Addressing the ADME Challenges of Compound Loss in a PDMS-Based Gut-on-Chip Microphysiological System

Organ Model: Intestine (Caco2)

Application: ADME-Tox

Abstract: Microphysiological systems (MPSs) are promising in vitro technologies for physiologically relevant predictions of the human absorption, distribution, metabolism, and excretion (ADME) properties of drug candidates. However, polydimethylsiloxane (PDMS), a common material used in MPSs, can both adsorb and absorb small molecules, thereby compromising experimental results. This study aimed to evaluate the feasibility of using the PDMS-based Emulate gut-on-chip to determine the first-pass intestinal drug clearance. In cell-free PDMS organ-chips, we assessed the loss of 17 drugs, among which testosterone was selected as a model compound for further study based on its substantial ad- and absorptions to organ chips and its extensive first-pass intestinal metabolism with well-characterized metabolites. A gut-on-chip model consisting of epithelial Caco-2 cells and primary human umbilical vein endothelial cells (HUVECs) was established. The barrier integrity of the model was tested with reference compounds and inhibition of drug efflux. Concentration-time profiles of testosterone were measured in cell-free organ chips and in gut-on-chip models. A method to deduce the metabolic clearance was provided. Our results demonstrate that metabolic clearance can be determined with PDMS-based MPSs despite substantial compound loss to the chip. Overall, this study offers a practical protocol to experimentally assess ADME properties in PDMS-based MPSs.

Identification of pharmacological inducers of a reversible hypometabolic state for whole organ preservation

Organ Model: Liver, Intestine (Caco2)

Application: Organ transplantation

Abstract:

Drugs that induce reversible slowing of metabolic and physiological processes would have great value for organ preservation, especially for organs with high susceptibility to hypoxia-reperfusion injury, such as the heart. Using whole-organism screening of metabolism, mobility, and development in Xenopus, we identified an existing drug, SNC80, that rapidly and reversibly slows biochemical and metabolic activities while preserving cell and tissue viability. Although SNC80 was developed as a delta opioid receptor activator, we discovered that its ability to slow metabolism is independent of its opioid modulating activity as a novel SNC80 analog (WB3) with almost 1,000 times less delta opioid receptor binding activity is equally active. Metabolic suppression was also achieved using SNC80 in microfluidic human organs-on-chips, as well as in explanted whole porcine hearts and limbs, demonstrating the cross-species relevance of this approach and potential clinical relevance for surgical transplantation. Pharmacological induction of physiological slowing in combination with organ perfusion transport systems may offer a new therapeutic approach for tissue and organ preservation for transplantation, trauma management, and enhancing patient survival in remote and low-resource locations.

Direct therapeutic effect of sulfadoxine-pyrimethamine on nutritional deficiency-induced enteric dysfunction in a human intestine chip

Organ Model: Intestine (Duodenum)

Application: Metabolic Disease

Abstract:

Background

Sulfadoxine-pyrimethamine (SP) antimalarial therapy has been suggested to potentially increase the birth weight of infants in pregnant women in sub-Saharan Africa, independently of malarial infection. Here, we utilized female intestinal organoid-derived cells cultured within microfluidic Organ Chips to investigate whether SP could directly impact intestinal function and thereby improve the absorption of essential fats and nutrients crucial for fetal growth.

Methods

Using a human organ-on-a-chip model, we replicated the adult female intestine with patient organoid-derived duodenal epithelial cells interfaced with human intestinal endothelial cells. Nutrient-deficient (ND) medium was perfused to simulate malnutrition, resulting in the appearance of enteric dysfunction indicators such as villus blunting, reduced mucus production, impaired nutrient absorption, and increased inflammatory cytokine secretion. SP was administered to these chips in the presence or absence of human peripheral blood mononuclear cells (PBMCs).

Findings

Our findings revealed that SP treatment effectively reversed multiple intestinal absorptive abnormalities observed in malnourished female Intestine Chips, as validated by transcriptomic and proteomic analyses. SP also reduced the production of inflammatory cytokines and suppressed the recruitment of PBMCs in ND chips.

Interpretation

Our results indicate that SP could potentially increase birth weights by preventing enteric dysfunction and suppressing intestinal inflammation. This underscores the potential of SP as a targeted intervention to improve maternal absorption, subsequently contributing to healthier fetal growth. While SP treatment shows promise in addressing malabsorption issues that can influence infant birth weight, we did not model pregnancy in our chips, and thus its usefulness for treatment of malnourished pregnant women requires further investigation through clinical trials.

Developing an adult stem cell derived microphysiological intestinal system for predicting oral prodrug bioconversion and permeability in humans

Organ Model: Intestine (Duodenum)

Application: ADME-Tox

Abstract: Microphysiological systems (MPS) incorporating human intestinal organoids have shown the potential to faithfully model intestinal biology with the promise to accelerate development of oral prodrugs. We hypothesized that an MPS model incorporating flow, shear stress, and vasculature could provide more reliable measures of prodrug bioconversion and permeability. Following construction of jejunal and duodenal organoid MPS derived from 3 donors, we determined the area under the concentration-time (AUC) curve for the active drug in the vascular channel and characterized the enzymology of prodrug bioconversion. Fosamprenavir underwent phosphatase mediated hydrolysis to amprenavir while dabigatran etexilate (DABE) exhibited proper CES2- and, as anticipated, not CES1-mediated de-esterification, followed by permeation of amprenavir to the vascular channel. When experiments were conducted in the presence of bio-converting enzyme inhibitors (orthovanadate for alkaline phosphatase; bis(p-nitrophenyl)phosphate for carboxylesterase), the AUC of the active drug decreased accordingly in the vascular channel. In addition to functional analysis, the MPS was characterized through imaging and proteomic analysis. Imaging revealed proper expression and localization of epithelial, endothelial, tight junction and catalytic enzyme markers. Global proteomic analysis was used to analyze the MPS model and 3 comparator sources: an organoid-based transwell model (which was also evaluated for function), Matrigel embedded organoids and finally jejunal and duodenal cadaver tissues collected from 3 donors. Hierarchical clustering analysis (HCA) and principal component analysis (PCA) of global proteomic data demonstrated that all organoid-based models exhibited strong similarity and were distinct from tissues. Intestinal organoids in the MPS model exhibited strong similarity to human tissue for key epithelial markers via HCA. Quantitative proteomic analysis showed higher expression of key prodrug converting and drug metabolizing enzymes in MPS-derived organoids compared to tissues, organoids in Matrigel, and organoids on transwells. When comparing organoids from MPS and transwells, expression of intestinal alkaline phosphatase (ALPI), carboxylesterase (CES)2, cytochrome P450 3A4 (CYP3A4) and sucrase isomaltase (SI) was 2.97-, 1.2-, 11.3-, and 27.7-fold higher for duodenum and 7.7-, 4.6-, 18.1-, and 112.2-fold higher for jejunum organoids in MPS, respectively. The MPS approach can provide a more physiological system than enzymes, organoids, and organoids on transwells for pharmacokinetic analysis of prodrugs that account for 10% of all commercial medicines.

Cytokine induced inflammatory bowel disease model using organ-on-a-chip technology

Organ Model: Intestine (Caco2)

Application: Immune Cell Recruitment

Abstract: Over 2 million people in North America suffer from inflammatory bowel disease (IBD), a chronic and idiopathic inflammatory condition. While previous research has primarily focused on studying immune cells as a cause and therapeutic target for IBD, recent findings suggest that non-immune cells may also play a crucial role in mediating cytokine and chemokine signaling, and therefore IBD symptoms. In this study, we developed an organ-on-a-chip co-culture model of Caco2 epithelial and HUVEC endothelial cells and induced inflammation using pro-inflammatory cytokines TNF-α and IFN-γ. We tested different concentration ranges and delivery orientations (apical vs. basal) to develop a consistently inducible inflammatory response model. We then measured pro-inflammatory cytokines and chemokines IL-6, IL-8, and CXCL-10, as well as epithelial barrier integrity. Our results indicate that this model 1. induces IBD-like cytokine secretion in non-immune cells and 2. decreases barrier integrity, making it a feasible and reliable model to test the direct actions of potential anti-inflammatory therapeutics on epithelial and endothelial cells.

Modelling Cancer-Microbe Interactions with Organoids and Organ-On-Chip

Lena Schorr is a PhD student in the Epithelium Microenvironment Interaction Laboratory (EMIL) led by Dr. Jens Puschhof in the Microbiome & Cancer Division of the German Cancer Research Center in Heidelberg. She is investigating microbe-host interactions using organoid and organ-on-chip models to further elucidate the role of intracellular bacteria in colorectal cancer progression and metastasis.  

Fusobacterium nucleatum, a prominent bacterium that is enriched in colorectal cancer, is her target bacterium of interest. She studied biology and immunology at the FAU in Erlangen, Germany, did research stays at the Rockefeller University and Roche and is now in the DKFZ International PhD Program. She has been awarded several stipends such as the DAAD scholarship and was a participant and team leader in iGEM, the world’s largest synthetic biology competition. 

This presentation is from a featured session at Netherlands MPS Day, which took place on 11/15/2023.

Constructing a Complex Culture System of the Celiac Disease Intestinal Mucosal Barrier on Chip

Featured session at Netherlands MPS Day, which took place on 11/15/2023.

SCRIB controls apical contractility during epithelial differentiation

Organ Model: Intestine (Caco2)

Application: Model Development

Abstract: Although mutations in the SCRIB gene lead to multiple morphological organ defects in vertebrates, the molecular pathway linking SCRIB to organ shape anomalies remains elusive. Here, we study the impact of SCRIB-targeted gene mutations during the formation of the gut epithelium in an organ-on-chip model. We show that SCRIB KO gut-like epithelia are flatter with reduced exposed surface area. Cell differentiation on filters further shows that SCRIB plays a critical role in the control of apical cell shape, as well as in the basoapical polarization of myosin light chain localization and activity. Finally, we show that SCRIB serves as a molecular scaffold for SHROOM2/4 and ROCK1 and identify an evolutionary conserved SHROOM binding site in the SCRIB carboxy-terminal that is required for SCRIB function in the control of apical cell shape. Our results demonstrate that SCRIB plays a key role in epithelial morphogenesis by controlling the epithelial apical contractility during cell differentiation.

Improving In Vitro Cancer Modeling with Chip-A1

Webinar Abstract

Oncology drug candidates are currently the least likely type of therapeutic to succeed in clinical trials, with only 5.1% of Phase I candidates going on to receive FDA approval1. Understanding a tumor’s microenvironment is key to regulating cancer progression and developing more effective therapies—and Chip-A1 will give researchers this capability. 

In this August 15, 2023 webinar, Luke Dimasi, Senior Director of Product Management at Emulate, provided an overview of the Chip-A1 Accessible Chip, a new Organ-Chip consumable that expands the applications of Organ-on-a-Chip technology by allowing users to create thicker, multilayered tissues within the epithelial culture chamber and directly treat the tissue with topical or aerosolized drugs. Following this introduction, Elee Shimshoni, PhD, postdoctoral researcher at MIT, discussed how she and her former team from the Wyss Institute at Harvard used a prototype of Chip-A1 and found that it offered a new approach for studying epithelial-stromal interactions in Barrett’s Esophagus as well as the broader underlying mechanisms associated with esophageal cancer progression. 

During this webinar, the speakers discussed: 

  • New capabilities of Chip-A1 that expand the applications of Organ-on-a-Chip technology 
  • Why Chip-A1 improves in vitro modeling of epithelial-stromal interactions and tumor microenvironments 
  • A case study of using Chip-A1 to model Barrett’s Esophagus
  • How a Barrett’s Esophagus Chip-A1 model could potentially serve as a tool for personalized drug-response assessments between different patients or genetic subpopulations

About Organ-on-a-Chip Technology 

Organ-on-a-Chip technology is poised to deliver a paradigm shift in drug discovery. By emulating human physiology, Organ-Chips have the potential to increase the predictive power of preclinical modeling and advance more drugs to the clinic. Learn more about Organ-on-a-Chip technology by downloading our free eBook.

Improving Gene Therapy Development with Organ-on-a-Chip Technology

With the potential to bring cures to some of the world’s most devastating diseases, including cancer, muscular atrophy, and blindness, gene therapy is a cutting-edge area of research that is generating a tremendous amount of excitement. Download this free eBook to learn what gene therapy is, how it works, and why Organ-on-a-Chip technology could help gene therapy reach widespread use.