Microfabrication techniques such as 3D printing, have enabled the synthesis of highly specialised and controlled microchips with well-defined chemical and physical environments. The integration of such chips with modern cell culture techniques has created a new way to mimic the in vivo. Growing excitement around these microphysiological models of human organs on microfluidic cell culture chips has led to a marked interest and investment in such technologies, yet many challenges remain ahead, not least of all scalability and adaptation for high throughput screening. Join us for Cambridge Healthtech Institute’s Inaugural Organ-on-a-Chip and MicroPhysiological Systems symposium, where this promising preclinical model will be explored and evaluated, where future steps towards more accurate and reliable preclinical trials will be taken.

Recommended All Access Package:

27 November: Organ-on-a-Chip and MicroPhysiological Systems

27 November Dinner Course: SC3: The Origins, Optimization and Application of Organ-on-a-Chip Systems

28-29 November: Optimizing Leads and Predicting Drug Toxicity

29-30 November: 3D Cellular Models

29 November Dinner Course: SC5: Humanized Mouse Models: Technology and Applications in Preclinical Assessment of Cancer Immunotherapy

Tuesday 27 November

7:00 Registration and Morning Coffee

SINGLE SYSTEM MODELS

8:20 Welcome Remarks

Joel Hornby, BSc, Conference Director, Cambridge Healthtech Institute

8:25 Chairperson’s Opening Remarks

James J. Hickman PhD, Professor, NanoScience Technology Center, University of Central Florida

8:30 Stem Cell Based MicroPhysiological Organ-on-a-Chip Systems as in vitro Models of Human Tissue with Physiological Structure and Function

Peter Loskill, PhD, Assistant Professor for Experimental Regenerative Medicine, Department of Women’s Health, Research Institute for Women’s Health, Faculty of Medicine, Eberhard Karls University Tübingen

Drug discovery and development to date has relied on animal models, which fail to resemble human physiology. The discovery of human induced pluripotent stem cells (hiPSC) has led to the emergence of a new paradigm of drug screening using patient- and disease-specific organ/tissue-models. One promising approach is the organ-on-a-chip system that integrates hiPSC-derived tissues in microphysiological environments and combines human genetic background, in vivo-like tissue structure, physiological functionality, and “vasculature-like” perfusion.

9:00 Lung Alveolar Models Based on Organs-on-Chip Technologies

Olivier Guenat, PhD, Head Organs-on-Chip Technologies, ARTORG Center, University of Bern

Organs-on-chips are able to recapitulate the in vivo cellular environment in unprecedented way and are widely expected to better predict drug’s response in humans than standard in vitro models. We will present several lung-on-chip models using primary cells from patients that closely recapitulate the cellular environment of the lung parenchyma. These functional models, which mimic the lung alveolar barrier as well as the lung microvasculature, are used to evaluate the effects of various compounds used for respiratory diseases, such as pulmonary fibrosis.

9:30 Development of Organ-on-a-Chip Systems for Disease Modeling In the Vasculature, Liver & Gut

Lech_MatthewMatthew Lech, ScM., Senior Scientist, Inflammation & Immunology Research Unit, Pfizer

The current drug development process results in many clinical trial failures due, in part, to poor translation from early stage development with simple in vitro assays through in vivo preclinical models which do to properly replicate human disease. Companies can do little to recover the effort and cost associated with a clinical trial failure. We are developing three human organ-on-a-chip systems for improved drug development; the liver, vasculature and gut.

10:00 Coffee Break

MULTI SYSTEM MODELS

10:30 Building Phenotypic Body-on-a-Chip Models for Preclinical Toxicological and Efficacy Evaluations Utilizing Stem Cell Derived Disease Models

James_HickmanJames J. Hickman, PhD, Professor, NanoScience Technology Center, University of Central Florida

The utilization of human-on-a-chip systems that incorporate iPSCs allows for understanding different disease states by constructing them utilizing patient cells. We are constructing systems in serum-free medium with functional readouts that employs a pumpless platform. Our group has been constructing these systems with up to 6 organs and demonstrated up to 28 days evaluation of drugs and compounds, that have shown similar response to results seen from clinical data or literature reports.

11:00 Emulating the Gut-Liver Axis - Organ-on-Chip in Preclinical Research

Alexander S. Mosig, PhD, Assistant Professor, Center for Sepsis Control and Care, Jena University Hospital

To investigate the mechanism of infection-related organ dysfunction we developed a microfluidically perfused model of the human gut-liver axis. The in vitro model comprises tissue resident and circulating immune cells to emulate essential components of the human immune system. Biochip-integrated sensors allow continuous monitoring of environmental conditions and allow quantification of tissue integrity. A synthetic microbiome is being integrated to emulate microbiome-host interaction under physiological and pathophysiological conditions.

11:30 Enjoy Lunch on Your Own

TRANSLATIONAL AND APPLIED SYSTEMS

13:25 Chairperson’s Remarks

Peter Loskill, PhD, Assistant Professor for Experimental Regenerative Medicine, Department of Women’s Health, Research Institute for Women’s Health, Faculty of Medicine, Eberhard Karls University Tübingen

13:30 Modeling Rare Diseases with Organs-on-Chips

Tagle_DaniloDanilo A. Tagle, MS, PhD, Associate Director for Special Initiatives, National Center for Advancing Translational Sciences, National Institutes of Health

Developing drugs for rare diseases can be challenging due to the small, heterogeneous patient populations, few disease experts and expert centers, variable and long time-frames for disease progression, a poor understanding of disease natural history, and a lack of prior clinical studies. Recent technological advances, particularly in the area of organs-on-chips and induced pluripotent stem cells (iPSCs) have created opportunities to create a paradigm shift in therapy development, especially in the area of rare disease research.

14:00 Reducing Attrition in Drug Discovery through the Use of Human Translational Cellular Models

Wendy_RowanWendy Rowan, PhD, FRSB, Associate GSK Fellow, Scientific Leader Target Sciences, GlaxoSmithKline

Preclinical efficacy and toxicology data derived from in vitro and animal models often fails to translate to clinical trials, resulting in high rates of attrition and falling Research and Development productivity. The development of more predictive human in vitro systems represents one of the most urgent challenges facing the pharmaceutical industry. In a fast moving field, advances in bioengineering are showing the potential for transforming the outcome of drug discovery.

14:30 Maximizing the Impact of MicroPhysiological Systems with in vitro-in vivo Translation

Murat_CiritMurat Cirit, PhD, Director, Biological Engineering, MIT

Microphysiological systems (MPS) hold promise for improving therapeutic drug approval rates by providing more physiological, human-based, in vitro assays for preclinical drug development activities compared to traditional in vitro and animal models. The full impact of MPS technologies will be realized only when robust approaches for in vitro-in vivo (MPS-to-human) translation are developed and utilized and explain how the burgeoning field of quantitative systems pharmacology (QSP) can fill that need.

15:00 Refreshment Break

15:30 Credibility/Validity of Complex in vitro Models: Crowdsourcing Strategies to Facilitate Acceptance and Use

Sofia_BLeiteSofia Batista Leite, PhD, Scientific/Technical Project Officer, Chemical Safety and Alternative Methods, ECVAM, European Commission – Joint Research Centre (EC-JRC)

Complex in vitro models aim to represent higher-level anatomical and physiological aspects of human biology. Combined with their fast developing technology, these models are very attractive to multiple research and regulatory areas. Further implementation could be achieved by establishing a standardized framework for their assessment. More than 500 people responded to an EURL-ECVAM survey sharing their opinion on if/how such framework should/could be established. The survey results will be presented.

16:00 A Human Heart-Liver Platform to Study Acute and Chronic Cardiotoxicity upon Hepatic Biotransformation

Anne_RiuAnne Riu, PhD, Project Manager, Research and Innovation, L’Oréal

Regulation of cosmetic testing has spurred efforts to develop new methods for systemic toxicity, however, in vitro assays are often lacking xenobiotic metabolism. A heart-liver system was developed to study metabolism-dependent cardiotoxicity combining functional cardiac and metabolically competent hepatic modules maintained under flow for 14 days. The system was characterized with a set of reference compounds and then evaluated with cosmetic ingredients.

16:30 GUT ON-A-CHIP: Intestinal barrier model for studying host microbe-immune responses

van_de_Steeg_EvitaEvita van de Steeg, PhD, Senior Scientist, Human Biology & Microbiology, TNO Pharma Leiden

The majority of screening and predictive models do not reflect the physiology of the human intestinal tract properly, resulting in low translational value to the clinical situation. We were able to fix human intestinal tissue in a microfluidic chip and maintain its functionality under physiological conditions for 24h. The new 3D printed chip is easy to use and will provide us with a higher throughput system.

17:00 Close of Symposium


18:0020:30 Recommended Dinner Short Course*

SC3: The Origins, Optimization and Application of Organ-on-a-Chip Systems

* Separate registration required.


WPE-CAG-Web