Charlie Taylor, PhD CpTaylor Consulting Chelsea, MI, USA

Slides:



Advertisements
Similar presentations
PHARMACOKINETIC.
Advertisements

Regulatory Framework Leigh Shaw, Director.
Daniel Everitt, MD; Erica Egizi MPH Global Alliance for TB Drug Development, New York Helen Winter, PhD University of Otago, New Zealand 2012 International.
Improving Candidate Quality Through the Prediction of Clinical Outcome.
1 PK/PD modeling within regulatory submissions Is it used? Can it be used and if yes, where? Views from industry 24 September 2008.
Pipeline Session: NBI Human Pharmacokinetics of NBI-98854: A Selective Inhibitor of VMAT2 with an Attractive PK and Safety Profile for Hyperkinetic.
JMV 1843 pharmacological profile
Pharmacotherapy in the Elderly Judy Wong
Neonatal/Juvenile Animal Safety Studies Kenneth L. Hastings, Dr.P.H., D.A.B.T. Office of New Drugs, CDER.
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?
Drug-Like Properties: Optimizing Pharmacokinetics and Safety During Drug Discovery Li Di and Edward H. Kerns ACS Short Course.
Food and Drug Administration Preclinical safety data for “first in human” (FIH) clinical trials in healthy volunteer subjects Oncology Drug Advisory Committee.
Office of Clinical Pharmacology and Biopharmaceutics IDSA/ISAP/FDA Workshop 4/16/04 1 Improvement in Dose Selection: FDA Perspective IDSA/ISAP/FDA Workshop.
Gokaraju Rangaraju College of Pharmacy
Virtual Drug Development in Southern California, A Pre-Clinical Focus in vitro tests to support IND submissions David Johnson, Ph.D. Director, DMPK MicroConstants,
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.
Office of Clinical Pharmacology and Biopharmaceutics IDSA/ISAP/FDA Workshop 4/16/04 In Vitro/Animal Models to Support Dosage Selection: FDA Perspective.
Required steps Clinical protocol preparation Informed consent Approval of the ethical committee Phase I Healthy volunteers Objectives: 1 - To identify.
Exploratory IND Studies
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.
Week 6- Bioavailability and Bioequivalence
PK/PD Modeling in Support of Drug Development Alan Hartford, Ph.D. Associate Director Scientific Staff Clinical Pharmacology Statistics Merck Research.
Nonlinear Pharmacokinetics
Concepts and Applications of Pharmacokinetics
Modeling and simulation (M&S) was employed to recommend doses for human Phase I studies of a direct Factor Xa (FXa) inhibitor, CS Predicted human.
Regulatory requirements Drs. Jan Welink Training workshop: Assessment of Interchangeable Multisource Medicines, Kenya, August 2009.
APPLYING PRE-CLINICAL DATA TO CLINICAL STUDIES-I Edward A. Sausville, M.D., Ph.D. Developmental Therapeutics Program National Cancer Institute October.
1 Axcan Public Presentation for the FDA Pharmaceutical Science and Clinical Pharmacology Advisory Committee Meeting July 23, 2008.
Background Objectives Data Simulation in human Conclusion -F14512, a polyamine-vectorized anti-cancer drug which combines an epipodophyllotoxin core targeting.
PLASMA HALF LIFE ( t 1/2 ).  Minimum Effective Concentration (MEC): The plasma drug concentration below which a patient’s response is too small for clinical.
EMEA London Pharmacokinetic- pharmacodynamic integration in veterinary drug development: an overview P.L. Toutain National Veterinary School ;Toulouse.
FDA Case Studies Pediatric Oncology Subcommittee March 4, 2003.
Pipeline Session: NBI Selective Inhibitor of VMAT2 with an Attractive PK and Safety Profile for Hyperkinetic Movement Disorders.
Preclinical Models to Support Dosage Selection
The New Drug Development Process (www. fda. gov/cder/handbook/develop
Drug Administration Pharmacokinetic Phase (Time course of ADME processes) Absorption Distribution Pharmaceutical Phase Disintegration of the Dosage Form.
Rivaroxaban Has Predictable Pharmacokinetics (PK) and Pharmacodynamics (PD) When Given Once or Twice Daily for the Treatment of Acute, Proximal Deep Vein.
Phase I Issues for Novel TB Drugs Dakshina M. Chilukuri, Ph.D. Office of Clinical Pharmacology and Biopharmaceutics, FDA OPEN FORUM ON KEY ISSUES IN TB.
Regulatory Aspects of PK/PD – (modelling) Karolina Törneke Senior expert, member of the CVMP.
INTRODUCTION CLINICAL PHARMACOKINETICS
BIOPHARMACEUTICS.
© Paradigm Publishing, Inc.1 Chapter 2 Basic Concepts of Pharmacology.
European Patients’ Academy on Therapeutic Innovation The key principles of pharmacology.
Do We Need to Optimize Protein Binding in Drug Discovery? NEDMDG Summary Meeting Xingrong Liu, Ph.D. Genentech.
1 Biopharmaceutics Dr Mohammad Issa Saleh. 2 Biopharmaceutics Biopharmaceutics is the science that examines this interrelationship of the physicochemical.
IPCP Core C Activities Aprepitant Dose-exposure Relationship in the Monkey Jeffrey S. Barrett, Ph.D., FCP The Children’s Hospital of Philadelphia Division.
Pharmacokinetics (PK) and Pharmacodynamics (PD) of Rivaroxaban: A Comparison of Once- and Twice-daily Dosing in Patients Undergoing Total Hip Replacement.
PRECLINICAL PHARMACOLOGY & TOXICOLOGY OF ANTI-NEOPLASTIC AGENTS  THE NCI PERSPECTIVE Joseph E. Tomaszewski, Ph.D. Toxicology & Pharmacology Branch.
| 1 Application of a Bayesian strategy for monitoring multiple outcomes in early oncology clinical trials Application of a Bayesian strategy for monitoring.
Viagra (sildenafil citrate): Extensive Clinical and Post-Marketing Experience Michael Sweeney, MD Senior Medical Director Pfizer Inc.
The process of drug development. Drug development 0,8 – 1 mld. USD.
Source: Frank M. Balis Concentration and Effect vs. Time Conc./ Amount Effect [% of E MAX ] Time Central Compartment Peripheral Compartment Effect Compartment.
MULTIPLE DOSAGE REGIMEN
for Human Pharmaceuticals Kyung-Chul Choi D.V.M., Ph.D.
The Stages of a Clinical Trial
Eucrisa™ - Crisaborole
Drug Discovery &Development
Senior Medical Director, Cardiovascular
Biopharmaceutics Dr Mohammad Issa Saleh.
5 Pharmacodynamics.
Clinical Pharmacokinetics
1 Concentration-time curve
Therapeutic Drug Monitoring chapter 1 part 1
Efficacy, Potency and Safety of Drugs
unbound concentration µM
Considerations for Successful Biopharmaceutical Product Development: Discovery to Proof of Concept -A Panel Discussion Stanley C. McDermott, PharmD, MS,
Presentation transcript:

Charlie Taylor, PhD CpTaylor Consulting Chelsea, MI, USA Dosage Determination from Preclinical to Proof-of-Concept Trials, (Including Toxicology) Charlie Taylor, PhD CpTaylor Consulting Chelsea, MI, USA

Choosing Doses for POC: Preclinical and early studies that enable dose selection Reasons for drug failure in clinical phase 2-3 Need to choose both low (ineffective) and high (maximum tolerated) doses within dose range Biomarkers (one endpoint: animal → human translation) PK/PD modeling – EC50 as a target for efficacy or AEs Toxicology/toxicokinetics – daily AUC(0-24) as a limit Putting it together – visualizing multiple datasets Human population PK modeling – determine which doses best fit the constraints

Sequence of Studies Needed Prior to Clinical Proof-of-Concept * Preclinical in vitro studies: action at drug target (pharmacology) Preclinical in vivo pain models: indicate treatment of disease Safety pharmacology: animal studies for adverse effects Preclinical (& human liver microsome) metabolism studies Animal toxicology & toxicokinetic studies to identify safety issues Clinical Phase I studies of pharmacokinetics and tolerance in healthy human volunteers (Optional) Biomarker studies with both animal models and humans to establish proof of pharmacology in vivo (apart from efficacy) * * * * * * Requires in vivo unbound plasma drug concentration or daily drug exposure to help choose human efficacy DOSES

Drug Development Failures – UK ’64-’85 Prentis et al. (1988) Brit J Clin Pharmacol 25:387-396

Determine Initial Phase 2 Dose Range ?? Clinical doses MUST encompass both low end (lack of efficacy) and high end (maximum tolerance) Data from animal efficacy, animal safety, biomarker and human tolerance ALL must be considered The peak unbound plasma drug concentration (animal studies), daily exposure (AUC0-24 - tox) and human multiple-dose PK each need consideration How to consider all these factors??

One Approach: Biomarkers – Surrogate HUMAN Endpoints for Efficacy Defines drug action in vivo Examples: Imaging, Adverse Event or Mechanism e.g. PET to measure receptor occupancy in CNS e.g. Nystagmus, dizziness, balance platform e.g. Experimental pain model w/ volunteers e.g. Electrographic response (EEG, retinogram, TMS) Biomarkers Allow no-go decision prior to proof of efficacy, for example: Poor oral drug absorption or lack of CNS penetration Lack of receptor occupancy at highest safe dose

Hypothetical Human Biomarker: Criterion: 75% drug receptor occupancy in human brain @ high dose This criterion met at animal effective dose (animal PET study) Drug displacement of PET ligand in human brain: 18F-x-drugamine given IV in tracer amount If greatest human volunteer dose of experimental drug reaches < 30% occupancy, NO-GO If greatest human volunteer dose > 75% occupancy, GO (further development) Caveat: Criteria must be selected based on results with a prior known compound – Otherwise, risk of poor validation

Toxicology Findings (non-pharmacology) are Based on Daily Drug Exposure (AUC0-24) Repeated-dose animal tox studies determine lowest toxic dose and greatest no-effect (daily) dose Toxicokinetics determine drug exposure (AUC0-24) in mg•hr/mL at greatest no-effect dose e.g. Drug X has 8 hr half-life; Cmax and AUC are determined from plasma drug samples taken 0, 1, 4, 7, 12, and 24 hr after single oral dose at steady-state Similar human pharmacokinetic data and PK modeling determine human drug exposure (AUC0-24) @ doses Analysis is adjusted for different drug binding of plasma proteins between species

Calculation of Animal Drug Exposure - Toxicokinetic AUC(0-24) Samples of drug in plasma of animal tox species Begin sampling after reaching repeated dose steady state Orange symbols are mean from n = 8 Mathematical fit to curves of oral absorption & elimination Measure area under curve for 0-24 hr = Drug Exposure Cmax = 10.8 mg/mL AUC(0-24) = 136 mg•hr/mL Dose = 50 mg/kg/day

Measured Drug Exposure in Rat Tox Studies Toxicokinetic Parameters in Multidose Oral Toxicity Studies Species Durat. Dose (mg/kg) Plasma Concentration (g/mL) AUC(0‑24) (g·hr/mL) Male Female Rat 2 Week Gavage 500 1250 2500 Diet 13.5 27.6 47.4 11.5 25.9 50.7 9.92 25.2 40.7 10.7 19.0 32.4 120.0 332.0 626.0 199.0 491.0 921.0 102.0 334.0 602.0 181.0 336.0 606.0

Animal Toxicology: Human Exposure Limits Are Set by Daily Drug Exposure (AUC0-24) RAT DOG MOUSE No-Effect Dose Limit: 200 mg•hr/mL – determines maximum permissible human exposure

PK Modeling of Drug in Human Plasma (daily dosing of 50 mg oral) Human Cmax Limit based on Animal Toxicology (Max no-effect dose AUC0-24) (mg/mL) AUC0-24

CYP2D6 Heterogeneity – Ca2+ Channel Blocker Smith & Jones (1999) Curr Opin Drug Discov Devl 2:33-41

Q: How to Predict Human Efficacious and Adverse Drug Doses Based on Animal Efficacy, Animal AEs and Human Pharmacokinetic Data?? A: Compare plasma drug Cmax obtained in animal pharmacology tests using a Napiergram to human Pharmacokinetic Cmax data

“Napiergram” Named for John Napier of Merchistoun (aka Marvelous Merchiston, Scotland) Inventor of Napier’s bones (slide rule), popularization of logarithms and the decimal point Also: used a pet black rooster to tell fortunes and devine truths Napiergram: graphic comparison of log10 unbound plasma drug concentrations associated with pharmacology and with safety concerns John Napier (1550-1617)

From Dose:Response experiments: Obtain ED5, ED50, ED95

Transform Pharmacology from ED50 to EC50

Napiergram: Many Pharmacology Datasets – Animal Cmax for doses with 5% 50% & 95% effect Cmax = 2,500 nM or 0.5 mg/mL (unbound) Cmin = 125 nM or 0.026 mg/mL (unbound)

PK Modeling of Drug in Human Plasma (daily dosing of 50 mg oral) (mg/mL) Animal Adverse Limit (EC50 for ataxia) Cmax (hi dose) Cmax (mid dose) Actual Human PK – mid dose Cmax (low dose) EC20 for Efficacy in Animal Model

Phase 2 Dose Selection (final chapter) Requires Deliberation from team of experts: Animal tox, Pharmacokinetics, PK/PD modeling, Clinical research, Preclinical pharmacology, (Biomarkers) Who pays the clinical trial bills? Clinical Research Despite planning, dosage and regimen often are readjusted during Phase 2 (toleration, efficacy or new safety findings) Dosages MUST continue to include both low (ineffective) and maximal tolerated dosages to provide basis for FDA approval Dose toleration may vary between healthy volunteers and patients with serious disease

SUMMARY: Preclinical Studies to Determine Phase 2 Dose Selection In vitro and in vivo animal pharmacology – target Cmax for therapy and adverse effects Animal toxicology & toxicokinetic studies – determines maximal human drug exposure (AUC0-24) Phase 1 Clinical trials determines human pharmacokinetics & drug exposure Napiergram – allows consideration of Cmax from multiple animal datasets & compare to human PK Phase 2 dose adjustment is common!

Example “Drug Killer” Problems Poor Oral Absorption (F < 25%) Poor Aqueous Solubility Poor Elimination Kinetics (t1/2 < 4 hr or t1/2 > 36 hr) Nonlinear Elimination Kinetics (e.g. blocked clearance at high doses) Extensive metabolism to active or toxic compound Excessive plasma protein binding (> 99%) Metabolism by variable CyP450 (CYP2D6, CYP2C19) Cardiac Q-T interval prolongation (hERG channel block) Genotoxic compound (Ames positive) Hepatic toxicity PK Tox