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.

  • Article
  • Published:

A tumor-on-a-chip for in vitro study of CAR-T cell immunotherapy in solid tumors

Abstract

Our limited understanding of cancer–immune interactions remains a critical barrier to advancing chimeric antigen receptor (CAR)-T cell therapy for solid malignancies. Here, we present a microengineered system that enables vascularization of human tumor explants and their controlled perfusion with immune cells to model the activity of CAR-T cells in the tumor microenvironment. Using vascularized human lung adenocarcinoma tumors, we first demonstrate the ability of our tumor-on-a-chip system to simulate, visualize and interrogate CAR-T cell function. We then test a chemokine-directed CAR-T cell engineering strategy in a model of malignant pleural mesothelioma and validate our findings in a matching in vivo mouse model. Finally, we describe a potential therapeutic target that can be pharmacologically modulated to increase the efficacy of CAR-T cells in lung adenocarcinoma, for which we present biomarkers identified by global metabolomics analysis. Our microphysiological system provides promising in vitro technology to advance the development of adoptive cell therapies for cancer and other diseases.

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: Microengineered system for in vitro transplantation and prolonged maintenance of human solid tumors.
Fig. 2: In vitro modeling of CAR-T–tumor interactions.
Fig. 3: Flow cytometric analysis of CAR-T cell phenotype and scRNA-seq analysis of microengineered tumor model.
Fig. 4: In vitro and in vivo analysis of human meso-CAR-T cells armored with CCR2.
Fig. 5: Transplantation and CAR-T cell treatment of human mesothelioma explants.
Fig. 6: Identification of therapeutic targets through analysis of ligand-receptor interactions in CAR-T cell-treated tumor models.

Similar content being viewed by others

Data availability

The scRNA-seq datasets generated and analyzed during the current study are available at the NCBI Gene Expression Omnibus, under accession number GSE240121 (ref. 110). All other relevant data supporting the key findings of this study are available within the article and its Supplementary Information files. The raw images are available in Dryad111, a general public repository, under the https://doi.org/10.5061/dryad.mw6m9068f. Source data are provided with this paper.

References

  1. Newick, K., O’Brien, S., Moon, E. & Albelda, S. M. CAR T cell therapy for solid tumors. Annu Rev. Med. 68, 139–152 (2017).

    Article  PubMed  CAS  Google Scholar 

  2. Shah, N. N. & Fry, T. J. Mechanisms of resistance to CAR T cell therapy. Nat. Rev. Clin. Oncol. 16, 372–385 (2019).

    PubMed  PubMed Central  CAS  Google Scholar 

  3. Lim, W. A. & June, C. H. The principles of engineering immune cells to treat cancer. Cell 168, 724–740 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Joyce, J. A. & Fearon, D. T. T cell exclusion, immune privilege, and the tumor microenvironment. Science (1979) 348, 74–80 (2015).

    CAS  Google Scholar 

  5. Ramakrishna, S., Barsan, V. & Mackall, C. Prospects and challenges for use of CAR T cell therapies in solid tumors. Expert Opin. Biol. Ther. 20, 503–516 (2020).

    Article  PubMed  CAS  Google Scholar 

  6. Sackstein, R., Schatton, T. & Barthel, S. R. T-lymphocyte homing: an underappreciated yet critical hurdle for successful cancer immunotherapy. Lab. Invest. 97, 669–697 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Martinez, M. & Moon, E. K. CAR T cells for solid tumors: new strategies for finding, infiltrating, and surviving in the tumor microenvironment. Front. Immunol. 10, 128 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Zitvogel, L., Pitt, J. M., Daillère, R., Smyth, M. J. & Kroemer, G. Mouse models in oncoimmunology. Nat. Rev. Cancer 2016 16:12 16, 759–773 (2016).

    CAS  Google Scholar 

  9. Gengenbacher, N., Singhal, M. & Augustin, H. G. Preclinical mouse solid tumour models: status quo, challenges and perspectives. Nat. Rev. Cancer 17, 751–765 (2017).

    Article  PubMed  CAS  Google Scholar 

  10. Moon, E. K. et al. Multifactorial T-cell hypofunction that is reversible can limit the efficacy of chimeric antigen receptor–transduced human T cells in solid tumors. Clin. Cancer Res. 20, 4262–4273 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Chuprin, J. et al. Humanized mouse models for immuno-oncology research. Nat. Rev. Clin. Oncol. 3, 192–206 (2023).

    Article  Google Scholar 

  12. Ma, X. et al. Interleukin-23 engineering improves CAR T cell function in solid tumors. Nat. Biotechnol. 38, 448–459 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Avanzi, M. P. et al. Engineered tumor-targeted T cells mediate enhanced anti-tumor efficacy both directly and through activation of the endogenous immune system. Cell Rep. 23, 2130–2141 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Klampatsa, A. et al. Analysis and augmentation of the immunologic bystander effects of CAR T cell therapy in a syngeneic mouse cancer model. Mol. Ther. Oncolytics 18, 360–371 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Evgin, L. et al. Oncolytic virus–mediated expansion of dual-specific CAR T cells improves efficacy against solid tumors in mice. Sci. Transl. Med. 14, 2231 (2022).

    Article  Google Scholar 

  16. Duncan, B. B., Dunbar, C. E. & Ishii, K. Applying a clinical lens to animal models of CAR-T cell therapies. Mol. Ther. Methods Clin. Dev. 27, 17–31 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Elsallab, M., Bravery, C. A., Kurtz, A. & Abou-El-Enein, M. Mitigating deficiencies in evidence during regulatory assessments of advanced therapies: a comparative study with other biologicals. Mol. Ther. Methods Clin. Dev. 18, 269–279 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Abou-el-Enein, M. et al. Evidence generation and reproducibility in cell and gene therapy research: a call to action. Mol. Ther. Methods Clin. Dev. 22, 11–14 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. McKean, M. et al. Safety and early efficacy results from a phase 1, multicenter trial of PSMA-targeted armored CAR T cells in patients with advanced mCRPC. J. Clin. Oncol. 40, 94 (2022).

    Article  Google Scholar 

  20. Kloss, C. C. et al. Dominant-negative TGF-β receptor enhances PSMA-targeted human CAR T cell proliferation and augments prostate cancer eradication. Mol. Ther. 26, 1855–1866 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Mak, I. W. Y., Evaniew, N. & Ghert, M. Lost in translation: animal models and clinical trials in cancer treatment. Am. J. Transl. Res 6, 114 (2014).

    PubMed  PubMed Central  Google Scholar 

  22. Hegde, P. S. & Chen, D. S. Top 10 challenges in cancer immunotherapy. Immunity 52, 17–35 (2020).

    Article  PubMed  CAS  Google Scholar 

  23. Vanmeerbeek, I., Naulaerts, S. & Garg, A. D. Reverse translation: the key to increasing the clinical success of immunotherapy?. Genes Immun. 24, 217–219 (2023).

    Article  PubMed  Google Scholar 

  24. Mollica, H. et al. A 3D pancreatic tumor model to study T cell infiltration. Biomater. Sci. 9, 7420–7431 (2021).

    Article  PubMed  CAS  Google Scholar 

  25. Wallstabe, L. et al. ROR1-CAR T cells are effective against lung and breast cancer in advanced microphysiologic 3D tumor models. JCI Insight 4, e126345 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Ayuso, J. M. et al. Microfluidic tumor-on-a-chip model to evaluate the role of tumor environmental stress on NK cell exhaustion. Sci. Adv. 7, 1–15 (2021).

    Article  Google Scholar 

  27. Sontheimer-Phelps, A., Hassell, B. A. & Ingber, D. E. Modelling cancer in microfluidic human organs-on-chips. Nat. Rev. Cancer 19, 65–81 (2019).

    Article  PubMed  CAS  Google Scholar 

  28. Nguyen, H. T. et al. Patient-specific vascularized tumor model: blocking monocyte recruitment with multispecific antibodies targeting CCR2 and CSF-1R. Biomaterials 312, 122731 (2025).

    Article  PubMed  CAS  Google Scholar 

  29. Pavesi, A. et al. A 3D microfluidic model for preclinical evaluation of TCR-engineered T cells against solid tumors. JCI Insight 2, e89762 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Maulana, T. I. et al. Breast cancer-on-chip for patient-specific efficacy and safety testing of CAR-T cells. Cell Stem Cell 31, 989–1002.e9 (2024).

    Article  PubMed  CAS  Google Scholar 

  31. Hidalgo, M. et al. Patient-derived xenograft models: an emerging platform for translational cancer research. Cancer Discov. 4, 998–1013 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Day, C. P., Merlino, G. & Van Dyke, T. Preclinical mouse cancer models: a maze of opportunities and challenges. Cell 163, 39–53 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Egeblad, M., Nakasone, E. S. & Werb, Z. Tumors as organs: complex tissues that interface with the entire organism. Dev. Cell 18, 884–901 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Li, L. et al. Laminin γ2–mediating T cell exclusion attenuates response to anti–PD-1 therapy. Sci. Adv. 7, eabc8346 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Klampatsa, A., Dimou, V. & Albelda, S. M. Mesothelin-targeted CAR-T cell therapy for solid tumors. Expert Opin. Biol. Ther. 21, 473–486 (2021).

    Article  PubMed  CAS  Google Scholar 

  36. Tanyi, J. et al. Phase I study of autologous T cells bearing fully-humanized chimeric antigen receptors targeting mesothelin in mesothelin- expressing cancers (314). Gynecol. Oncol. 166, S164–S165 (2022).

    Article  Google Scholar 

  37. MacKay, M. et al. The therapeutic landscape for cells engineered with chimeric antigen receptors. Nat. Biotechnol. 38, 233–244 (2020).

    Article  PubMed  CAS  Google Scholar 

  38. Saez-Ibañez, A. R. et al. Landscape of cancer cell therapies: trends and real-world data. Nat. Rev. Drug Discov. 21, 631–632 (2022).

    Article  PubMed  Google Scholar 

  39. Hassan, R. et al. Mesothelin immunotherapy for cancer: ready for prime time?. J. Clin. Oncol. 34, 4171–4179 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Pastan, I. & Hassan, R. Discovery of mesothelin and exploiting it as a target for immunotherapy. Cancer Res. 74, 2907–2912 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Mariathasan, S. et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 554, 544–548 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. O’Brien, S. M. et al. Function of human tumor-infiltrating lymphocytes in early-stage non–small cell lung cancer. Cancer Immunol. Res. 7, 896–909 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Ganesan, A. P. et al. Tissue-resident memory features are linked to the magnitude of cytotoxic T cell responses in human lung cancer. Nat. Immunol. 18, 940–950 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Klampatsa, A. et al. Phenotypic and functional analysis of malignant mesothelioma tumor-infiltrating lymphocytes. Oncoimmunology 8, 1–12 (2019).

    Article  Google Scholar 

  45. Hall, J. C. et al. Precise probes of type II interferon activity define the origin of interferon signatures in target tissues in rheumatic diseases. Proc. Natl Acad. Sci. USA. 109, 17609–17614 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Boroughs, A. C. et al. A distinct transcriptional program in human CAR T cells bearing the 4-1BB signaling domain revealed by scRNA-seq. Mol. Ther. 28, 1–16 (2020).

    Article  Google Scholar 

  47. Szabo, P. A. et al. Single-cell transcriptomics of human T cells reveals tissue and activation signatures in health and disease. Nat. Commun. 10, 4706 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Guo, X. et al. Global characterization of T cells in non-small-cell lung cancer by single-cell sequencing. Nat. Med. 24, 978–985 (2018).

    Article  PubMed  CAS  Google Scholar 

  49. Uslu, U., Castelli, S. & June, C. H. CAR T cell combination therapies to treat cancer. Cancer Cell 42, 1319–1325 (2024).

    Article  PubMed  CAS  Google Scholar 

  50. Craddock, J. A. et al. Enhanced tumor trafficking of GD2 chimeric antigen receptor T cells by expression of the chemokine receptor CCR2b. J. Immunother. 33, 780–788 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Gueugnon, F. et al. Identification of novel markers for the diagnosis of malignant pleural mesothelioma. Am. J. Pathol. 178, 1033–1042 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Servais, E. L. et al. Mesothelin overexpression promotes mesothelioma cell invasion and MMP-9 secretion in an orthotopic mouse model and in epithelioid pleural mesothelioma patients. Clin. Cancer Res. 18, 2478–2489 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Bagley, S. J., Desai, A. S., Linette, G. P., June, C. H. & O’Rourke, D. M. CAR T-cell therapy for glioblastoma: recent clinical advances and future challenges. Neuro Oncol. 20, 1429–1438 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Grosser, R., Cherkassky, L., Chintala, N. & Adusumilli, P. S. Combination immunotherapy with CAR T cells and checkpoint blockade for the treatment of solid tumors. Cancer Cell 36, 471–482 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Efremova, M., Vento-Tormo, M., Teichmann, S. A. & Vento-Tormo, R. CellPhoneDB: inferring cell–cell communication from combined expression of multi-subunit ligand–receptor complexes. Nat. Protoc. 15, 1484–1506 (2020).

    Article  PubMed  CAS  Google Scholar 

  56. Choi, J., Enis, D. R., Koh, K. P., Shiao, S. L. & Pober, J. S. T lymphocyte-endothelial cell interactions. Annu Rev. Immunol. 22, 683–709 (2004).

    Article  PubMed  CAS  Google Scholar 

  57. Motz, G. T. et al. Tumor endothelium FasL establishes a selective immune barrier promoting tolerance in tumors. Nat. Med. 20, 607–615 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Larson, R. C. & Maus, M. V. Recent advances and discoveries in the mechanisms and functions of CAR T cells. Nat. Rev. Cancer 21, 145–161 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Casneuf, T. et al. Effects of daratumumab on natural killer cells and impact on clinical outcomes in relapsed or refractory multiple myeloma. Blood Adv. 1, 2105–2114 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. St.Clair, E. W. et al. Clinical efficacy and safety of baminercept, a lymphotoxin β receptor fusion protein, in primary Sjögren’s syndrome: results from a phase II randomized, double-blind, placebo-controlled trial. Arthritis Rheumatol. 70, 1470–1480 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Tokunaga, R. et al. CXCL9, CXCL10, CXCL11/CXCR3 axis for immune activation: a target for novel cancer therapy. Cancer Treat. Rev. 63, 40–47 (2018).

    Article  PubMed  CAS  Google Scholar 

  62. Nagarsheth, N., Wicha, M. S. & Zou, W. Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat. Rev. Immunol. 17, 559–572 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Casrouge, A. et al. Evidence for an antagonist form of the chemokine CXCL10 in patients chronically infected with HCV. J. Clin. Invest. 121, 308–317 (2011).

    Article  PubMed  CAS  Google Scholar 

  64. Morello, A., Sadelain, M. & Adusumilli, P. S. Mesothelin-targeted CARs: driving T cells to solid tumors. Cancer Discov. 6, 133–146 (2016).

    Article  PubMed  CAS  Google Scholar 

  65. Kachala, S. S. et al. Mesothelin overexpression is a marker of tumor aggressiveness and is associated with reduced recurrence-free and overall survival in early-stage lung adenocarcinoma. Clin. Cancer Res. 20, 1020–1028 (2014).

    Article  PubMed  CAS  Google Scholar 

  66. Carpenito, C. et al. Control of large, established tumor xenografts with genetically retargeted human T cells containing CD28 and CD137 domains. Proc. Natl Acad. Sci. USA. 106, 3360–3365 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Zhao, Y. et al. Multiple injections of electroporated autologous T cells expressing a chimeric antigen receptor mediate regression of human disseminated tumor. Cancer Res. 70, 9053–9061 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Santos, A. M., Jung, J., Aziz, N., Kissil, J. L. & Puré, E. Targeting fibroblast activation protein inhibits tumor stromagenesis and growth in mice. J. Clin. Invest. 119, 3613–3625 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  69. Villhauer, E. B. et al. 1-[[(3-Hydroxy-1-adamantyl)amino]acetyl]-2-cyano-(S)-pyrrolidine: a potent, selective, and orally bioavailable dipeptidyl peptidase IV inhibitor with antihyperglycemic properties. J. Med. Chem. 46, 2774–2789 (2003).

    Article  PubMed  CAS  Google Scholar 

  70. Hu, P. et al. Pharmacokinetics and pharmacodynamics of vildagliptin in healthy Chinese volunteers. J. Clin. Pharm. 49, 39–49 (2009).

    Article  Google Scholar 

  71. He, Y. L. Clinical pharmacokinetics and pharmacodynamics of vildagliptin. Clin. Pharmacokinet. 51, 147–162 (2012).

    Article  PubMed  CAS  Google Scholar 

  72. Pathria, P., Louis, T. L. & Varner, J. A. Targeting tumor-associated macrophages in cancer. Trends Immunol. 40, 310–327 (2019).

    Article  PubMed  CAS  Google Scholar 

  73. Sautès-Fridman, C., Petitprez, F., Calderaro, J. & Fridman, W. H. Tertiary lymphoid structures in the era of cancer immunotherapy. Nat. Rev. Cancer 19, 307–325 (2019).

    Article  PubMed  Google Scholar 

  74. Schumacher, T. N. & Thommen, D. S. Tertiary lymphoid structures in cancer. Science 375, eabf9419 (2022).

    Article  PubMed  CAS  Google Scholar 

  75. Srivastava, S. et al. Immunogenic chemotherapy enhances recruitment of CAR-T cells to lung tumors and improves antitumor efficacy when combined with checkpoint blockade. Cancer Cell 39, 193–208 (2021).

    Article  PubMed  CAS  Google Scholar 

  76. Barrett, R. & Puré, E. Cancer-associated fibroblasts: key determinants of tumor immunity and immunotherapy. Curr. Opin. Immunol. 64, 80–87 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  77. Sahai, E. et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat. Rev. Cancer 20, 174–186 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Claesson-Welsh, L., Dejana, E. & McDonald, D. M. Permeability of the endothelial barrier: identifying and reconciling controversies. Trends Mol. Med. 27, 314–331 (2021).

    Article  PubMed  CAS  Google Scholar 

  79. Tian, L. et al. Mutual regulation of tumour vessel normalization and immunostimulatory reprogramming. Nature 544, 250–254 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  80. Barreira Da Silva, R. et al. Dipeptidylpeptidase 4 inhibition enhances lymphocyte trafficking, improving both naturally occurring tumor immunity and immunotherapy. Nat. Immunol. 16, 850–858 (2015).

    Article  PubMed  CAS  Google Scholar 

  81. Nishina, S. et al. Dipeptidyl peptidase 4 inhibitors reduce hepatocellular carcinoma by activating lymphocyte chemotaxis in mice. Cmgh 7, 115–134 (2019).

    PubMed  Google Scholar 

  82. Ohnuma, K., Hatano, R. & Morimoto, C. DPP4 in anti-tumor immunity: going beyond the enzyme. Nat. Immunol. 16, 791–792 (2015).

    Article  PubMed  CAS  Google Scholar 

  83. Young, R. M. et al. Next-generation CAR T-cell therapies. Cancer Discov. 12, 1625–1633 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  84. Sterner, R. C. & Sterner, R. M. CAR-T cell therapy: current limitations and potential strategies. Blood Cancer J. 11, 69 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  85. Hou, A. J., Chen, L. C. & Chen, Y. Y. Navigating CAR-T cells through the solid-tumour microenvironment. Nat. Rev. Drug Discov. 2021 20:7 20, 531–550 (2021).

    Article  CAS  Google Scholar 

  86. Gill, S., Maus, M. V. & Porter, D. L. Chimeric antigen receptor T cell therapy: 25 years in the making. Blood Rev. 30, 157–167 (2016).

    Article  PubMed  CAS  Google Scholar 

  87. Kumar, C. S. et al. Molecular characterization of the murine interferon γ receptor cDNA. J. Biol. Chem. 264, 17939–17946 (1989).

    Article  PubMed  CAS  Google Scholar 

  88. Mathieu, C. & Degrande, E. Vildagliptin: a new oral treatment for type 2 diabetes mellitus. Vasc. Health Risk Manag 4, 1349 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  89. House, I. G. et al. Macrophage-derived CXCL9 and CXCL10 are required for antitumor immune responses following immune checkpoint blockade. Clin. Cancer Res. 26, 487–504 (2020).

    Article  PubMed  CAS  Google Scholar 

  90. Laskowski, T. J., Biederstädt, A. & Rezvani, K. Natural killer cells in antitumour adoptive cell immunotherapy. Nat. Rev. Cancer 22, 557–575 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  91. Mensurado, S. et al. The emerging roles of γδ T cells in cancer immunotherapy. Nat. Rev. Clin. Oncol. 3, 178–191 (2023).

    Article  Google Scholar 

  92. Klichinsky, M. et al. Human chimeric antigen receptor macrophages for cancer immunotherapy. Nat. Biotechnol. 38, 947–953 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  93. Tang, H. et al. Facilitating T cell infiltration in tumor microenvironment overcomes resistance to PD-L1 blockade. Cancer Cell 29, 285–296 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  94. Tumeh, P. C. et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 515, 568–571 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  95. Rafiq, S., Hackett, C. S. & Brentjens, R. J. Engineering strategies to overcome the current roadblocks in CAR T cell therapy. Nat. Rev. Clin. Oncol. 17, 147–167 (2020).

    Article  PubMed  Google Scholar 

  96. Srivastava, S. & Riddell, S. R. Chimeric antigen receptor T cell therapy: challenges to bench-to-bedside efficacy. J. Immunol. 200, 459–468 (2018).

    Article  PubMed  CAS  Google Scholar 

  97. Park, S. E., Georgescu, A., Oh, J. M., Kwon, K. W. & Huh, D. Polydopamine-based interfacial engineering of extracellular matrix hydrogels for the construction and long-term maintenance of living three-dimensional tissues. ACS Appl. Mater. Interfaces 11, 23919–23925 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. Moon, E. K. et al. Expression of a functional CCR2 receptor enhances tumor localization and tumor eradication by retargeted human T cells expressing a mesothelin-specific chimeric antibody receptor. Clin. Cancer Res. 17, 4719–4730 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  99. Satija, R., Farrell, J. A., Gennert, D., Schier, A. F. & Regev, A. Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol. 33, 495–502 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  100. Aran, D. et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat. Immunol. 20, 163–172 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  101. Trapnell, C. et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 32, 381–386 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  102. Cao, J. et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature 566, 496–502 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  103. Vento-Tormo, R. et al. Single-cell reconstruction of the early maternal–fetal interface in humans. Nature 563, 347–353 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  104. Gu, Z., Gu, L., Eils, R., Schlesner, M. & Brors, B. circlize implements and enhances circular visualization in R. Bioinformatics 30, 2811–2812 (2014).

    Article  PubMed  CAS  Google Scholar 

  105. FRENS, G. Controlled nucleation for the regulation of the particle size in monodisperse gold suspensions. Nat. Phys. Sci. 241, 20–22 (1973).

    Article  CAS  Google Scholar 

  106. Chen, G. et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature 560, 382–386 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  107. Chambers, M. C. et al. A cross-platform toolkit for mass spectrometry and proteomics. Nat. Biotechnol. 30, 918–920 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  108. Agrawal, S. et al. EL-MAVEN: A fast, robust, and user-friendly mass spectrometry data processing engine for metabolomics. Methods Mol. Biol. 1978, 301–321 (2019).

    Article  PubMed  CAS  Google Scholar 

  109. Pang, Z. et al. Using MetaboAnalyst 5.0 for LC–HRMS spectra processing, multi-omics integration and covariate adjustment of global metabolomics data. Nat. Protoc. 2022 17:8 17, 1735–1761 (2022).

    CAS  Google Scholar 

  110. Liu, H. & Huh, D. D. Microengineered transplantation of human solid tumors for in vitro studies of CAR T immunotherapy. GSE240121. Gene Expression Omnibus. https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE240121 (2025).

  111. Liu, H. et al. Microengineered transplantation of human solid tumors for in vitro studies of CAR T immunotherapy. Dryad https://doi.org/10.5061/dryad.mw6m9068f (2025).

  112. Muri, J. et al. The thioredoxin-1 system is essential for fueling DNA synthesis during T-cell metabolic reprogramming and proliferation. Nat. Commun. 9, 1851 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  113. Weinreich, M. A. et al. KLF2 transcription-factor deficiency in T cells results in unrestrained cytokine production and upregulation of bystander chemokine receptors. Immunity 31, 122–130 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  114. Hayashi, K., Jutabha, P., Endou, H., Sagara, H. & Anzai, N. LAT1 Is a critical transporter of essential amino acids for immune reactions in activated human T cells. J. Immunol. 191, 4080–4085 (2013).

    Article  PubMed  CAS  Google Scholar 

  115. Tenbrock, K., Juang, Y.-T., Tolnay, M. & Tsokos, G. C. The cyclic adenosine 5′-monophosphate response element modulator suppresses IL-2 production in stimulated T cells by a chromatin-dependent mechanism. J. Immunol. 170, 2971–2976 (2003).

    Article  PubMed  CAS  Google Scholar 

  116. Rajah, R., Valentinis, B. & Cohen, P. Insulin-like growth factor (IGF)-binding protein-3 induces apoptosis and mediates the effects of transforming growth factor-β1 on programmed cell death through a p53- and IGF-independent mechanism. J. Biol. Chem. 272, 12181–12188 (1997).

    Article  PubMed  CAS  Google Scholar 

  117. Stein, S. et al. NDRG1 is necessary for p53-dependent apoptosis. J. Biol. Chem. 279, 48930–48940 (2004).

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

We thank M. Luo, A. Georgescu, J. M. Oh, N. Matisioudis, A. C. Huang, J. E. Wu, J.-C. Beltra, J. R. Giles, C. Alanio, D. Oldridge, E. Niemeyer, Y. C. Choi and C. R. Kagan for their intellectual input and technical assistance. We also thank R. Pellegrino Da Silva, F. Mafra, J. M. Smiler, J. P. Garifallou and M. V. Gonzalez from the Center for Applied Genomics at The Children’s Hospital of Philadelphia, and J. Schug and O. Smirnova from the Next-Generation Sequencing Core at the University of Pennsylvania for their contributions to scRNA-seq; T. Skinner and D. Martinez from the Pathology Core Laboratory at The Children’s Hospital of Philadelphia for their technical assistance in immunohistochemistry; and S. Singhal, M. Culligan and J. Friedberg from the Hospital of the University of Pennsylvania and the Temple University Hospital for their help with mesothelioma patient explants. This work was supported by the Cancer Research Institute (DDH), the National Institutes of Health (NIH) (1DP2HL127720-01 to D.D.H.; F32DK127843 to W.D.L.; R01CA163591 and DP1DK113643 to J.D.R.), the National Science Foundation (CMMI:15-48571) (D.D.H.), the Ministry of Trade, Industry & Energy of the Republic of Korea (D.D.H.), the GRDC Cooperative Hub through the National Research Foundation of Korea funded by the Ministry of Science and ICT (RS-2023-00259341) (T.K., D.D.H.), the Paul Allen Foundation (J.D.R.), Ludwig Cancer Research (J.D.R.) and the University of Pennsylvania. H.L. is a recipient of the Penn Institute for Regenerative Medicine Postdoctoral Fellowship and the Penn Center for Engineering MechanoBiology Pilot Grant Award.

Author information

Authors and Affiliations

Authors

Contributions

H.L. designed the research, performed most of the experiments, analyzed the data, created the figures and wrote the manuscript. E.N.-O., X.S., M.L., M.C.M., J.J.B., S.K., E.K.M. and S.M.A. provided materials, performed flow cytometry experiments and analyzed the data. Z.C. and E.J.W. provided assistance in the design of research and the analysis of the scRNA-seq data. W.D.L. and J.D.R. performed the experiments and provided assistance in the analysis of the metabolomics data. X.D., J.C., Y.L., A.W., Z.O., J.P., J.Y.P., A.L. and H.H. provided assistance in the experiments. S.A.A., Y.S., D.M.R., S.K., G.S.W., W.G. and T.K. provided assistance in the experiments and analyzed the data. Z.X. and E.P. provided materials and analyzed the data. D.D.H. designed the research, analyzed the data and wrote the manuscript.

Corresponding author

Correspondence to Dan Dongeun Huh.

Ethics declarations

Competing interests

D.D.H. is a co-founder of Vivodyne and holds equity in the company. D.D.H. and H.L. are inventors on a patent application for tumor-on-a-chip technology. E.J.W. holds equity and has other ownership interests in Arseal Bio, Danger Bio and Surface Oncology. E.J.W. has consulting or advisory role at Danger Bio, Jaenssen, Marengo Therapeutics, NewLimit, Pluto Immunotherapeutics, Related Sciences, Santa Ana Bio, Surface Oncology and Synthekine. S.M.A. is a scientific founder and holds equity in Capstan Therapeutics. S.M.A. is on the scientific advisory boards of Verismo and Bio4t2. E.P. is a scientific founder and holds equity in Capstan Therapeutics. E.P. is on the scientific advisory boards of Parthenon Therapeutics and POINT Biopharma. E.P. is an inventor (University of Pennsylvania) on a patent (10329355) and patent application for the 4G5 FAP CAR (Patent Applications 20210087294 and 20210087295). E.P. is an inventor (University of Pennsylvania) on a patent for the use of CAR-T therapy in heart disease (US Provisional Patent Application 62/563,323 filed 26 September 2017, WIPO Patent Application PCT/US2018/052605). J.D.R. is an advisor and stockholder in Colorado Research Partners, L.E.A.F. Pharmaceuticals, Bantam Pharmaceuticals, Barer Institute and Rafael Pharmaceuticals; a paid consultant of Pfizer and Third Rock Ventures; a founder, director, and stockholder of Farber Partners, Serien Therapeutics and Sofro Pharmaceuticals; a founder and stockholder in Empress Therapeutics; inventor of patents held by Princeton University; and a director of the Princeton University-PKU Shenzhen collaboration. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Biotechnology thanks Michael Birnbaum, Justin Eyquem, Gordana Vunjak-Novakovic 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.

Extended data

Extended Data Fig. 1 Single-cell trajectory analysis of CAR-T and tumor cells in the meso-tumor model.

a, UMAP plot showing CAR-T cell subpopulations in the meso-tumor model re-clustered for trajectory analysis. b, Transition of CAR-T cell phenotype along the pseudotime trajectory. c-h, Pseudotime dynamics of key marker expression plotted by cell type (c,e,g) and location (d,f,h). c,d, Biphasic regulation of gene markers associated with T cell activation (for example, GZMB, CCL3, IL2RA, IFNG). These markers begin to increase with the emergence of early activated CAR-T cells in the stroma and peak when the cells exhibit the phenotype of proliferative effector T cells within the tumors, which is followed by a plateau or gradual decrease with the progression of the cytotoxic activities of the CAR-T cells while they are still in the tumors. e,f, The activated effector CAR-T cells in the tumors show monotonically increasing expression of stress markers, such as AGR2, IGFBP1, S100P, and SPP1, demonstrating exposure and responses to increasing cellular stress in the CAR-T cells as they gain effector function, enter the tumor environment, and engage cancer cells. g,h, Among transcription factors correlated with the phenotypic changes of CAR-T cells are KLF2 and TXNIP. These markers exhibit almost monotonically decreasing expression, suggesting that their downregulation may be required for antigenic activation of CAR-T cells due to their established roles in regulating T cell effector functions and glucose consumption112,113. Increased expression of CREM and DDIT3 is closely associated with the transition of CAR-T cells towards effector lineages, consistent with their reported role as negative regulators of effector functions114,115. i, UMAP plots of tumor cell clusters in the meso-tumor model (left) and pseudotime trajectories (right). j,k, Dynamic expression of select markers by tumor cells over pseudotime. At the beginning of the pseudotime trajectory, the cells show the highest expression of genes activated by IFN-É£ (for example, STAT1, WARS), verifying their phenotype as tumor cells stimulated by effector CAR-T cell-produced IFN-É£. These genes show a cycle of downregulation and subsequent recovery, which is reversed in SERPINA1, FOS, and other transcription factors associated with survival promotion. Another notable feature is the induction of genes implicated in tumor cell apoptosis or inhibition of cancer progression (for example, MTRNR2L8, NDRG1)116,117 and their continuous increase prior to entering the apoptotic states, illustrating persistent deleterious effects of tumor-infiltrating CAR-T cells. Transcriptomic signatures of tumor cells also include markers that promote tumor development and progression (for example, MUC5AC, NEAT1), which may be interpreted as a mechanism to protect/recover stressed and apoptotic tumor cells affected by CAR-T cells. Increased induction of these genes is indeed visible at the late stages of phenotypic transition as the tumor cells move towards the apoptotic state.

Extended Data Fig. 2 Analysis of ligand-receptor interactions in a model of human lung adenocarcinoma tumors infused with meso-CAR-T cells.

a, Chord diagrams showing an overview of intercellular communication and the number of identified interactions in the meso-tumor (left) and control (middle) groups. Each chord represents a bundle of paired and statistically significant ligand-receptor interactions between a particular pair of cell types (right). The width of the chord indicates the number of interacting ligand-receptor pairs. A random-permutation test was used in the CellPhoneDB method to calculate empirical p values. b-e, Chord diagrams of cell pair-specific ligand-receptor interactions with p-values < 0.05 and total mean expression > 0.35. A random-permutation test was used in the CellPhoneDB method to calculate empirical p values.

Extended Data Fig. 3 Pharmacological inhibition of CAR-T-endothelial interactions mediated by CD38-PECAM1 signaling.

a, Visualization of the interacting pairs of ITGAL-ICAM1 and CD38-PECAM1 in the meso-tumor model (left) and violin plots comparing the expression of interacting genes of interest mediating the crosstalk between CAR-T cells and endothelial cells (right). n = 2574 CAR-T cells and 306 endothelial cells in the Meso group, and 3832 CAR-T cells and 1278 endothelial cells in the Control group. Boxplots overlaid show minimum, 25th percentile, mean (black line), 75th percentile, and maximum. b, Experimental timeline for CAR-T cell infusion and drug treatment. c, Representative fluorescence micrographs of single meso-tumors infused with meso-CAR-T cells without drug treatment (Untreated), treated with Daratumumab (0.5 or 10 μg/ml), or IgG isotype control antibody (10 μg/ml IgG). Blood vessels are not shown in these images. Scale bars, 250 μm. d,e, Quantification of T cell area (d) and normalized tumor area (e) over time. Data are presented as mean ± s.e.m. In d, n = 5 independent engineered tissue constructs for the untreated-meso-CAR-T only group; n = 6 independent engineered tissue constructs for the other groups treated with Daratumumab or IgG isotype control. One-way ANOVA with Tukey’s multiple comparisons test was used for statistical analysis. In e, n = 3 independent engineered tissue constructs at all time points for all treatment groups. Two-way ANOVA with Tukey’s multiple comparisons test was used for statistical analysis. Dara, daratumumab. CAR-T cells derived from one healthy donor were tested. These cells were administered at 2.5×105 cells per device.

Source data

Extended Data Fig. 4 Pharmacological inhibition of CAR-T-endothelial interactions mediated by LTB-LTBR signaling.

a, Visualization of the interacting LTB-LTBR pair in the meso-tumor model (left) and violin plots comparing the expression of interacting genes of interest mediating the crosstalk between CAR-T cells and endothelial cells (right). n = 2574 CAR-T cells and 306 endothelial cells in the Meso group, and 3832 CAR-T cells and 1278 endothelial cells in the Control group. Boxplots overlaid show minimum, 25th percentile, mean (black line), 75th percentile, and maximum. b, Experimental timeline for CAR-T cell infusion and drug treatment. c, Representative fluorescence micrographs of single meso-tumors infused with meso-CAR-T cells without drug treatment (Untreated), treated with Baminercept (0.5 or 10 μg/ml), or IgG isotope control antibody (10 μg/ml IgG). Blood vessels are not shown in these images. Scale bars, 250 μm. d,e, Quantification of (d) T cell area and (e) normalized tumor area over time. Data are presented as mean ± s.e.m. In d, n = 5 independent engineered tissue constructs for the group without drug treatment (Untreated) and the group treated with low concentration of Baminercept; n = 4 independent engineered tissue constructs for the group treated with high concentration of Baminercept; n = 6 independent engineered tissue constructs for the group treated with IgG isotype control. One-way ANOVA with Tukey’s multiple comparisons test was used for statistical analysis. In e, n = 3 independent engineered tissue constructs at all time points for all treatment groups. Two-way ANOVA with Tukey’s multiple comparisons test was used for statistical analysis. Bami, baminercept. CAR-T cells derived from one healthy donor were tested. These cells were administered at 2.5×105 cells per device.

Source data

Extended Data Fig. 5 Discovery of therapy biomarkers through metabolomic analysis of tumor-on-a-chip.

a, Workflow of untargeted, global metabolomic analysis using vascular perfusate from our meso-CAR-T cell-infused meso-tumor model treated with LAF237. Created with Biorender.com. The goal of this analysis was to identify metabolic signatures that correlate with the therapeutic outcome of the DPP4 inhibition method. Specific conditions considered included media only, pre-infusion, and CAR-T cell infusion groups with no LAF237 treatment (ctrl), 50 nM LAF237 (low), and 1,000 nM LAF237 (high) – examined at days 2, 7, 11, and 16 post initial infusion (Supplementary Table 2). b, Plot of partial least squares discriminant analysis (PLS-DA) scores. N = 4 independent engineered tissue constructs for each group. Our analysis revealed 205 differentially regulated metabolites (Supplementary Fig. 29). PLS-DA was performed to identify a fraction of these metabolites (FDR < 0.05) that can distinguish the tested conditions at different time points (Supplementary Fig. 30). c, Heatmap of 96 metabolites identified by multivariate analysis whose levels of expression were significantly changed by LAF237 treatment. 65 of these were upregulated and the other 31 metabolites were downregulated in LAF237-treated groups, compared to CAR-T cell-only control. N = 4 independent engineered tissue constructs for each group. The source of metabolites is color-coded in their labels – blue, amino acid metabolism; red, carbohydrate metabolism; green, nucleotide metabolism. The color gradient of the scale bar indicates the relative abundance of metabolites, with red and blue indicating higher and lower concentrations, respectively. Two-way ANOVA was used for statistical analysis, with multiple testing correction using false discovery rate. p < 0.05 was considered significant. d, Plot of significantly changed metabolic pathways identified by pathway impact analysis of significantly upregulated metabolites in the high-dose LAF237 group (Supplementary Fig. 31). The upregulated metabolites had significant effects on beta-alanine metabolism, citrate cycle, and other key processes, suggesting substantial changes in central carbon metabolism. Hypergeometric test was used as the enrichment method, with false discovery rate adjustment for significance. e-g, Comparison of normalized concentrations of select metabolites across all conditions. One-way ANOVA with Fisher’s LSD post-hoc test was used for statistical analysis. Key findings include elevated levels of carbohydrate metabolism products, such as cis-aconitic acid in the TCA cycle, at day 16 with 1,000 nM LAF237. This high-dose treatment also correlated with increased amino acid and nucleic acid metabolism, shown by higher levels of xanthine, L-proline, fumaric acid, and beta-alanine. Pyruvic acid levels were negatively correlated with drug treatment. h, Receiver operating characteristic (ROC) curves for biomarker prediction models to identify metabolites that can differentiate more efficacious CAR-T cell treatment with high-dose LAF237 from control CAR-T cell treatment without LAF237 at day 16 post initial infusion (Supplementary Fig. 32). Different numbers of constituent metabolite features per model are indicated by different colors. i, List of top metabolites yielded by the 25-feature prediction model for day 16 shown in h and ranked according to their predictive accuracy. j, Comparison of normalized concentrations of metabolites selected from i. One-way ANOVA with Fisher’s LSD post-hoc test was used for statistical analysis. Higher CAR-T therapy efficacy with 1,000 nM LAF237 was predicted to significantly correlate with reduced production of 1,2,3-propanetricarboxylic acid, 2-hydroxybutyric acid, 3-acetyl-2,7-naphthyridine, L-cystine and increased production of 3-nitrotyrosine and 2-hydroxyethanesulfonate at day 16 post initial infusion (Supplementary Fig. 33). k, ROC curves for biomarker prediction models to identify metabolites that can differentiate more efficacious CAR-T cell treatment with high-dose LAF237 from control treatment at all time points post-infusion. l, List of top metabolites yielded by the 15-feature prediction model shown in k and ranked according to their predictive accuracy. The predictions shown in i and l shared metabolites (labeled blue) that were upregulated (5-methoxytryptophan, ibudilast, vanilpyruvic acid) or downregulated (2-hydroxybutyric acid, inosine, glutaminylglutamine, 2-aminoacrylic acid) due to administration of 1,000 nM LAF237 during CAR-T cell infusion (Supplementary Fig. 33). m, Comparison of normalized concentrations of metabolites selected from l. One-way ANOVA with Fisher’s LSD post-hoc test was used for statistical analysis. Boxplots show minimum, 25th percentile, mean (yellow closed diamond), 75th percentile, and maximum. N = 4 independent engineered tissue constructs for each group. Note that post-hoc pairwise comparison was calculated between the high-dose (high) and control (ctrl) groups at all time points with *p < 0.05.

Supplementary information

Supplementary Information

Supplementary Figures 1–33.

Reporting Summary

Supplementary Table

Supplementary Table 1: List of significant interactions in CellPhoneDB analysis. Supplementary Table 2: Peak intensities of metabolites. Supplementary Table 3: Summary of MSEA and pathway impact analysis.

Supplementary Video 1

3D reconstruction of blood vessels wrapping around a tumor transplant in the device.

Supplementary Video 2

3D reconstruction of blood vessels penetrating a tumor transplant in the device.

Supplementary Video 3

Visualization of the perfusability of vascularized human lung tumor explants using 75- kDa FITC-Dextran.

Supplementary Video 4

Visualization of the perfusability of vascularized human lung tumor explants using 1-µm fluorescent microbeads.

Supplementary Video 5

Visualization of the perfusability of vascularized lung tumor spheroids using 1-µm fluorescent microbeads.

Supplementary Video 6

Comparison of initial CAR-T cell infusion between the meso-tumor and control groups.

Supplementary Video 7

Extravasation and directional migration of CAR-T cells in the meso-tumor model.

Supplementary Video 8

Behavior of CAR-T cells in the control tumor group.

Supplementary Video 9

Time-lapse imaging of CAR-T cell-infused lung tumor growth without LAF237.

Supplementary Video 10

Time-lapse imaging of CAR T cell-infused lung tumor growth treated with 50 nM of LAF237.

Supplementary Video 11

Time-lapse imaging of CAR T cell-infused lung tumor growth treated with 1000 nM of LAF237.

Source data

Source Data Figures 1–6 and Extended Data Figures 3–4

Statistical source data for Fig. 1 f Branch length and 1 f Vessel diameter. Statistical source data for Figs. 2i, 2j, 2k, 2p, 2q IL-2, 2q IFN-gamma, 2r Caspase3, and 2r LDH. Statistical source data for Figs. 3b, 3c, 3 d percentage of effector memory CAR T cells, and 3 d numbers of effector memory CAR T cells. Statistical source data for Figs. 4e, 4 f, 4i, 4j, 4k, 4 l, and 4 m. Statistical source data for Fig. 5c Vessel area, 5c Branch length, 5c Number of junctions, 5c Vessel diameter, 5e, 5 f, 5 g number of Tem cells, 5 g number of Tcm cells, 5 h, 5i, 5j, and 5k. Statistical source data for Fig. 6i Tumor area, 6i T cell area, 6j CXCL10, 6j CXCL11, and 6k. Statistical source data for Extended Data Figs. 3 d and 3e. Statistical source data for Extended Data Figs. 4 d and 4e.

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

Liu, H., Noguera-Ortega, E., Dong, X. et al. A tumor-on-a-chip for in vitro study of CAR-T cell immunotherapy in solid tumors. Nat Biotechnol (2025). https://doi.org/10.1038/s41587-025-02845-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41587-025-02845-z

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer