RE: CPMT Pharmacokinetic Analyses of Fixed-Dose Drug Combinations for Pediatric Tuberculosis

Similar documents
2401 Gillham Road Kansas City, Missouri Phone (816)

Workflow for PBPK Modeling to Support Pediatric Research and Development

Extrapolation in Pediatric Product Development: Practical Application of the Principle of Scientific Necessity

Reflection paper on the use of extrapolation in the development of medicines for paediatrics

Model based approaches to target special populations with rational formulation and clinical design strategies

Injectable modified release products

Optimizing the Development of Biosimilars Using PK/PD: Recent Scientific and Regulatory Advances

COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP) GUIDELINE ON THE NON-CLINICAL DEVELOPMENT OF FIXED COMBINATIONS OF MEDICINAL PRODUCTS

New TB Drugs Approval in Thailand

Merging the Science of Quantitative Pharmacology into Routine Patient Care: Focus on Digital Tools

The views and opinions expressed in the following PowerPoint slides are

Is Confirmatory PK/PD Modeling Possible?

Drug Development: Why Does it Cost so Much? Lewis J. Smith, MD Professor of Medicine Director, Center for Clinical Research Associate VP for Research

COMMITTEE FOR PROPRIETARY MEDICINAL PRODUCTS (CPMP)

Development of paediatric formulations - points to consider

Guidance for Industry

COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP)

Special Considerations and Utility of Modeling and Simulation for Pediatric Medical Countermeasures: Introduction

MEDICINES CONTROL COUNCIL

Guideline on the qualification and reporting of physiologically based pharmacokinetic (PBPK) modelling and simulation

Abstract. Technical Aspects. Applying GastroPlus for Extensions of Biowaivers for BCS Class II Compounds 2

Clinically Relevant Specifications (CRS): A Regulatory Perspective

Guideline on the reporting of physiologically based pharmacokinetic (PBPK) modelling and simulation

PART I PAST, PRESENT, AND FUTURE OF PEDIATRIC DRUG DEVELOPMENT COPYRIGHTED MATERIAL

Cytochrome P450 Genotype Panel

Basic Principles for Conducting Phase I Trials in the Japanese Population Prior to Global Clinical Trials

fact sheet 3 Introduction to Biosimilars & Regulatory Requirements

PIP s Pup s and Problems

The Role of a Clinical Statistician in Drug Development By: Jackie Reisner

Model-based rationale for drug combinations in tuberculosis

BIOSTATISTICAL METHODS FOR TRANSLATIONAL & CLINICAL RESEARCH

Multi-Regional Studies and Bridging Studies. Simon Day, PhD Johns Hopkins University

Pharmacokinetic / pharmacodynamic modeling and simulation in the design and analysis of Randomized Concentration-Controlled Trials (RCCTs)

NDA Advisory Services Ltd

Regulatory Perspective

Outline CLINICALLY RELEVANT SPECIFICATIONS. ISPE Process Validation Conference September 2017 Bethesda, MD

International Consortium For Innovation & Quality in Pharmaceutical Development

From Discovery to Development of new Drugs. and pitfalls along the way. by Kim Dekermendjian, PhD in Medicine BD & Key Account manager

Mechanistic IVIVC Using the Simcyp ADAM Model. Make SCIENCE out of IVIVC

Comments and suggestions from reviewer

Drug Discovery and Development PHG 311. Prof. Dr. Amani S. Awaad

DIAGNOSTICS ASSESSMENT PROGRAMME

S9 Nonclinical Evaluation for Anticancer Pharmaceuticals

Use of Mechanistic Population Based PBPK Models in the Establishment and Application of IVIVCs. Nikunjkumar Patel, Simcyp Limited

Re: Docket Number FDA-2017-D-0154 Considerations in Demonstrating Interchangeability With a Reference Product; Guidance for Industry, Draft Guidance

BIOSTATISTICAL METHODS

The Investigator s Brochure: A multidisciplinary document

Clinical Pharmacology of Antimicrobials in Children

Pediatric Exclusivity and Drug Development Requirements in the Overall Pediatric Population. Prepared by Beckloff Associates, Inc.

Regulatory Perspective on Developing Long Acting ARVs for HIV Treatment/Prevention. FDA Division of Antiviral Products

A. TRIAL IDENTIFICATION

M4E(R2) - Common technical document for the registration of pharmaceuticals for human use Efficacy

Guideline for Bioequivalence Studies of Generic Products. December 22, 1997

Bioavailability and Bioequivalence Studies

Acceptance of Foreign Clinical Data A U.S. Perspective

S9 Implementation Working Group ICH S9 Guideline: Nonclinical Evaluation for Anticancer Pharmaceuticals Questions and Answers

9/7/17. Implementing the ARK Immunoassay for Therapeutic Drug Monitoring of High-Dose Methotrexate. Disclosure. Learning Objectives

Question and Answer Guide Regarding Notification on Practical Operations of Electronic Study Data Submissions

Role of Academic Investigators in Drug Development

What s New in GCP? FDA Draft Guidance Details FIH Multiple Cohort Trials

Subgroups along the development axis

Agreed with W. Cornell Graduate Program and Tri-I

Modeling & simulation in pediatric drug development and regulation

Celerion s Symposia Series: Bridging the Gap from Phase I to Proof-of-Concept. San Francisco, CA Tue 8 th, Apr 2014

September 12, Division of Dockets Management (HFA-305) Food and Drug Administration 5630 Fishers Lane, Room 1061 Rockville, MD 20852

Introduction to Pharmacology. Prepared by: Shaikh Abusufyan, M. Pharm (Pharmacology)

Role of PBPK based virtual trials modeling in generic product development and regulation

First UNGAP meeting Food-Drug Interactions Regulatory Aspects

In vivo predictive biopharmaceutics tools for oral drug delivery

An Introduction to Clinical Research and Development

Chin Koerner Executive Director US Regulatory and Development Policy

Guideline on the use of pharmacogenetic methodologies in the pharmacokinetic evaluation of medicinal products

Session 2 summary Designs & Methods. Pairwise comparisons approach. Dose finding approaches discussed: Guiding principles for good dose selection

8. Clinical Trial Assessment Phase II

BIOEQUIVALENCE TRIAL INFORMATION FORM (Medicines and Allied Substances Act [No. 3] of 2013 Part V Section 39)

Pharmacokinetics. Processes, Mathematics, and Applications. Second Edition. Peter G. Welling. Institut de Recherche Jouveinal

M4E(R2) - Common technical document for the registration of pharmaceuticals for human use Efficacy

Pediatric Drug Development:

Guideline for the conduct of efficacy studies for nonsteroidal anti-inflammatory drugs

Clinical Pharmacology

Translating TB Therapy Response:

Results to be Presented at LDN WORLD Symposium in February Initiation of Repeat-Dose Pompe Study Anticipated in 3Q13

Microdose Radiopharmaceutical Diagnostic Drugs: Nonclinical Study Recommendations Guidance for Industry

Recommendation on elements required to support the medical plausibility and the assumption of significant benefit for an orphan designation

Organic Pharmaceutical Chemistry: Prodrugs

Line extension of immediate release products

Introduction to Drug Design and Discovery

The Critical Path to TB Drug Regimens Initiative

Office for Human Subject Protection. University of Rochester

ACG Public Forum. Join ACG, the FDA, and EMA for a discussion on biosimilars and IBD. Monday, 12:45 pm 2:15 pm

Investigational New Drug Application

Dose Exposure Response Relationships: the Basis of Effective Dose-Regimen Selection

Introduction to clinical trials

Preclinical pharmacokinetics and pharmacodynamics of AG-519, an allosteric pyruvate kinase activator

Regulatory considerations for initiating paediatric trials with RSV antivirals

Non-clinical Assessment Requirements

Role of Pharmacometrics in Drug Development and Regulatory Review:PMDA perspectives

Making PK Analysis Easier: The New ADaM Data Standard ADNCA

Nanomedicine for Improved Efficacy of Tuberculosis Drugs Pharmacokinetic importance

PHARMACOKINETICS. Dr. M.Mothilal Assistant professor

Transcription:

Suzanne Hill, M.D. Essential Medicines and Pharmaceutical Policies World Health Organization Geneva, Switzerland RE: CPMT-08-011 Pharmacokinetic Analyses of Fixed-Dose Drug Combinations for Pediatric Tuberculosis Dear Dr. Hill, We appreciate the opportunity to respond to the thoughtful comments on the aforementioned study report that were submitted by reviewers from the Gates Founation [Richard Elliott, Michael Kimerling and Jan Gheuens as provided via electronic communication from Saul Morris]. The sections that follow represent our response to these comments/queries. 1. How predictive is this model if you perform in silico modeling for adults and compared with real clinical PK data (e.g. Cmax; AUC, %time>desirable conc.)? Data detailing maximum plasma concentrations (Cmax) were extracted from pediatric pharmacokinetic studies and overlaid on the relevant figures provided in the aforementioned technical study report [symbols represent individual data or mean data with or without the standard deviation as reflected in the original studies]. To minimize complexity, we have not segregated the data based on the nature of the study; however, these data derive from studies of varied design (e.g. single-dose vs. steady state, drug alone vs. in combination) with widely varying subject populations. Furthermore, it should be noted that numerous data points were extracted from studies utilizing a sparse sampling design thus they may represent a close approximation to, but not the actual, Cmax. Finally, it should be noted that the analytical methods used to generate the data in the original studies varied from simple microbiological assays to traditional chromatographic techniques. Upon review of the figure, several observations can be made. The most notable is that there exists a substantial degree of inter-individual variability in the observed exposure estimates for the anti-tubercular agents that were simulated. In all cases, the predicted estimates (shaded regions) are reflected in, but do not wholly encompass, the observed values. Ethambutol serves as the exception where the predicted concentrations fully encompass the observed values. Notably, many of the observed ethambutol concentrations derive from the singular study that was robust enough to provide pharmacokinetic parameter estimates used in the simulations (thus, this would be expected via circular reasoning). In contrast, isoniazid concentrations markedly exceed the bounds defined in the simulations. This is not unexpected based on the multitude of factors that influence the biodisposition of the compound (including the well described polymorphism in N-acetyltransferase). In this case, the number of existing studies that Kansas City, Missouri 64108 Phone (816) 234-3000 www.childrens-mercy.org

provide adequate information on the impact of development, genetics and pathophysiology on isoniazid pharmacokinetics is strikingly limited. Highly predictive simulations would require parameter estimates generated from well designed studies that simply do not exist. Similarly, the paucity of comprehensive pediatric trials on pyrazinamide pharmacokinetics results in observed values that appear to indicate the absence of a dose-exposure relationship in children despite the fact that this relationship is reasonably well established in adults. Apart from the limitations to the data used to drive the simulations, the simplistic nature of the simulations, as set forth by the scope of work, accounts for models with varying predictive power. Our commission was to perform an initial set of steady-state

simulations for each drug as a singular entity (absent consideration for drug-drug interactions) based on information in the primary medical literature and contained in WHO technical reports. 2. What would the data look like if you modeled twice a day dosing. Would the advantages outweigh the inconvenience? The ad hoc working group on paediatric tuberculosis constituted by WHO developed the initial drug/dosing table which was used as the basis for our simulations. It was the conclusion of this working group that a once-daily dosing regimen for a multi-drug formulation afforded the greatest chance for treatment adherence, especially in the developing world. Thus, for these initial, preliminary simulations, WHO suggested that we evaluate a once daily dosing regimen. Certainly, in silico approaches can be used to simulate twice daily dosing regimens which have the potential to improve inter-dose systemic exposure relative to expected pharmacodynamic surrogates. This could ostensibly improve therapeutic efficacy if adherence rates were 100%. Given many of the challenges associated with directly supervised TB treatment in the developing world, we can not reliably address whether any potential benefit associated with treatment regimens employing twice daily dosing outweigh convenience. 3. Page 2: Figure 1: may want to put units on Y axis (mcg/ml) Any aspect of the technical report can be amended at the request of the sponsor. We concur that clarity for each illustration in the technical report is important. 4. Figure 4. Are calculated values for infants or children? Both infants and children are considered in the pyrazinamide simulations as the pharmacokinetic parameters derived from available studies that did not segregate children by age. 5. Might be nice to put in a (adult) toxicity line for the Cmax plots for each drug (where known), so can put exposure predictions in context with safety. 6. What is known about the safety in infants/children for the doses used in these calculations? Should cut off values be added? These simulations were in no way intended to infer safety. Moreover, the number of existing studies detailing the relationship between concentration and toxicity are, in our opinion, insufficient to demarcate a specific threshold plasma concentration that can be reliably associated with the production of adverse events from any of the drugs administered in normal therapeutic doses. This not withstanding, the plasma concentration illustrations contained within the technical report are constructed in such a fashion to enable reviewers to visually interpolate any plasma concentrations that they might feel are associated with adverse events / toxicity. 7. Questions around some of the assumptions: Assumptions 1 &2: Do these assumptions take into account differential hepatic function of infants/children compared to adults? Have these assumptions proven to be true in any real life examples?

Assumption 3: What about Rif induction of CYP2D6 and CYP3A4? I see on bottom of page 15 some reference to CYP induction, but seems like one should be able to use whatever is know from adult Rif induction and, for a first approximation, build that into the model. May not be perfect, but will be a little closer to reality. Assumption 5: Is this true in adults? Assumptions 6-8: Any evidence for this assumption? Assumptions are employed out of necessity when the existing medical literature are unavailable or incomplete. As is the case for any pharmacokinetic simulation of plasma drug concentrations, one must assume that pharmacokinetic parameter estimates derived from a particular, generally small population can be generalized to represent the majority of individuals in that population. To that end we were careful to identify pharmacokinetic parameters that most closely reflected the population of interest (where available). Drug-drug interactions were not considered given the absence of relevant data from pediatric populations where the intersection of developmental and constitutive expression of drug metabolizing enzyme / transporter activity can determine the direction and magnitude of any drug-drug interaction. In children, this undertaking is far more complex than would be the case for adults where constitutive enzyme expression is established and relatively static. As indicated in the report, the validity of selected assumptions remains questionable as for the case of rifampin absorption. Previous experimental data appear to indicate that selected rifamycins may undergo active transport at the level of the intestine but the extent and magnitude to which this occurs in humans has not been established. 8. How well do these simulations predict what is known in the literature? Are there other variables (e.g. GI volumes, liver function, GI tract permeability differences) that could be incorporated into the model for improved predictivity? Known developmental differences in drug disposition have been previously summarized (Kearns GL, et al. N Engl J Med 2003;349:1157-1167) and certainly, have the ability to impact the biodisposition of antitubercular drugs. With the possible exception of developmental road maps for renal function and some of the major drug metabolizing enzymes, existing developmental physiology literature does not afford the degree of granularity necessary to effectively parameterize a pharmacokinetic/pharmacodynamic model in a quantitative fashion. At best, one can incorporate directional changes which, in the case of several of the simulations contained in this report, have been incorporated when possible (ie. afforded by some existing information). In the absence of clear data, introducing hypothetical differences in the model could serve to compound variability in an artificial manner and thereby, further compromise any predictive value in the simulations. Importantly, the limitations of the simulations discussed herein illustrate the gaps that need to be filled before we can reliably identify the ideal formulation for use in children. These initial simulations as performed are intended to provide a starting point in the development of a multi-drug formulation of conventional antitubercular drugs that are currently recommended by WHO for the management of TB in paediatric patients. As such, they provide a necessary framework upon which to further build strategies for dose selection and ultimately, the prospective study of such drug formulations in patients who receive them as treatment. Given the urgency to ensure the availability of effective treatments that can be reliably used in developing countries, it is hoped that these

simulations, all of which are based on fundamental clinical pharmacology principles, will provide the insight necessary to move the paediatric TB initiative forward. Kind regards, Susan M. Abdel-Rahman, Pharm.D. Associate Professor of Pediatrics Director, Developmental PK/PD Core Laboratory The Children s Mercy Hospitals and Clinics, Kansas City, MO Gregory L. Kearns, Pharm.D., Ph.D. Marion Merrell Dow / Missouri Chair in Pediatric Medical Research Chairman, Department of Medical Research Associate Chairman, Department of Pediatrics The Children s Mercy Hospitals and Clinics, Kansas City, MO