Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Integrating organoids and organ-on-a-chip devices

Abstract

Organoids and organs-on-chips are two rapidly emerging 3D cell culture techniques that aim to bridge the gap between in vitro 2D cultures and animal models to enable clinically relevant drug discovery and model human diseases. Despite their similar goals, they use different approaches and exhibit varying requirements for implementation. Integrative approaches promise to provide improved cellular fidelity in the format of a device that can control the geometry of the organoid and provide flow, mechanical and electrical stimuli. In this Review, we discuss recent integrative approaches in the areas of intestine, kidney, lung, liver, pancreas, brain, retina, heart and tumour. We start by defining the two fields and describe how they emerged from the fields of tissue engineering, regenerative medicine and stem cells. We compare the scales at which the two methods operate and briefly describe their achievements, followed by studies integrating organoids and organ-on-a-chip devices. Finally, we define implementation limitations and requirements for translation of the integrated devices, including determining the differentiation stage at which an organoid should be placed into an organ-on-a-chip device, providing perfusable vasculature within the organoid and overcoming limitations of cell line and batch-to-batch variability.

Key points

  • Organoids and organs-on-chips (OoCs) aim to improve drug testing and disease modelling, but integration examples are still scarce.

  • The benefits of integration include organ-specific cellular hierarchy and structural fidelity; microscopic features from OoCs guiding tissue morphological formation; better reproducibility and scale-up capacities; and biocompatible built-in sensors for in situ functional readouts and industrially compatible culture formats.

  • A key challenge is vascularizing organoids with tissue-specific endothelial cells and aligning different cell types in organoids with appropriate flow in scalable, integrated devices.

  • In parallel, advances in computer vision and deep learning will be needed to enhance data processing and analysis. Addressing cell line variability and establishing validation criteria for OoC–organoid integrated devices is critical for commercial and translational success.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Main approaches in organoids and OoC systems.
Fig. 2: Timeline and examples of multi-well devices for integration of organoids and OoC devices.
Fig. 3: Advances and challenges in the integration of organoids and OoC systems.

Similar content being viewed by others

References

  1. Loskill, P., Hardwick, R. N. & Roth, A. Challenging the pipeline. Stem Cell Rep. 16, 2033–2037 (2021).

    Google Scholar 

  2. Fogel, D. B. Factors associated with clinical trials that fail and opportunities for improving the likelihood of success: a review. Contemp. Clin. Trials Commun. 11, 156–164 (2018).

    Google Scholar 

  3. Hwang, T. J. et al. Failure of investigational drugs in late-stage clinical development and publication of trial results. JAMA Intern. Med. 176, 1826–1833 (2016).

    Google Scholar 

  4. Birgersdotter, A., Sandberg, R. & Ernberg, I. Gene expression perturbation in vitro-a growing case for three-dimensional (3D) culture systems. Semin. Cancer Biol. 15, 405–412 (2005).

    Google Scholar 

  5. Debnath, J. & Brugge, J. S. Modelling glandular epithelial cancers in three-dimensional cultures. Nat. Rev. Cancer 5, 675–688 (2005).

    Google Scholar 

  6. Hu, W. & Lazar, M. A. Modelling metabolic diseases and drug response using stem cells and organoids. Nat. Rev. Endocrinol. 18, 744–759 (2022).

    Google Scholar 

  7. Huch, M. & Koo, B. K. Modeling mouse and human development using organoid cultures. Development 142, 3113–3125 (2015).

    Google Scholar 

  8. Vunjak-Novakovic, G., Ronaldson-Bouchard, K. & Radisic, M. Organs-on-a-chip models for biological research. Cell 184, 4597–4611 (2021).

    Google Scholar 

  9. Pasca, S. P. et al. A nomenclature consensus for nervous system organoids and assembloids. Nature 609, 907–910 (2022).

    Google Scholar 

  10. Sato, T. et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 459, 262–265 (2009).

    Google Scholar 

  11. Aloia, L. et al. Epigenetic remodelling licences adult cholangiocytes for organoid formation and liver regeneration. Nat. Cell Biol. 21, 1321–1333 (2019).

    Google Scholar 

  12. Wilson, H. V. A new method by which sponges may be artificially reared. Science 25, 912–915 (1907).

    Google Scholar 

  13. Weiss, P. & Taylor, A. C. Reconstitution of complete organs from single-cell suspensions of chick embryos in advanced stages of differentiation. Proc. Natl Acad. Sci. USA 46, 1177–1185 (1960).

    Google Scholar 

  14. Martin, G. R. Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells. Proc. Natl Acad. Sci. USA 78, 7634–7638 (1981).

    Google Scholar 

  15. Thomson, J. A. et al. Embryonic stem cell lines derived from human blastocysts. Science 282, 1145–1147 (1998).

    Google Scholar 

  16. Takahashi, K. et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861–872 (2007).

    Google Scholar 

  17. Eiraku, M. et al. Self-organized formation of polarized cortical tissues from ESCs and its active manipulation by extrinsic signals. Cell Stem Cell 3, 519–532 (2008).

    Google Scholar 

  18. Mariani, J. et al. FOXG1-dependent dysregulation of GABA/glutamate neuron differentiation in autism spectrum disorders. Cell 162, 375–390 (2015).

    Google Scholar 

  19. Pasca, A. M. et al. Functional cortical neurons and astrocytes from human pluripotent stem cells in 3D culture. Nat. Methods 12, 671–678 (2015).

    Google Scholar 

  20. Eiraku, M. et al. Self-organizing optic-cup morphogenesis in three-dimensional culture. Nature 472, 51–56 (2011).

    Google Scholar 

  21. Takebe, T. et al. Vascularized and functional human liver from an iPSC-derived organ bud transplant. Nature 499, 481–484 (2013).

    Google Scholar 

  22. Little, M. H. & Combes, A. N. Kidney organoids: accurate models or fortunate accidents. Genes Dev. 33, 1319–1345 (2019).

    Google Scholar 

  23. Yoshihara, E. et al. Immune-evasive human islet-like organoids ameliorate diabetes. Nature 586, 606–611 (2020).

    Google Scholar 

  24. Lee, J. H. et al. Anatomically and functionally distinct lung mesenchymal populations marked by Lgr5 and Lgr6. Cell 170, 1149–1163.e12 (2017).

    Google Scholar 

  25. Drost, J. et al. Organoid culture systems for prostate epithelial and cancer tissue. Nat. Protoc. 11, 347–358 (2016).

    Google Scholar 

  26. Roth, J. G. et al. Advancing models of neural development with biomaterials. Nat. Rev. Neurosci. 22, 593–615 (2021).

    Google Scholar 

  27. Qazi, T. H. et al. Programming hydrogels to probe spatiotemporal cell biology. Cell Stem Cell 29, 678–691 (2022).

    Google Scholar 

  28. Ao, Z. et al. One-stop microfluidic assembly of human brain organoids to model prenatal cannabis exposure. Anal. Chem. 92, 4630–4638 (2020).

    Google Scholar 

  29. Andersen, J. et al. Generation of functional human 3D cortico-motor assembloids. Cell 183, 1913–1929.e26 (2020). This article describes how to build a mutli-organ system using the assembloid approach to connect the cerebral cortex or the hindbrain or spinal cord organoids with human skeletal muscle spheroids.

    Google Scholar 

  30. Birey, F. et al. Assembly of functionally integrated human forebrain spheroids. Nature 545, 54–59 (2017).

    Google Scholar 

  31. Huh, D. et al. Reconstituting organ-level lung functions on a chip. Science 328, 1662–1668 (2010). This article was the first to define the ‘organ-on-a-chip’ term, describing a microfluidic device that recapitulated endothelial–epithelial interface for studies of barrier function.

    Google Scholar 

  32. Viravaidya, K., Sin, A. & Shuler, M. L. Development of a microscale cell culture analog to probe naphthalene toxicity. Biotechnol. Prog. 20, 316–323 (2004).

    Google Scholar 

  33. Phan, D. T. T. et al. A vascularized and perfused organ-on-a-chip platform for large-scale drug screening applications. Lab Chip 17, 511–520 (2017).

    Google Scholar 

  34. Ma, L. D. et al. Design and fabrication of a liver-on-a-chip platform for convenient, highly efficient, and safe in situ perfusion culture of 3D hepatic spheroids. Lab Chip 18, 2547–2562 (2018).

    Google Scholar 

  35. Zhao, Y. et al. A platform for generation of chamber-specific cardiac tissues and disease modeling. Cell 176, 913–927.e8 (2019).

    Google Scholar 

  36. Lind, J. U. et al. Instrumented cardiac microphysiological devices via multimaterial three-dimensional printing. Nat. Mater. 16, 303–308 (2017). This article describes how to instrument an OoC device via 3D printing to measure contraction force via non-invasive electrical readouts.

    Google Scholar 

  37. Jalili-Firoozinezhad, S. et al. A complex human gut microbiome cultured in an anaerobic intestine-on-a-chip. Nat. Biomed. Eng. 3, 520–531 (2019).

    Google Scholar 

  38. Musah, S. et al. Mature induced-pluripotent-stem-cell-derived human podocytes reconstitute kidney glomerular-capillary-wall function on a chip. Nat. Biomed. Eng. 1, 0069 (2017).

    Google Scholar 

  39. Booth, R. & Kim, H. Characterization of a microfluidic in vitro model of the blood-brain barrier (muBBB). Lab Chip 12, 1784–1792 (2012).

    Google Scholar 

  40. Lee, J. H., Gu, Y., Wang, H. & Lee, W. Y. Microfluidic 3D bone tissue model for high-throughput evaluation of wound-healing and infection-preventing biomaterials. Biomaterials 33, 999–1006 (2012).

    Google Scholar 

  41. Carson, D. et al. Nanotopography-induced structural anisotropy and sarcomere development in human cardiomyocytes derived from induced pluripotent stem cells. ACS Appl. Mater. Interfaces 8, 21923–21932 (2016).

    Google Scholar 

  42. Mandrycky, C. J., Howard, C. C., Rayner, S. G., Shin, Y. J. & Zheng, Y. Organ-on-a-chip systems for vascular biology. J. Mol. Cell Cardiol. 159, 1–13 (2021).

    Google Scholar 

  43. Somers, S. M., Spector, A. A., DiGirolamo, D. J. & Grayson, W. L. Biophysical stimulation for engineering functional skeletal muscle. Tissue Eng. Part. B Rev. 23, 362–372 (2017).

    Google Scholar 

  44. Cook, D. et al. Lessons learned from the fate of AstraZeneca’s drug pipeline: a five-dimensional framework. Nat. Rev. Drug Discov. 13, 419–431 (2014).

    Google Scholar 

  45. Zhao, Y., Wang, E. Y., Lai, F. B. L., Cheung, K. & Radisic, M. Organs-on-a-chip: a union of tissue engineering and microfabrication. Trends Biotechnol. 41, 410–424 (2023).

    Google Scholar 

  46. McKinley, K. L., Longaker, M. T. & Naik, S. Emerging frontiers in regenerative medicine. Science 380, 796–798 (2023).

    Google Scholar 

  47. Reza, H. A., Okabe, R. & Takebe, T. Organoid transplant approaches for the liver. Transpl. Int. 34, 2031–2045 (2021).

    Google Scholar 

  48. Xiang, Y. et al. Fusion of regionally specified hPSC-derived organoids models human brain development and interneuron migration. Cell Stem Cell 21, 383–398.e7 (2017).

    Google Scholar 

  49. Rajasekar, S. et al. IFlowPlate — a customized 384‐well plate for the culture of perfusable vascularized colon organoids. Adv. Mater. 32, e2002974 (2020).

    Google Scholar 

  50. Fuchs, S. et al. In-line analysis of organ-on-chip systems with sensors: integration, fabrication, challenges, and potential. ACS Biomater. Sci. Eng. 7, 2926–2948 (2021).

    Google Scholar 

  51. Aleman, J., Kilic, T., Mille, L. S., Shin, S. R. & Zhang, Y. S. Microfluidic integration of regeneratable electrochemical affinity-based biosensors for continual monitoring of organ-on-a-chip devices. Nat. Protoc. 16, 2564–2593 (2021).

    Google Scholar 

  52. Li, T. L. et al. Stretchable mesh microelectronics for the biointegration and stimulation of human neural organoids. Biomaterials 290, 121825 (2022).

    Google Scholar 

  53. McDonald, M. et al. A mesh microelectrode array for non-invasive electrophysiology within neural organoids. Biosens. Bioelectron. 228, 115223 (2023).

    Google Scholar 

  54. Zachos, N. C. et al. Human enteroids/colonoids and intestinal organoids functionally recapitulate normal intestinal physiology and pathophysiology. J. Biol. Chem. 291, 3759–3766 (2016).

    Google Scholar 

  55. Kim, H. J., Huh, D., Hamilton, G. & Ingber, D. E. Human gut-on-a-chip inhabited by microbial flora that experiences intestinal peristalsis-like motions and flow. Lab Chip 12, 2165–2174 (2012).

    Google Scholar 

  56. Kasendra, M. et al. Development of a primary human small intestine-on-a-chip using biopsy-derived organoids. Sci. Rep. 8, 2871 (2018).

    Google Scholar 

  57. Sontheimer-Phelps, A. et al. Human colon-on-a-chip enables continuous in vitro analysis of colon mucus layer accumulation and physiology. Cell. Mol. Gastroenterol. Hepatol. 9, 507–526 (2020).

    Google Scholar 

  58. Gjorevski, N. et al. Tissue geometry drives deterministic organoid patterning. Science 375, eaaw9021 (2022). This paper describes how to control organoid shape and patterning by confined culture in photo-patterned structures from adhesion peptide-modified poly(ethylene glycol) hydrogels.

    Google Scholar 

  59. Nikolaev, M. et al. Homeostatic mini-intestines through scaffold-guided organoid morphogenesis. Nature 585, 574–578 (2020).

    Google Scholar 

  60. Faria, J., Ahmed, S., Gerritsen, K. G. F., Mihaila, S. M. & Masereeuw, R. Kidney-based in vitro models for drug-induced toxicity testing. Arch. Toxicol. 93, 3397–3418 (2019).

    Google Scholar 

  61. Xie, R. et al. h-FIBER: microfluidic topographical hollow fiber for studies of glomerular filtration barrier. ACS Cent. Sci. 6, 903–912 (2020).

    Google Scholar 

  62. Lin, N. Y. C. et al. Renal reabsorption in 3D vascularized proximal tubule models. Proc. Natl Acad. Sci. USA 116, 5399–5404 (2019).

    Google Scholar 

  63. Vriend, J. et al. Flow stimulates drug transport in a human kidney proximal tubule-on-a-chip independent of primary cilia. Biochim. Biophys. Acta Gen. Subj. 1864, 129433 (2020).

    Google Scholar 

  64. Vormann, M. K. et al. Nephrotoxicity and kidney transport assessment on 3D perfused proximal tubules. AAPS J. 20, 90 (2018).

    Google Scholar 

  65. Sakolish, C. et al. Predicting tubular reabsorption with a human kidney proximal tubule tissue-on-a-chip and physiologically-based modeling. Toxicol. In Vitro 63, 104752 (2020).

    Google Scholar 

  66. Wang, J. et al. A virus-induced kidney disease model based on organ-on-a-chip: pathogenesis exploration of virus-related renal dysfunctions. Biomaterials 219, 119367 (2019).

    Google Scholar 

  67. Takasato, M. et al. Kidney organoids from human iPS cells contain multiple lineages and model human nephrogenesis. Nature 526, 564–568 (2015).

    Google Scholar 

  68. Morizane, R. et al. Nephron organoids derived from human pluripotent stem cells model kidney development and injury. Nat. Biotechnol. 33, 1193–1200 (2015).

    Google Scholar 

  69. Freedman, B. S. et al. Modelling kidney disease with CRISPR-mutant kidney organoids derived from human pluripotent epiblast spheroids. Nat. Commun. 6, 8715 (2015).

    Google Scholar 

  70. Homan, K. A. et al. Flow-enhanced vascularization and maturation of kidney organoids in vitro. Nat. Methods 16, 255–262 (2019). This paper is an example of an integrative study, which demonstrated that kidney organoids cultured under flow had more mature podocytes and tubules with enhanced polarity than static culture had.

    Google Scholar 

  71. Lee, H. N. et al. Effect of biochemical and biomechanical factors on vascularization of kidney organoid-on-a-chip. Nano Converg. 8, 35 (2021).

    Google Scholar 

  72. Schutgens, F. et al. Tubuloids derived from human adult kidney and urine for personalized disease modeling. Nat. Biotechnol. 37, 303–313 (2019).

    Google Scholar 

  73. Aceves, J. O. et al. 3D proximal tubule-on-chip model derived from kidney organoids with improved drug uptake. Sci. Rep. 12, 14997 (2022).

    Google Scholar 

  74. Dye, B. R. et al. In vitro generation of human pluripotent stem cell derived lung organoids. eLife 4, e05098 (2015).

    Google Scholar 

  75. Chen, Y. W. et al. A three-dimensional model of human lung development and disease from pluripotent stem cells. Nat. Cell Biol. 19, 542–549 (2017).

    Google Scholar 

  76. Tindle, C. et al. Adult stem cell-derived complete lung organoid models emulate lung disease in COVID-19. eLife 10, e66417 (2021).

    Google Scholar 

  77. Li, C. et al. Human airway and nasal organoids reveal escalating replicative fitness of SARS-CoV-2 emerging variants. Proc. Natl Acad. Sci. USA 120, e2300376120 (2023).

    Google Scholar 

  78. Stucki, A. O. et al. A lung-on-a-chip array with an integrated bio-inspired respiration mechanism. Lab Chip 15, 1302–1310 (2015).

    Google Scholar 

  79. Benam, K. H. et al. Small airway-on-a-chip enables analysis of human lung inflammation and drug responses in vitro. Nat. Methods 13, 151–157 (2016).

    Google Scholar 

  80. Park, S. & Young, E. W. K. E-FLOAT: extractable floating liquid gel-based organ-on-a-chip for airway tissue modeling under airflow. Adv. Mater. Technol. 6, 2100828 (2021).

    Google Scholar 

  81. Park, J. Y. et al. Development of a functional airway-on-a-chip by 3D cell printing. Biofabrication 11, 015002 (2018).

    Google Scholar 

  82. Soleas, J. P. et al. Assembly of lung progenitors into developmentally-inspired geometry drives differentiation via cellular tension. Biomaterials 254, 120128 (2020).

    Google Scholar 

  83. Martignoni, M., Groothuis, G. M. & de Kanter, R. Species differences between mouse, rat, dog, monkey and human CYP-mediated drug metabolism, inhibition and induction. Expert Opin. Drug Metab. Toxicol. 2, 875–894 (2006).

    Google Scholar 

  84. Jang, K.-J. et al. Reproducing human and cross-species drug toxicities using a liver-chip. Sci. Transl. Med. 11, eaax5516 (2019).

    Google Scholar 

  85. Sung, J. H. & Shuler, M. L. 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 9, 1385–1394 (2009).

    Google Scholar 

  86. Tao, T. et al. Microengineered multi-organoid system from hiPSCs to recapitulate human liver-islet axis in normal and type 2 diabetes. Adv. Sci. 9, e2103495 (2022).

    Google Scholar 

  87. Jin, Y. et al. Vascularized liver organoids generated using induced hepatic tissue and dynamic liver-specific microenvironment as a drug testing platform. Adv. Funct. Mater. 28, 1801954 (2018).

    Google Scholar 

  88. Bovard, D. et al. A lung/liver-on-a-chip platform for acute and chronic toxicity studies. Lab Chip 18, 3814–3829 (2018).

    Google Scholar 

  89. Natarajan, V. & Simoneau, C. R. Modelling T-cell immunity against hepatitis C virus with liver organoids in a microfluidic coculture system. Open Biol. 12, 210320 (2022).

    Google Scholar 

  90. Skardal, A. et al. Multi-tissue interactions in an integrated three-tissue organ-on-a-chip platform. Sci. Rep. 7, 8837 (2017).

    Google Scholar 

  91. Teng, Y., Zhao, Z., Tasnim, F., Huang, X. & Yu, H. A scalable and sensitive steatosis chip with long-term perfusion of in situ differentiated HepaRG organoids. Biomaterials 275, 120904 (2021).

    Google Scholar 

  92. Takebe, T. et al. Massive and reproducible production of liver buds entirely from human pluripotent stem cells. Cell Rep. 21, 2661–2670 (2017).

    Google Scholar 

  93. Rajasekar, S. et al. Subtractive manufacturing with swelling induced stochastic folding of sacrificial materials for fabricating complex perfusable tissues in multi-well plates. Lab Chip 22, 1929–1942 (2022).

    Google Scholar 

  94. Koui, Y. et al. An in vitro human liver model by iPSC-derived parenchymal and non-parenchymal cells. Stem Cell Rep. 9, 490–498 (2017).

    Google Scholar 

  95. Kang, Y. B., Eo, J., Mert, S., Yarmush, M. L. & Usta, O. B. Metabolic patterning on a chip: towards in vitro liver zonation of primary rat and human hepatocytes. Sci. Rep. 8, 8951 (2018).

    Google Scholar 

  96. Huch, M. et al. Unlimited in vitro expansion of adult bi-potent pancreas progenitors through the Lgr5/R-spondin axis. EMBO J. 32, 2708–2721 (2013).

    Google Scholar 

  97. Shik Mun, K. et al. Patient-derived pancreas-on-a-chip to model cystic fibrosis-related disorders. Nat. Commun. 10, 3124 (2019).

    Google Scholar 

  98. Tao, T. et al. Engineering human islet organoids from iPSCs using an organ-on-chip platform. Lab Chip 19, 948–958 (2019).

    Google Scholar 

  99. Aamodt, K. I. & Powers, A. C. Signals in the pancreatic islet microenvironment influence β-cell proliferation. Diabetes, Obes. Metab. 19, 124–136 (2017).

    Google Scholar 

  100. Lancaster, M. A. et al. Cerebral organoids model human brain development and microcephaly. Nature 501, 373–379 (2013).

    Google Scholar 

  101. Eichmuller, O. L. & Knoblich, J. A. Human cerebral organoids — a new tool for clinical neurology research. Nat. Rev. Neurol. 18, 661–680 (2022).

    Google Scholar 

  102. Camp, J. G. et al. Human cerebral organoids recapitulate gene expression programs of fetal neocortex development. Proc. Natl Acad. Sci. USA 112, 15672–15677 (2015).

    Google Scholar 

  103. Nascimento, J. M. et al. Human cerebral organoids and fetal brain tissue share proteomic similarities. Front. Cell Dev. Biol. 7, 303 (2019).

    Google Scholar 

  104. Giandomenico, S. L., Sutcliffe, M. & Lancaster, M. A. Generation and long-term culture of advanced cerebral organoids for studying later stages of neural development. Nat. Protoc. 16, 579–602 (2021).

    Google Scholar 

  105. Kelley, K. W. & Pașca, S. P. Human brain organogenesis: toward a cellular understanding of development and disease. Cell 185, 42–61 (2022).

    Google Scholar 

  106. Sakaguchi, H. et al. Generation of functional hippocampal neurons from self-organizing human embryonic stem cell-derived dorsomedial telencephalic tissue. Nat. Commun. 6, 8896 (2015).

    Google Scholar 

  107. Kiral, F. R. et al. Generation of ventralized human thalamic organoids with thalamic reticular nucleus. Cell Stem Cell 30, 677–688.e5 (2023).

    Google Scholar 

  108. Ozone, C. et al. Functional anterior pituitary generated in self-organizing culture of human embryonic stem cells. Nat. Commun. 7, 10351 (2016).

    Google Scholar 

  109. Nickels, S. L. et al. Reproducible generation of human midbrain organoids for in vitro modeling of Parkinson’s disease. Stem Cell Res. 46, 101870 (2020).

    Google Scholar 

  110. Muguruma, K., Nishiyama, A., Kawakami, H., Hashimoto, K. & Sasai, Y. Self-organization of polarized cerebellar tissue in 3D culture of human pluripotent stem cells. Cell Rep. 10, 537–550 (2015).

    Google Scholar 

  111. Xiang, Y. et al. hESC-derived thalamic organoids form reciprocal projections when fused with cortical organoids. Cell Stem Cell 24, 487–497.e7 (2019).

    Google Scholar 

  112. Abud, E. M. et al. iPSC-derived human microglia-like cells to study neurological diseases. Neuron 94, 278–293.e9 (2017).

    Google Scholar 

  113. Pham, M. T. et al. Generation of human vascularized brain organoids. Neuroreport 29, 588–593 (2018).

    Google Scholar 

  114. Sun, X.-Y. et al. Generation of vascularized brain organoids to study neurovascular interactions. eLife 11, e76707 (2022).

    Google Scholar 

  115. Wang, Y. I., Abaci, H. E. & Shuler, M. L. Microfluidic blood–brain barrier model provides in vivo-like barrier properties for drug permeability screening. Biotechnol. Bioeng. 114, 184–194 (2017).

    Google Scholar 

  116. Wolff, A., Antfolk, M., Brodin, B. & Tenje, M. In vitro blood–brain barrier models — an overview of established models and new microfluidic approaches. J. Pharm. Sci. 104, 2727–2746 (2015).

    Google Scholar 

  117. Park, W., Lee, J. S., Gao, G., Kim, B. S. & Cho, D. W. 3D bioprinted multilayered cerebrovascular conduits to study cancer extravasation mechanism related with vascular geometry. Nat. Commun. 14, 7696 (2023).

    Google Scholar 

  118. Wang, Y., Wang, L., Zhu, Y. & Qin, J. Human brain organoid-on-a-chip to model prenatal nicotine exposure. Lab Chip 18, 851–860 (2018).

    Google Scholar 

  119. Salmon, I. et al. Engineering neurovascular organoids with 3D printed microfluidic chips. Lab Chip 22, 1615–1629 (2022).

    Google Scholar 

  120. Karzbrun, E., Kshirsagar, A., Cohen, S. R., Hanna, J. H. & Reiner, O. Human brain organoids on a chip reveal the physics of folding. Nat. Phys. 14, 515–522 (2018).

    Google Scholar 

  121. Skylar-Scott, M. A. et al. Biomanufacturing of organ-specific tissues with high cellular density and embedded vascular channels. Sci. Adv. 5, eaaw2459 (2019).

    Google Scholar 

  122. Cui, K. et al. Brain organoid-on-chip system to study the effects of breast cancer derived exosomes on the neurodevelopment of brain. Cell Regen. 11, 7 (2022).

    Google Scholar 

  123. Ao, Z. et al. Understanding immune‐driven brain aging by human brain organoid microphysiological analysis platform. Adv. Sci. 9, 2200475 (2022). This article describes the incorporation of immune cells within the microfluidic chip with brain organoids to mimic immune-driven brain ageing.

    Google Scholar 

  124. Ao, Z. et al. Human spinal organoid-on-a-chip to model nociceptive circuitry for pain therapeutics discovery. Anal. Chem. 94, 1365–1372 (2022).

    Google Scholar 

  125. Marcos, L. F., Wilson, S. L. & Roach, P. Tissue engineering of the retina: from organoids to microfluidic chips. J. Tissue Eng. 12, 20417314211059876 (2021).

    Google Scholar 

  126. Achberger, K. et al. Merging organoid and organ-on-a-chip technology to generate complex multi-layer tissue models in a human retina-on-a-chip platform. eLife 8, e46188 (2019).

    Google Scholar 

  127. Lindsey, S. E., Butcher, J. T. & Yalcin, H. C. Mechanical regulation of cardiac development. Front. Physiol. 5, 318 (2014).

    Google Scholar 

  128. Drakhlis, L. et al. Human heart-forming organoids recapitulate early heart and foregut development. Nat. Biotechnol. 39, 737–746 (2021).

    Google Scholar 

  129. Lewis-Israeli, Y. R. et al. Self-assembling human heart organoids for the modeling of cardiac development and congenital heart disease. Nat. Commun. 12, 5142 (2021).

    Google Scholar 

  130. Shirure, V. S., Hughes, C. C. & George, S. C. Engineering vascularized organoid-on-a-chip models. Annu. Rev. Biomed. Eng. 23, 141–167 (2021).

    Google Scholar 

  131. Garreta, E. et al. Rethinking organoid technology through bioengineering. Nat. Mater. 20, 145–155 (2021).

    Google Scholar 

  132. Lemoine, M. D. et al. Human induced pluripotent stem cell-derived engineered heart tissue as a sensitive test system for QT prolongation and arrhythmic triggers. Circ. Arrhythmia Electrophysiol. 11, e006035 (2018).

    Google Scholar 

  133. Riaz, M. et al. Muscle LIM protein force-sensing mediates sarcomeric biomechanical signaling in human familial hypertrophic cardiomyopathy. Circulation 145, 1238–1253 (2022).

    Google Scholar 

  134. Mills, R. J. et al. BET inhibition blocks inflammation-induced cardiac dysfunction and SARS-CoV-2 infection. Cell 184, 2167–2182.e22 (2021). This article describes how a two-post heart-on-a-chip platform can be rapidly adopted to mimic cytokine storm conditions and rapidly screen cardioprotective drugs in the settings of COVID-19-induced inflammation.

    Google Scholar 

  135. Leonard, A. et al. Afterload promotes maturation of human induced pluripotent stem cell derived cardiomyocytes in engineered heart tissues. J. Mol. Cell. Cardiol. 118, 147–158 (2018).

    Google Scholar 

  136. Hinson, J. T. et al. HEART DISEASE. Titin mutations in iPS cells define sarcomere insufficiency as a cause of dilated cardiomyopathy. Science 349, 982–986 (2015).

    Google Scholar 

  137. Tiburcy, M. et al. Defined engineered human myocardium with advanced maturation for applications in heart failure modeling and repair. Circulation 135, 1832–1847 (2017).

    Google Scholar 

  138. Nunes, S. S. et al. Biowire: a platform for maturation of human pluripotent stem cell-derived cardiomyocytes. Nat. Methods 10, 781–787 (2013).

    Google Scholar 

  139. Mathur, A. et al. Human iPSC-based cardiac microphysiological system for drug screening applications. Sci. Rep. 5, 8883 (2015).

    Google Scholar 

  140. Lai, B. F. L. et al. InVADE: integrated vasculature for assessing dynamic events. Adv. Funct. Mater. 27, 1703524 (2017).

    Google Scholar 

  141. Hoang, P. et al. Engineering spatial-organized cardiac organoids for developmental toxicity testing. Stem Cell Rep. 16, 1228–1244 (2021).

    Google Scholar 

  142. Abilez, O. J. et al. Micropatterned organoids enable modeling of the earliest stages of human cardiac vascularization. Preprint at bioRxiv https://doi.org/10.1101/2022.07.08.499233 (2022).

    Article  Google Scholar 

  143. Hoang, P., Wang, J., Conklin, B. R., Healy, K. E. & Ma, Z. Generation of spatial-patterned early-developing cardiac organoids using human pluripotent stem cells. Nat. Protoc. 13, 723–737 (2018).

    Google Scholar 

  144. Silva, A. C. et al. Co-emergence of cardiac and gut tissues promotes cardiomyocyte maturation within human iPSC-derived organoids. Cell Stem Cell 28, 2137–2152.e6 (2021).

    Google Scholar 

  145. Hofbauer, P. et al. Cardioids reveal self-organizing principles of human cardiogenesis. Cell 184, 3299–3317.e22 (2021).

    Google Scholar 

  146. Ma, Z. et al. Self-organizing human cardiac microchambers mediated by geometric confinement. Nat. Commun. 6, 7413 (2015).

    Google Scholar 

  147. Kupfer, M. E. et al. In situ expansion, differentiation, and electromechanical coupling of human cardiac muscle in a 3D bioprinted, chambered organoid. Circ. Res. 127, 207–224 (2020).

    Google Scholar 

  148. Liu, C. et al. Drug screening model meets cancer organoid technology. Transl. Oncol. 13, 100840 (2020).

    Google Scholar 

  149. Arima, Y., Nobusue, H. & Saya, H. Targeting of cancer stem cells by differentiation therapy. Cancer Sci. 111, 2689–2695 (2020).

    Google Scholar 

  150. LeSavage, B. L., Suhar, R. A., Broguiere, N., Lutolf, M. P. & Heilshorn, S. C. Next-generation cancer organoids. Nat. Mater. 21, 143–159 (2022).

    Google Scholar 

  151. Sun, Y. Tumor microenvironment and cancer therapy resistance. Cancer Lett. 380, 205–215 (2016).

    Google Scholar 

  152. Pinho, D., Santos, D., Vila, A. & Carvalho, S. Establishment of colorectal cancer organoids in microfluidic-based system. Micromachines 12, 497 (2021).

    Google Scholar 

  153. Shin, T. H., Kim, M., Sung, C. O., Jang, S. J. & Jeong, G. S. A one-stop microfluidic-based lung cancer organoid culture platform for testing drug sensitivity. Lab Chip 19, 2854–2865 (2019).

    Google Scholar 

  154. Fang, G. & Lu, H. Enabling peristalsis of human colon tumor organoids on microfluidic chips. Biofabrication 14, https://doi.org/10.1088/1758-5090/ac2ef9 (2021).

  155. Haque, M. R. et al. Patient-derived pancreatic cancer-on-a-chip recapitulates the tumor microenvironment. Microsyst. Nanoeng. 8, 36 (2022).

    Google Scholar 

  156. Miller, C. P., Tsuchida, C., Zheng, Y., Himmelfarb, J. & Akilesh, S. A 3D human renal cell carcinoma-on-a-chip for the study of tumor angiogenesis. Neoplasia 20, 610–620 (2018).

    Google Scholar 

  157. Shirure, V. S. et al. Tumor-on-a-chip platform to investigate progression and drug sensitivity in cell lines and patient-derived organoids. Lab Chip 18, 3687–3702 (2018).

    Google Scholar 

  158. Schuster, B. et al. Automated microfluidic platform for dynamic and combinatorial drug screening of tumor organoids. Nat. Commun. 11, 5271 (2020).

    Google Scholar 

  159. Lai Benjamin, F. L. et al. Recapitulating pancreatic tumor microenvironment through synergistic use of patient organoids and organ-on-a-chip vasculature. Adv. Funct. Mater. 30, 2000545 (2020).

    Google Scholar 

  160. Mazzocchi, A. R., Rajan, S. A., Votanopoulos, K. I., Hall, A. R. & Skardal, A. In vitro patient-derived 3D mesothelioma tumor organoids facilitate patient-centric therapeutic screening. Sci. Rep. 8, 2886 (2018).

    Google Scholar 

  161. Betge, J. et al. The drug-induced phenotypic landscape of colorectal cancer organoids. Nat. Commun. 13, 3135 (2022).

    Google Scholar 

  162. Usman, O. H. et al. Genomic heterogeneity in pancreatic cancer organoids and its stability with culture. NPJ Genomic Med. 7, 71 (2022).

    Google Scholar 

  163. Miura, Y. et al. Generation of human striatal organoids and cortico-striatal assembloids from human pluripotent stem cells. Nat. Biotechnol. 38, 1421–1430 (2020).

    Google Scholar 

  164. Son, J. et al. Electrophysiological monitoring of neurochemical-based neural signal transmission in a human brain–spinal cord assembloid. ACS Sens. 7, 409–414 (2022).

    Google Scholar 

  165. Kim, E. et al. Creation of bladder assembloids mimicking tissue regeneration and cancer. Nature 588, 664–669 (2020).

    Google Scholar 

  166. McAleer, C. W. et al. Multi-organ system for the evaluation of efficacy and off-target toxicity of anticancer therapeutics. Sci. Transl. Med. 11, eaav1386 (2019). This paper describes a pumpless multi-organ platform used to predict drug efficacy and metabolic conversion in liver, tumour and heart systems while recirculating serum-free medium between the compartments.

    Google Scholar 

  167. Yin, F. et al. HiPSC-derived multi-organoids-on-chip system for safety assessment of antidepressant drugs. Lab Chip 21, 571–581 (2021).

    Google Scholar 

  168. Wikswo, J. P. et al. Scaling and systems biology for integrating multiple organs-on-a-chip. Lab Chip 13, 3496–3511 (2013).

    Google Scholar 

  169. Ronaldson-Bouchard, K. et al. A multi-organ chip with matured tissue niches linked by vascular flow. Nat. Biomed. Eng. 6, 351–371 (2022). This article describes a body-on-a-chip system in which matured human heart, liver, bone and skin tissue niches are linked by recirculating flow through an endothelialized channel.

    Google Scholar 

  170. Radisic, M. et al. Oxygen gradients correlate with cell density and cell viability in engineered cardiac tissue. Biotechnol. Bioeng. 93, 332–343 (2006).

    Google Scholar 

  171. Mansour, A. A. et al. An in vivo model of functional and vascularized human brain organoids. Nat. Biotechnol. 36, 432–441 (2018).

    Google Scholar 

  172. Revah, O. et al. Maturation and circuit integration of transplanted human cortical organoids. Nature 610, 319–326 (2022).

    Google Scholar 

  173. Zhang, S., Wan, Z. & Kamm, R. D. Vascularized organoids on a chip: strategies for engineering organoids with functional vasculature. Lab Chip 21, 473–488 (2021).

    Google Scholar 

  174. Chiu, L. L., Montgomery, M., Liang, Y., Liu, H. & Radisic, M. Perfusable branching microvessel bed for vascularization of engineered tissues. Proc. Natl Acad. Sci. USA 109, E3414–E3423 (2012).

    Google Scholar 

  175. Haase, K. et al. Physiologic flow-conditioning limits vascular dysfunction in engineered human capillaries. Biomaterials 280, 121248 (2022). This paper demonstrates the importance of interstitial flow in promoting early blood vessel formation and continuous flow through the vessels for maintenance and remodelling of the formed vasculature.

    Google Scholar 

  176. Zhang, B. et al. Biodegradable scaffold with built-in vasculature for organ-on-a-chip engineering and direct surgical anastomosis. Nat. Mater. 15, 669–678 (2016).

    Google Scholar 

  177. Barrile, R. et al. Organ-on-chip recapitulates thrombosis induced by an anti-CD154 monoclonal antibody: translational potential of advanced microengineered systems. Clin. Pharmacol. Ther. 104, 1240–1248 (2018).

    Google Scholar 

  178. Schulla, L. S. et al. Development of a novel microfluidic co-culture model to study organoid vascularization. Preprint at bioRxiv https://doi.org/10.1101/2022.03.25.485813 (2022).

    Article  Google Scholar 

  179. Landau, S. et al. Tissue-level mechanosensitivity: predicting and controlling the orientation of 3D vascular networks. Nano Lett. 18, 7698–7708 (2018).

    Google Scholar 

  180. Ingber, D. E. Human organs-on-chips for disease modelling, drug development and personalized medicine. Nat. Rev. Genet. 23, 467–491 (2022).

    Google Scholar 

  181. Korolj, A., Wu, H. T. & Radisic, M. A healthy dose of chaos: using fractal frameworks for engineering higher-fidelity biomedical systems. Biomaterials 219, 119363 (2019).

    Google Scholar 

  182. Doi, K. et al. Enhanced podocyte differentiation and changing drug toxicity sensitivity through pressure-controlled mechanical filtration stress on a glomerulus-on-a-chip. Lab Chip 23, 437–450 (2023).

    Google Scholar 

  183. Zhang, S. Y. & Mahler, G. J. A glomerulus and proximal tubule microphysiological system simulating renal filtration, reabsorption, secretion, and toxicity. Lab chip 23, 272–284 (2023).

    Google Scholar 

  184. Roth, J. G. et al. Spatially controlled construction of assembloids using bioprinting. Nat. Commun. 14, 4346 (2023).

    Google Scholar 

  185. Misun, P. M., Rothe, J., Schmid, Y. R., Hierlemann, A. & Frey, O. Multi-analyte biosensor interface for real-time monitoring of 3D microtissue spheroids in hanging-drop networks. Microsyst. Nanoeng. 2, 16022 (2016).

    Google Scholar 

  186. Gebreyesus, S. T. et al. Streamlined single-cell proteomics by an integrated microfluidic chip and data-independent acquisition mass spectrometry. Nat. Commun. 13, 37 (2022).

    Google Scholar 

  187. Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676 (2006).

    Google Scholar 

  188. Burridge, P. W., Holmstrom, A. & Wu, J. C. Chemically defined culture and cardiomyocyte differentiation of human pluripotent stem cells. Curr. Protoc. Hum. Genet. 87, 21.3.1–21.3.15 (2015).

    Google Scholar 

  189. Rausch, V., Sala, V., Penna, F., Porporato, P. E. & Ghigo, A. Understanding the common mechanisms of heart and skeletal muscle wasting in cancer cachexia. Oncogenesis 10, 1 (2021).

    Google Scholar 

  190. Petersson, C. et al. Current approaches for predicting human PK for small molecule development candidates: findings from the IQ human PK prediction working group survey. AAPS J. 24, 85 (2022).

    Google Scholar 

  191. Gintant, G., Sager, P. T. & Stockbridge, N. Evolution of strategies to improve preclinical cardiac safety testing. Nat. Rev. Drug Discov. 15, 457–471 (2016).

    Google Scholar 

  192. Han, J. J. FDA modernization act 2.0 allows for alternatives to animal testing. Artif. Organs 47, 449–450 (2023).

    Google Scholar 

  193. Landon‐Brace, N. et al. An engineered patient‐derived tumor organoid model that can be disassembled to study cellular responses in a graded 3D microenvironment. Adv. Funct. Mater. 31, 2105349 (2021).

    Google Scholar 

  194. Barker, N. et al. Lgr5+ve stem cells drive self-renewal in the stomach and build long-lived gastric units in vitro. Cell Stem Cell 6, 25–36 (2010).

    Google Scholar 

  195. Xia, Y. et al. Directed differentiation of human pluripotent cells to ureteric bud kidney progenitor-like cells. Nat. Cell Biol. 15, 1507–1515 (2013).

    Google Scholar 

  196. Zhang, L. et al. Establishing estrogen-responsive mouse mammary organoids from single Lgr5+ cells. Cell Signal. 29, 41–51 (2017).

    Google Scholar 

  197. Miller, J. S. et al. Rapid casting of patterned vascular networks for perfusable engineered three-dimensional tissues. Nat. Mater. 11, 768–774 (2012).

    Google Scholar 

  198. Ronaldson-Bouchard, K. et al. Advanced maturation of human cardiac tissue grown from pluripotent stem cells. Nature 556, 239–243 (2018).

    Google Scholar 

  199. Wu, Q. et al. Flexible 3D printed microwires and 3D microelectrodes for heart-on-a-chip engineering. Biofabrication 15, 035023 (2023).

    Google Scholar 

  200. Zhang, B. & Radisic, M. Organ-on-a-chip devices advance to market. Lab Chip 17, 2395–2420 (2017).

    Google Scholar 

  201. Homan, K. A. Industry adoption of organoids and organs-on-chip technology: toward a paradox of choice. Adv. Biol. 7, 2200334 (2023).

    Google Scholar 

Download references

Acknowledgements

Our work is funded by Canadian Institutes of Health Research (CIHR) Foundation grant FDN-167274, Natural Sciences and Engineering Research Council of Canada (NSERC) Discovery grant (RGPIN 326982-10), NSERC-CIHR Collaborative Health Research grant (CHRP 493737-16), US National Institutes of Health grant 2R01 HL076485 and a Stem Cell Network Impact Award (IMP-C4R1-3). M.R. was supported by the Killam Fellowship and Canada Research Chair. Y.Z. was supported by a CIHR postdoctoral award. S.L. was supported by a Rothschild, Zuckerman, and EMBO (ALTF 530-2022) fellowship.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: M.R., Y.Z., S.L. and S.O. Writing – original draft: Y.Z., S.L., S.O., C.L., R.X.Z.L., B.F.L.L., Q.W., J.K., K.C., S.R., K.J., B.Z. and M.R. Writing – review and editing: M.R., Y.Z., S.L. and S.O. Visualization: Y.Z., S.L., S.O. and K.C. Supervision: M.R. and B.Z. Project administration: M.R. Funding acquisition: M.R. and B.Z.

Corresponding author

Correspondence to Milica Radisic.

Ethics declarations

Competing interests

M.R., Y.Z. and B.Z. are inventors on an issued US patent for Biowire technology that is licensed to Valo Health; they receive royalties for this invention. B.Z. and S.R. are co-founders and hold equity in OrganoBiotech. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Review Bioengineering thanks Kimberly Homan, who co-reviewed with Julien Roth; Ryuji Morizane; and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related links

Bioconvergence hub: https://bico.com/what-we-do/

Comprehensive In Vitro Pro-arrhythmia Assay (CIPA): https://cipaproject.org/

Human Cell Atlas: https://www.humancellatlas.org/

IQ consortium: https://iqconsortium.org/

United Network for Organ Sharing: https://unos.org/

Valo Health, an AI company, acquiring the heart-on-a-chip company TARA Biosystems: https://www.valohealth.com/press/valo-health-acquires-tara-biosystems-creating-first-of-its-kind-vertically-integrated-cardiovascular-platform

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhao, Y., Landau, S., Okhovatian, S. et al. Integrating organoids and organ-on-a-chip devices. Nat Rev Bioeng 2, 588–608 (2024). https://doi.org/10.1038/s44222-024-00207-z

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s44222-024-00207-z

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research