2017 Archived Content

WPE-PTX


The conference on Optimizing Leads and Predicting Drug Toxicity looks at the scientific and technological progress being made to better optimize drug candidates and accurately predict drug related toxicities. What assays and models are being used, how reliable and predictable is the data, and how is this information translated into knowledge that can impact decision-making? Hear experiences shared by experts and join the interactive sessions and panel discussions for active networking, brainstorming and collaborating.

Recommended Package:


Final Agenda

Wednesday 15 November

7:00 Registration and Morning Coffee

Innovative Tools for Drug Toxicity Testing

8:25 Chairperson’s Opening Remarks

Christopher Goldring, Ph.D., Senior Lecturer, Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool

8:30 Evaluation of Lead Optimization and Toxicity Risk Assessment with Human-on-a-Chip Systems

James_HickmanJames J. Hickman, Ph.D., Founding Director, NanoScience Technology Center and Professor, Nanoscience Technology, Chemistry, Biomolecular Science, Material Science and Electrical Engineering, University of Central Florida

Phenotypic screening systems for lead optimization have the advantage of providing useful information without a known target. Human-on-chip systems can potentially establish a therapeutic index before evaluation in animal models or clinical trials and also give the possibility of target identification. These innovative platforms can make lead optimization more efficient which can lead to a greater number of successful clinical trials and de-risk the process.

9:00 All Optical Cardiac Toxicity Testing with Optogenetics and Calcium Imaging

Matthew_DanielsMatthew J. Daniels, MA, Ph.D., MRCP, Wellcome Trust Intermediate Clinical Fellow, Principal Investigator, Division of Cardiovascular Medicine, and BHF Oxbridge Centre of Regenerative Medicine, Oxford University

Detection of QT prolongation pre-clinically requires measurements reflecting the action potential duration (APD). Since the APD is influenced by beat frequency methods to control the cell are also needed. Normally this is achieved with electrical stimulation and patch clamp. These require contact with the cell and with the dish, constraining throughput. We replace those with genetically encoded tools to make this possible in a microscope.

9:30 2017 – Growing Confidence in Use of hiPSC-Cardiomyocyte Assays in Early Drug Hazard Identification

Ard C.H. Teisman, Ph.D., Scientific Director, Global Safety Pharmacology, Discovery Sciences, Janssen Research & Development 

 Selecting the best candidates for a drug-pipeline is a desirable but challenging process. Safety assessment from preclinical models is often essential within this selection process. Nevertheless, accurately predicting clinical effects remains a challenge with many of the existing preclinical models. Human stem cells at least bridge the “species difference gap”, and therefore hiPSC-cardiomyocyte are becoming more accepted within safety testing and drug candidate selection.  

10:00 Grand Opening Coffee Break in the Exhibit Hall with Poster Viewing

10:45 Update on IMI MIP-DILI Consortium Efforts: Stem Cells and More

Chris_GoldringChristopher Goldring, Ph.D., Senior Lecturer, Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool

Drug-induced injury is a major cause of drug attrition, leading to the withdrawal of potentially valuable therapies. A particular problem is drug-induced liver injury (DILI), and our work in the MIP-DILI consortium (Mechanism-Based Integrated Systems for the Prediction of Drug-Induced Liver Injury) will form the basis of this talk, which will cover the range of different model systems (simple and complex cell models, including stem cells) that have been evaluated for their relevance to man as well as mechanisms of DILI. 

11:15 Evaluation of Toxicogenomics Models Using the Literature on Human Toxicity

Miguel AndradeMiguel Andrade, Ph.D., Professor, Bioinformatics, Johannes Gutenberg University of Mainz and Adjunct Director, Institute of Molecular Biology

Toxicogenomics resources describe the effects of toxic compounds in system models that can be in vitro (including human cell lines) or in vivo (including rat and mouse whole organs). A problem remains that is to evaluate how these models recapitulate toxicity in humans. I will present approaches that we proposed that evaluate toxicogenomics models comparing compound-induced gene expression profiles with the literature on human toxicity.

11:45 Enjoy Lunch on Your Own

 

Understanding Mechanisms Underlying Drug Toxicity

13:45 Chairperson’s Remarks

Jordi Mestres, Ph.D., Head, Systems Pharmacology Group, IMIM-UPF Joint Research Programme on Biomedical Informatics; Associate Professor, Universitat Pompeu Fabra (UPF), Spain

13:50 In silico Prediction of Hepatotoxicity – Use of eTOX in vivo Data for Development of New in silico Tools

Alexander_AmbergAlexander Amberg, Ph.D., Computational Toxicologist, R&D Preclinical Safety, Sanofi

Within the eTOX consortium, in vivo data were extracted from legacy toxicity reports of 13 participating companies. With this approach, a database containing high detail toxicity results (histopathology, clinical chemistry, etc.) from 1414 compounds was compiled. This presentation will show how hepatotoxicity data was extracted from this database, and how it was used for the development of new in silico tools for the prediction of DILI.

14:20 Drug Safety Prediction in the Era of Precision Medicine

Jordi_MestresJordi Mestres, Ph.D., Head, Systems Pharmacology Group, IMIM-UPF Joint Research Programme on Biomedical Informatics; Associate Professor, Universitat Pompeu Fabra (UPF), Spain


14:50 Refreshment Break in the Exhibit Hall with Poster Viewing

15:30 The Emerging Role of MicroRNAs in Toxicity Detection

Rachel Church, Ph.D., Research Assistant Professor, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, and Head, Hamner-UNC Organ Injury Biomarker Core

Safety concerns, identified when pharmaceutical compounds reach clinical trials, are a major cause for drug attrition, highlighting the need for novel biomarkers with improved translatability. MicroRNAs (miRNAs) are non-coding RNAs that post-transcriptionally regulate gene expression, are highly conserved across species, and released into biofluids (blood, urine) in response to organ toxicity. The potential for miRNAs to serve as translational biomarkers of organ toxicity will be discussed.

16:00 Interference of Drugs with Canalicular Bile Formation

Bruno_StiegerBruno Stieger, Ph.D., Group Leader, Department of Clinical Pharmacology and Toxicology, University Hospital, Zurich

Bile is rich in phosphatidylcholine and bile salts. The latter are detergents and are cytotoxic. Canalicular bile formation involves an array of ABC-transporters. Interference of drugs with selected ABC-transporters may lead to the impairment of canalicular bile formation followed by accumulation of bile salts in hepatoctyes. Such an accumulation leads to drug-induced cholestasis and, if persistent, to severe liver disease. This presentation will present evidence that some drugs may inhibit BSEP and/or MDR3, two key transporters for canalicular bile formation.

16:30 Breakout Discussion Groups

This session features various discussion groups that are led by a moderator/s who ensures focused conversations around the key issues listed. Attendees choose to join a specific group and the small, informal setting facilitates sharing of ideas and active networking.

TOPIC: Cardiotoxicity Assessments: New Tools and Strategies
Moderators:
James J. Hickman, Ph.D., Founding Director, NanoScience Technology Center and Professor, Nanoscience Technology, Chemistry, Biomolecular Science, Material Science and Electrical Engineering, University of Central Florida
Matthew J. Daniels, MA, Ph.D., MRCP, Wellcome Trust Intermediate Clinical Fellow, Principal Investigator, Division of Cardiovascular Medicine, and BHF Oxbridge Centre of Regenerative Medicine, Oxford University

  • Maturity of human cardiomyocytes
  • Functional Measurements as opposed to biomarker and viability assays
  • Integration with other organs
  • Use of induced pluripotent stem cells for cardiotoxicity assessments

TOPIC: Early Predictions of Hepatotoxicity and Idiosyncratic Toxicity
Moderator: Christoph Funk, Ph.D., Head Mechanistic ADME, Roche Innovation Center Basel, F. Hoffmann-La Roche

  • Experimental approaches: Mechanistic tools (reactive metabolites, transporter inhibition, mitochondrial toxicity) versus holistic tools (3D cultures; organs on chips)
  • In vitro to in vivo extrapolation (role of compound concentration, host-specific factors)

TOPIC: Lead Optimization for Drug Safety
Moderator: Bruno Stieger, Ph.D., Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Zurich, Switzerland

  • Understanding drug transport and role of drug transporters
  • Role of reactive drug metabolites in drug toxicity
  • Methodologies: What experimental systems are available?

 

17:30 Welcome Reception in the Exhibit Hall with Poster Viewing

18:30 Close of Day and Dinner Short Course Registration

19:00 Recommended Dinner Short Course*

(SC1) New Technologies for Improving Drug Safety Screening

*Separate registration required

Thursday 16 November

 8:00 Registration and Morning Coffee

Lead Optimization and Early Safety Risk Assessments

8:25 Chairperson’s Remarks

Nigel Greene, Ph.D., Director, Head of Predictive Compound Safety and ADME, AstraZeneca

8:30 Summary of Breakout Discussions

Participants: Moderators of the Breakout Discussions Hosted on Nov 15

9:00 Impact of Secondary Pharmacology Data: Integration of Translational Information in Research to Better Mitigate the Risk of Adverse Clinical Effects

Bérengère_DumotierBérengère Dumotier, Ph.D., Secondary Pharmacology Expert, Safety Pharmacology, Novartis Pharma AG

Adverse Drug Reactions (ADRs) still often cause drug development failure or withdrawal. Considering the molecular pathways involved in the most severe clinical side effects is an integrated part of the early drug development phases. How to best share translational information based on analysis of marketed drugs plasma exposure/ADR relationship and use this information to minimize the risk of severe adverse effects in later phases will be the main topics of this presentation.

 Zeclinics 9:30 ZeGlobalTox - An Innovative Approach to Address Organ Drug Toxicity Using Zebrafish

Davide_DAmicoDavide D'Amico, Ph.D., CEO, Management Board, ZeClinics

Assessments of drug-induced organ-toxicity using zebrafish larvae can provide high predictivity and effectively bridge the gap between preclinical in vitro safety evaluation and rodent models. ZeGlobalTox is an innovative assay that integrates sequential cardio-, neuro- and hepatotoxicity assessment in the same animal, allowing a strong impact in 3R implementation. 

10:00 Coffee Break in the Exhibit Hall with Poster Viewing

10:45 Implications of Reactive Metabolite Formation for Lead Optimization in Drug Discovery

Andreas_BrinkAndreas Brink, Ph.D., Principle Scientist Drug Metabolism, Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd.

Bioactivation of drugs to chemically reactive metabolites is a potential safety liability. A practice often applied to minimize the bioactivation potential of lead structures is to guide rational drug design based on the detection of stable trapping products of reactive metabolites with e.g. glutathione. This contribution illustrates this strategy, exemplified by experiences at F. Hoffmann-La Roche over the last decade.

11:15 Target Tissue-Focused ADMET

Christoph_FunkChristoph Funk, Ph.D., Head Mechanistic ADME, Roche Innovation Center Basel, F. Hoffmann-La Roche

Active targeting of drugs to particular target organs such as liver, tumor and brain can be achieved by targeting drug transporters or specific enzymes for prodrug activation. This approach can significantly enlarge the therapeutic window by increasing efficacy at reduced drug exposure/safety. However, target-tissue specific ADMET properties, such as polymorphism, regulation and ontogeny of involved mechanisms, disease state as well as compound distribution, have to be assessed in order to reliably predict safety and efficacy in different patient populations.

11:45 Using 21st Century Science in the Design of Safer Medicines

Nigel Greene, Ph.D., Director, Head of Predictive Compound Safety and ADME, AstraZeneca

Technological advances in the last decade have greatly enhanced our ability to screen larger and larger numbers of compounds for a variety of safety endpoints of concern in pharmaceutical development. These large data sets have led to an increased understanding of trends in chemical features and properties that lead to an increased likelihood of seeing adverse events. This presentation will provide an overview of these properties of compounds and the challenges of using high-throughput assays in the design of novel therapeutics.

12:15 Close of Conference


WPE-CAG-Web