Lillian L. Siu Princess Margaret Hospital

Slides:



Advertisements
Similar presentations
Inhibition of Poly (ADP-Ribose) Polymerase (PARP) by ABT-888 in Patients With Advanced Malignancies: Results of a Phase 0 Trial Shivaani Kummar, MD National.
Advertisements

AbstractSchema Conclusion Pharmacokinetic profile of the base-excision repair inhibitor Methoxyamine-HCl (TRC102; MX) given as an one-hour intravenous.
Phase 1/2 Study of Weekly MLN9708, an Investigational Oral Proteasome Inhibitor, in Combination with Lenalidomide and Dexamethasone in Patients with Previously.
Bayesian Adaptive Methods
Novel AKT Inhibitor Afuresertib in Combination with Bortezomib and Dexamethasone Demonstrates Favorable Safety Profile and Significant Clinical Activity.
HCC Journal Club September 2009 Statistical Topic: Phase I studies Selected article: Fong, Boss, Yap, Tutt, Wu, et al. Inhibition of Poly(ADP-Ribose) Polymerase.
Modified Megestrol The Clinical Trials by : Carolina R. Akib
Richardson PG et al. Proc ASH 2013;Abstract 535.
Roberts AW et al. Proc ASH 2014;Abstract 325.
U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES NATIONAL INSTITUTES OF HEALTH Working with FDA: Biological Products and Clinical Development Critical Path.
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
Phase I trials: A New era in OnCology drug development
Re-Examination of the Design of Early Clinical Trials for Molecularly Targeted Drugs Richard Simon, D.Sc. National Cancer Institute linus.nci.nih.gov/brb.
Novel Trial Designs for Early Phase Drug Development Elizabeth Garrett-Mayer, PhD Associate Professor Director of Biostatistics Hollings Cancer Center.
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.
(a.k.a. Phase I trials) Dose Finding Studies. Dose Finding  Dose finding trials: broad class of early development trial designs whose purpose is to find.
“Simple” CRMs for ordinal and multivariate outcomes Elizabeth Garrett-Mayer, PhD Emily Van Meter Hollings Cancer Center Medical University of South Carolina.
Phase 1 Clinical Studies First-In-Human (FIH) Pharmacologically-Guided Dose Escalation Jerry M. Collins, Ph.D. Developmental Therapeutics Program Division.
Thymidine phosphorylase (TP) upregulation Dose- and time-dependent upregulation of TP in human colon cancer xenografts PaclitaxelDocetaxel.
Ponatinib as Initial Therapy for Patients with Chronic Myeloid Leukemia in Chronic Phase (CML-CP) Cortes JE et al. Proc ASH 2013;Abstract 1483.
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.
Investigational Drugs in the hospital. + What is Investigational Drug? Investigational or experimental drugs are new drugs that have not yet been approved.
Results: In the presence of PD measurements, PK data provided little additional information. By a limited PD sampling the number of patients on target.
Weekly MLN9708, an Investigational Oral Proteasome Inhibitor, in Relapsed/Refractory Multiple Myeloma: Results from a Phase I Study After Full Enrollment.
Experimental Design and Statistical Considerations in Translational Cancer Research (in 15 minutes) Elizabeth Garrett-Mayer, PhD Associate Professor of.
APPLYING PRE-CLINICAL DATA TO CLINICAL STUDIES-I Edward A. Sausville, M.D., Ph.D. Developmental Therapeutics Program National Cancer Institute October.
Palumbo A et al. Proc ASH 2014;Abstract 175.
FDA Case Studies Pediatric Oncology Subcommittee March 4, 2003.
1Bachelot T et al. Proc SABCS 2010;Abstract S1-6.
Is the Continual Reassessment Method Superior to the Standard “3+3” dose escalation scheme? Alexia Iasonos 1 Elyn R. Riedel 1, David.
BASED ON PROTOCOL VERSION 1 SEPTEMBER 2012 A new study evaluating an investigational drug to treat patients with HER2-positive metastatic gastroesophageal.
Ibrutinib in Combination with Bendamustine and Rituximab Is Active and Tolerable in Patients with Relapsed/Refractory CLL/SLL: Final Results of a Phase.
An Ongoing Phase 3 Study of Bosutinib (SKI-606) versus Imatinib in Patients with Newly Diagnosed Chronic Phase Chronic Myeloid Leukemia Gambacorti-Passerini.
Ibrutinib, Single Agent or in Combination with Dexamethasone, in Patients with Relapsed or Relapsed/Refractory Multiple Myeloma (MM): Preliminary Phase.
Rivaroxaban Has Predictable Pharmacokinetics (PK) and Pharmacodynamics (PD) When Given Once or Twice Daily for the Treatment of Acute, Proximal Deep Vein.
Lenalidomide Is Safe and Active in Waldenstrom Macroglobulinemia (WM) 1 Updated Results from a Multicenter, Open-Label, Dose-Escalation Phase 1b/2 Study.
Updated Results of a Phase I First-in-Human Study of the BCL-2 Inhibitor ABT-199 (GDC-0199) in Patients with Relapsed/Refractory (R/R) Chronic Lymphocytic.
Epic: A Phase 3 Trial of Ponatinib Compared with Imatinib in Patients with Newly Diagnosed Chronic Myeloid Leukemia in Chronic Phase (CP-CML) Lipton JH.
C LINICAL R ESEARCH. CONTENTS Drug Development Process Pre – Clinical Studies Clinical Trials Phase I Phase II Phase III Phase IV 2.
Phase I Trials of Chemotherapy and Targeted Agents
C-1 Pegfilgrastim (Neulasta  ) Oncologic Drugs Advisory Committee Pediatric Subcommittee October 20, 2005 Amgen Inc.
Modified release products. Considerations in the evaluation of modified release products Requirements for preparing extended release products. The bioavailability.
12 th Annual CTOS Meeting 2006 AP23573 Induced Long-term Stability in 2 Patients with Desmoplastic Small Round Cell Tumor (#561) Scott Schuetze, Warren.
European Patients’ Academy on Therapeutic Innovation The key principles of pharmacology.
EORTC OSN/CTOS11 Safety of Caelyx combined with ifosfamide in previously untreated adult patients with advanced or metastatic soft tissue sarcomas. Final.
A Multi-Center Phase I/II Trial of Carfilzomib and Pomalidomide with Dexamethasone (Car-Pom-d) in Patients with Relapsed/Refractory Multiple Myeloma Shah.
Phase II Trial of R-CHOP plus Bortezomib Induction Therapy Followed by Bortezomib Maintenance for Previously Untreated Mantle Cell Lymphoma: SWOG 0601.
Erlotinib plus Gemcitabine Compared with Gemcitabine Alone in Patients with Advanced Pancreatic Cancer: A Phase III Trial of the National Cancer Institute.
MM-005: A Phase 1, Multicenter, Open-Label, Dose-Escalation Study to Determine the Maximum Tolerated Dose for the Combination of Pomalidomide, Bortezomib,
| 1 Application of a Bayesian strategy for monitoring multiple outcomes in early oncology clinical trials Application of a Bayesian strategy for monitoring.
Acute Toxicity Studies Single dose - rat, mouse (5/sex/dose), dog, monkey (1/sex/dose) 14 day observation In-life observations (body wt., food consumption,
Romidepsin in Association with CHOP in Patients with Peripheral T-Cell Lymphoma: Final Results of the Phase Ib/II Ro-CHOP Study Dupuis J et al. Proc ASH.
The process of drug development. Drug development 0,8 – 1 mld. USD.
ELIZABETH GARRETT-MAYER (SOME SLIDES BY PAT LORUSSO OF KARMANOS CANCER INSTITUTE WAYNE STATE UNIVERSITY) Phase I Trials of Chemotherapy and Targeted Agents.
The Stages of a Clinical Trial
University of Southern California, Norris Comprehensive Cancer Center
Oki Y et al. Proc ASH 2013;Abstract 252.
Broadening Eligibility Criteria to Make Clinical Trials More Representative Joint Recommendations of the American Society of Clinical Oncology and Friends.
Dose-finding designs incorporating toxicity data from multiple treatment cycles and continuous efficacy outcome Sumithra J. Mandrekar Mayo Clinic Invited.
Statistical Methods for Biotechnology Products II
CTD Module 4: Non-Clinical Studies SPC Relevant Scientific information
Seymour JF et al. Proc ASH 2013;Abstract 872.
Pomalidomide plus Low-Dose Dexamethasone in Myeloma Refractory to Both Bortezomib and Lenalidomide: Comparison of Two Dosing Strategies in Dual-Refractory.
Yang Liu, Anne Chain, Rebecca Wrishko,
Dose setting for a Phase I Clinical Study
Theis Lange and Shanmei Liao
Presentation transcript:

Lillian L. Siu Princess Margaret Hospital Phase I Trials Lillian L. Siu Princess Margaret Hospital

Definition(s) of a phase I trial First evaluation of a new cancer therapy in humans First-in-human, first-in-kind (e.g. the first compound ever evaluated in humans against a new molecular target), single-agent First-in-human, but not first-in-kind (i.e. others agents of the same class have entered human testing), single-agent

Definition(s) of a phase I trial First evaluation of a new cancer therapy in humans Investigational agent + investigational agent Investigational agent + approved agent(s) Approved agent + approved agent(s) Approved or investigational agent with pharmacokinetic focus (e.g. adding of CYP inhibitor to enhance drug levels) Approved or investigational agent with pharmacodynamic focus (e.g. evaluation using functional imaging) Approved or investigational agent with radiotherapy

Objectives of a phase I trial Primary objective: Identify dose-limiting toxicities (DLTs) and the recommended phase II dose (RPTD) Secondary objectives: Describe the toxicity profile of the new therapy in the schedule under evaluation Assess pharmacokinetics (PK) Assess pharmacodynamic effects (PD) in tumor and/or surrogate tissues Document any preliminary evidence of objective antitumor activity

Definitions of key concepts in phase I trials Dose-limiting toxicity (DLT): Toxicity that is considered unacceptable (due to severity and/or irreversibility) and limits further dose escalation Specified using standardized grading criteria, e.g. Common Terminology Criteria for Adverse Event (CTCAE v3.0; v4.0 release in May 2009) DLT is defined in advance prior to beginning the trial and is protocol-specific Typically defined based on toxicity seen in the first cycle

Definitions of key concepts in phase I trials Examples of DLTs – chronic (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 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 I trials Examples of DLTs – intermittent dosing: Generally can tolerate higher degrees of toxicity because the interval between treatments allows for rest and recovery Examples: Grade 3 or worse non-hematologic toxicity despite supportive measures ANC < 0.5 x 109/L for > 5 or 7 days Febrile neutropenia (ANC < 1 x 109/L, fever > 38.5C) Platelets < 25 x 109/L or thrombocytopenic bleeding Inability to re-treat patient within 2 weeks of scheduled treatment

Definitions of key concepts in phase I trials Maximum administered dose (MAD), maximum tolerated dose: confusing More important term: Recommended phase II 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 II trials

Phase I trial design: standard 3+3 design MAD 3 pts Dose DLT 3 pts 3 pts + DLT Recommended dose (some call this MTD in US) 3 pts Adapted from E. Eisenhauer

Definitions of key concepts in phase I trials Optimal biological dose (OBD): Dose associated with a pre-specified desired effect on a biomarker Examples: Dose at which > XX% of patients have inhibition of a key target in tumor/surrogate tissues Dose at which > XX% of patients achieve a pre- specified immunologic parameter Challenge with defining OBD is that the “desired effect on a biomarker” is generally not known or validated before initiation of the phase I trial

Definitions of key concepts in phase I trials Pharmacokinetics (PK): “what the body does to the drug” absorption, distribution, metabolism and excretion PK parameters: Cmax, AUC (drug exposure), t1/2, Clearance, etc. Pharmacodynamics (PD): “what the drug does to the body” e.g. nadir counts, non-hematologic toxicity, molecular correlates, imaging endpoints

Phase I 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 I 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?

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 gastrointestinal toxicity more predictable Hepatic and renal toxicities – large false positive Toxicologic parameters: LD10 – lethal dose in 10% of animals TDL (toxic dose low) – lowest dose that causes any toxicity in animals NOAEL – no observed adverse effect level

Phase I trials: starting dose 1/10 of the LD10 in rodents or (depending on sensitivity of the species) 1/6 or 1/3 of the TDL in large animals Unless preclinical studies suggest a very steep dose/toxicity curve

Conversion of animal dose to human equivalent doses (HED) Species To convert animal dose in mg/kg to dose in mg/m2, multiply by Km below: To convert animal dose in mg/kg to HED in mg/kg, either: Divide animal dose by Multiple animal dose by Human 37 - Child (20 kg) 25 Mouse 3 12.3 0.08 Hamster 5 7.4 0.13 Rat 6 6.2 0.16 Ferret 7 5.3 0.19 Guinea pig 8 4.6 0.22 Rabbit 12 3.1 0.32 Dog 20 1.8 0.54 Primates: Monkeys Marmoset Squirrel monkey Baboon Micro-pig 27 1.4 0.73 Mini-pig 35 1.1 0.95

Phase I 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 I patient population “Conventional” eligibility criteria- examples: Advanced solid tumors unresponsive to standard therapies or for which there is no known effective treatment Performance status (e.g. ECOG 0 or 1) Adequate organ functions (e.g. ANC, platelets, Creatinine, AST/ALT, bilirubin) Specification about prior therapy allowed Specification about time interval between prior therapy and initiation of study treatment No serious uncontrolled medical disorder or active infection

Phase I patient population “Agent-specific” eligibility criteria - examples: Restriction to certain patient populations – must have strong scientific rationale Specific organ functions: For example – cardiac function restrictions (e.g. QTc < 450-470 ms, LVEF > 45%, etc) if preclinical data or prior clinical data of similar agents suggest cardiac risks For example – no recent (6-12 months) history of acute MI/unstable angina, cerebrovascular events, venous thromboembolism; no uncontrolled hypertension; no significant proteinuria, for antiangiogenic agents Prohibited medications if significant risk of interaction with study drug

Phase I 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?

Cohort dose escalation: standard 3+3 design Many phase I trials accrue additional patients at the RPTD to obtain more safety, PK, PD data (but this expansion cohort does not equal to a phase II trial)

Phase I 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 I trial basic principles Start 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 I trial assumption The higher the dose, the greater the likelihood of efficacy Dose-related acute toxicity is regarded as a surrogate for efficacy The highest safe dose is the dose most likely to be efficacious This dose-effect assumption is primarily for cytotoxic agents and may not apply to molecularly targeted agents

Dose-response: efficacy and toxicity Therapeutic window

P1T Designs for Targeted Agents (till 2003) Reasons for halting dose escalation, targeted agents given as single-agents Reason -2003 2007-2008 Toxicity 36 (60%) 20 (63%) PK (+/- other) 8 (13%) 4 (13%) Others Design, maximum planned dose 5 Limited drug Supply 4 Other phase I results 2 Drug activity observed 1 Not stated 4 Total 60 32 Parulekar and Eisehauer, JNCI, 2004 Le Tournea, Lee, Siu, JNCI, 2009

Phase I Trial Design : Modified Fibonacci Dose Escalation (Rule-based) Attributed to a merchant from the 13th century Doses increase by: 100%, 66%, 50%, 40%, 33%, etc. Standard “3+3” design: 3 patients per cohort, escalating to 6 if DLT occurs Dose escalate until DLT observed and MTD/RPTD defined Advantages: relatively safe, straightforward, clinician-friendly Disadvantages: lacks statistical foundation and precision, potentially treating a large proportion of patients at sub-therapeutic doses, time consuming

Phase I trial design: standard 3+3 design 3 pts Dose 3 pts DLT 3 pts + DLT 3 pts Recommended dose Starting dose Eisenhauer et al.

Phase I Trial Design: Accelerated Titration Design (Rule-based) First proposed by Simon et al (J Natl Cancer Inst 1997) Several variations exist: usual is doubling dose in single-patient cohorts till Grade 2 toxicity then revert to standard 3+3 design using a 40% dose escalation intrapatient dose escalation allowed in some variations More rapid initial escalation

Phase I trial design: accelerated titration 1 pt Dose DLT 3 pts 3 pts + DLT 3 pts Recommended dose 1 pt Gr 2 toxicity Starting dose

Phase I Trial Design: Modified Continual Reassessment Method (MCRM; Model-based) Bayesian method Pre-study probabilities based on preclinical or clinical data of similar agents At each dose level, add clinical data to better estimate the probability of MTD being reached Fixed dose levels, so that increments of escalation are still conservative

Example: Pre-set dose levels of 10, 20, 40, 80, 160, 250, 400 Phase I Trial Design: Modified Continual Reassessment Method (MCRM; Model-based) Example: Pre-set dose levels of 10, 20, 40, 80, 160, 250, 400 If after each dose level, the statistical model predicts a MTD higher than the next pre-set dose level, then dose escalation is allowed to the next pre-set dose level Advantages: Allows more dose levels to be evaluated with a smaller number of patients More patients treated at or closer to “therapeutic” dose Disadvantages: Does not save time, not easily implemented if without access to biostatistician support

Phase I Trial Design: Dose Escalation with Overdose Control (EWOC; Model-based) Bayesian method After each cohort of patients, the posterior distribution is updated with DLT data to obtain d (probability of DLT at dose d). The recommended dose is the one with the highest posterior probability of DLT in the “ideal dosing” category The overdose control mandates that any dose that has > 25% chance of being in the “over-dosing” or “excessive over-dosing” categories, or > 5% chance of being in the “excess-overdosing” category, is not considered for dosing

Excessive Over-Dosing Estimated MTD Based on Bayesian Logistic Method (2-parameter evaluation with over-dose control) EXAMPLE of Probability of DLTs (Bayesian design) Under-Dosing (This % should be minimal) Ideal Dosing (This bar should be the highest percentage) Over-Dosing (This bar should be below 25%) Excessive Over-Dosing (This bar should be 0%) 7% 60% 30% 3% 44% 52% 4% 0% Drug at 0.5mg 7% 66% 27% Drug at 0.75 mg 35% 64% Drug at 1.0 mg

Phase I Trials 2007-8: use of new designs 246 published papers 208 phase I cancer clinical trials -12 trials with no planned dose escalation 20 no access to the dose escalation method used 176 evaluable phase I clinical trials 170 traditional 3+3 design or variations (96.4%): 162 traditional 3+3 design 1 traditional 3+3 design with intrapatient dose escalation 7 ATD 6 model-based designs (3.6%): 5 CRM 1 TITE-CRM Le Tourneau, Lee, Siu, JNCI, 2009

Phase I 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?

Endpoints in phase I trials DLT and other toxicity – safety and tolerability Pharmacokinetics Pharmacodynamics – biological correlates, imaging endpoints Preliminary antitumor activity

Response Rates and Deaths from Toxic Events in Phase I Oncology Trials Involving the First Use of Agent in Humans (Horstman et al, NEJM 352, 2005) Trial No. of Trials No. of Patients Assessed for Response Overall Response Rate* % No. of Patients Assessed for Toxic Events Deaths from Toxic Events no. % Total First use of an agent in humans 117 3164 4.8 3498 9 (0.26) Cytotoxic chemotherapy First use of an agent in humans 43 1298 5.0 1422 7 (0.49) Immunomodulator First use of an agent in humans 16 404 7.4 431 1 (0.23) Receptor or signal transduction First use of an agent in humans 27 742 3.8 853 1 (0.12) Antiangiogenesis First use of an agent in humans 8 200 7.0 228 Gene transfer First use of an agent in humans Vaccine First use of an agent in humans 23 520 3.1 564

Pitfalls of phase I trials Chronic toxicities usually cannot be assessed Cumulative toxicities usually cannot be identified Uncommon toxicities will be missed

Phase I trials and infrequent toxicities Probability of overlooking a toxicity: POT(p) = (1-p)n; n = sample size, p = true toxicity rate

The successful phase I team Scientists Fellows Trial nurses Data coordinators Investigators Lab personnel: reference, PK, PD Pharmacists Biostatisticians Radiologists