Abstract
The development and wider adoption of adoptive cell therapies is constrained by complex and costly manufacturing processes and by inconsistent efficacy across patients. Here we discuss how microfluidic and other fluidic devices can be implemented at each stage of cell manufacturing for adoptive cell therapies, from the harvesting and isolation of the cells to their editing, culturing and functional selection. We suggest that precise and controllable microfluidic systems can streamline the development of these therapies by offering scalability in cell production, bolstering the efficacy and predictability of the therapies and improving their cost-effectiveness and accessibility for broader populations of patients with cancer.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$29.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 digital issues and online access to articles
$119.00 per year
only $9.92 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout
Similar content being viewed by others
References
Rosenberg, S., Spiess, P. & Lafreniere, R. A new approach to the adoptive immunotherapy of cancer with tumor-infiltrating lymphocytes. Science 233, 1318–1321 (1986).
Rosenberg, S. A. Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. N. Engl. J. Med. 319, 1676–1680 (1988).
Gross, G., Waks, T. & Eshhar, Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc. Natl Acad. Sci. USA 86, 10024–10028 (1989).
Sheykhhasan, M., Manoochehri, H. & Dama, P. Use of CAR T-cell for acute lymphoblastic leukemia (ALL) treatment: a review study. Cancer Gene Ther. 29, 1080–1096 (2022).
World’s first TIL therapy approved. Nat. Biotechnol. 42, 347–350 (2024).
Creelan, B. C. et al. Tumor-infiltrating lymphocyte treatment for anti-PD-1-resistant metastatic lung cancer: a phase 1 trial. Nat. Med. 27, 1410–1418 (2021).
Mariuzza, R. A., Agnihotri, P. & Orban, J. The structural basis of T-cell receptor (TCR) activation: an enduring enigma. J. Biol. Chem. 295, 914–925 (2020).
Puig-Saus, C. et al. Neoantigen-targeted CD8+ T cell responses with PD-1 blockade therapy. Nature 615, 697–704 (2023).
Wang, Z. & Chen, K. Understanding the heterogeneous immune repertoire of brain metastases for designing next-gen therapeutics. Brain-X 33, e33 (2023).
Stanojevic, M. et al. Identification of novel HLA-restricted preferentially expressed antigen in melanoma peptides to facilitate off-the-shelf tumor-associated antigen-specific T-cell therapies. Cytotherapy 23, 694–703 (2021).
Li, J. et al. The screening, identification, design and clinical application of tumor-specific neoantigens for TCR-T cells. Mol. Cancer 22, 141 (2023).
Sterner, R. C. & Sterner, R. M. CAR-T cell therapy: current limitations and potential strategies. Blood Cancer J. 11, 69 (2021).
Pan, K. et al. CAR race to cancer immunotherapy: from CAR T, CAR NK to CAR macrophage therapy. J. Exp. Clin. Cancer Res. 41, 119 (2022).
Akatsuka, Y. TCR-like CAR-T cells targeting MHC-bound minor histocompatibility antigens. Front. Immunol. 11, 257 (2020).
DiNofia, A. M. & Grupp, S. A. Will allogeneic CAR T cells for CD19+ malignancies take autologous CAR T cells ‘off the shelf’? Nat. Rev. Clin. Oncol. 18, 195–196 (2021).
Depil, S., Duchateau, P., Grupp, S. A., Mufti, G. & Poirot, L. ‘Off-the-shelf’ allogeneic CAR T cells: development and challenges. Nat. Rev. Drug Discov. 19, 185–199 (2020).
Caldwell, K. J., Gottschalk, S. & Talleur, A. C. Allogeneic CAR cell therapy—more than a pipe dream. Front. Immunol. 11, 618427 (2021).
Lv, Z., Luo, F. & Chu, Y. Strategies for overcoming bottlenecks in allogeneic CAR-T cell therapy. Front. Immunol. 14, 1199145 (2023).
Elsallab, M. & Maus, M. V. Expanding access to CAR T cell therapies through local manufacturing. Nat. Biotechnol. 41, 1698–1708 (2023).
Mock, U. et al. Automated manufacturing of chimeric antigen receptor T cells for adoptive immunotherapy using CliniMACS Prodigy. Cytotherapy 18, 1002–1011 (2016).
Aleksandrova, K. et al. Functionality and cell senescence of CD4/CD8-selected CD20 CAR T cells manufactured using the automated CliniMACS Prodigy® platform. Transfus. Med. Hemotherapy 46, 47–54 (2019).
Shah, M., Krull, A., Odonnell, L., De Lima, M. J. & Bezerra, E. Promises and challenges of a decentralized CAR T-cell manufacturing model. Front. Transplant. 2, 1238535 (2023).
Blache, U., Popp, G., Dünkel, A., Koehl, U. & Fricke, S. Potential solutions for manufacture of CAR T cells in cancer immunotherapy. Nat. Commun. 13, 5225 (2022).
Ran, T., Eichmuller, S. B., Schmidt, P. & Schlander, M. Cost of decentralized CAR T-cell production in an academic nonprofit setting. Int. J. Cancer 147, 3438–3445 (2020).
Sackmann, E. K., Fulton, A. L. & Beebe, D. J. The present and future role of microfluidics in biomedical research. Nature 507, 181–189 (2014).
Adriani, G. et al. Microfluidic models for adoptive cell-mediated cancer immunotherapies. Drug Discov. Today 21, 1472–1478 (2016).
Paterson, K., Zanivan, S., Glasspool, R., Coffelt, S. B. & Zagnoni, M. Microfluidic technologies for immunotherapy studies on solid tumours. Lab. Chip 21, 2306–2329 (2021).
Kim, H., Kim, S., Lim, H. & Chung, A. J. Expanding CAR-T cell immunotherapy horizons through microfluidics. Lab. Chip 24, 1088–1120 (2024).
van den Berg, J. H. et al. Tumor infiltrating lymphocytes (TIL) therapy in metastatic melanoma: boosting of neoantigen-specific T cell reactivity and long-term follow-up. J. Immunother. Cancer 8, e000848 (2020).
Baldan, V., Griffiths, R., Hawkins, R. E. & Gilham, D. E. Efficient and reproducible generation of tumour-infiltrating lymphocytes for renal cell carcinoma. Br. J. Cancer 112, 1510–1518 (2015).
Kongkaew, T. et al. TIL expansion with high dose IL-2 or low dose IL-2 with anti-CD3/anti-CD28 stimulation provides different quality of TIL-expanded T cell clones. J. Immunol. Methods 503, 113229 (2022).
Qiu, X. et al. Microfluidic channel optimization to improve hydrodynamic dissociation of cell aggregates and tissue. Sci. Rep. 8, 2774 (2018).
Lombardo, J. A., Aliaghaei, M., Nguyen, Q. H., Kessenbrock, K. & Haun, J. B. Microfluidic platform accelerates tissue processing into single cells for molecular analysis and primary culture models. Nat. Commun. 12, 2858 (2021).
Lutz, E. R. et al. Superior efficacy of CAR-T cells using Marrow-Infiltrating Lymphocytes (MILsTM) as compared to Peripheral Blood Lymphocytes (PBLs). Blood 134, 4437 (2019).
Kröger, N. et al. (eds) The EBMT/EHA CAR-T Cell Handbook (Springer, 2022).
Jeon, H. et al. Fully-automated and field-deployable blood leukocyte separation platform using multi-dimensional double spiral (MDDS) inertial microfluidics. Lab. Chip 20, 3612–3624 (2020).
Zeming, K. K., Salafi, T., Chen, C.-H. & Zhang, Y. Asymmetrical deterministic lateral displacement gaps for dual functions of enhanced separation and throughput of red blood cells. Sci. Rep. 6, 22934 (2016).
Lezzar, D. L. et al. A high-throughput microfluidic device based on controlled incremental filtration to enable centrifugation-free, low extracorporeal volume leukapheresis. Sci. Rep. 12, 13798 (2022).
Mishra, A. et al. Ultrahigh-throughput magnetic sorting of large blood volumes for epitope-agnostic isolation of circulating tumor cells. Proc. Natl Acad. Sci. USA 117, 16839–16847 (2020).
Lin, E. et al. High-throughput microfluidic labyrinth for the label-free isolation of circulating tumor cells. Cell Syst. 5, 295–304.e4 (2017).
Sarioglu, A. F. et al. A microfluidic device for label-free, physical capture of circulating tumor cell clusters. Nat. Methods 12, 685–691 (2015).
Renier, C. et al. Label-free isolation of prostate circulating tumor cells using Vortex microfluidic technology. Npj Precis. Oncol. 1, 15 (2017).
Zhu, S., Jiang, F., Han, Y., Xiang, N. & Ni, Z. Microfluidics for label-free sorting of rare circulating tumor cells. Analyst 145, 7103–7124 (2020).
Hao, S.-J., Wan, Y., Xia, Y.-Q., Zou, X. & Zheng, S.-Y. Size-based separation methods of circulating tumor cells. Adv. Drug Deliv. Rev. 125, 3–20 (2018).
Urbanska, M. et al. A comparison of microfluidic methods for high-throughput cell deformability measurements. Nat. Methods 17, 587–593 (2020).
Lenshof, A. & Laurell, T. Continuous separation of cells and particles in microfluidic systems. Chem. Soc. Rev. 39, 1203–1217 (2010).
Di Carlo, D., Irimia, D., Tompkins, R. G. & Toner, M. Continuous inertial focusing, ordering and separation of particles in microchannels. Proc. Natl Acad. Sci. USA 104, 18892–18897 (2007).
Xiang, N. & Ni, Z. High-throughput blood cell focusing and plasma isolation using spiral inertial microfluidic devices. Biomed. Microdevices 17, 110 (2015).
Mutlu, B. R. et al. Non-equilibrium inertial separation array for high-throughput, large-volume blood fractionation. Sci. Rep. 7, 9915 (2017).
Gomis, S. et al. Single-cell tumbling enables high-resolution size profiling of retinal stem cells. ACS Appl. Mater. Interfaces 10, 34811–34816 (2018).
Sadeqi Nezhad, M., Abdollahpour-Alitappeh, M., Rezaei, B., Yazdanifar, M. & Seifalian, A. M. Induced pluripotent stem cells (iPSCs) provide a potentially unlimited T cell source for CAR-T cell development and off-the-shelf products. Pharm. Res. 38, 931–945 (2021).
Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676 (2006).
Nakagawa, M. et al. Generation of induced pluripotent stem cells without Myc from mouse and human fibroblasts. Nat. Biotechnol. 26, 101–106 (2008).
Guan, J. et al. Chemical reprogramming of human somatic cells to pluripotent stem cells. Nature 605, 325–331 (2022).
Park, I.-H., Lerou, P. H., Zhao, R., Huo, H. & Daley, G. Q. Generation of human-induced pluripotent stem cells. Nat. Protoc. 3, 1180–1186 (2008).
Mandal, P. K. & Rossi, D. J. Reprogramming human fibroblasts to pluripotency using modified mRNA. Nat. Protoc. 8, 568–582 (2013).
Ghaedi, M. & Niklason, L. E. in Organoids Vol. 1576 (ed. Turksen, K.) 55–92 (Springer, 2016).
Giulitti, S. et al. Direct generation of human naive induced pluripotent stem cells from somatic cells in microfluidics. Nat. Cell Biol. 21, 275–286 (2019).
Ashby, K. M. & Hogquist, K. A. A guide to thymic selection of T cells. Nat. Rev. Immunol. 24, 103–117 (2024).
Wang, Z. et al. 3D-organoid culture supports differentiation of human CAR+ iPSCs into highly functional CAR T cells. Cell Stem Cell 29, 515–527.e8 (2022).
Lipsitz, Y. Y., Timmins, N. E. & Zandstra, P. W. Quality cell therapy manufacturing by design. Nat. Biotechnol. 34, 393–400 (2016).
Fernández, A. et al. Optimizing the procedure to manufacture clinical-grade NK cells for adoptive immunotherapy. Cancers 13, 577 (2021).
Wang, Z. et al. Nanoparticle amplification labeling for high-performance magnetic cell sorting. Nano Lett. 22, 4774–4783 (2022).
Sutermaster, B. A. & Darling, E. M. Considerations for high-yield, high-throughput cell enrichment: fluorescence versus magnetic sorting. Sci. Rep. 9, 227 (2019).
Binek, A. et al. Flow cytometry has a significant impact on the cellular metabolome. J. Proteome Res. 18, 169–181 (2019).
Riddell, A., Gardner, R., Perez-Gonzalez, A., Lopes, T. & Martinez, L. Rmax: a systematic approach to evaluate instrument sort performance using center stream catch. Methods 82, 64–73 (2015).
Pfister, G., Toor, S. M., Sasidharan Nair, V. & Elkord, E. An evaluation of sorter induced cell stress (SICS) on peripheral blood mononuclear cells (PBMCs) after different sort conditions - are your sorted cells getting SICS? J. Immunol. Methods 487, 112902 (2020).
Faraghat, S. A. et al. High-throughput, low-loss, low-cost, and label-free cell separation using electrophysiology-activated cell enrichment. Proc. Natl Acad. Sci. USA 114, 4591–4596 (2017).
Wu, M. et al. Acoustofluidic separation of cells and particles. Microsyst. Nanoeng. 5, 32 (2019).
Nawaz, A. A. et al. Acoustofluidic fluorescence activated cell sorter. Anal. Chem. 87, 12051–12058 (2015).
Li, P. & Ai, Y. Label-free multivariate biophysical phenotyping-activated acoustic sorting at the single-cell level. Anal. Chem. 93, 4108–4117 (2021).
Staunstrup, N. H. et al. Comparison of electrostatic and mechanical cell sorting with limited starting material. Cytom. A 101, 298–310 (2022).
Green, B. J. et al. PillarX: a microfluidic device to profile circulating tumor cell clusters based on geometry, deformability and epithelial state. Small 18, 2106097 (2022).
Ocanas, R. et al. Minimizing the ex vivo confounds of cell-isolation techniques on transcriptomic-profiles of purified microglia. Preprint at https://doi.org/10.1101/2021.07.15.452509 (2021).
Webb, C. et al. Using microfluidics for scalable manufacturing of nanomedicines from bench to GMP: a case study using protein-loaded liposomes. Int. J. Pharm. 582, 119266 (2020).
Nitta, N. et al. Intelligent image-activated cell sorting. Cell 175, 266–276.e13 (2018).
Zhao, L. et al. Deep learning of morphologic correlations to accurately classify CD4+ and CD8+ T cells by diffraction imaging flow cytometry. Anal. Chem. 94, 1567–1574 (2022).
Alvarez-Breckenridge, C. et al. Microenvironmental landscape of human melanoma brain metastases in response to immune checkpoint inhibition. Cancer Immunol. Res. 10, 996–1012 (2022).
Geens, M. et al. The efficiency of magnetic-activated cell sorting and fluorescence-activated cell sorting in the decontamination of testicular cell suspensions in cancer patients. Hum. Reprod. 22, 733–742 (2007).
Moore, D. K. et al. Isolation of B-cells using Miltenyi MACS bead isolation kits. PLoS ONE 14, e0213832 (2019).
Wang, Z., Sargent, E. H. & Kelley, S. O. Ultrasensitive detection and depletion of rare leukemic B cells in T cell populations via immunomagnetic cell ranking. Anal. Chem. 93, 2327–2335 (2021).
Mair, B. et al. High-throughput genome-wide phenotypic screening via immunomagnetic cell sorting. Nat. Biomed. Eng. 3, 796–805 (2019).
Aldridge, P. M. et al. Prismatic deflection of live tumor cells and cell clusters. ACS Nano 12, 12692–12700 (2018).
Wang, Z. et al. Efficient recovery of potent tumour-infiltrating lymphocytes through quantitative immunomagnetic cell sorting. Nat. Biomed. Eng. 6, 108–117 (2022).
Wang, Z. et al. Isolation of tumour-reactive lymphocytes from peripheral blood via microfluidic immunomagnetic cell sorting. Nat. Biomed. Eng. 7, 1188–1203 (2023).
Zhang, Y. et al. Interfacial polymerization produced magnetic particles with nano-filopodia for highly accurate liquid biopsy in the PSA gray zone. Adv. Mater. 35, e2303821 (2023).
Adams, J. D., Kim, U. & Soh, H. T. Multitarget magnetic activated cell sorter. Proc. Natl Acad. Sci. USA 105, 18165–18170 (2008).
Wang, Z. et al. Phenotypic targeting using magnetic nanoparticles for rapid characterization of cellular proliferation regulators. Sci. Adv. 10, eadj1468 (2024).
Dudley, M. E., Wunderlich, J. R., Shelton, T. E., Even, J. & Rosenberg, S. A. Generation of tumor-infiltrating lymphocyte cultures for use in adoptive transfer therapy for melanoma patients. J. Immunother. 26, 332–342 (2003).
Riddell, S. R. et al. Restoration of viral immunity in immunodeficient humans by the adoptive transfer of T cell clones. Science 257, 238–241 (1992).
Atsavapranee, E. S., Billingsley, M. M. & Mitchell, M. J. Delivery technologies for T cell gene editing: applications in cancer immunotherapy. EBioMedicine 67, 103354 (2021).
Vera, J. F. et al. Accelerated production of antigen-specific T cells for preclinical and clinical applications using gas-permeable rapid expansion cultureware (G-Rex). J. Immunother. 33, 305–315 (2010).
FDA Investigating Serious Risk of T-Cell Malignancy Following BCMA-Directed or CD19-Directed Autologous Chimeric Antigen Receptor (CAR) T Cell Immunotherapies (FDA, 2023); https://www.fda.gov/vaccines-blood-biologics/safety-availability-biologics/fda-investigating-serious-risk-t-cell-malignancy-following-bcma-directed-or-cd19-directed-autologous
Geng, T. & Lu, C. Microfluidic electroporation for cellular analysis and delivery. Lab Chip 13, 3803–3821 (2013).
Zhan, Y., Wang, J., Bao, N. & Lu, C. Electroporation of cells in microfluidic droplets. Anal. Chem. 81, 2027–2031 (2009).
Chakrabarty, P. et al. Microfluidic mechanoporation for cellular delivery and analysis. Mater. Today Bio 13, 100193 (2022).
Ding, X. et al. High-throughput nuclear delivery and rapid expression of DNA via mechanical and electrical cell-membrane disruption. Nat. Biomed. Eng. 1, 0039 (2017).
Toner, M. Gene delivery: suddenly squeezed and shocked. Nat. Biomed. Eng. 1, 0047 (2017).
Hur, J. et al. Microfluidic cell stretching for highly effective gene delivery into hard-to-transfect primary cells. ACS Nano 14, 15094–15106 (2020).
Hur, J. et al. Genetically stable and scalable nanoengineering of human primary T cells via cell mechanoporation. Nano Lett. 23, 7341–7349 (2023).
Li, X. et al. High-throughput and efficient intracellular delivery method via a vibration-assisted nanoneedle/microfluidic composite system. ACS Nano 17, 2101–2113 (2023).
Zhuo, C. et al. Spatiotemporal control of CRISPR/Cas9 gene editing. Signal Transduct. Target. Ther. 6, 238 (2021).
Leung, A. K. K., Tam, Y. Y. C., Chen, S., Hafez, I. M. & Cullis, P. R. Microfluidic mixing: a general method for encapsulating macromolecules in lipid nanoparticle systems. J. Phys. Chem. B 119, 8698–8706 (2015).
Billingsley, M. M. et al. In vivo mRNA CAR T cell engineering via targeted ionizable lipid nanoparticles with extrahepatic tropism. Small 20, 2304378 (2024).
Golubovskaya, V. et al. CAR-NK cells generated with mRNA-LNPs kill tumor target cells in vitro and in vivo. Int. J. Mol. Sci. 24, 13364 (2023).
Liu, Y. et al. In situ MUC1-specific CAR engineering of tumor-supportive macrophages stimulates tumoricidal immunity against pancreatic adenocarcinoma. Nano Today 49, 101805 (2023).
Alkan, F. & Lacin, N. A cationic stearamide-based solid lipid nanoparticle for delivering Yamanaka factors: evaluation of the transfection efficiency. ChemistryOpen 9, 1181–1189 (2020).
Lee, K. S., Boccazzi, P., Sinskey, A. J. & Ram, R. J. Microfluidic chemostat and turbidostat with flow rate, oxygen and temperature control for dynamic continuous culture. Lab. Chip 11, 1730–1739 (2011).
Mozdzierz, N. J. et al. A perfusion-capable microfluidic bioreactor for assessing microbial heterologous protein production. Lab. Chip 15, 2918–2922 (2015).
Kwon, T. et al. Microfluidic cell retention device for perfusion of mammalian suspension culture. Sci. Rep. 7, 6703 (2017).
Sin, W.-X. et al. A high-density microfluidic bioreactor for the automated manufacturing of CAR T cells. Nat. Biomed. Eng. https://doi.org/10.1038/s41551-024-01219-1 (2024).
Bohrer, L. R. et al. CGMP compliant microfluidic transfection of induced pluripotent stem cells for CRISPR-mediated genome editing. Stem Cells 41, 1037–1046 (2023).
Krishna, S. et al. Stem-like CD8 T cells mediate response of adoptive cell immunotherapy against human cancer. Science 370, 1328–1334 (2020).
Yossef, R. et al. Phenotypic signatures of circulating neoantigen-reactive CD8+ T cells in patients with metastatic cancers. Cancer Cell 41, 2154–2162.e5 (2023).
Siddiqui, I. et al. Intratumoral Tcf1+ PD-1+ CD8+ T cells with stem-like properties promote tumor control in response to vaccination and checkpoint blockade immunotherapy. Immunity 50, 195–211.e10 (2019).
Liu, B., Zhang, Y., Wang, D., Hu, X. & Zhang, Z. Single-cell meta-analyses reveal responses of tumor-reactive CXCL13+ T cells to immune-checkpoint blockade. Nat. Cancer 3, 1123–1136 (2022).
Greenman, R. et al. Shaping functional avidity of CAR T cells: affinity, avidity and antigen density that regulate response. Mol. Cancer Ther. 20, 872–884 (2021).
Zhou, Y. et al. Evaluation of single-cell cytokine secretion and cell-cell interactions with a hierarchical loading microwell chip. Cell Rep. 31, 107574 (2020).
Lei, K. et al. Cancer-cell stiffening via cholesterol depletion enhances adoptive T-cell immunotherapy. Nat. Biomed. Eng. 5, 1411–1425 (2021).
Waugh, R. E., Lomakina, E., Amitrano, A. & Kim, M. Activation effects on the physical characteristics of T lymphocytes. Front. Bioeng. Biotechnol. 11, 1175570 (2023).
Schmidt, R. et al. CRISPR activation and interference screens decode stimulation responses in primary human T cells. Science 375, eabj4008 (2022).
Labib, M. et al. Identification of druggable regulators of cell secretion via a kinome-wide screen and high-throughput immunomagnetic cell sorting. Nat. Biomed. Eng. https://doi.org/10.1038/s41551-023-01135-w (2023).
Rossi, J. et al. Preinfusion polyfunctional anti-CD19 chimeric antigen receptor T cells are associated with clinical outcomes in NHL. Blood 132, 804–814 (2018).
Xue, Q. et al. Single-cell multiplexed cytokine profiling of CD19 CAR-T cells reveals a diverse landscape of polyfunctional antigen-specific response. J. Immunother. Cancer 5, 85 (2017).
Bai, Z. et al. Single-cell antigen-specific landscape of CAR T infusion product identifies determinants of CD19-positive relapse in patients with ALL. Sci. Adv. 8, eabj2820 (2022).
De Rutte, J. et al. Suspendable hydrogel nanovials for massively parallel single-cell functional analysis and sorting. ACS Nano 16, 7242–7257 (2022).
Udani, S. et al. Associating growth factor secretions and transcriptomes of single cells in nanovials using SEC-seq. Nat. Nanotechnol. 19, 354–363 (2024).
Koo, D. et al. Defining T cell receptor repertoires using nanovial-based binding and functional screening. Proc. Natl Acad. Sci. USA 121, e2320442121 (2024).
Hoffmann, M. & Slansky, J. T-cell receptor affinity in the age of cancer immunotherapy. Mol. Carcinog. https://doi.org/10.1002/mc.23212 (2020).
Ashby, J. F. et al. Microfluidic T cell selection by cellular avidity. Adv. Healthc. Mater. 11, 2200169 (2022).
Cox, M. C. et al. Application of LDH assay for therapeutic efficacy evaluation of ex vivo tumor models. Sci. Rep. 11, 18571 (2021).
Bronevetsky, Y. Directly test individual T cell function with fewer cells on the berkeley lights lightningTM platform. Cytotherapy 22, S119–S120 (2020).
Valdez, J. et al. 184 Discovery, cloning and functional validation of a neoantigen specific patient derived TCR on the berkeley lights platform, with implications in personalized cancer immunotherapy. Regular and Young Investigator Award Abstracts A196–A196 (BMJ Publishing Group, 2022); https://doi.org/10.1136/jitc-2022-SITC2022.0184
Good, C. R. et al. An NK-like CAR T cell transition in CAR T cell dysfunction. Cell 184, 6081–6100.e26 (2021).
Luah, Y. H., Wu, T. & Cheow, L. F. Identification, sorting and profiling of functional killer cells via the capture of fluorescent target-cell lysate. Nat. Biomed. Eng. https://doi.org/10.1038/s41551-023-01089-z (2023).
Kirouac, D. C. et al. Deconvolution of clinical variance in CAR-T cell pharmacology and response. Nat. Biotechnol. 41, 1606–1617 (2023).
Mittelheisser, V. et al. Evidence and therapeutic implications of biomechanically regulated immunosurveillance in cancer and other diseases. Nat. Nanotechnol. https://doi.org/10.1038/s41565-023-01535-8 (2024).
Wu, P.-H. et al. A comparison of methods to assess cell mechanical properties. Nat. Methods 15, 491–498 (2018).
Gossett, D. R. et al. Hydrodynamic stretching of single cells for large population mechanical phenotyping. Proc. Natl Acad. Sci. USA 109, 7630–7635 (2012).
Otto, O. et al. Real-time deformability cytometry: on-the-fly cell mechanical phenotyping. Nat. Methods 12, 199–202 (2015).
Byun, S. et al. Characterizing deformability and surface friction of cancer cells. Proc. Natl Acad. Sci. USA 110, 7580–7585 (2013).
Tse, H. T. K. et al. Quantitative diagnosis of malignant pleural effusions by single-cell mechanophenotyping. Sci. Transl. Med. 5, 212ra163 (2013).
Nawaz, A. A. et al. Intelligent image-based deformation-assisted cell sorting with molecular specificity. Nat. Methods 17, 595–599 (2020).
Soteriou, D. et al. Rapid single-cell physical phenotyping of mechanically dissociated tissue biopsies. Nat. Biomed. Eng. 7, 1392–1403 (2023).
Jung, P., Zhou, X., Iden, S., Bischoff, M. & Qu, B. T cell stiffness is enhanced upon formation of immunological synapse. eLife 10, e66643 (2021).
Mhaidly, R. & Verhoeyen, E. Humanized mice are precious tools for preclinical evaluation of CAR T and CAR NK cell therapies. Cancers 12, 1915 (2020).
Yue, H. & Bai, L. Progress, implications and challenges in using humanized immune system mice in CAR-T therapy—application evaluation and improvement. Anim. Models Exp. Med. 7, 3–11 (2024).
Wang, Z. Assessing tumorigenicity in stem cell-derived therapeutic products: a critical step in safeguarding regenerative medicine. Bioengineering 10, 857 (2023).
Wu, L. et al. Organoids/organs-on-a-chip: new frontiers of intestinal pathophysiological models. Lab. Chip 23, 1192–1212 (2023).
Si, L. et al. A human-airway-on-a-chip for the rapid identification of candidate antiviral therapeutics and prophylactics. Nat. Biomed. Eng. 5, 815–829 (2021).
Bein, A. et al. Nutritional deficiency in an intestine-on-a-chip recapitulates injury hallmarks associated with environmental enteric dysfunction. Nat. Biomed. Eng. 6, 1236–1247 (2022).
Maulana, T. I. et al. Immunocompetent cancer-on-chip models to assess immuno-oncology therapy. Adv. Drug Deliv. Rev. 173, 281–305 (2021).
Peng, Y. & Lee, E. Microphysiological systems for cancer immunotherapy research and development. Adv. Biol 8, e2300077 (2023).
Ando, Y. et al. Evaluating CAR-T cell therapy in a hypoxic 3D tumor model. Adv. Healthc. Mater 8, e1900001 (2019).
Pavesi, A. et al. A 3D microfluidic model for preclinical evaluation of TCR-engineered T cells against solid tumors. JCI Insight 2, e89762 (2017).
Chouaib, S., Noman, M. Z., Kosmatopoulos, K. & Curran, M. A. Hypoxic stress: obstacles and opportunities for innovative immunotherapy of cancer. Oncogene 36, 439–445 (2017).
Lee, S. W. L. et al. Characterizing the role of monocytes in T cell cancer immunotherapy using a 3D microfluidic model. Front. Immunol. 9, 416 (2018).
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, eabc2331 (2021).
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).
Nguyen, O. T. P. et al. An immunocompetent microphysiological system to simultaneously investigate effects of anti-tumor natural killer cells on tumor and cardiac microtissues. Front. Immunol. 12, 781337 (2021).
Buoncervello, M. et al. Inflammatory cytokines associated with cancer growth induce mitochondria and cytoskeleton alterations in cardiomyocytes. J. Cell. Physiol. 234, 20453–20468 (2019).
Totzeck, M., Michel, L., Lin, Y., Herrmann, J. & Rassaf, T. Cardiotoxicity from chimeric antigen receptor-T cell therapy for advanced malignancies. Eur. Heart J. 43, 1928–1940 (2022).
Nenna, A. et al. Cardiotoxicity of Chimeric Antigen Receptor T-cell (CAR-T) therapy: pathophysiology, clinical implications and echocardiographic assessment. Int. J. Mol. Sci. 23, 8242 (2022).
Kerns, S. J. et al. Human immunocompetent Organ-on-Chip platforms allow safety profiling of tumor-targeted T-cell bispecific antibodies. eLife 10, e67106 (2021).
Maulana, T. I. et al. Solid tumor-on-chip model for efficacy and safety assessment of CAR-T cell therapy. Preprint at https://doi.org/10.1101/2023.07.13.548856 (2023).
Nahon, D. M. et al. Standardizing designed and emergent quantitative features in microphysiological systems. Nat. Biomed. Eng. 8, 941–962 (2024).
Zhuang, R. Z., Lock, R., Liu, B. & Vunjak-Novakovic, G. Opportunities and challenges in cardiac tissue engineering from an analysis of two decades of advances. Nat. Biomed. Eng. 6, 327–338 (2022).
Ronaldson-Bouchard, K. et al. A multi-organ chip with matured tissue niches linked by vascular flow. Nat. Biomed. Eng. 6, 351–371 (2022).
Novak, R. et al. Robotic fluidic coupling and interrogation of multiple vascularized organ chips. Nat. Biomed. Eng. 4, 407–420 (2020).
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).
Buchanan, B. C. & Yoon, J.-Y. Microscopic imaging methods for organ-on-a-chip platforms. Micromachines 13, 328 (2022).
Shpigelman, J. et al. Generation and application of a reporter cell line for the quantitative screen of extracellular vesicle release. Front. Pharmacol. 12, 668609 (2021).
Orita, K., Sawada, K., Koyama, R. & Ikegaya, Y. Deep learning-based quality control of cultured human-induced pluripotent stem cell-derived cardiomyocytes. J. Pharmacol. Sci. 140, 313–316 (2019).
Zheng, Z. et al. Unsupervised deep learning of bright-field images for apoptotic cell classification. Signal Image Video Process. 17, 3657–3664 (2023).
Harrison, P. J. et al. Evaluating the utility of brightfield image data for mechanism of action prediction. PLoS Comput. Biol. 19, e1011323 (2023).
Das, J. et al. Reagentless biomolecular analysis using a molecular pendulum. Nat. Chem. 13, 428–434 (2021).
Langer, A. et al. Protein analysis by time-resolved measurements with an electro-switchable DNA chip. Nat. Commun. 4, 2099 (2013).
Wang, M. et al. A wearable electrochemical biosensor for the monitoring of metabolites and nutrients. Nat. Biomed. Eng. 6, 1225–1235 (2022).
Chang, D. et al. A high-dimensional microfluidic approach for selection of aptamers with programmable binding affinities. Nat. Chem. 15, 773–780 (2023).
Ye, C. et al. A wearable aptamer nanobiosensor for non-invasive female hormone monitoring. Nat. Nanotechnol. 19, 330–337 (2024).
Cell Sorting Applications on the MACSQuant® Tyto® Cell Sorter (Miltenyi Biotec, 2025); https://www.miltenyibiotec.com/US-en/products/macs-flow-cytometry/cell-sorter/macsquant-tyto-cell-sorting-applications.html?query=:relevance:allCategoriesOR:10000422%23OnJlbGV2YW5jZTphbGxDYXRlZ29yaWVzT1I6MTAwMDA0MjI%3D
Byun, C. K., Abi-Samra, K., Cho, Y. & Takayama, S. Pumps for microfluidic cell culture. Electrophoresis 35, 245–257 (2014).
Zhang, C., Xing, D. & Li, Y. Micropumps, microvalves and micromixers within PCR microfluidic chips: advances and trends. Biotechnol. Adv. 25, 483–514 (2007).
Baysoy, A., Bai, Z., Satija, R. & Fan, R. The technological landscape and applications of single-cell multi-omics. Nat. Rev. Mol. Cell Biol. 24, 695–713 (2023).
Kong, J. et al. Network-based machine learning approach to predict immunotherapy response in cancer patients. Nat. Commun. 13, 3703 (2022).
Ghassemi, S. et al. Rapid manufacturing of non-activated potent CAR T cells. Nat. Biomed. Eng. 6, 118–128 (2022).
Ahmadi, M. et al. Accelerating CAR T cell manufacturing with an automated next-day process. Preprint at https://doi.org/10.1101/2024.06.04.596536 (2024).
Fraietta, J. A. et al. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat. Med. 24, 563–571 (2018).
Arcangeli, S. et al. CAR T cell manufacturing from naive/stem memory T lymphocytes enhances antitumor responses while curtailing cytokine release syndrome. J. Clin. Invest. 132, e150807 (2022).
Pan, J. et al. High efficacy and safety of low-dose CD19-directed CAR-T cell therapy in 51 refractory or relapsed B acute lymphoblastic leukemia patients. Leukemia 31, 2587–2593 (2017).
Garcia, J. et al. Naturally occurring T cell mutations enhance engineered T cell therapies. Nature 626, 626–634 (2024).
Maldini, C. R., Ellis, G. I. & Riley, J. L. CAR T cells for infection, autoimmunity and allotransplantation. Nat. Rev. Immunol. 18, 605–616 (2018).
Ledford, H. Support for risky cancer therapy. Nature https://www.nature.com/articles/nature.2017.22304.pdf (2017).
Schuster, S. J. et al. Long-term clinical outcomes of tisagenlecleucel in patients with relapsed or refractory aggressive B-cell lymphomas (JULIET): a multicentre, open-label, single-arm, phase 2 study. Lancet Oncol. 22, 1403–1415 (2021).
Papadouli, I. et al. EMA review of axicabtagene ciloleucel (Yescarta) for the treatment of diffuse large B-cell lymphoma. Oncologist 25, 894–902 (2020).
Kite Receives US FDA Approval of Manufacturing Process Change Resulting in Reduced Median Turnaround Time for Yescarta® CAR T-Cell Therapy (Gilead, 2024); https://www.gilead.com/news-and-press/press-room/press-releases/2024/1/kite-receives-us-fda-approval-of-manufacturing-process-change-resulting-in-reduced-median-turnaround-time-for-yescarta-car-tcell-therapy
US Food and Drug Administration Approves Bristol Myers Squibb’s Breyanzi (Lisocabtagene Maraleucel), a New CAR T Cell Therapy for Adults with Relapsed or Refractory Large B-Cell Lymphoma (BMS, 2021); https://news.bms.com/news/details/2021/U.S.-Food-and-Drug-Administration-Approves-Bristol-Myers-Squibbs-Breyanzi-lisocabtagene-maraleucel-a-New-CAR-T-Cell-Therapy-for-Adults-with-Relapsed-or-Refractory-Large-B-cell-Lymphoma/default.aspx
Wang, M. et al. KTE-X19 CAR T-cell therapy in relapsed or refractory mantle-cell lymphoma. N. Engl. J. Med. 382, 1331–1342 (2020).
US FDA Approves Kite’s TecartusTM, the First and Only CAR T Treatment for Relapsed or Refractory Mantle Cell Lymphoma (Gilead, 2020); https://www.gilead.com/news-and-press/press-room/press-releases/2020/7/us-fda-approves-kites-tecartus-the-first-and-only-car-t-treatment-for-relapsed-or-refractory-mantle-cell-lymphoma
Munshi, N. C. et al. Idecabtagene vicleucel in relapsed and refractory multiple myeloma. N. Engl. J. Med. 384, 705–716 (2021).
San-Miguel, J. et al. Cilta-cel or standard care in lenalidomide-refractory multiple myeloma. N. Engl. J. Med. 389, 335–347 (2023).
Acknowledgements
This contribution was supported in part by the National Cancer Institute of the National Institutes of Health (grants nos. 1R01CA260170 and 1R01CA277507). It was also supported in part by the McCormick Catalyst Fund at Northwestern University. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health or the other funding agencies. Z.W. was supported by a Banting Postdoctoral Scholarship from the Canadian Institutes of Health Research (application no. 489318).
Author information
Authors and Affiliations
Contributions
Z.W. and S.O.K. conceived, wrote and edited the manuscript.
Corresponding author
Ethics declarations
Competing interests
Z.W. and S.O.K. are co-founders of CTRL Therapeutics, which is commercializing microfluidic technologies for cellular therapy. S.O.K. has received research funds from Amgen and Moderna outside of the submitted work.
Peer review
Peer review information
Nature Biomedical Engineering thanks Aram Chung, Dino Di Carlo 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.
Supplementary information
Supplementary Information
Supplementary tables.
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.
About this article
Cite this article
Wang, Z., Kelley, S.O. Microfluidic technologies for enhancing the potency, predictability and affordability of adoptive cell therapies. Nat. Biomed. Eng (2025). https://doi.org/10.1038/s41551-024-01315-2
Received:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41551-024-01315-2