Wednesday, 9 September 2020

08:00

Conference Registration and Morning Coffee


Session Title: Conference Opening Session

09:00

Amy  ShenKeynote Presentation

Detection of Antibodies Against SARS-CoV-2 Spike Protein by Gold Nanospikes in an Optomicrofluidic Chip
Amy Shen, Professor and Provost, Okinawa Institute of Science and Technology Graduate University, Japan

The ongoing global pandemic of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has led to active research in its associated diagnostics and medical treatments. While quantitative reverse transcription polymerase chain reaction (qRT--PCR) is the most reliable method to detect viral genes of SARS-CoV-2, serological tests for specific antiviral antibodies are also important as they identify false negative qRT--PCR responses, track how effectively the patient's immune system is fighting the infection, and are potentially helpful for plasma transfusion therapies. In this work, based on the principle of localized surface plasmon resonance (LSPR), we develop an optomicrofluidic sensing platform with gold nanospikes, fabricated by electrodeposition, to detect the presence and amount of antibodies specific to the SARS-CoV-2 spike protein in 1 uL of human plasma diluted in 1 mL of buffer solution, within ~30~min. The target antibody concentration can be correlated with the LSPR wavelength peak shift of gold nanospikes caused by the local refractive index change due to the antigen-antibody binding. This label-free microfluidic platform achieves a limit of detection of ~0.08~ng/mL (~0.5~pM), falling under the clinical relevant concentration range. We demonstrate that our opto-microfluidic platform offers a promising point-of-care testing tool to complement standard serological assays and make SARS-CoV-2 quantitative diagnostics easier, cheaper, and faster.

09:30

Lorena DiéguezKeynote Presentation

Using Microfluidics for Non-Invasive Cancer Diagnosis
Lorena Diéguez, Leader of the Medical Devices Research Group, INL- International Iberian Nanotechnology Laboratory, Portugal

Microfluidics presents numerous advantages for the handling of biological samples, as it provides careful control of fluids in the microscale. When it comes to biomarkers enrichment, microfluidics has demonstrated superior sensitivity and enhanced recovery compared to traditional methods. Incorporating sensors, lab-on-a-chip technologies offer efficient characterization of disease biomarkers from body fluids, making microfluidics ideal for clinical practice, enabling high throughput, portability, and automation. Early dissemination of cancer is difficult to detect by traditional imaging and pathological methods. While the presence of cancer material in body fluids is well known, current techniques for the isolation, analysis and characterization of these biomarkers are not efficient enough to be fully applied in clinical routine. In this talk, we present our work for isolation and multiplex analysis of cancer biomarkers from body fluids based on microfluidics, and biosensors towards personalized medicine and earlier diagnosis of cancer.

10:00

Robert VriesKeynote Presentation

Novel Organoid Models to Develop Drug Treatment Strategies
Robert Vries, CEO, HUB Organoids, Netherlands

Organoids such as IPSC derived brain organoids (Lancaster et al Nature 2013) or our adults epithelial stem cell derived organoids (Sato et al., Nature 2009, 2011) are proving to be a major breakthrough in preclinical models. The new patient like models are fundamental change in the way drug discovery and development can be performed. The development of the HUB Organoids started in the lab of Hans Clevers with the discovery of the identity of adult stem cells in human epithelial tissues such as intestine and liver (Barker et al., Nature 2007; Huch et al., Nature 2013). With the identification of these stem cells, we were able to develop a culture system that allowed for the virtually unlimited, genetically and phenotypically stable expansion of the epithelial cells from animals including humans, both from healthy and diseased tissue (Sato et al., Nature 2009, 2011; Gastroenterology 2011; Huch et al., Nature 2013, Cell 2015; Boj et al., Cell 2015).

We have now generated HUB organoid models from most epithelial organs. Recently, we and others have demonstrated that the in vitro response of organoids correlates with the clinical outcome of the patient from which the organoid was derived (Dekkers et al., Sci Trans Med 2016; Sachs et al., Cell 2018; Vlachogiannis et al., Science 2018). In addition, we have developed a coculture system using HUB Organoids and the immune system to study this interaction and drugs that target the role of the immune system in cancer and other diseases.

We have recently developed new models to study intestinal and lung barrier function and transport of the epithelium of these organs. These experiments show how organoids can be used to study mechanism that underly barrier function disruption in IBD or COPD. Furthermore, we have developed new models to study the interaction between immune system and epithelium. The combination of the new coculture models and assay development to study the epithelium allows us new insights into disease mechanisms and drug treatment strategies.

10:30

Morning Coffee Break and Networking in the Exhibit Hall

11:00

Valérie TalyKeynote Presentation

Droplet-based Microfluidics for Cancer Research
Valérie Taly, CNRS Research Director, Professor and Group leader Translational Research and Microfluidics, Université Paris Cité, France

Droplet-based microfluidics has led to the development of highly powerful tools with great potential in High-Throughput Screening where individual assays are compartmentalized within aqueous droplets acting as independent microreactors. Thanks to the combination of a decrease of assay volume and an increase of throughput, this technology goes beyond the capacities of conventional screening systems. Added to the flexibility and versatility of platform designs, such progresses in the manipulation of sub-nanoliter droplets has allowed to dramatically increase experimental level of control and precision. The presentation will aim at demonstrating through selected example, the great potential of this technology for biotechnology and cancer research. A first part of the presentation will exemplify how microfluidic systems can be used to compartmentalize and assay various types of cells without deleterious effects on their viability within complex and controlled platforms. The application of microfluidic systems for different cell-based assays will be demonstrated. Illustrative examples of droplet-based microfluidic platforms with high potential impact for cancer research will be presented. We will also show how by combining microfluidic systems and clinical advances in molecular diagnostic we have developed an original method to perform millions of single molecule PCR in parallel to detect and quantify a minority of target sequences in complex mixture of DNA with a sensitivity unreachable by conventional procedures. To demonstrate the pertinence of our procedures to overcome clinical oncology challenges, the results of clinical studies will be presented.

11:30

Emmanuel DelamarcheKeynote Presentation

Self-Coalescing Flows: A Powerful Method For Integrating Biochemical Reactions In Portable Diagnostic Devices
Emmanuel Delamarche, Manager Precision Diagnostics, IBM Research - Zürich, Switzerland

Diagnostics are ubiquitous in healthcare because they support prevention, monitoring, and treatment of diseases. Specifically, point-of-care diagnostics (POCDs) are particularly attractive for identifying diseases near patients, quickly, and in many settings and scenarios. POCDs can also trace exposure and acquired immunity of populations exposed to infectious diseases and screen metabolic deficiencies of individuals, who may be exposed to severe drug side effects. However, a long-standing challenge with POCDs is the need to integrate reagents in closed devices for a large number of potential applications. Following our previous contributions on developing capillary-driven microfluidic chips for highly miniaturized immunoassays, controlling and monitoring flow with nanoliter precision, and securing diagnostics against counterfeiting with dynamic optical security codes, we recently demonstrated how to shape and fold liquids inside microfluidic chambers to dissolve reagents with extreme precision. In this presentation, I will explain the underlying concept of this method, called self-coalescing flows, and will illustrate how it can be used to perform various assays, ranging from enzymatic assays, to immunoassays and molecular assays. Despite self-coalescing flows being still an open research topic in fluid physics, their implementation is surprisingly facile and robust and therefore may benefit the entire community working on POCDs.

12:00

Regina LuttgeKeynote Presentation

Nervous Systems-on-a-Chip: From Technology to Applied Biomedical Sciences
Regina Luttge, Professor, Eindhoven University of Technology, Netherlands

Challenges in eavesdropping on the complex cell signaling of the human central nervous system is an essential driver for the development of advanced in vitro technologies, called Brain-on-a-Chip. Developments in Brain-on-a-Chip technology focus primarily on the implementation of cortical cells from human stem cell source in a 3D cultured microenvironment. The aim of a recently launched EU project CONNECT is to mimic the in vivo functions of the nervous system in one connected chip system. The creation of new neurodegenerative disease models in this project brings together the knowledge accumulated among neuroscientists, stem cell experts and engineers to investigate the origins and possible treatments for Parkinson's disease. In this presentation, we will discuss in detail the technical approach of a nervous system on a chip as a unique tool for modelling the neural pathway of connected tissues on the brain-gut axis. In addition to design criteria for these microliter-sized physiological cell culture systems, the presentation will focus on guidance of the growth process of axon protrusions and the local control of cell differentiation processes while maintaining physiological conditions.

12:30

Networking Lunch in the Exhibit Hall, Exhibits and Poster Viewing


Session Title: Technologies and Applications in the Organs-on-Chip Space

14:00

Peter ErtlKeynote Presentation

Joint-on-a-Chip: Monitoring the Onset and Progression of Inflammatory Responses in a Personalized Rheumatoid Arthritis Model
Peter Ertl, Professor of Lab-on-a-Chip Systems, Vienna University of Technology, Austria

Rheumatoid arthritis is an autoimmune disease that causes inflamed joints through recruitment of immune cells, synovitis as well as degradation of cartilage. As a result, patients suffer from intense joint pain, are limited in their daily activities and have a lower life expectancy. To date almost 1% of the population worldwide is affected by arthritis, but no cure is available and pain reduction remains the treatment of choice. Although a number of drugs have shown to decrease disease progression, remission is achieved in less than 50% of patients, indicating a patient-specific drug response. Consequently, a personalized disease model is urgently needed to identify suitable drug combinations and concentrations for each individual patient. In this presentation the development of a sensor-integrated joint-on-a-chip platform capable of mimicking the physiological environment of a human diarthrodial joint is presented and observed patient variability discussed.

14:30

Séverine Le GacKeynote Presentation

Organ-on-a-Chip Platforms for Assisted Reproductive Technologies
Séverine Le Gac, Professor, Applied Microfluidics for Bioengineering Research, MESA+ Institute for Nanotechnology, University of Twente, Netherlands

Organ-on-a-chip platforms are currently considered as the next-generation in vitro models for various fields of applications such as drug and toxicity screening, disease modeling, tissue regeneration, metabolic studies, etc. Key-advantages offered by these platforms compared to standard in vitro models are, for instance, the possibility to accurately control the cellular microenvironment and to implement dynamic culture conditions in a microfluidic format, to emulate the architecture and/or the function of targeted organs by combining specific microstructures with cells, and to stimulate the cells in the device, e.g., chemically, mechanically, electrically, with an excellent spatiotemporal control. In my presentation, I will present current work from our group in the field of organ-on-a-chip with a particular focus on the field of assisted reproductive technologies (ART), with devices for the in vitro culture of mammalian embryos, the in vitro production of bovine embryos (oviduct-on-a-chip platform) and the ex vivo culture of testis tissues.

15:00

Developing Novel High-Throughput Organ-on-a-Chip Tissue Models for Drug Discovery and Unlocking Their Full Potential Using High-Content Imaging and Analysis
Silvia Bonilla García, Scientist, MIMETAS B.V., Netherlands
Kenneth Seistrup, Application Scientist, Molecular Devices, Denmark

In this presentation, scientific experts from MIMETAS and Molecular Devices will introduce the culture and interrogation of complex 3D tissues models in the OrganoPlate®, a unique 3D organ-on-a-chip platform, including models for gut-on-a-chip, angiogenesis and immuno-oncology. We will show examples of Organ-on-a-Chip tissues for use in disease modeling and drug safety testing and will present the new OrganoPlate® Graft, which allows vascularization of spheroids, organoids and PDX explants, and offers a powerful solution for the study of angiogenesis, an important focus for cancer therapeutics. These complex 3D tissue cultures can be interrogated in a high-throughput manner using powerful cellular imaging and analysis systems. We will show how Molecular Device’s ImageXpress imaging systems play a vital role in the development and analysis of 3D tissue models, such as those built on the OrganoPlate® platform, by enabling translatable assays and readouts for healthy and diseased human organ models as well as quantitative characterization of complex phenotypic effects.

15:30

Afternoon Coffee Break in the Exhibit Hall

16:00

The Glomerulus-on-a-Chip as a Platform for Disease Modeling, Drug Screening, Biomarker Discovery and mechanistic Studies
Stefano Da Sacco, Assistant Professor of Urology, GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics in Urology, Keck School of Medicine – University of Southern California, United States of America

Within the kidney, the glomerulus is the structure in charge of the renal ultrafiltration. Loss of glomerular function leads to renal damage and could end with irreversible damage. The major roadblocks to advancing new drugs and therapeutics designed specifically to preserve glomerular function stem from the inability to effectively develop multicellular in vitro models that can accurately mimic the architecture of the glomerular filtration barrier. We have recently developed a barrier-free, human based glomerulus-on-a-chip system that closely replicates the glomerular filtration barrier and its functions. In this session we will present our platform and discuss our newer insights on modeling of various diseases including membranous nephropathy, Alport Syndrome and diabetic nephropathy as well as describe potential applications ranging from mechanistic studies, biomarker discovery and personalized medicine.

16:30

Blood Vessels-on-Chips
Andries D. van der Meer, Associate Professor, Scientific lead, Organ-on-Chip Center Twente, University of Twente, Netherlands

The total surface area of blood vessel wall in the human body is more than 1,000 m2, equal to more than five tennis courts. The vessel wall is not simply a physical barrier between blood and surrounding tissue, it is also a remarkably plastic structure, which plays a key role in the pathophysiology of many diseases. Organs-on-chips are ideal platforms to model blood vessel biology in vitro, because they allow researchers to set defined and dynamic patterns of flow, pressure, geometry and levels of oxygen, all of which are essential aspects of blood vessel physiology. Moreover, organs-on-chips enable the controlled co-culture between blood vessel tissue and neighboring tissues and matrices, ultimately allowing studies of disease mechanisms. In this talk, I will show how blood vessel tissue, primarily endothelial cells, can be integrated in organs-on-chips to realistically mimic blood vessel physiology. Moreover, I will show how exposing these blood vessels to activating stimuli, like inflammatory cytokines, can induce vascular adhesion molecule expression, vascular leakage and thrombosis. Finally, I will discuss how organ-on-chip technology allows the engineering of ‘personalized’ in vitro models of blood vessels by mimicking 3D vessel structures based on medical imaging, perfusion of blood samples spiked with disease-related cytokines and incorporation of vascular tissue derived from human induced pluripotent stem cells.

17:00

James HickmanKeynote Presentation

Body on a Chip: Human Microscale Models for Drug Development
James Hickman, Professor, Nanoscience Technology, Chemistry, Biomolecular Science and Electrical Engineering, University of Central Florida; Chief Scientist, Hesperos, United States of America

The preclinical drug development process is inefficient at selecting drug candidates for human clinical trials, since only 11% of drug candidates selected for clinical trials exit with regulatory approval. Current technology is based on isolated human cells and animal surrogates.  We believe that a “human” multiorgan model based on physiologically based pharmacokinetics-pharmacodynamic (PBPK-PD) models that house interconnected modules with tissue mimics of various organs.  The system captures key aspects of human physiology that would potentially reduce drug attrition in clinical trials and decrease the cost of development. Integrated, multi-organ microphysiological systems (MPS) based on human tissues (also known as “body-on-a-chip”) could be important tools to improve the selection of drug candidates exiting preclinical trials for those drug most likely to earn regulatory approval from clinical trials. This methodology integrates microsystems fabrication technology and surface modifications with protein and cellular components, for initiating and maintaining self-assembly and growth into biologically, mechanically and electronically interactive functional multi-component systems.

I will describe such systems being constructed at Hesperos and at UCF that are guided in their design by a PBPK model. They are “self-contained” in that they can operate independently and do not require external pumps as is the case with man other microphysiological systems. They are “low cost”, in part, because of the simplicity and reliability of operation. They maintain a ratio of fluid (blood surrogate) to cells that is more physiologic than in many other in vitro systems allowing the observation of the effects of not only drugs but their metabolites. While systems can be sampled to measure the concentrations of drugs, metabolites, or biomarkers, they also can be interrogated in situ for functional responses such as electrical activity, force generation, or integrity of barrier function. Operation up to 28 days has been achieved allowing observation of both acute and chronic responses with serum free media. We have worked with various combinations of internal organ modules (liver, fat, neuromuscular junction, skeletal muscle, cardiac, bone marrow, blood vessels and brain) and barrier tissues (eg skin, GI tract, blood brain barrier, lung, and kidney). The use of microelectrode arrays to monitor electrically active tissues (cardiac and neuronal) and micro cantilevers (muscle) have been demonstrated. Most importantly these technical advances allow prediction of both a drug’s potential efficacy and toxicity (side-effects) in pre-clinical studies. This talk will also give results of six workshops held at NIH to explore what is needed for validation and qualification of these new systems.

17:30

Close of Day 1 of the Conference

Thursday, 10 September 2020

08:00

Morning Coffee


Session: Organoids, Organ-on-a-Chip and Tissue-on-a-Chip -- Where are these Fields Heading?

09:00

David HayKeynote Presentation

Development of an Automated Tissue-Engineering Platform to Produce Human Liver Tissue for Basic and Applied Research
David Hay, Chair of Tissue Engineering, MRC Centre for Regenerative Medicine, University of Edinburgh, United Kingdom

Liver disease represents an increasing cause of global morbidity and mortality. Currently, liver transplant is the only treatment curative for end stage liver disease. Donor organs cannot meet the demand and therefore scalable treatments and new disease models are required to improve clinical intervention. Pluripotent stem cells represent a renewable source of human tissue. Recent advances in three-dimensional cell culture have provided the field with more complex systems that better mimic liver physiology and function. Despite these improvements, current cell based models are variable in performance and expensive to manufacture at scale. This is due, in part, to the use of poorly defined or cross-species materials within the process, severely affecting technology translation. To address this issue, we have developed an automated and economical platform to produce liver tissue at scale for modelling disease and small molecule screening. Stem cell derived liver spheres were formed by combining hepatic progenitors with endothelial cells and stellate cells, in the ratios found within the liver. The resulting tissue permitted the study of human liver biology ‘in the dish ‘and could be scaled for screening.Going forward we believe that this resource will not only serve as an in vitro resource and may have an important role to play in supporting failing liver function in humans.

09:30

Exploring Gut-Vagus Nerve Interactions on a Microfluidic Chip
Lena Sophie Koch, PhD Candidate , University of Twente, Netherlands

As a first step towards better understanding the gut-brain axis, the intestinal epithelium and the vagus nerve were differentiated from human pluripotent stem cells. Using a microfluidic chip to spatially arrange these units, enabled insight in their convergence and interactions.

10:00

Microfluidic ChipShopIt’s the Economy – Industrial Aspects of Organ-on-a-Chip Device Manufacturing
Holger Becker, Chief Scientific Officer, Microfluidic ChipShop

While academic activities in the organ-on-a-chip field have multiplied in recent years, it becomes apparent that translating academic results into commercially viable products can be challenging. This is even more true for such devices which require a generically multidisciplinary approach, combining application know-how with surface chemistry, microfabrication and materials technology. In this presentation, we will give an overview over available solutions for such products and explain classical pitfalls on the way from the academic laboratory bench to an industrial product.

10:30

Morning Coffee Break and Networking in the Exhibit Hall

11:00

HepaChip-MP – An organ-like Perfusable Cell Culture System in Multi-well Plate Format for Toxicity Testing
Marius Busche, Researcher, NMI, Germany

We present a microfluidic, continuously perfused in vitro model of the liver-sinusoid in multiwellplate format (HepaChip-MP). Electrodes integrated within the system enable active and simultaneous assembly of only viable liver cells into sinusoid-like structures within 24 independent cultivation chambers. Automated filling by a pipetting robot and tubeless gravitational driven perfusion yields an easy-to-use system operable by non-expert end-users. Reduced vitality after diclofenac treatment could be measured and metabolic functions such as CYP activity and CYP induction were demonstrated. Integrated oxygen sensors allow to measure oxygen concentration and cellular respiration inside the culture chamber.

11:30

Microfabrication Technologies For Engineering a Joint on Chip
Marcel Karperien, Professor, University of Twente, Netherlands

Osteoarthritis (OA) is a degenerative joint disease affecting more than 130.000.000 patients world-wide. The etiology of the disease is still poorly understood. While initially considered as a disease of cartilage it is now clear that the disease involves all tissues in the joint. A disease trigger in each of these tissues can initiate the onset of disease and each of these triggers converge over time in a similar disease presentation. Despite tremendous efforts in the recent past, the disease can still not be treated.  It is believed that lack of translational power of currently used in vitro and in vivo animal models can at least in part this lack of success. It has also become clear that currently used animal models poorly reflect the complexity of human disease nor do they allow the detailed study of the complex interactions between the different joint tissues at various stages of disease. To address these shortcomings my group has started the development of a joint-on-chip. The joint-on-chip has a modular chip design (Piluso et al., 2019). We are engineering chips for each individual tissue, i.e. cartilage, subchondral bone, synovium, ligament and/or meniscus which together form the joint. The individual chips are connected to each other through blood vessel mimicking microfluidic channels  as well as with a chip mimicking the intra-articular space. This latter chip will contain features allowing non-invasive imaging / sensing of inter tissue communication. Since movement is a critical and an essential feature in every joint, the individual chips can all be independently actuated mimicking both compression and shear stress. Prototype chips of the synovium and the cartilage including actuation have become available (Paggi et al., 2020). Additionally we developed various strategies for introducing cell laden membranes composed of natural polymers and/or tissue constructs in the chip. Finally, we have developed sensors that can assess local matrix metalloproteinase activity, a key factor driving joint degeneration on chip. In my presentation I will discuss various engineering aspects of our microfabrication technology platforms needed for recreating a representative and functional human joint. It is expected that these efforts will help us studying the pathophysiology of osteoarthritis and will accelerate the development of dearly needed osteoarthritis disease modifying treatments.

12:00

Vascular Organization in Engineered Tissues: Tuning the Mechanical and Growth Factor Environment
Jeroen Rouwkema, Associate Professor, University of Twente, Netherlands

Engineered tissues are interesting platforms for investigating tissue responses to changing environments including for instance changes in gravity. Additionally, they are highly suitable to test potential drug compounds. However, to achieve a physiological response, an included vascular network is often desired.

In order to achieve multiscale organized vascular networks within engineered tissues, we are developing methodologies to spatially control the organization of vascular cells within tissue analogues. A potent approach in this is to spatially control the mechanical environment, as well as the availability of angiogenic growth factors. By developing transparent ex ovo chicken chorioallantoic membrane (CAM) platforms, we are building tools to investigate the effect of perturbations on in vivo vascular development and organization. This provides us with valuable information that can be translated to an in vitro tissue engineering setting. With this approach, our aim is to locally control tissue remodeling and maturation, resulting in a vascular network that resembles a vascular tree.

12:30

Networking Lunch in the Exhibit Hall and Networking with Exhibitors

13:30

InTESTine Barrier Chip: A Microfluidic ex vivo Model to Study Intestinal Permeability and Host-Microbe Interactions in the Human Intestinal Tract
Hossein Eslami Amirabadi, Scientist, TNO, Netherlands

We developed a microfluidic chip, InTESTine Barrier Chip, with a 3D clicking mechanism that fixes intestinal biopsies between two microfluidic channels. This novel technology enables us to study intestinal processes such as drug absorption and immune responses.

14:00

A Novel System of Infusion to Provide Drugs Within ex vivo Skin Models
Emma Raude, Researcher, LAAS-CNRS/Genoskin, France

We characterize a novel microfluidic system of infusion to provide high molecular weight molecules within ex vivo skin models. The results position the system as a new relevant tool to study the efficacy and/or toxicity of intravenously administered biotherapeutics directly on human skin.

14:30

Coffee Break

15:00

A Biomimetic Human Gut-on-a-Chip For Oral Drug Delivery Testing
Nayere Taebnia, Researcher, Technical University of Denmark, Denmark

This presentation introduces a biomimetic human gut-on-a-chip platform, fabricated using stereolithographic high-resolution 3D printing, which recapitulates the complex geometry of the native tissue and advances our understanding of intestinal physiology and its role in drug uptake and personalized medicine.

15:30

3D-printed Microbioreactor with Integrated Impedance Spectroscopy for Cell Barrier Monitoring
Georg Linz, Researcher, DWI Leibniz-Institut for Interactive Materials, Germany

Today's cell culture experiments often suffer from the limited commercially available housings for flow culture experiments. In recent years additive manufacturing helped researchers to design cell culture bioreactors with integrated online analytics. We present a novel 3D-printed bioreactor enabling epithelial cell culture experiments under homogeneous flow conditions and online barrier monitoring with four integrated electrodes for electrical impedance spectroscopy (EIS).
Transparent indium tin oxide (ITO)-glass as current-injecting electrodes allow direct visualization of the cells and EIS measurement simultaneously. The bioreactor's design takes the importance of homogeneous electric fields into account by placing the voltage pick-up electrodes in the homogeneous electrical field. Subsequent equivalent electric circuit fitting of the impedance data results in cell layer resistance and capacitance.

In the first part of the talk, the bioreactor's design will be presented, including a Comsol simulation of the fluid and electrical fields. In the second part, cultivation results will be shown where epithelial Caco-2 cells were cultivated, and EIS monitored the reaction of the cell layer to different chemicals that alter the cell layer physiology.

16:00

A Novel Soft Lithography Technique for Porous Membrane Fabrication as a Permeable Barrier in Gut- and Lung-on-Chip Devices
Mohammad Jouybar, Researcher, Politecnico di Milano, Italy

To mimic the epithelium-endothelium barrier in microfluidic devices, the fabrication of porous membranes through soft-lithography is a common option. Thus far, SU8 photoresist micropillars are the prevailing master molds. However, they suffer from the fragile behavior of pillars and need sophisticated clean room facility. In this presentation, we introduce a spin-coatable epoxy-based mold of micropillars to overcome the limitations of former techniques.

Both intestinal and lung epithelium cell lines were cultured in microchannels to validate the functionality of the porous membrane. Subsequently, to evaluate the contribution of mechanical stimulation with focus on flow, to the 3D morphogenesis of intestinal epithelium, three different culture methods were tested: pump perfusion, tilting rocker perfusion, and steady culture. The primary results will be presented in this presentation.

17:00

Modeling Immune Mediated Beta Cell Destruction in Human Type 1 Diabetes with Organoids
Matthias von Herrath, Vice President and Senior Medical Officer, Novo Nordisk, Professor, La Jolla Institute, United States of America

In the past 15 years we have been studying the pathology of human type 1 diabetes with access to donor pancreata through the human pancreatic organ donor consortium (nPOD). These studies have led to several findings, for example that certain cytokines are generated by beta cells themselves, sometimes under stress, and also that there are probably key factors that render beta cells susceptible to immune attacks. Mechanistically, the importance and meaning of these observations needs to be addressed in a suitable and easily manipulable in vitro system consisting of human islets and immune cells. We have built such a system in collaboration with the company InSphero and will discuss emerging findings.

17:30

Close of Conference