image
ORGAN MODELS

Lifelike, functional
human organ tissues.

ellipse

Imagine testing on 10,000 identical biopsies.

We cultivate human tissues in 3D microfluidics to recreate the complex functions and behaviors of native human organs — all at incredible throughput.
background
background
background

Stunning realism.

With each human tissue containing 200,000 to 500,000 cells, our models reconstitute the cell-, tissue-, and organ-scale phenotypes and dynamic functions of native human tissues. Our large, dense models capture critical biochemical and mechanical signaling to model nature's complex interactions.

background
We cultivate human organ models of health and disease. (Shown: lung airway tissue infected with Pseudomonas bacteria.)
background
High-resolution imaging enables precise understanding of cell and tissue physiology in health and disease.
background
Our models mimic the complex functions of the human immune system and its contribution to disease & inflammation.
backgroundbackground
Human tissue biopsy compared to Vivodyne lab-grown human tissues.
iconicon
ellipse
ellipse

Fully perfusable
vascular networks.

Each of our models contains tissue-specific, self-assembled 3D vascular networks that can be fully perfused. This allows us to deliver large and small therapeutics intravascularly, mimicking the physiological transport, trafficking, and cellular transmigration to the tissue parenchyma.

ellipse

Predictive and clinically relevant human data.

We capture data with live and terminal endpoints including secretory sampling, 3D confocal imaging for single-cell phenomics, and single-cell transcriptomics and proteomics, along with flow cytometry, histology, and other traditional modalities.

background
backgroundbackround
ellipse

Consistently
reproducible.

Our proprietary human organ models are cultivated with Vivodyne-optimized formulations in our robotic platforms with full automation and sub-second precision. This level of repeatability allows the models to be reproduced time and time again with exceptional consistency.

Healthy Control
background
Chemotherapeutic
elllipse
Regimen 1
background
Chemotherapeutic
ellipse
Regimen 2
background
ellipse
ellipse

A selection of our optimized tissue models.

Our portfolio of human models of health and disease provide translatable, predictive data of human responses and outcomes. We additionally offer our partners the ability to co-develop new models for discovery & development in novel disease areas.
Bone Marrow

Our bone marrow-on-a-chip facilitates the production of blood cells needed to oxygenate tissues, regulate hemostasis, and provide immunity.

The multi-lineage development of myeloid and erythroid cells from hematopoietic stem and progenitor stem cells (HSPCs) recapitulates the native process of hematopoiesis.

This model supports the screening of anti-cancer drugs for hemotoxicity and the modeling of disease states such as erythropenia, anemia, neutropenia, and immunosuppression.

Small/Large Airway

Our large and small airway models replicate the hallmark features of the bronchi and bronchioles within the lung, comprising beating ciliated cells, mucus producing goblet cells, mature epithelial basal cells and protective club cells.

Lung cell differentiation and mucociliary beating is achieved via culture with an air-liquid interface.

The model faithfully recapitulates neutrophil extravasation in response to inflammatory stimuli such as bacterial infection, cigarette smoke, aerosolized particles, and viruses, and can be leveraged to model chronic obstructive pulmonary disease (COPD).

Liver

Our liver-on-a-chip enables deep analysis of native liver functions such as drug metabolism, protein synthesis, waste clearance and blood glucose regulation.

The fully vascularized liver model supports urea synthesis and albumin production, the formation of robust bile canaliculi, quantification of drug-induced liver toxicity (DILI) biomarkers such as alanine aminotransferase (ALT), and high-content secretome analysis.

This is a ready-to-deploy platform for predicting DILI and drug metabolism and modeling diseases such as hepatitis, fatty liver disease and cirrhosis.

Solid Tumor

In our solid-tumor models, co-cultures of tumor spheroids with parenchymal and stromal cells provide a physiologically relevant niche for modeling cancer biology.

By incorporating key components of the tumor microenvironment, we model the microarchitecture of cancerous tissue, tumor growth, angiogenesis, epithelial-mesenchymal transition (EMT), and metastasis.

The solid-tumor-on-a-chip enables screening of anticancer drugs and next generation immunotherapies.

Pancreas

Our islet-on-a-chip captures the pancreas’ ability to secrete enzymes for digestion and produce hormones for regulating blood glucose levels. The model recapitulates in vivo glucose-stimulated insulin secretion and incorporates all the major hormone producing endocrine cells (alpha, beta, delta, and PP cells) of the pancreas.

This platform supports testing and screening of diabetes drugs for enhancing insulin release and may be leveraged to monitor autoimmune pathogenesis and beta cell destruction.

Intestine

Our intestine-on-a-chip encompasses villi-like structures and the major intestinal cell types including enterocytes, enteroendocrine cells, goblet cells, and paneth cells.

The formation of mature enteroids is achieved via epithelial barrier formation with intestinal barrier function, mucus production, and the development of complex gut-microbiota, all of which contribute to the intestine’s role in food digestion and nutrient absorption.

The model reproduces key functions of the intestine, pathological features of IBD and innate immune responses, allowing for the testing of drug pharmacokinetics/pharmacodynamics (PK/PD), modeling of inflammatory and infectious diseases, and screening of drug compounds for gastrointestinal toxicity.

Placental Barrier

Our model mimics the bi-layered architecture and hemodynamic environment of the native placenta barrier. This regulates the transport of materials between the mother and the fetus during pregnancy.

The model incorporates cytotrophoblasts, which undergo syncytialization to form a cell monolayer with tight intercellular junctions, fetal endothelial cells separated by a basement membrane, and microvilli on the apical surface, which serve to increase the surface area of the maternal-fetal interface and act as sites for the expression of active membrane transporters associated with drug and xenobiotic transfer.

Leverage this to evaluate the PK/PD of maternally administered drugs to improve preclinical evaluation of drug safety in pregnancy.

Uterus (Placental Implantation)

This model recapitulates the adhesion of embryos to the endometrium and subsequent invasion into the maternal tissue, mimicking early implantation and placentation observed in early pregnancy.

We reconstruct the three-dimensional structural organization of the maternal-fetal interface, simulating the invasion of trophoblasts into the maternal uterus and the subsequent arterial remodeling that is necessary for improved blood circulation.

This platform provides insights into implantation abnormalities and disorders of placentation, enabling the development of therapeutics for early pregnancy and the evaluation of drug compounds' off-target effects.

Skeletal Muscle

Our skeletal muscle-on-a-chip recapitulates the anisotropic structure and mechanical properties of native muscle, comprising of realistic muscle that undergoes stimulated contraction.

We establish a microenvironment that supports the myogenic differentiation of mesenchymal stromal cells and the formation of aligned multinucleated myotubes and myofibers with directional organization.

This platform faithfully models oxidative muscle injury and cachexia, enabling screening of therapies for muscle injuries or drug toxicity against skeletal muscle.

Blood-Brain-Barrier

The blood-brain barrier (BBB)-on-a-chip comprises specialized microvascular networks, mural cells, and astrocytes that together mimic the barrier function of the human BBB to separate blood from brain interstitial fluids.

Small molecule and immune cell transport across the BBB model is regulated by tight junctions, endothelial cell-expressed transporters, transcytosis, leukocyte adhesion molecules, and signaling across the incorporated cell populations.

This platform can be leveraged to screen the transport of drugs targeted to the brain for neurological and psychiatric diseases and to model BBB dysfunction implicated in diseases such as multiple sclerosis, stroke and epilepsy.

ellipse
ellipse
ellipse
ellipse
ellipse

Harness predictive human data in your pipeline.

We partner with teams from the top 10 largest pharmaceutical companies to improve the pace, predictability and breadth of their drug development programs.

Explore Partnerships

Let's dive deeper.

background

Partner With Us

Let's explore how our collaborative solutions can amplify your research, drive breakthroughs, and reshape the future of medicine.

Our Platform

A scalable infrastructure & AI platform for sophisticated, large-scale experimentation on lab-grown human organ tissues.

ellipse