Utah Life Science Summit

Slides:



Advertisements
Similar presentations
The Drug Discovery Process
Advertisements

Ramana S. Uppoor, M.Pharm., Ph.D., R.Ph.
Regulatory Framework Leigh Shaw, Director.
Establishing Preclinical Development Operations in China Ming Guo, Ph.D. VP Pharmaceutical Sciences & Manufacturing SABPA Pacific Forum Nov. 4, 2006.
U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES NATIONAL INSTITUTES OF HEALTH Working with FDA: Biological Products and Clinical Development Critical Path.
The Application of the Scientific Method: Preclinical Trials Copyright PEER.tamu.edu.
Special Topics in IND Regulation
Pharmaceutical Development and Review Process Rev. 10/21/2014 APGO Interaction with Industry: A Medical Student Guide.
Stefan Franzén Introduction to clinical trials.
Neonatal/Juvenile Animal Safety Studies Kenneth L. Hastings, Dr.P.H., D.A.B.T. Office of New Drugs, CDER.
Career Opportunities for PharmDs in the Pharmaceutical Industry: Research & Development.
COMPARABILITY PROTOCOLS ACPS March 12-13, 2003 Stephen K. Moore, Ph.D. Chemistry Team Leader CDER/Office of New Drug Chemistry Co-Chair, Comparability.
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?
Recommendations on integrated safety summaries from Phase 1 studies
Interchangeability and study design Drs. Jan Welink Training workshop: Training of BE assessors, Kiev, October 2009.
FDA Nasal BA/BE Guidance Overview
Venkata Ramana S. Uppoor, M.Pharm., Ph.D., R.Ph.
Tanzania, August, 2006 Dr. Barbara Sterzik, BfArM, Bonn 1 Guidelines and Tools available TRS 937 and BTIF (Bioequivalence Trial Information Form)
Documentation of bioequivalence Drs. J. Welink Workshop on WHO prequalification requirements for reproductive health medicines, Jakarta, October 2009.
Guidance for Industry M4S: The CTD-Safety
Bioequivalence Studies Dr Sanet Aspinall, PhD Managing Director AddClin Research Pretoria 20 March 2009.
Figure 4.1 NEW PRODUCT DEVELOPMENT PROCESS Finance Corporate strategy and portfolio decisions Regulatory affairs Marketing and sales + market research.
Nonclinical Studies Subcommittee Advisory Committee for Pharmaceutical Science CMC Issues for Screening INDs Eric B. Sheinin, Ph.D. Acting Deputy Director.
Pharmacokinetics (PK): What the body does to the drug? Most drugs: Enter the body by crossing barriers Distributed by the blood to the site of action Biotransform.
Stefan Franzén Introduction to clinical trials.
Virtual Drug Development in Southern California, A Pre-Clinical Focus in vitro tests to support IND submissions David Johnson, Ph.D. Director, DMPK MicroConstants,
ERT 420 BIOPHARMACEUTICAL ENGINEERING Semester 1 Academic Session 2012/2013 HUZAIRY HASSAN School Of Bioprocess Engineering Universiti Malaysia Perlis.
Essential Considerations for Your IND Submission: Objectives and Pitfall Avoidance in Your Preclinical Program Darren Warren.
Exploratory IND Studies
Nonclinical Perspective on Initiating Phase 1 Studies for Small Molecular Weight Compounds John K. Leighton, PH.D., DABT Supervisory Pharmacologist Division.
Clinical Pharmacy Part 2
 Founded in August 2009, in Nanjing, China  SFDA GLP and AAALAC certified DMPK CRO company  GLP bioanalytical support for small and macro molecule,
Concepts and Applications of Pharmacokinetics
Regulatory requirements Drs. Jan Welink Training workshop: Assessment of Interchangeable Multisource Medicines, Kenya, August 2009.
FARMAKOKINETIKA. INTRODUCTION Historically, pharmaceutical scientists have evaluated the relative drug availability to the body in vivo after giving a.
EMEA London Pharmacokinetic- pharmacodynamic integration in veterinary drug development: an overview P.L. Toutain National Veterinary School ;Toulouse.
Chapter 4 Pharmacokinetics Copyright © 2011 Delmar, Cengage Learning.
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.
Privacy Symposium / HIPAA Summit
CHEE 4401 Definitions drug - any substance that affects the structure or functioning of an organism pharmaceutics - the area of study concerned with the.
FDA Office of Orphan Products Development
Overview of Mass Spectrometry
LIQUID CHROMATOGRAPHY-MASS SPECTROMETRY
INTRODUCTION CLINICAL PHARMACOKINETICS
Malaysia, EVALUTION OF DOSSIERS IN WHO- PREQUALIFICATION PROJECT MULTISOURCE TB-DRUGS Evaluation of bioavailability/bioequivalence data Based,
European Patients’ Academy on Therapeutic Innovation The key principles of pharmacology.
Interchangeability and study design Drs. Jan Welink Training workshop: Assessment of Interchangeable Multisource Medicines, Kenya, August 2009.
PHT 415 BASIC PHARMACOKINETICS
IPCP Core C Activities Aprepitant Dose-exposure Relationship in the Monkey Jeffrey S. Barrett, Ph.D., FCP The Children’s Hospital of Philadelphia Division.
Medica Innova Siriporn Nonenoy, B.N.S. Kingkamol Kaenkratoke, M.Sc. Pharm Promporn Jumnongtanachot, M.Sc. Pharm Ariya Khunvichai, Ph.D. May 21 st, 2008.
In vitro - In vivo Correlation
3M Drug Delivery Systems 3 Introduction A family of hydrofluoroalkane-compatible excipients based on oligomeric lactic acid (OLA) has been proposed for.
The First Conference for Medicines Regulatory Authorities In Sudan and Neighboring Countries Khartoum December 2014 Alain PRAT, Technical Officer,
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.
Drug Discovery &Development
Regulatory– Terms & Definitions רגולציה - מונחים והגדרות
ICH-GCP Avinash Kondawar M. Pharm Lead CRA
Prof. Dr. Basavaraj K. Nanjwade
Mass Spectrometry Vs. Immunoassay
Innovative approach for the quantitative analysis of therapeutic monoclonal antibody (mAb), and simultaneous characterization of Anti-Drug Antibodies (ADA)
Hanneke van der Lee, MD, PhD
Biopharmaceutics Dr Mohammad Issa Saleh.
Clinical Pharmacokinetics
Drug Design and Drug Discovery
Bioequivalence trials: design, evaluation, regulatory requirements
Chapter 1 Introduction of Pharmacology [,fɑ:mə'kɔlədʒi] 药理学
Therapeutic Drug Monitoring chapter 1 part 1
Considerations for Successful Biopharmaceutical Product Development: Discovery to Proof of Concept -A Panel Discussion Stanley C. McDermott, PharmD, MS,
Presentation transcript:

Utah Life Science Summit State of the Industry - Growth and Success through Outsourcing/Partnering Regulated Bioanalysis for the Pharmaceutical Industry Scott A. Reuschel, M.S.F.S Tandem Labs – Salt Lake City, UT A Labcorp Company

Who are we? Who or what is Tandem Labs and what do we do?

Company Overview Our 33rd year of operation 1981 Northwest Toxicology established in SLC, UT (clinical toxicology) 1985 Certified drugs-of-abuse testing lab (NIDA, DoD, SAMSHA) – GC/MS 1994 Established bioanalytical division, SLC, UT – GLP bioanalytical, GC/MS, LC/MS 1998 Established 2nd site in NJ – Discovery PK/GLP bioanalytical, LC/MS 2004 Divested drug testing division – Renamed company to Tandem Labs 2008 Acquired by Laboratory Corporation of America® Holdings 2009 Established 3rd site in San Diego, CA – GLP Immunoanalytical 2010 Acquired BA division of Enthalpy Analytical - RTP, NC (4th Tandem site) – GLP bioanalytical, LC/MS 200+ scientific/support staff; ~50 mass spectrometers across all sites Contract Research Organization (CRO) – Regulated Bioanalysis

Tandem Labs Locations

Regulated Bioanalysis What regulatory agencies have authority over the work performed at Tandem Labs? Not regulated by Clinical Laboratory Improvement Amendments (CLIA)

Regulated Bioanalysis What regulatory agencies have authority over the work performed at Tandem Labs? Not regulated by Clinical Laboratory Improvement Amendments (CLIA) Are regulated by the Food and Drug Administration (FDA)

Regulated Bioanalysis What regulatory agencies have authority over the work performed at Tandem Labs? Not regulated by Clinical Laboratory Improvement Amendments (CLIA) Are regulated by the Food and Drug Administration (FDA) Are subject to the Code of Federal Regulations (CFR) 21 CFR Part 58 – Good Laboratory Practice for Non-Clinical Laboratory Studies 21 CFR Part 11 – Electronic Records, Electronic Signatures

Regulated Bioanalysis What regulatory agencies have authority over the work performed at Tandem Labs? Not regulated by Clinical Laboratory Improvement Amendments (CLIA) Are regulated by the Food and Drug Administration (FDA) Are subject to the Code of Federal Regulations (CFR) 21 CFR Part 58 – Good Laboratory Practice for Non-Clinical Laboratory Studies 21 CFR Part 11 – Electronic Records, Electronic Signatures Also subject to: ICH (International Conference on Harmonization) - Guideline for Good Clinical Practice (GCP) – informed consent, patient confidentiality, blinding

Regulated Bioanalysis What regulatory agencies have authority over the work performed at Tandem Labs? Are subject to additional regulations and guidelines from various international regulatory authorities, including:

Regulated Bioanalysis What regulatory agencies have authority over the work performed at Tandem Labs? Are subject to additional regulations and guidelines from various international regulatory authorities, including: US FDA – Guidance for Industry: Bioanalytical Method Validation

Regulated Bioanalysis What regulatory agencies have authority over the work performed at Tandem Labs? Are subject to additional regulations and guidelines from various international regulatory authorities, including: US FDA – Guidance for Industry: Bioanalytical Method Validation European Medicines Agency (EMA): Guideline on Bioanalytical Method Validation

Regulated Bioanalysis What regulatory agencies have authority over the work performed at Tandem Labs? Are subject to additional regulations and guidelines from various international regulatory authorities, including: US FDA – Guidance for Industry: Bioanalytical Method Validation European Medicines Agency (EMA): Guideline on Bioanalytical Method Validation Brazilian Health Surveillance Agency (ANVISA): Bioanalytical Guidance RDC 27/2012

Regulated Bioanalysis What regulatory agencies have authority over the work performed at Tandem Labs? Are subject to additional regulations and guidelines from various international regulatory authorities, including: US FDA – Guidance for Industry: Bioanalytical Method Validation European Medicines Agency (EMA): Guideline on Bioanalytical Method Validation Brazilian Health Surveillance Agency (ANVISA): Bioanalytical Guidance RDC 27/2012 Japanese Ministry of Health, Labour and Welfare (MHLW): Draft Guideline on Bioanalytical Method Validation in Pharmaceutical Development

Regulated Bioanalysis What is the purpose of all this regulated bioanalysis?

Pharmacodynamics (PD) and Pharmacokinetics (PK) Tandem Labs helps pharmaceutical companies obtain the necessary information to make assessments regarding PD/PK of new drugs that are being developed. Pharmacodynamics (PD) is the study of the biochemical and physiological effects of drugs on the body (i.e. what the drug does to a body). Pharmacokinetics (PK) describes the drug concentration/time course in body fluids resulting from administration of a certain drug dose (i.e. what the body does to a drug). Tandem Labs use mass spectrometry to provide both qualitative and quantitative information to our pharmaceutical partners during all phases of the drug development process.

Drug Discovery and Development Timeline 1 - 5+ yrs 2-3 yrs 1 yr 2 yrs 3 yrs 1-2 yrs Discovery Preclinical Phase I Phase II Phase III Review Phase IV IND to FDA NDA to FDA

Discovery Drug Development Qualitative Analysis (by Mass Spectrometry) In Vitro High-throughput screening (lead generation, identification, and optimization) Metabolite Identification (cytochrome P450 enzymes) Quantitative Analysis (by Mass Spectrometry) Solubility, plasma protein binding, permeability, plasma stability, metabolic stability In Vivo (preliminary animal studies) Determine basic PK parameters (e.g. half-life, oral bioavailability , clearance and tissue distribution) Qualified Assays / “Fit for Purpose”; less stringent acceptance criteria, “quick and dirty”

Preclinical Drug Development Quantitative Analysis (by Mass Spectrometry) Exploratory Toxicology (non-GLP) Dose range finding studies Different species and methods of administration Multiple dosing regimens Definitive Toxicology (GLP) – validated assays General toxicology studies Immunogenicity studies with non-human primates (NHPs) Dose formulation analysis Safety is the key focus

Clinical Drug Development Clinical (Phases I-IV) Quantitative Analysis (by Mass Spectrometry) Validated Assays – not GLP; however, conducted under the principles of GLP First in Human Single Ascending Dose (SAD) Multiple Ascending Dose (MAD) Food Effects (fed vs. fasted) Special Populations Elderly, renal impaired, hepatic impaired Drug-Drug Interaction (DDI) Bioequivalence (BE)

Quantitative Analysis by LC/MS Method Development Extraction conditions, chromatography conditions, MS parameters Very challenging; dictated by the chemistry of the analytes, maximum sensitivity often required, instability, non-specific binding, tight time pressures. Method Validation Highly regulated; A/P, stability, robustness, selectivity, matrix effects, etc. Constantly evolving requirements; additional tests Sample Analysis Tight time pressures, sample dilution, sample discrepancies with clinics, ISR

PK/PD Parameters ADME (Absorption, Distribution, Metabolism, Excretion) Dose Concentration Area under the curve (AUC) Accumulation Bioavailability Clearance (CL) Half life (t ½ ) Cmax

PK/PD Parameters

PK/PD Parameters

Mass Spectrometry in Drug Development Why is Mass Spectrometry such a good tool for these applications? Selective Allows the discrimination of a target analyte to the exclusion of other interferences. Sensitive Can routinely detect analytes at ng/mL, pg/mL and sub pg/mL levels. Compatible with other separation techniques “hyphenated methods” GC-MS, LC-MS, UPLC-MS, CE-MS, etc. Robust / High throughput Versatile

Mass Spectrometry in Drug Development Why is Mass Spectrometry such a good tool for these applications? Selectivity Biological samples are first extracted (PPE, SPE, LLE, SLE) Chromatographic Separation (GC, HPLC, UPLC, Microflow LC) Multiple MS Techniques Full Scan SIM MS/MS (SRM, Product Ion, Precursor Ion, Neutral Loss) TOF/MS HRAM

Mass Spectrometry in Drug Development MS/MS (tandem MS, triple-quadrupole MS) and Selected Reaction Monitoring (SRM) is by far the most commonly used technique for quantitative bioanalysis for PK/PD testing.

Mass Spectrometry in Drug Development LC ionization techniques have also revolutionized quantitative MS bioanalysis for PK/PD testing. Ionization is required for mass spectrometry (charged species; m/z) GC/MS applications were limited to volatile, thermally stable compounds for ionization. Atmospheric ionization techniques (ESI, APCI) removed these limitations, allowing for LC/MS applications to expand to a wider variety of compounds.

Mass Spectrometry in Drug Development

Mass Spectrometry in Drug Development Types of compounds analyzed with LC/MS techniques: Small Molecules (MW < 800 amu) Traditional drugs, NCEs Large Molecules (MW > 800 amu; up to 10-40,000 Da) Biologics Peptides Oligonuclieotides SiRNA Lipids Biomarkers Proteins

Mass Spectrometry in Drug Development

Mass Spectrometry in Drug Development

Acknowledgements Tandem Labs - Salt Lake City, UT Laixin Wang, PhD Min Meng, PhD Troy Voelker, PhD Juan Rogness, MS Life Science Cluster, Utah Governor's Office of Economic Development Kevin Jessing

Q & A 33