Zephyrnet Logo

Organs-on-chips: a decade of innovation

Date:

Main text

Cell culture and animal model systems are essential for preclinical drug testing. However, the lack of complexity in conventional cell cultures, together with the time-consuming nature of animal studies, limit their predictability of clinical response and exacerbate the high cost of drug discovery and development. Microphysiological systems (MPS), which include OOC technology [

1.

  • Leung C.M.
  • et al.
A guide to the organ-on-a-chip.

], are defined as complex, multicellular (and now multi-organ), in vitro culture systems that mimic the physiological properties of a specific organ or tissue that govern function [

2.

  • Wang Y.I.
  • et al.
Multiorgan microphysiological systems for drug development: strategies, advances, and challenges.

]. Several pivotal MPS studies have expanded our understanding of biological systems with respect to disease treatment and prevention, and give promise to OOC technology that bridges the gap between in vitro experiments and animal studies [

3.

  • Sin A.
  • et al.
The design and fabrication of three-chamber microscale cell culture analog devices with integrated dissolved oxygen sensors.

,

4.

  • Low L.A.
  • et al.
Organs-on-chips: into the next decade.

]. A landmark 2010 study by Huh and colleagues described an OOC model of the alveolar–capillary interface of the human lung that demonstrated lung physiology in vitro [

5.

  • Huh D.
  • et al.
Reconstituting organ-level lung functions on a chip.

]. More than a decade following this foundational work, a study by Ronaldson-Bourchard and colleagues demonstrated how multiple organs can be integrated to study disease progression and drug toxicities systemically [

6.

  • Ronaldson-Bouchard K.
  • et al.
A multi-organ chip with matured tissue niches linked by vascular flow.

], highlighting the exciting potential of OOC models in drug development. We describe here advances in OOC technology over the past decade by spotlighting the work of two key opinion leaders, Donald Ingber [

5.

  • Huh D.
  • et al.
Reconstituting organ-level lung functions on a chip.

] and Gordana Vunjak-Novakovic [

6.

  • Ronaldson-Bouchard K.
  • et al.
A multi-organ chip with matured tissue niches linked by vascular flow.

], in a ‘then and now’ synopsis. These studies add layers of complexity to OOC technology in faithfully recapitulating structure and function both at the organ level, by incorporating biophysics/motion, and at the systems level by interconnecting multiple organs through recirculating vascular flow (Figure 1).

Figure 1

Figure 1Organ-on-a-chip (OOC) innovation over the past 10 years.

Show full caption

Advances in OOC technology have spanned recapitulating single organ function and disease states to integrating multiple organ chips together to model systemic interactions and perform drug studies. The green box summarizes general OOC features, and the blue boxes highlight possible applications for single and interconnected organ chips. Abbreviations: PD, pharmacodynamics; PK, pharmacokinetics. Figure created with BioRender (BioRender.com).

Huh and colleagues described an OOC model that reproduces the functional alveolar–capillary interface of the human lung. The lung OOC is composed of two microfluidic chambers separated by a porous membrane. Human cells were seeded in extracellular matrix (ECM)-coated chambers, with alveolar epithelial cells in the top channel and pulmonary microvascular endothelial cells in the bottom channel. The novelty of this OOC model is the ability to replicate dynamic mechanical breathing by providing vacuum pressure alongside the cell culture chambers. The authors characterized the lung OOC model by creating an air–liquid interface that resulted in surfactant production as seen in vivo. Epithelial and endothelial junctional proteins, transepithelial/endothelial electrical resistance (TEER) measurements, and protein transport were measured to assess barrier integrity. Furthermore, the alveolar–capillary unit replicated organ-level responses such as pulmonary inflammation, as measured by cytokine production, endothelial activation, and leukocyte infiltration. Live bacteria were incorporated into the epithelial channel to recapitulate a lung infection. Interestingly, the breathing motions increased the inflammatory responses to nanoparticles in the alveolar layer, as measured by reactive oxygen species (ROS) production and endothelial activation. In addition, breathing motions resulted in a fourfold increase in nanoparticle uptake and transport to the endothelial compartment. This study was one of the first to integrate mechanical forces into such a complex, multi-cell-type ex vivo experimental model system, and although the impact of physical motions on organ functionality is significant, the underlying biology is still not well understood.

An explosion of OOC designs across academic labs and spin-off companies [

7.

  • Zhang B.
  • Radisic M.
Organ-on-a-chip devices advance to market.

] occurred during the 2010s, prompting efforts to translate a proof-of-principle idea into a drug development reality. OOC technologies matured from recapitulating single organs to linking multiple organ tissues [

8.

  • Park D.
  • et al.
Integrating organs-on-chips: multiplexing, scaling, vascularization, and innervation.

] to recreate a human ‘body-on-a-chip’, which was accelerated by Defense Advanced Research Projects Agency (DARPA) and National Institutes of Health (NIH) funding initiatives awarded to several pioneers in the field (including Ingber). In 2022, researchers led by Vunjak-Novakovic published a multi-tissue chip comprising human heart, liver, bone, and skin organs connected by vascular flow [

6.

  • Ronaldson-Bouchard K.
  • et al.
A multi-organ chip with matured tissue niches linked by vascular flow.

]. Each tissue niche maintained its unique environment, with supporting stromal cells, a selectively permeable endothelial barrier, and vascular flow containing circulating immune cells, cytokines, and extracellular vesicles. Ronaldson-Bouchard and coworkers showed that their multi-tissue chip engineered from human iPSCs supported crosstalk between organs by detecting heart tissue-derived exosomes in other organs and CD14+ monocytes that infiltrated the injured tissue. Importantly, the authors showed that selective infiltration of CD14+ cells to the injured organ only occurred when the endothelial barrier was intact. Furthermore, proteomic analyses revealed that the multi-tissue chip with a functional endothelial barrier most closely matched human tissue. The authors then performed pharmacokinetic/pharmacodynamic (PK/PD) studies of doxorubicin treatment and analyzed toxicity. They reported that miR-1 (a known biomarker for predicting doxorubicin-induced cardiotoxicity) was significantly upregulated in the multi-tissue chip. In addition, the expression of early miRNA biomarkers of doxorubicin cardiomyopathy in the multi-tissue chip most closely matched the patient data, as compared to each tissue individually or the multi-tissue chip without the endothelial barrier, highlighting the potential of these systems for improved testing of therapies and identification of early biomarkers. It is important to note that the significance of the endothelial barrier in recapitulating physiological tissue responses was not fully realized in 2010. Biological and engineering advances over a decade have resulted in an appreciation for designing systems today that maintain media and culture environments for proper tissue maturation to reproduce in vivo functional responses.

Back in 2010, Huh and colleagues discussed the possibility that biomimetic microsystems could replace animal testing, which was an ambitious prospect at the time. Although there has been significant progress toward this goal, considerable work remains for the OOC field to transform the drug discovery pipeline, including improvements in areas such as cell sourcing, platform fabrication, integration across organs, orthogonal assay development, and cross-comparison of animal, tissue chip, and human data. These factors need to be validated and standardized, together with clear establishment of the context of use, in partnership with regulatory bodies [

9.

  • Hargrove-Grimes P.
  • et al.
Microphysiological systems: stakeholder challenges to adoption in drug development.

]. In December 2022 a major step toward dramatically reducing animal testing occurred with the passing of the FDA Modernization Act 2.0. This legislation eliminates the federal mandate of animal testing for any new FDA-approved drugs, which has been in effect since 1938, and comes with financial support for an FDA-wide New Alternative Methods Program. Several organizations (e.g., the US Health and Environmental Sciences Institute and the UK National Centre for Replacement, Reduction, and Refinement) have been working with the FDA to expand the use of MPS for drug testing. In addition, companies such as Emulate Inc., which started out of the group of Don Ingber shortly after the 2010 article, raised considerable venture capital funding to commercialize OOC technology and demonstrate translational relevance [

10.

  • Ewart L.
  • et al.
Performance assessment and economic analysis of a human liver-chip for predictive toxicology.

]. We foresee that the next decade of OOCs will break the mold of traditional drug testing and improve assessments of drug efficacy and safety.

spot_img

Latest Intelligence

spot_img