Clinical and Experimental Reproductive Medicine

Search

Close

Song and Jeong: State-of-the-art in high throughput organ-on-chip for biotechnology and pharmaceuticals

State-of-the-art in high throughput organ-on-chip for biotechnology and pharmaceuticals

Suk-Heung Song1, Sehoon Jeong2
Received February 14, 2024       Revised May 28, 2024       Accepted July 5, 2024
Abstract
Modern drug discovery is driven by high demand in the pharmaceutical industry to test growing libraries of compounds against potential targets. High-throughput screening (HTS) is characterized by fully automated experimentation that leverages robotic liquid handling systems, analytical techniques, and advanced computing and statistics, including the recent integration of artificial intelligence. To align with this trend, it is crucial to develop and implement new HTS platforms that offer improved predictivity and physiological relevance. In recent years, microphysiological systems, commonly known as organ-on-chip (OoC) systems, have progressed from a theoretical concept to a powerful alternative to conventional in vitro and animal models. High-throughput OoC (HT-OoC) systems could represent the disruptive technology sought by pharmaceutical companies to address their enormous research and development (R&D) expenses. In this study, we provide a brief overview of commercial products utilizing modern HT-OoC systems in drug discovery and development. Additionally, we discuss recent trends in R&D aimed at industrialization.
Introduction
Introduction
The biopharmaceutical industry utilizes its scientific and industrial expertise to advance basic science research and develop safe, effective treatments that can be made accessible to patients. This industry is uniquely equipped to shoulder the risks associated with furthering research into these treatments. The 2021 report of the Pharmaceutical Research and Manufacturers of America indicates that, on average, developing a single new medicine takes between 10 to 15 years and costs US dollar (USD) 2.6 billion, factoring in the costs of numerous failures. However, only 12% of new molecular entities that enter clinical trials ultimately receive approval from the U.S. Food and Drug Administration (FDA). Today, approximately 7,000 rare diseases are documented, yet treatments are available for only about 5% of them. The biopharmaceutical pipeline currently includes nearly 260 vaccines aimed at treating and preventing diseases, including dozens targeting coronavirus disease 2019 (COVID-19).
The substantial investments made by biopharmaceutical companies over the past several decades regarding new technologies, research, and prior vaccine development prepared the industry to respond to the COVID-19 pandemic swiftly, without compromising safety or efficacy. However, recent studies indicate that pharmaceutical companies face escalating costs to bring new drugs to market, as high as USD 2.6 billion per drug. These rising expenses have contributed to a relatively low annual average of 32 new molecular entities and biologic license application approvals by the FDA over the past two decades, with further decreases in recent years [1-4].
New drug discovery involves the screening of numerous candidate chemicals. Thousands of initial hits on high-throughput screening (HTS) are subsequently tested and refined through a series of assays, yielding a few hundred qualified leads. To manage resources and costs, alternative drug development paradigms that emphasize a “quick win, fast fail” approach have been proposed. This strategy is intended to resolve technical uncertainties early in the development process [5]. Its adoption requires the development and implementation of new assays and testing systems, such as HTS, that offer significantly improved drug predictivity and physiological relevance.
In recent years, organ-on-chip (OoC) systems have progressed from a theoretical concept to a powerful alternative to conventional in vitro and animal models [6]. These systems incorporate human tissues that exhibit physiological structure and function within a microphysiological environment that is precisely controlled and features vasculature-like perfusion [7]. Researchers have created a range of OoC systems, from single-organ platforms (such as those for vascular, heart, or lung tissue) to highly intricate multi-organ systems that integrate multiple tissues within a single circulatory flow [8-10]. Several academic studies have demonstrated the considerable potential of OoC systems as in vitro disease models that offer high physiological relevance [11].
Recently, OoC technologies have transitioned from primarily academic uses to tangible industrial applications [12]. Several start-up companies are now entering the biotechnology market with these innovations. To apply OoC technologies in early drug discovery and development, it is essential to automate the systems to increase throughput. Thus, the industrialization of OoC systems necessitates the establishment of high-throughput, physiologically relevant systems.
High throughput organ-on-chip systems
High throughput organ-on-chip systems
Cells in vivo are exposed to a complex, three-dimensional (3D) environment that includes circulating molecules, neighboring cells, and the extracellular matrix (ECM). Organ-on-chip (OoC) is a powerful technology that provides superior human relevance through microfluidic techniques. Since most platform technologies rely on one or a few chips within dedicated environments, OoCs are primarily envisioned for use in preclinical testing. In contrast, high throughput organ-on-chip (HT-OoC) platforms combine physiological relevance with the scalability of a single chip. This combination enables automation, facilitating the scaling and streamlining of research processes. Recent OoC systems combine individual devices, which contain only one or a few tissues per chip, into modules that integrate multiple chips. To apply OoC technology to HT system, a limited number of OoCs have been adapted for high throughput experimentation by using parallelization to increase the number of replicates per chip.
Currently, most commercially available OoC products are in use as a limited number of parallelized OoC setups that function as HT-OoC systems. In practice, the development of HT-OoC systems has focused on batch processing for perfusion, sampling, and cell injection; scaling up using standard well plates (384-, 96-, 64-, and 40-well); sensor integration; and online analysis. A handful of companies are at the global forefront of HT-OoC platforms. For hydrogel patterning-based HT-OoC, these include AIM Biotech, MIMETAS, and Qureator Inc. For membrane-based HT-OoC, Emulate, Dynamic42 GmbH, PREDICT96-ALI (Draper Laboratory), Bi/ond, and AlveoliX AG predominate. Leaders in non-sacrificial or sacrificial material-based HT-OoC include Nortis Bio and Dr. Jennifer Lewis’ platforms (Wyss Institute). The area of multi-chamber–based HT-OoC (compatible with transwells) is led by TissUse GmbH, CN Bio, and Kirkstall Ltd., while companies engaging in microwell-/milliwell-based HT-OoC include Curi Bio, Hesperos Inc., and MISO Chip.
1. Multi-well plate HT-OoC
1. Multi-well plate HT-OoC
Recent advances in phenotypic tissue and disease modeling are central to the development of therapies in the 21st century. The OrganoPlate technology is available in various formats to accommodate different culture setups, facilitating the development of complex 3D tissue, organ, and disease models. This technology has been demonstrated to be robust and amenable to screening assays, making it one of the most versatile platforms available [13-15]. As shown in Table 1, the OrganoPlate HT-OoC system offers a comprehensive solution for in vitro tissue culture applications [16]. The platform supports the creation of unique tissue cultures and applications through several microfluidic designs and dedicated instruments. Each plate features a single in-gel culture channel and two perfusion channels. This configuration enables the cultivation of one or multiple tissues within an ECM and up to two perfused tubules adjacent to an ECM of choice, without the need for artificial membranes. Compounds and stimuli can be directly applied to both the apical and basolateral sides of the culture. With this level of direct access, the platform enables perfusion and supports a variety of barrier integrity, transport, and migration assays.
2. OrganoPlate with organoid models
2. OrganoPlate with organoid models
In Table 2, the term “organoid” indicates ready-to-use 3D tissue models that are utilized for drug exposure, transport, and permeability studies [17]. Due to their superior physiological resemblance to tissues compared to cell lines, primary tissue, or induced pluripotent stem cell-derived cells, these tubules represent an ideal model for examining the mechanistic toxicity of both small and large molecules, as well as for assessing their absorption, metabolism, and secretion.
3. Multi-chip plate HT-OoC systems
3. Multi-chip plate HT-OoC systems
As shown in Table 3, multi-chip plates enable the recreation of complex human biology and the accurate prediction of human drug responses. They are purpose-designed to more accurately recapitulate the physiology and function of human organs and tissues in vitro [18]. The familiar plate-based format contains multiple chips to facilitate rapid adoption, increase throughput, and decrease costs [19,20].
4. Multi-array HT-OoC
4. Multi-array HT-OoC
Multi-array HT-OoC is advancing the development of next-generation human medicines by integrating human cells, systems, and data. Table 4 showcases tissue-specific multi-array plates featuring human induced pluripotent stem cells, supporting all actors within the drug discovery pipeline—from pharmaceutical companies engaged in drug discovery to the individuals depending on new therapies to be both safe and effective [21-23]. Specifically, the optical-array 96-well plate is rapidly advancing, aiming to fulfill the demand for more accurate, effective, and scalable preclinical screening tools that can predict a drug’s potential toxic effects in humans.
5. Tissue-specific design of HT-OoC
5. Tissue-specific design of HT-OoC
The development of tissue-specific microphysiological systems, commonly known as organ chips [24-30], is revolutionizing the study of biology and the advancement of drug development, therapies, and cures for those in greatest need (Table 5). These tissue-specific designs of OoC technologies provide human-relevant models for complex biological processes. Multiple validated organ chip models now exist, allowing researchers to more accurately replicate human biology. This is achieved by integrating human cell sources, an organ-specific microenvironment, and tissue-relevant mechanical forces. These models can simulate the functions of various organs, such as the liver, intestine, brain, kidney, and lung. Each model also facilitates robust data collection throughout the course of a study, including imaging, effluent analysis, biomarker analysis, etc.
Challenges addressed by HT-OoC
Challenges addressed by HT-OoC
The uniqueness and power of OoC technology lie in its capacity to support the reverse-engineering of all living human tissues using microengineered devices. OoC technologies replicate the intricate biological interactions and physiological functions of these tissues in a manner that has not been achievable using traditional cell culture techniques. This marks a major progression in our capacity to model and comprehend the complex processes of human physiological systems.
Generally, OoC devices are constructed from clear silicone rubber, with variable size and design. These devices contain microfabricated 3D chambers lined with human cells of various types. These cells are arranged and cultivated to form structures sufficiently complex to emulate the essential functions of a living organ. OoC technologies are increasingly being applied across different fields. Most chip models that incorporate two or more types of tissue or organs can effectively provide a supply of patient-specific cells, blood, or diseased cells. This creates an environment conducive to clinical research based on HT-OoC or high-throughput organ-on-a-plate methodologies. Additionally, these technologies are being developed as new drug development platforms and fostering research on reproductive OoC models relevant to both human and animal reproductive medicine.
The primary fields of industrial HT-OoC systems are attempting to address the following challenges, as outlined in Table 6.
1. Cancer research
1. Cancer research
Although animal models can effectively model certain behaviors of human cancers, they fall short in allowing researchers to examine the effects of specific tissue components and investigate the role of the tumor microenvironment [31]. Consequently, we continue to face a shortage of effective and safe treatments for many types of cancer, due in part to the lack of models that accurately reflect cancer progression in humans. OoC technologies enable researchers to replicate the human tumor microenvironment in vitro, facilitating mechanistic studies of tumor cell behavior as well as drug efficacy and safety. Co-culturing endothelial cells within OoC systems produces key cell-cell interactions, while the application of media flow and tissue-relevant stretching simulates the mechanical forces encountered by cancer cells within the body. These highly tunable OoC systems allow for the precise modulation of various cellular, molecular, chemical, and biophysical properties to explore their effects on cancer progression and behavior. For instance, cancer progression is a complex process that remains incompletely understood, partially due to the limitations of animal and in vitro models in replicating the human tumor microenvironment. OoC technology can be employed to model the early stages of cancer metastasis, including the intravasation of tumor cells into the vasculature. By accurately modeling and adjusting the tumor microenvironment, researchers can gain a deeper understanding of, and ultimately influence, various facets of cancer progression.
2. Gene therapy research
2. Gene therapy research
Developing safe and effective viral vectors for gene therapy is both time-consuming and challenging. Gene therapy holds promise for treating both inherited and acquired diseases; however, predicting responses is difficult due to the complex mechanisms involved and the scarcity of clinical data or predictive non-clinical models. A key challenge in gene therapy is the development of gene delivery vehicles. Conventional in vitro models are limited in complexity, often producing non-physiological responses. OoC technology that models the intravenous infusion of gene therapy vectors can be utilized to evaluate the transport of these vectors from the vasculature into the target tissue [32,33]. The application of adeno-associated virus (AAV) transduction for human tissue-based OoC systems allows for rapid testing and iteration of AAV designs within a human-relevant model of the liver sinusoid, thereby accelerating vector optimization.
3. Immunology research
3. Immunology research
Human inflammation mechanisms remain poorly understood, largely due to the challenge of modeling complex immune responses in vitro and the fundamental differences between animal and human immune systems [34]. More human-relevant models of inflammation and the immune response are required to advance our understanding and facilitate the development of treatments for inflammatory diseases. OoC technology offers a promising avenue for constructing more human-relevant models of inflammation. This technology enables the incorporation of the cellular diversity observed in vivo into a tissue-specific microenvironment that more closely emulates the complex cell-cell interactions involved in inflammation. Additionally, it enables the control and examination of the individual contributions of various factors to the inflammatory process. These factors include resident and circulating immune cells, inflammatory cytokines, cell-cell interactions, and organ-relevant mechanical forces. In particular, the complex mechanisms of the human immune response in inflammatory bowel disease require further elucidation. This disease is characterized by dysregulation of colon immune cell recruitment, cytokine-mediated disruption of the colonic barrier, and neuroinflammation, the last of which involves both the neuronal compartment and brain microvascular endothelial-like cells.
4. Infectious disease research
4. Infectious disease research
The COVID-19 pandemic has demonstrated that infectious diseases can have profound global implications due to their rapid spread and unpredictable nature [35]. OoC technology, with its intricate 3D architecture and the mechanical forces induced by flow and stretch, offers a more physiologically relevant model for studying infectious diseases. This technology enables the examination of host-pathogen interactions, the investigation of bacterial and viral infection progression, and the evaluation of treatment efficacy for these infections.
5. Neuroscience research
5. Neuroscience research
The human brain and nervous system are notoriously complex [36,37]. To accelerate the development of safe and increasingly effective treatments for neurodegenerative diseases, a more physiologically relevant model of the human brain is needed. OoC technology can provide a more representative model of the human brain’s neurovascular unit, enabling a more accurate understanding of neurodegenerative disease mechanisms. This includes elucidating the inner workings of the blood-brain barrier and increasing the likelihood that preclinical drug studies will translate to human responses. Although animal models have provided many insights into human neurophysiology, differences between species have often limited the successful translation of neurodegenerative therapies to clinical settings. Conventional in vitro models typically lack the necessary cellular complexity, leading to discrepancies in permeability and transporter expression that hinder clinical translation. Even advanced in vitro models, such as brain organoids, usually do not include microglia, media flow, or vasculature, which are critical for studying a disease’s impact on the blood-brain barrier. Recent brain OoC studies have made strides by incorporating microglia, which are essential for modeling neuroinflammation. These studies also include both the neuronal compartment and brain microvascular endothelial-like cells, featuring models such as blood-brain barrier-penetrating OoC and neuroinflammation OoC.
6. Toxicology assessment research
6. Toxicology assessment research
Biopharmaceutical companies face numerous challenges when developing human drugs, with the assessment of potential toxicity during the clinical stage being particularly daunting [38,39]. Despite passing preclinical safety evaluations in animal models, approximately half of all drugs fail during human trials due to toxicity. Traditional models lack the predictive value necessary to confidently advance drug candidates to clinical testing. The use of HT-OoC systems for earlier prediction of human responses can lead to safer drug candidates and diminish the reliance on animal testing, in contexts such as assessing hepatotoxicity for drug-induced liver injury.
7. Microbiome research
7. Microbiome research
Understanding the biology and function of the gut microbiome is crucial [40,41]. The human gut microbiome plays a pivotal role in immune system regulation, inflammatory bowel disease, and cancer, while impacting drug absorption and metabolism. OoC systems enable the culture of human intestinal cells in direct contact with human gut microbiota. These systems could serve as a platform for clarifying the role of the microbiome in both healthy and disease physiology, and they may accelerate the development of microbiome-related therapies, probiotics, and nutraceuticals. The colon intestine-chip allows for the co-culture of a complex human microbiome with primary intestinal epithelium over several days. This technology provides a means to investigate the microbiome’s role in maintaining barrier function, explore the effects of shear forces and peristaltic-like cyclic stretch, and study the impact of drugs on the intestinal microenvironment.
8. Research in human and animal reproductive medicine
8. Research in human and animal reproductive medicine
Over the past decade, numerous studies have demonstrated the feasibility of developing OoC models of intractable reproductive diseases. These models improve our understanding of disease pathophysiology and support the advancement of clinical and pharmaceutical interventions. Specifically, OoC technology has been instrumental in studying the female reproductive system [42].
A newer OoC model for reproductive medicine has recently been developed, one that replicates the interface between maternal tissue and placental cells during the critical moments of early pregnancy when the embryo implants in the uterus [43,44]. Reproductive OoC technology is still in its infancy. A defining characteristic of the reproductive system is its highly complex and dynamically regulated secretome, which is crucial for maintaining physiological homeostasis and is implicated in various disease processes. To create realistic reproductive OoC models, it will be necessary to capture this complexity and to quantitatively measure, characterize, and validate the complex soluble environment of the engineered constructs. As such, the potential for using these models in medium- or even HTS has not yet been established. Nonetheless, the potential for advancing and applying reproductive OoC technology holds great promise.
Outlook
Outlook
Globally, North America and the European Union are expected to remain the largest-growing markets for OoC in the coming years. This growth is primarily due to the increasing number of manufacturing companies and the rising demand for HT-OoC systems. Additionally, factors such as increased research and development (R&D) spending and the growing trend of outsourcing R&D activities are likely to contribute to the expansion of the OoC market in these regions. In particular, the increasing number of clinical trials conducted in these areas is a key driver of market growth. North America leads the world in the number of clinical trials performed, with various studies currently underway. The growing adoption and preference for personalized medicine in the European region are key factors driving the European OoC market forward. Many developments in personalized medicine utilize OoC technology to achieve optimal results. OoC technology, which combines biology and engineering, allows pharmaceutical and biotechnology companies to simulate human tissue and organ functions without the use of animals. Consequently, many OoC system product companies are focusing on the implementation and scaling of OoC technologies, particularly HT-OoC systems, across various sectors of pharmaceuticals and healthcare. Advancements in 3D bioprinting and 3D microfabrication are aiding in tissue modeling and the creation of organ-like structures. These developments are crucial for the advancement of HT-OoC technologies and have a broad range of applications, including micro-robotics, drug delivery, and therapeutics. Such innovations are instrumental in driving market growth.
In the coming years, the OoC market is anticipated to experience global growth, driven by a surge in market activities including product launches, collaborations, mergers, and acquisitions.
Notes
Notes

Conflict of interest

There is no conflict of interest with Ginkgo Bioworks Inc.

Author contributions

Conceptualization: SHS. Data curation: SHS. Investigation: SJ. Writing-original draft: SHS. Writing-review & editing: SHS, SJ. Approval of final manuscript: SHS, SJ.

Table 1.
OrganoPlate options for high-throughput screening
Type Feature Image
Two‑lane 96 Each chip contains one in-gel culture channel and one perfusion channel, enabling the culture of perfused tubules adjacent to a chosen ECM without the need for artificial membranes. The design provides direct access to the apical tubule lumen, facilitating the perfusion and addition of cells, compounds, and stimuli. Key features include: cerm-2024-06954i1.tif
 ·96 Independent tissue culture chips
 ·Two adjacent channels per chip
 ·Direct access to the apical lumen of tubular cultures
Three‑lane 40 Each chip includes one in-gel culture channel and two perfusion channels. This design enables the cultivation of one or multiple cultures within an ECM and up to two perfused tubules adjacent to an ECM of choice, without the need for artificial membranes. Compounds and stimuli can be directly applied to both the apical and basolateral sides of the culture. Key features include: cerm-2024-06954i2.tif
 ·40 Independent tissue culture chips
 ·Three adjacent channels per chip
 ·Direct access to the apical and basal tubule lumen
Three‑lane 64 The three-lane 64 system is designed to facilitate automation workflows. Each chip contains a single in-gel culture channel, flanked by perfusion channels on both sides. This enables the establishment of complex (co)cultures in multiple configurations. Compounds and stimuli can be directly applied to both the apical and basolateral sides of the culture. Key features include: cerm-2024-06954i3.tif
 ·64 Independent tissue culture chips designed for automated workflows
 ·Three adjacent channels per chip
 ·Direct access to the apical and basal tubule lumen
Graft Each chip comprises an in-gel culture channel, two perfusion channels, and an open grafting chamber. This platform facilitates perfusion as well as the addition of cells, compounds, and stimuli, while offering direct access to vascularized tissues. Key features include: cerm-2024-06954i4.tif
 ·64 Independent tissue culture chips
 ·Three adjacent channels, with one open grafting chamber per chip
 ·Direct access to the apical and basal tubule and vessel lumen

ECM, extracellular matrix.

Table 2.
OrganoPlate with 3D tissue
Type Feature Image
Organoid Organoid models are derived from ASCs, unlike many other organoid technologies that utilize embryonic or primary cells. This distinction is crucial because ASC-derived organoids do not necessitate the reprogramming or transformation of stem cells. Moreover, the intrinsic characteristics of the donor, including specific disease attributes, are preserved when cultured ex vivo. Consequently, the regenerative potential of patient-derived ASCs can be harnessed to replicate organ functionality in a laboratory setting. Key features include: cerm-2024-06954i5.tif
 ·64 Ready-to-use ASC-derived perfused organoid tubules
 ·Recovery medium and culture medium
 ·ECM: optimized collagen I
Collagen The collagen plate comes pre-seeded with a validated and optimized batch of rat-tail collagen in the central channel. It is designed to promote optimal tubule formation and maintain barrier integrity with most epithelial and endothelial cells. Built on the OrganoPlate three-lane platform with either 40 or 64 chips, this ready-to-use solution adds speed and robustness to a workflow, eliminating the need for ECM handling. Additionally, it can be stored at room temperature for up to 3 months. Endothelial or epithelial cells can be seeded on demand to create either one or two leak-tight perfused tubules. cerm-2024-06954i6.tif
 ·Includes 40 or 64 ready-to-use chips pre-seeded with collagen I
This option broadens screening capabilities and enables the development of more robust 3D in vitro cell models with the speed and flexibility of 2D models.
Colon Caco‑2 Each tissue culture chip contains one in-gel culture channel and two perfusion channels, one of which is a perfused Caco-2 tubule. Compounds and stimuli can be directly applied to both the apical and basolateral sides of the culture. With this direct access, the platform enables perfusion and supports various barrier functions and transport assays. Key features include: cerm-2024-06954i7.tif
 ·An OrganoPlate three-lane 40 or three-lane 64 setup, including 40 or 64 chips with ready-to-use Caco-2 tubules
 ·Direct access to the apical and basal tubule lumen
 ·Caco-2 cell line: human Caucasian colon adenocarcinoma, wild-type
 ·ECM: collagen I​
BBB HBMEC This platform utilizes primary HBMEC cells that express all relevant markers, transporters, receptors, and enzymes. Constructed on the OrganoPlate system with a three-lane 40 or 64 configuration, this platform enables the immediate de-risking of compound toxicity. Barrier integrity can be assessed by measuring fluorescent dye leakage or by employing a faster and more sensitive method using the OrganoPlate shaker. Key features include: cerm-2024-06954i8.tif
 ·40 or 64 ready-to-use HBMEC perfused tubules
 ·Organ medium: HBMEC-BM
 ·HBMEC cell line: human brain microvascular endothelial cells
 ·ECM: collagen I​
With this option, researchers can assess the permeability and active transport of ​compounds with direct apical and basal access.​ They can also incorporate a neuronal component, obtaining a complete​ neurovascular unit, using an optimized and consistent batch of human primary BBB cells cultured alongside a validated batch of collagen I.
Blood vessel HUVEC Each tissue culture chip contains one in-gel culture channel and two perfusion channels, one of which is a perfused HUVEC tubule. Compounds and stimuli can be directly applied to both the apical and basolateral sides of the culture. With this direct access, the platform enables perfusion and supports various barrier functions and transport assays. Key features include: cerm-2024-06954i9.tif
 ·An OrganoPlate three-lane 40 or three-lane 64 setup, including 40 or 64 chips with ready-to-use HUVEC tubules
 ·Direct access to the apical and basal tubule lumen
 ·HUVEC cell line: human umbilical vein endothelial cells
 ·ECM: collagen I
Angiogenesis HUVEC Each tissue culture chip contains one in-gel culture channel and two perfusion channels, one of which is a perfused HUVEC tubule. Compounds and stimuli can be directly applied to both the apical and basolateral sides of the culture. With this direct access, the platform enables the investigation of microvascular damage, tumor angiogenesis, fibrosis, and sprout formation. Key features include: cerm-2024-06954i10.tif
 ·An OrganoPlate three-lane 64 setup, including:
  - 64 HUVEC tubules ready to sprout
  - Sprout initiation mix: organ medium HUVEC-AN-I
 ·Perfused 3D microvasculature
 ·Tip-stalk cell hierarchy and anastomosis
 ·HUVEC cell line: human umbilical vein endothelial cells
 ·ECM: collagen I
Vascular bed HUVEC Constructed on the OrganoPlate platform, this model features 64 chips in parallel, allowing for the immediate integration of the tissue of interest. Conditions may be optimized for tissue vascularization. This ready-to-use vascular bed not only improves the model relevance but also facilitates access to the core of the tissue explants. Key features include: cerm-2024-06954i11.tif
 ·64 Pre-formed HUVEC vascular beds
 ·Culture medium
 ·Sprouting initiation mix: organ medium HUVEC VB-I
 ·Prepared and fully quality-controlled​
 ·Ready for use after overnight recovery

3D, three-dimensional; ASC, adult stem cell; ECM, extracellular matrix; 2D, two-dimensional; BBB, blood-brain barrier; HBMEC, human brain microvascular endothelial cell; HUVEC, human umbilical vein endothelial cell.

Table 3.
Multi-chip plates for high-throughput screening
Type Features Image
Barrier plate Epithelial and endothelial cells may be cultured at an air-liquid interface or in the presence of media. Circular flow perfusion on the basolateral side recreates a physiologically relevant microenvironment within each of the plate’s 12 chips. Uses include: cerm-2024-06954i12.tif
 ·Primary cells
 ·Induced pluripotent stem cells
 ·Immortalized cell lines
 ·Circulating immune cells
 ·Organoids
Dual-organ plate Six independently grown biological barrier models (e.g., lung- or gut-on-a-chip) are generated by culturing primary human cells on the apical and basolateral sides of a well-plate insert. Uses include: cerm-2024-06954i13.tif
 ·Primary cells
 ·Induced pluripotent stem cells
 ·Immortalized cell lines
 ·Circulating immune cells
 ·Precision-cut tissue slices
 ·Organoids
Liver plate Three-dimensional scaffolds are embedded within multi-chip liver-12 and -48 plates, which are continuously perfused with cell culture medium during experiments. cerm-2024-06954i14.tif
Perfusion provides a continuous supply of oxygen and nutrients for long-term liver-on-a-chip culture, maintaining cell phenotypes and functions for at least 4 weeks.
 ·The design aims to provide optimal conditions for primary human hepatocytes and non-parenchymal cells, facilitating deep mechanistic insights into drug or disease mechanisms.
Table 4.
Tissue-specific multi-array plate with human induced pluripotent stem cells
Type Feature Image
Multi-array plate A platform for human-relevant 3D-engineered muscle tissue analysis cerm-2024-06954i15.tif
By facilitating parallel analysis of 3D-engineered muscle tissues with adult-like functional profiles, the multi-array plate enables the discovery, safety, and efficacy testing of new therapeutics. Applications include:
 ·Force frequency
 ·Rate of contraction/systole
 ·Rate of relaxation/diastole
 ·Time to max twitch (peak)
 ·Maximal twitch amplitude
 ·Full width at half-max
Nanoarray plate Nanoarray topography offers cells and tissues a biomimetic surface, improving the physiological relevance of experiments by promoting improved structural and phenotypic development compared to cells cultured on conventional dishes. cerm-2024-06954i16.tif
Nanoarray plates are available in standard microplate formats featuring No. 1.5 glass-bottom wells to facilitate high-quality imaging (384-well and 96-well).
Optical-array 96-well plate The optical-array 96-well plate enables high-throughput analysis of electrophysiology and calcium transients within a standard SBS titer-plate format (85.48 mm×127.76 mm) compliant plate. cerm-2024-06954i17.tif
The information-rich output supports multiple applications:
 ·Calcium flux
 ·Cardiomyocyte screening
 ·Cardiotoxicity
 ·Potassium assays
 ·Ion channel

3D, three-dimensional; SBD, society for biomolecular screening.

Table 5.
Tissue-specific organ chips
Type Feature Image
Multi-organ chips cerm-2024-06954i18.tif cerm-2024-06954i19.tif
Colon intestine‑chip · A comprehensive colonic barrier model cerm-2024-06954i20.tif
Mechanical forces applied to the colon intestine-chip create an environment that closely resembles in vivo conditions. Under dynamic conditions, cells differentiate into characteristic populations and structures, establishing the intestinal barrier and forming microvilli. This contrasts with conventional cell culture, which features limited and largely undifferentiated cell populations, as well as an absence of physical stimuli.
Duodenum intestine‑chip · An improved model of the human duodenum cerm-2024-06954i21.tif
The duodenum intestine-chip integrates primary human duodenal organoids and small intestine microvascular endothelial cells with mechanical forces that mimic intestinal peristalsis. Within this dynamic microenvironment, cells achieve a well-polarized state and exhibit in vivo-like morphology, functionality, and gene expression. Additionally, this system permits access to the apical surface.
Kidney-chip · A physiologically relevant kidney model cerm-2024-06954i22.tif
Inside the kidney-chip, cells exhibit an in vivo-like phenotype characterized by high differentiation, normal epithelial cell polarity, and morphology, as well as functional transporter activity. This facilitates a physiological analysis of both healthy kidney function and the nephrotoxic effects of drug candidates. Long-term culture permits multiple measurements over time for mechanistic studies, biomarker discovery, and the investigation of nutrient metabolism.
Liver‑chip · A validated, comprehensive preclinical model of the human liver cerm-2024-06954i23.tif
The human liver-chip faithfully replicates the in vivo physiological functions of the human liver by incorporating essential microenvironmental features, including three-dimensional multicellular architecture and vascular flow. This offers a model of the liver that is more relevant to human biology compared to traditional sandwich cultures, spheroids, and animal models, each of which lacks relevant features necessary to appropriately model the human liver.
Lung‑chip · A human-based co-culture model incorporating relevant physical forces cerm-2024-06954i24.tif
The lung-chip, unlike commonly used cell line models, integrates a co-culture of primary human lung epithelial and endothelial cells within a dynamic microenvironment. Cell-cell interactions, flow, and (when relevant) stretch improve functionality, resulting in in vivo-like cell differentiation, cilia behavior, and mucociliary clearance along with a tight epithelial barrier.
Table 6.
Major challenges addressed by HT-OoC systems
Challenge OoC application HT-OoC product
Cancer research Complex tissues in a relevant in vitro microenvironment MIMETAS, Emulate
Angiogenesis for microvascular damage, tumor, and fibrosis
Gene therapy research Cell-cell interactions for cell signaling and migration MIMETAS
Microbiome research Gut-on-chip MIMETAS, Emulate
Perfused tubular tissues against an extracellular matrix
Immunology research Liver-on-chip CN Bio, Curi Bio
Lung-on-chip Emulate
Skeletal muscle models
Cardiac models
Infectious disease research Gut/Liver-on-chip MIMETAS, CN Bio
Lung/Liver-on-chip
Neuroscience research Brain-on-chip Emulate
Toxicology assessment research Vascularization MIMETAS, Emulate
3D cell migration
Research in human and animal reproductive medicine Microphysiological model Vivodyne
Placenta-on-a-chip

HT-OoC, high-throughput organ-on-chip; OoC, organ-on-chip; 3D, three-dimensional.

References
References

1. Mullard A. New drugs cost US$2.6 billion to develop. Nat Rev Drug Discov 2014;13:877.
[Article]
2. DiMasi JA, Hansen RW, Grabowski HG. The price of innovation: new estimates of drug development costs. J Health Econ 2003;22:151-85.
[Article] [PubMed]
3. Mullard A. 2016 FDA drug approvals. Nat Rev Drug Discov 2017;16:73-6.
[Article] [PubMed]
4. Paul SM, Mytelka DS, Dunwiddie CT, Persinger CC, Munos BH, Lindborg SR, et al. How to improve R&D productivity: the pharmaceutical industry’s grand challenge. Nat Rev Drug Discov 2010;9:203-14.
[Article] [PubMed]
5. Reardon S. ‘Organs-on-chips’ go mainstream. Nature 2015;523:266.
[Article] [PubMed]
6. Bhatia SN, Ingber DE. Microfluidic organs-on-chips. Nat Biotechnol 2014;32:760-72.
[Article] [PubMed]
7. Sung JH, Shuler ML. A micro cell culture analog (microCCA) with 3-D hydrogel culture of multiple cell lines to assess metabolism-dependent cytotoxicity of anti-cancer drugs. Lab Chip 2009;9:1385-94.
[Article] [PubMed]
8. Mathur A, Ma Z, Loskill P, Jeeawoody S, Healy KE. In vitro cardiac tissue models: current status and future prospects. Adv Drug Deliv Rev 2016;96:203-13.
[Article] [PubMed] [PMC]
9. Huh D, Matthews BD, Mammoto A, Montoya-Zavala M, Hsin HY, Ingber DE. Reconstituting organ-level lung functions on a chip. Science 2010;328:1662-8.
[Article] [PubMed] [PMC]
10. Marx U, Andersson TB, Bahinski A, Beilmann M, Beken S, Cassee FR, et al. Biology-inspired microphysiological system approaches to solve the prediction dilemma of substance testing. ALTEX 2016;33:272-321.
[Article] [PubMed] [PMC]
11. Zhang B, Radisic M. Organ-on-a-chip devices advance to market. Lab Chip 2017;17:2395-420.
[Article] [PubMed]
12. Beilmann M, Boonen H, Czich A, Dear G, Hewitt P, Mow T, et al. Optimizing drug discovery by investigative toxicology: current and future trends. ALTEX 2019;36:289-313.
[Article] [PubMed]
13. Wevers NR, van Vught R, Wilschut KJ, Nicolas A, Chiang C, Lanz HL, et al. High-throughput compound evaluation on 3D networks of neurons and glia in a microfluidic platform. Sci Rep 2016;6:38856.
[Article] [PubMed] [PMC]
14. Gumuscu B, Albers HJ, van den Berg A, Eijkel JC, van der Meer AD. Compartmentalized 3D tissue culture arrays under controlled microfluidic delivery. Sci Rep 2017;7:3381.
[Article] [PubMed] [PMC]
15. Trietsch SJ, Naumovska E, Kurek D, Setyawati MC, Vormann MK, Wilschut KJ, et al. Membrane-free culture and real-time barrier integrity assessment of perfused intestinal epithelium tubes. Nat Commun 2017;8:262.
[Article] [PubMed] [PMC]
16. MIMETAS. OrganoPlate® [Internet]. MIMETAS; 2024 [cited 2024 Jul 30]. Available from: https://www.mimetas.com/en/organoplate-sup-sup-product-overview

17. MIMETAS. OrganoReady® [Internet]. MIMETAS; 2024 [cited 2024 Jul 30]. Available from: https://www.mimetas.com/en/organoready/

18. CN-BIO. PhysioMimix® multi-chip plates [Internet]. CN Bio Innovations Ltd.; 2024 [cited 2024 Jul 30]. Available from: https://cn-bio.com/consumables/multi-chip-plates

19. Domansky K, Inman W, Serdy J, Dash A, Lim MH, Griffith LG. Perfused multiwell plate for 3D liver tissue engineering. Lab Chip 2010;10:51-8.
[Article] [PubMed] [PMC]
20. Inman W, Domansky K, Serdy J, Owens B, Trumper D, Griffith LG. Design, modeling and fabrication of a constant flow pneumatic micropump. J Micromech Microeng 2007;17:891.
[Article]
21. Curibio. Mantarray™ platform for human-relevant 3D engineered muscle tissue analysis [Internet]. Curibio; 2024 [cited 2024 Jul 30]. Available from: https://www.curibio.com/biosystem-platforms-overview

22. Phan DT, Wang X, Craver BM, Sobrino A, Zhao D, Chen JC, et al. A vascularized and perfused organ-on-a-chip platform for large-scale drug screening applications. Lab Chip 2017;17:511-20.
[Article] [PubMed] [PMC]
23. Wang X, Phan DT, Zhao D, George SC, Hughes CC, Lee AP. An on-chip microfluidic pressure regulator that facilitates reproducible loading of cells and hydrogels into microphysiological system platforms. Lab Chip 2016;16:868-76.
[Article] [PubMed] [PMC]
24. Emulate. Organ-chip models: human-relevant models for complex biology [Internet]. Emulate; 2024 [cited 2024 Jul 30]. Available from: https://emulatebio.com/organ-chips

25. Varone A, Nguyen JK, Leng L, Barrile R, Sliz J, Lucchesi C, et al. A novel organ-chip system emulates three-dimensional architecture of the human epithelia and the mechanical forces acting on it. Biomaterials 2021;275:120957.
[Article] [PubMed]
26. Apostolou A, Panchakshari RA, Banerjee A, Manatakis DV, Paraskevopoulou MD, Luc R, et al. A novel microphysiological colon platform to decipher mechanisms driving human intestinal permeability. Cell Mol Gastroenterol Hepatol 2021;12:1719-41.
[Article] [PubMed] [PMC]
27. Nawroth JC, Lucchesi C, Cheng D, Shukla A, Ngyuen J, Shroff T, et al. A microengineered airway lung chip models key features of viral-induced exacerbation of asthma. Am J Respir Cell Mol Biol 2020;63:591-600.
[Article] [PubMed]
28. Kasendra M, Luc R, Yin J, Manatakis DV, Kulkarni G, Lucchesi C, et al. Duodenum intestine-chip for preclinical drug assessment in a human relevant model. Elife 2020;9:e50135.
[Article] [PubMed] [PMC]
29. Jang KJ, Otieno MA, Ronxhi J, Lim HK, Ewart L, Kodella KR, et al. Reproducing human and cross-species drug toxicities using a liver-chip. Sci Transl Med 2019;11:eaax5516.
[PubMed]
30. Levner D, Ewart L. Integrating liver-chip data into pharmaceutical decision-making processes. Expert Opin Drug Discov 2023;18:1313-20.
[Article] [PubMed]
31. Dourson M, Ewart L, Fitzpatrick SC, Barros SB, Mahadevan B, Hayes AW. The future of uncertainty factors with in vitro studies using human cells. Toxicol Sci 2022;186:12-7.
[Article] [PubMed] [PMC]
32. Manatakis DV, VanDevender A, Manolakos ES. An information-theoretic approach for measuring the distance of organ tissue samples using their transcriptomic signatures. Bioinformatics 2021;36:5194-204.
[Article] [PubMed] [PMC]
33. Sheyn D, Cohn-Yakubovich D, Ben-David S, De Mel S, Chan V, Hinojosa C, et al. Bone-chip system to monitor osteogenic differentiation using optical imaging. Microfluid Nanofluidics 2019;23:99.
[Article] [PubMed] [PMC]
34. Kerns SJ, Belgur C, Petropolis D, Kanellias M, Barrile R, Sam J, et al. Human immunocompetent organ-on-chip platforms allow safety profiling of tumor-targeted T-cell bispecific antibodies. Elife 2021;10:e67106.
[Article] [PubMed] [PMC]
35. Si L, Bai H, Rodas M, Cao W, Oh CY, Jiang A, et al. A human-airway-on-a-chip for the rapid identification of candidate antiviral therapeutics and prophylactics. Nat Biomed Eng 2021;5:815-29.
[Article] [PubMed] [PMC]
36. Pediaditakis I, Kodella KR, Manatakis DV, Le CY, Barthakur S, Sorets A, et al. A microengineered brain-chip to model neuroinflammation in humans. iScience 2022;25:104813.
[Article] [PubMed] [PMC]
37. Pediaditakis I, Kodella KR, Manatakis DV, Le CY, Hinojosa CD, Tien-Street W, et al. Modeling alpha-synuclein pathology in a human brain-chip to assess blood-brain barrier disruption. Nat Commun 2021;12:5907.
[Article] [PubMed] [PMC]
38. Ewart L, Apostolou A, Briggs SA, Carman CV, Chaff JT, Heng AR, et al. Performance assessment and economic analysis of a human liver-chip for predictive toxicology. Commun Med (Lond) 2022;2:154.
[Article] [PubMed] [PMC]
39. Ewart L, Apostolou A, Briggs SA, Carman CV, Chaff JT, Heng AR, et al. Qualifying a human liver-chip for predictive toxicology: performance assessment and economic implications. Biorxiv [Preprint] 2022 Aug 2. https://doi.org/10.1101/2021.12.14.472674
[Article]
40. Grassart A, Malarde V, Gobaa S, Sartori-Rupp A, Kerns J, Karalis K, et al. Bioengineered human organ-on-chip reveals intestinal microenvironment and mechanical forces impacting shigella infection. Cell Host Microbe 2019;26:435-44.
[Article] [PubMed]
41. Suligoj T, Vigsnæs LK, Abbeele PV, Apostolou A, Karalis K, Savva GM, et al. Effects of human milk oligosaccharides on the adult gut microbiota and barrier function. Nutrients 2020;12:2808.
[Article] [PubMed] [PMC]
42. Young RE, Huh DD. Organ-on-a-chip technology for the study of the female reproductive system. Adv Drug Deliv Rev 2021;173:461-78.
[Article] [PubMed] [PMC]
43. Park JY, Mani S, Clair G, Olson HM, Paurus VL, Ansong CK, et al. A microphysiological model of human trophoblast invasion during implantation. Nat Commun 2022;13:1252.
[Article] [PubMed] [PMC]
44. Vivodyne. Human organ models: lifelike, functional ‍human organ tissues [Internet]. Vivodyne; 2024 [cited 2024 Jul 30]. Available from: https://www.vivodyne.com/human-organ-models

Go to Top