out of bed > 50%) Organ function: ANC > 1500/uL; plts > 100 K/uL; hgb > 9 g/dl; creatinine < 1.5 g/dl, SGPT/SGOT 2 5 X ULN, total bilirubin < ULN. Mental: Able to comprehend and comply with study requirements.

Patient Eligibility Criteria: Exclusion The purpose of exclusion criteria are to minimize the risks of drug related toxicity, thus some are agent specific: Constitutional: Almost all phase I studies exclude pregnancy due to the lack of information on fetal effects, as well as other serious underlying medical diseases. Prior therapy: For myelosuppressive compounds, patients with extensive prior therapy are excluded. Cardiac: Patients may be excluded with known underlying diseases, or may require a MUGA, EKG, etc. Gastrointestinal: Studies of oral drugs exclude patients with refractory N/V or extensive bowel surgery.

Phase 1 Trials: Fundamental Questions At what dose do you start? What type of patients? How many patients per cohort? How quickly do you escalate? What are the endpoints?

Phase 1 Trial Basic Principles Begin with a safe starting dose Minimize the number of pts treated at sub toxic (and thus maybe sub therapeutic) doses Escalate dose rapidly in the absence of toxicity Escalate dose slowly in the presence of toxicity

Phase 1 Trial Assumptions The higher the dose, the greater the likelihood of efficacy Dose related acute toxicity regarded as a surrogate for efficacy Highest safe dose is dose most likely to be efficacious This dose effect assumption is primarily for cytotoxic agents and may not apply to molecularly targeted agents

Mechanism Based Toxicity With drugs that interact with a target on normal tissues, mechanism based toxicity is a type of biomarker: skin rash, diarrhea EGFR agents hypertension, proteinuria VEGF agents Overall, not unlike the myelosuppression and GI effects (mucositis, diarrhea) observed with traditional cytotoxic agents If a tissue based biomarker is difficult to obtain, mechanism based toxicity may establish biological activity and you don t need to biopsy the patient! This is why reaching an MTD with a biological agent may in fact still be relevant

Dose Escalation Scheme and Schedule Factors to be considered Are there significant inter species differences in tolerance? (may want faster escalation) Is the drug a typical cytotoxic agent or more targeted agent? What is the PK half life? (short vs. long) Does the therapeutic index appear narrow? (slower dose escalation) Is cumulative toxicity an issue? (may need to assess longer than 30 days for dose limiting toxicity) Is there schedule dependent toxicity or efficacy? (continuous vs. intermittent) What is the formulation? (can t give daily IV)

Frequently Used Dose Escalation Schemes Modified Fibonacci: sequentially smaller increments of dose escalation 100%, 67%, 50%, 40%, 33%, etc. Doubling method: doubling of the dose until any grade 2 drug related toxicity, then reversion to modified Fibonacci or fixed (25 40%) dose escalation. Pharmacologically guided: use of real time PK data to guide escalation with intent to achieve target AUC. Continual Reassessment Method (CRM): statisticallyguided dose escalation that uses real time toxicity data to predict the dose closest to the MTD. Accelerated Titration Design: a constellation of designs that utilize single patient toxicity guided escalations (intra patient dose escalation allowed).

Phase I Standard 3 + 3 Design Bottom line is that there are many ways to design a dose escalation scheme and a lot of flexibility as long as it is safe for patients!

Dose Escalation: Lessons Learned Adaptive dose escalation methods have demonstrated that cohort size can be toxicity based and not fixed at 3 or 6. With biological agents, its important to enroll sufficient numbers of patients to assess dose response relationships of PK and PD. Currently, the trend is towards standard cohort sizes during dose escalation with cohort expansion around the MTD to 10 12 patients There are also novel adaptive designs in development that can incorporate PK/PD data and enhance enrollment around biologically active dose levels

Clinical Development of Novel Agents Challenges 1. Difficulty in determining the optimal dose/schedule in phase I MTD versus biologically efficacious dose? 2. Absent or low level tumor regression as single agents in unselected patients Predictive biomarker may be needed early to enhance efficacy Problematic for making decisions regarding phase II trials 3. Need for large randomized trials to definitively assess clinical benefit This can result in underpowered randomized phase II trials or high risk phase III trials

Approaches That Have Been Utilized in Early Clinical Trials of Novel Agents Novel/rapid dose escalation Use of biological correlative studies to assess biological effects Alternative endpoints to the Maximum Tolerable Dose (MTD) Optimal Biological Dose (OBD) Functional imaging studies (PET, DCE MRI) to determine tumor derived biological effects noninvasively. Revision of response criteria to include disease stability as a clinical benefit Early phase I combination studies with chemotherapy ( Octopus trials).

Biological Correlative Studies: Why are They Important? In early clinical trials of targeted agents: Normal tissue may not be used as a surrogate for effects, as is the case with standard cytotoxic agents The relative lack of toxicity to normal tissue often means that deriving the MTD, or optimal biological dose may be difficult. Therefore, biological correlative studies may be used to derive the best dose and schedule of an agent, and To determine whether the drug is inducing a biological effect in the patient before advancing to phase II.

Chronic and cumulative toxicities usually cannot be assessed & may be missed Most patients do not stay on trial beyond 2 cycles Uncommon toxicities will be missed Too few patient numbers in a phase 1 trial Reason for toxicity evaluation & reporting through phase IV drug testing Exactly what tumor or tumor cell subset are you treating Heterogeneity Resistance Pitfalls of Phase 1 Trials

Phase I Trial Combination Trials Combination phase I trials: New drug A + Standard drug B Need to provide rationale: why add A to B? Need to think about overlapping toxicity in your definition of DLT Ideally keep standard drug dose fixed and escalate the new drug (e.g. 1/2, 2/3, full dose)

Process for determination of the Phase I combination trial design: pharmacodynamic (PD); pharmacodinetic (PK) 1. Provide explicit or implicit hypothesis justifying the combination, including a pharmacologic or biologic rationale. 2. State potential clinical results from phase I and hypotheses for future studies. 3a. If overlapping DLT(s)or plausible basis for PD leading to DLT(s) 3b. If no overlapping DLT and no plausible PD, but plausible PK interaction 3c. If no plausible basis for PK or PD interaction 4a. Use formal phase I with success/failure criteria stated a priori. 4b. Use a formal drug drug interaction design primary endpoint PK. 4c. No formal phase I required. Consider tolerability as first phase (run in) of phase II study. Paller, Bradbury, et al. Clin Cancer Res; 20(16); 1-8, 2014

IDSC Consensus Recommendations for Evaluation of Combination Therapies Recommendations 1 All phase I combination trials should state an explicit or implicit hypothesis justifying the combination, including a pharmacological or biological rationale that includes at least one of the following: in vitro data, in vivo data, or clinical data. The rationale may extrapolate from results with similar drugs and may be based on in silico analyses. The hypothesis supporting the combination should be clearly stated in the protocol. 2 The potential results and next steps of the development plan of the combination should be clearly described. The description should include two parts: the rationale for why the biologic or pharmacologic interactions should translate into clinical effects, and one or more examples of phase II studies to test the hypothesis. The phase II example(s) should follow the guidelines of the Phase II Consensus Recommendations of the NCI s IDSC CTD Task Force. 3 The design of combination phase I studies should address the following three factors: overlapping dose limiting toxicities (DLTs); a plausible mechanistic basis for a pharmacodynamic interaction leading to DLTs; and a plausible mechanistic basis for a pharmacokinetic interaction. 4 Selection of the clinical trial design should be based on the scientific rationale, underlying data and hypothesis for the combination, and the intended development plan for the combination A B C Combination therapies with overlapping DLTs or a plausible basis for a pharmacodynamic interaction leading to DLTs require formal phase I evaluation. The selected doses to be studied should be justified based on the specific phase II plans. Combinations without overlapping DLTs and without a plausible basis for pharmacodynamic interaction, but with a plausible pharmacokinetic interaction, should be studied using a formal drug-drug interaction design. The primary endpoint is pharmacokinetics. Crossover design is often optimal. Combinations without overlapping toxicities, without a plausible basis for a pharmacodynamic interaction leading to a DLT, and without a plausible basis for a pharmacokinetic interaction do not require a formal phase I study. Design of Phase I Combination Trials: Recommendations of the Clinical Trial Design Task Force of the NCI Investigational Drug Steering Committee: Clin Cancer Res 2014;20:4210 17.

Opportunities/Pitfalls: Opportunities Combination Therapies Pitfalls Validate novel biological hypotheses Unreliable pre clinical models Synergize anti tumor effect without synergizing toxicity Increase therapeutic index/window Synthetic lethality: optimize combination use of single agents with limited single agent activity Counteract primary and secondary resistance Develop novel indications for existing and approved drugs Optimal selection of drugs and targets to study in combination Optimal sequence and dose of combination therapy Risk overlapping toxicity Lack of standard design for phase 1 / 2 for combination therapies Competing interests of researchers, corporations and / or institutions to combine treatments Yap, Omlin & de Bono, JCO 2013

The Successful Phase 1 TEAM The Patient Investigators Scientists Fellows Data Coordinators Pharmacists Lab Personnel: pharmacology, reference, PK, PD Bio statisticians Radiologists, pathologists, IR Finance And many others.

Thank You!