Abstract
In vitro tumor models are essential tools for cancer research, offering key insights into not only tumor biology but also therapeutic responses. The transition from traditional two-dimensional to three-dimensional organoid systems marks a paradigm shift in cancer modeling. Although two-dimensional models have been instrumental in elucidating fundamental molecular and genetic mechanisms, they fail to accurately replicate the intricate three-dimensional architecture and dynamic microenvironment characteristic of human tumors. Here we outline how advanced organoid technologies now enable more faithful recapitulation of tumor heterogeneity that better mimic native tissue mechanics and biochemistry. We discuss emerging methods, including air–liquid interface cultures, microfluidic tumor-on-a-chip devices and high-content imaging integrated with machine learning, which collectively address longstanding challenges such as matrix variability and the limited incorporation of immune and vascular elements. These innovations promise to enhance reproducibility and scalability while providing unprecedented insights into tumor biology, cancer progression and therapeutic strategies.
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
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$259.00 per year
only $21.58 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
Gerlinger, M. et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 366, 883–892 (2012).
Navin, N. et al. Tumour evolution inferred by single-cell sequencing. Nature 472, 90–94 (2011).
Patel, A. P. et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science 344, 1396–1401 (2014).
Li, Z. et al. Hypoxia-inducible factors regulate tumorigenic capacity of glioma stem cells. Cancer Cell 15, 501–513 (2009).
Rofstad, E. K. & Halsør, E. F. Hypoxia-associated spontaneous pulmonary metastasis in human melanoma xenografts: involvement of microvascular hot spots induced in hypoxic foci by interleukin 8. Br. J. Cancer 86, 301–308 (2002).
Mani, S. A. et al. The epithelial–mesenchymal transition generates cells with properties of stem cells. Cell 133, 704–715 (2008).
Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).
McGranahan, N. & Swanton, C. Cancer evolution constrained by the immune microenvironment. Cell 170, 825–827 (2017).
Gupta, P. B., Pastushenko, I., Skibinski, A., Blanpain, C. & Kuperwasser, C. Phenotypic plasticity: driver of cancer initiation, progression, and therapy resistance. Cell Stem Cell 24, 65–78 (2019).
Tabassum, D. P. & Polyak, K. Tumorigenesis: it takes a village. Nat. Rev. Cancer 15, 473–483 (2015).
Cheon, D. -J. & Orsulic, S. Mouse models of cancer. Annu. Rev. Pathol. 6, 95–119 (2011).
Naba, A. et al. The matrisome: in silico definition and in vivo characterization by proteomics of normal and tumor extracellular matrices. Mol. Cell. Proteomics 11, M111.014647 (2012).
Mestas, J. & Hughes, C. C. W. Of mice and not men: differences between mouse and human immunology. J. Immunol. 172, 2731–2738 (2004).
Sutherland, R. M., McCredie, J. A. & Inch, W. R. Growth of multicell spheroids in tissue culture as a model of nodular carcinomas. J. Natl Cancer Inst. 46, 113–120 (1971).
Weaver, V. M. et al. Reversion of the malignant phenotype of human breast cells in three-dimensional culture and in vivo by integrin blocking antibodies. J. Cell Biol. 137, 231–245 (1997).
Cukierman, E., Pankov, R., Stevens, D. R. & Yamada, K. M. Taking cell-matrix adhesions to the third dimension. Science 294, 1708–1712 (2001).
Oishi, H., Tabibzadeh, N. & Morizane, R. Advancing preclinical drug evaluation through automated 3D imaging for high-throughput screening with kidney organoids. Biofabrication https://doi.org/10.1088/1758-5090/ad38df (2024).
Bozal, S. B. et al. Development of an automated 3D high content cell screening platform for organoid phenotyping. SLAS Discov. 29, 100182 (2024).
Carrel, A. On the permanent life of tissues outside of the organism. J. Exp. Med. 15, 516–528 (1912).
Gey, G. Tissue culture studies of the proliferative capacity of cervical carcinoma and normal epithelium. Cancer Res. 12, 264–265 (1952).
Lucey, B. P., Nelson-Rees, W. A. & Hutchins, G. M. Henrietta Lacks, HeLa cells, and cell culture contamination. Arch. Pathol. Lab. Med. 133, 1463–1467 (2009).
Soule, H. D., Vazguez, J., Long, A., Albert, S. & Brennan, M. A human cell line from a pleural effusion derived from a breast carcinoma. J. Natl Cancer Inst. 51, 1409–1416 (1973).
Young, R., Cailleau, R., Mackay, B. & Reeves, W. Jr. Establishment of epithelial cell line MDA-MB-157 from metastatic pleural effusion of human breast carcinoma. In Vitro 9, 239–245 (1974).
Cailleau, R., Young, R., Olive, M. & Reeves, W. Jr Breast tumor cell lines from pleural effusions. J. Natl Cancer Inst. 53, 661–674 (1974).
Cailleau, R., Olive, M. & Cruciger, Q. V. Long-term human breast carcinoma cell lines of metastatic origin: preliminary characterization. In Vitro 14, 911–915 (1978).
Gillet, J. P., Varma, S. & Gottesman, M. M. The clinical relevance of cancer cell lines. J. Natl Cancer Inst. 105, 452–458 (2013).
Valiente, M. et al. Brain metastasis cell lines panel: a public resource of organotropic cell lines. Cancer Res. 80, 4314–4323 (2020).
Shoemaker, R. H. The NCI60 human tumour cell line anticancer drug screen. Nat. Rev. Cancer 6, 813–823 (2006).
Ethier, S. P. et al. Development and implementation of the SUM breast cancer cell line functional genomics knowledge base. NPJ Breast Cancer 6, 30 (2020).
Weinberg, R. A. Oncogenes and the molecular biology of cancer. J. Cell Biol. 97, 1661–1662 (1983).
Shih, C. & Weinberg, R. A. Isolation of a transforming sequence from a human bladder carcinoma cell line. Cell 29, 161–169 (1982).
Land, H., Parada, L. F. & Weinberg, R. A. Tumorigenic conversion of primary embryo fibroblasts requires at least two cooperating oncogenes. Nature 304, 596–602 (1983).
Knudson, A. G. Jr. Mutation and cancer: statistical study of retinoblastoma. Proc. Natl Acad. Sci. USA 68, 820–823 (1971).
Steck, P. A. et al. Identification of a candidate tumour suppressor gene, MMAC1, at chromosome 10q23.3 that is mutated in multiple advanced cancers. Nat. Genet. 15, 356–362 (1997).
Levine, A. J. The tumor suppressor genes. Annu. Rev. Biochem. 62, 623–651 (1993).
Hahn, W. C. et al. Creation of human tumour cells with defined genetic elements. Nature 400, 464–468 (1999).
Lo, C. -M., Wang, H. -B., Dembo, M. & Wang, Y. -l Cell movement is guided by the rigidity of the substrate. Biophys. J. 79, 144–152 (2000).
Pelham, R. J. & Wang, Y. -L. Cell locomotion and focal adhesions are regulated by substrate flexibility. Proc. Natl Acad. Sci. USA 94, 13661–13665 (1997).
Ng, M. R., Besser, A., Brugge, J. S. & Danuser, G. Mapping the dynamics of force transduction at cell–cell junctions of epithelial clusters. eLife 3, e03282 (2014).
Sutherland, R. M. & Durand, R. E. Radiation response of multicell spheroids-an in vitro tumour model. Curr. Top. Radiat. Res. Q. 11, 87–139 (1976).
Maritan, S. M., Lian, E. Y. & Mulligan, L. M. An efficient and flexible cell aggregation method for 3D spheroid production. J. Vis. Exp. https://doi.org/10.3791/55544 (2017).
Nazari, S. S. Generation of 3D tumor spheroids with encapsulating basement membranes for invasion studies. Curr. Protoc. Cell Biol. 87, e105 (2020).
Carlsson, J. et al. Formation and growth of multicellular spheroids of human origin. Int. J. Cancer 31, 523–533 (1983).
Sutherland, R. M. Cell and environment interactions in tumor microregions: the multicell spheroid model. Science 240, 177–184 (1988).
Casciari, J. J., Sotirchos, S. V. & Sutherland, R. M. Glucose diffusivity in multicellular tumor spheroids. Cancer Res. 48, 3905–3909 (1988).
Lee, C. H., Yu, C. C., Wang, B. Y. & Chang, W. W. Tumorsphere as an effective in vitro platform for screening anti-cancer stem cell drugs. Oncotarget 7, 1215–1226 (2016).
Ishiguro, T. et al. Tumor-derived spheroids: relevance to cancer stem cells and clinical applications. Cancer Sci. 108, 283–289 (2017).
Froehlich, K. et al. Generation of multicellular breast cancer tumor spheroids: comparison of different protocols. J. Mammary Gland Biol. Neoplasia 21, 89–98 (2016).
Wei, X., Wu, Y., Chen, K., Wang, L. & Xu, M. Embedded bioprinted multicellular spheroids modeling pancreatic cancer bioarchitecture towards advanced drug therapy. J. Mater. Chem. B 12, 1788–1797 (2024).
Wang, Z. et al. Rapid tissue prototyping with micro-organospheres. Stem Cell Rep. 17, 1959–1975 (2022).
Bissell, M. J., Hall, H. G. & Parry, G. How does the extracellular matrix direct gene expression? J. Theor. Biol. 99, 31–68 (1982).
Li, M. L. et al. Influence of a reconstituted basement membrane and its components on casein gene expression and secretion in mouse mammary epithelial cells. Proc. Natl Acad. Sci. USA 84, 136–140 (1987).
Barcellos-Hoff, M. H., Aggeler, J., Ram, T. G. & Bissell, M. J. Functional differentiation and alveolar morphogenesis of primary mammary cultures on reconstituted basement membrane. Development 105, 223–235 (1989).
Muthuswamy, S. K., Li, D., Lelievre, S., Bissell, M. J. & Brugge, J. S. ErbB2, but not ErbB1, reinitiates proliferation and induces luminal repopulation in epithelial acini. Nat. Cell Biol. 3, 785–792 (2001).
Kleinman, H. K. et al. Basement membrane complexes with biological activity. Biochemistry 25, 312–318 (1986).
Tsai, K. K. et al. Screening of organoids derived from patients with breast cancer implicates the repressor NCOR2 in cytotoxic stress response and antitumor immunity. Nat. Cancer 3, 734–752 (2022).
Hebner, C., Weaver, V. M. & Debnath, J. Modeling morphogenesis and oncogenesis in three-dimensional breast epithelial cultures. Annu. Rev. Pathol. 3, 313–339 (2008).
Sachs, N. et al. Long‐term expanding human airway organoids for disease modeling. EMBO J. 38, e100300 (2019).
Sato, T. et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 459, 262–265 (2009).
Huch, M. et al. Long-term culture of genome-stable bipotent stem cells from adult human liver. Cell 160, 299–312 (2015).
Rosenbluth, J. M. et al. Organoid cultures from normal and cancer-prone human breast tissues preserve complex epithelial lineages. Nat. Commun. 11, 1711 (2020).
Di Lullo, E. & Kriegstein, A. R. The use of brain organoids to investigate neural development and disease. Nat. Rev. Neurosci. 18, 573–584 (2017).
Cho, A. N. et al. Microfluidic device with brain extracellular matrix promotes structural and functional maturation of human brain organoids. Nat. Commun. 12, 4730 (2021).
Lancaster, M. A. et al. Cerebral organoids model human brain development and microcephaly. Nature 501, 373–379 (2013).
Takebe, T. et al. Vascularized and functional human liver from an iPSC-derived organ bud transplant. Nature 499, 481–484 (2013).
Sekine, K. et al. Generation of human induced pluripotent stem cell-derived liver buds with chemically defined and animal origin-free media. Sci. Rep. 10, 17937 (2020).
Li, J. et al. Functional 3D human liver bud assembled from MSC-derived multiple liver cell lineages. Cell Transplant. 28, 510–521 (2019).
Zhang, R. R. et al. Human iPSC-derived posterior gut progenitors are expandable and capable of forming gut and liver organoids. Stem Cell Rep. 10, 780–793 (2018).
Yan, H. H. N. et al. A comprehensive human gastric cancer organoid biobank captures tumor subtype heterogeneity and enables therapeutic screening. Cell Stem Cell 23, 882–897 (2018).
van de Wetering, M. et al. Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell 161, 933–945 (2015).
Jacob, F. et al. A patient-derived glioblastoma organoid model and biobank recapitulates inter- and intra-tumoral heterogeneity. Cell 180, 188–204 (2020).
Drost, J. & Clevers, H. Organoids in cancer research. Nat. Rev. Cancer 18, 407–418 (2018).
Bleijs, M., van de Wetering, M., Clevers, H. & Drost, J. Xenograft and organoid model systems in cancer research. EMBO J. 38, e101654 (2019).
Hubert, C. G. et al. A three-dimensional organoid culture system derived from human glioblastomas recapitulates the hypoxic gradients and cancer stem cell heterogeneity of tumors found in vivo. Cancer Res. 76, 2465–2477 (2016).
Sato, T. et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett’s epithelium. Gastroenterology 141, 1762–1772 (2011).
Boj, S. F. et al. Organoid models of human and mouse ductal pancreatic cancer. Cell 160, 324–338 (2015).
Sachs, N. et al. A living biobank of breast cancer organoids captures disease heterogeneity. Cell 172, 373–386 (2018).
Kim, M. et al. Patient-derived lung cancer organoids as in vitro cancer models for therapeutic screening. Nat. Commun. 10, 3991 (2019).
Li, X. et al. Organoid cultures recapitulate esophageal adenocarcinoma heterogeneity providing a model for clonality studies and precision therapeutics. Nat. Commun. 9, 2983 (2018).
Lee, S. H. et al. Tumor evolution and drug response in patient-derived organoid models of bladder cancer. Cell 173, 515–528 (2018).
de Witte, C. J. et al. Patient-derived ovarian cancer organoids mimic clinical response and exhibit heterogeneous inter- and intrapatient drug responses. Cell Rep. https://doi.org/10.1016/j.celrep.2020.107762 (2020).
Li, Z. et al. Patient-derived renal cell carcinoma organoids for personalized cancer therapy. Clin. Transl. Med. 12, e970 (2022).
Broutier, L. et al. Human primary liver cancer-derived organoid cultures for disease modeling and drug screening. Nat. Med. 23, 1424–1435 (2017).
Kopper, O. et al. An organoid platform for ovarian cancer captures intra- and interpatient heterogeneity. Nat. Med. 25, 838–849 (2019).
Gao, D. et al. Organoid cultures derived from patients with advanced prostate cancer. Cell 159, 176–187 (2014).
Drost, J. et al. Organoid culture systems for prostate epithelial and cancer tissue. Nat. Protoc. 11, 347–358 (2016).
Calandrini, C. et al. An organoid biobank for childhood kidney cancers that captures disease and tissue heterogeneity. Nat. Commun. 11, 1310 (2020).
Sanchez-Fdez, A., Sharma, A. K., Tiriac, H. & Sicklick, J. K. Patient-derived sarcoma organoids offer a novel platform for personalized precision medicine. Ann. Surg. Oncol. 29, 7239–7241 (2022).
Xu, S., Tan, S. & Guo, L. Patient-derived organoids as a promising tool for multimodal management of sarcomas. Cancers https://doi.org/10.3390/cancers15174339 (2023).
Chen, Y. et al. Addressing modern diagnostic pathology for patient-derived soft tissue sarcosphere models in the era of functional precision oncology. Lab Invest. 103, 100039 (2023).
Meister, M. T. et al. Mesenchymal tumor organoid models recapitulate rhabdomyosarcoma subtypes. EMBO Mol. Med. 14, e16001 (2022).
Savary, C. et al. Fusion-negative rhabdomyosarcoma 3D organoids to predict effective drug combinations: a proof-of-concept on cell death inducers. Cell Rep. Med. 4, 101339 (2023).
Guillen, K. P. et al. A human breast cancer-derived xenograft and organoid platform for drug discovery and precision oncology. Nat. Cancer 3, 232–250 (2022).
Driehuis, E., Kretzschmar, K. & Clevers, H. Establishment of patient-derived cancer organoids for drug-screening applications. Nat. Protoc. 15, 3380–3409 (2020).
Khalili, S. et al. StarTrace: a multiplex organoid avatar drug testing platform for personalized medicine. Preprint at bioRxiv https://doi.org/10.1101/2025.02.12.637574 (2025).
Vlachogiannis, G. et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science 359, 920–926 (2018).
Drost, J. et al. Use of CRISPR-modified human stem cell organoids to study the origin of mutational signatures in cancer. Science 358, 234–238 (2017).
Geurts, M. H. & Clevers, H. CRISPR engineering in organoids for gene repair and disease modelling. Nat. Rev. Bioeng. 1, 32–45 (2023).
Drost, J. et al. Sequential cancer mutations in cultured human intestinal stem cells. Nature 521, 43–47 (2015).
Matano, M. et al. Modeling colorectal cancer using CRISPR-Cas9-mediated engineering of human intestinal organoids. Nat. Med. 21, 256–262 (2015).
Pasch, C. A. et al. Patient-derived cancer organoid cultures to predict sensitivity to chemotherapy and radiation. Clin. Cancer Res. 25, 5376–5387 (2019).
Ooft, S. et al. Prospective experimental treatment of colorectal cancer patients based on organoid drug responses. ESMO Open 6, 100103 (2021).
Thorel, L. et al. Patient-derived tumor organoids: a new avenue for preclinical research and precision medicine in oncology. Exp. Mol. Med. 56, 1531–1551 (2024).
Millen, R. et al. Patient-derived head and neck cancer organoids allow treatment stratification and serve as a tool for biomarker validation and identification. Med. 4, 290–310 (2023).
Berg, H. F. et al. Patient-derived organoids reflect the genetic profile of endometrial tumors and predict patient prognosis. Commun. Med. 1, 20 (2021).
Zeng, W. et al. Patient-derived xenografts of different grade gliomas retain the heterogeneous histological and genetic features of human gliomas. Cancer Cell Int. 20, 1 (2020).
Ding, S. et al. Patient-derived micro-organospheres enable clinical precision oncology. Cell Stem Cell 29, 905–917 (2022).
Nishiguchi, A. et al. In vitro 3D blood/lymph-vascularized human stromal tissues for preclinical assays of cancer metastasis. Biomaterials 179, 144–155 (2018).
Mao, M. et al. Human‐on‐leaf‐chip: a biomimetic vascular system integrated with chamber‐specific organs. Small 16, e2000546 (2020).
Lee, H., Park, W., Ryu, H. & Jeon, N. L. A microfluidic platform for quantitative analysis of cancer angiogenesis and intravasation. Biomicrofluidics 8, 054102 (2014).
Aleman, J. & Skardal, A. A multi‐site metastasis‐on‐a‐chip microphysiological system for assessing metastatic preference of cancer cells. Biotechnol. Bioeng. 116, 936–944 (2019).
Liu, X. et al. Tumor-on-a-chip: from bioinspired design to biomedical application. Microsyst. Nanoeng. 7, 50 (2021).
Jouybar, M. et al. Cancer-on-chip models for metastasis: importance of the tumor microenvironment. Trends Biotechnol. 42, 431–448 (2024).
Wörsdörfer, P. et al. Generation of complex human organoid models including vascular networks by incorporation of mesodermal progenitor cells. Sci. Rep. 9, 15663 (2019).
Tsai, S. et al. Development of primary human pancreatic cancer organoids, matched stromal and immune cells and 3D tumor microenvironment models. BMC Cancer 18, 335 (2018).
Haque, M. R. et al. Patient-derived pancreatic cancer-on-a-chip recapitulates the tumor microenvironment. Microsyst. Nanoeng. 8, 36 (2022).
Seino, T. et al. Human pancreatic tumor organoids reveal loss of stem cell niche factor dependence during disease progression. Cell Stem Cell 22, 454–467 (2018).
Neal, J. T. et al. Organoid modeling of the tumor immune microenvironment. Cell 175, 1972–1988 (2018).
Dijkstra, K. K. et al. Generation of tumor-reactive T cells by co-culture of peripheral blood lymphocytes and tumor organoids. Cell 174, 1586–1598 (2018).
Mu, P. et al. Newly developed 3D in vitro models to study tumor-immune interaction. J. Exp. Clin. Cancer Res. 42, 81 (2023).
Kozlowski, M. T., Crook, C. J. & Ku, H. T. Towards organoid culture without Matrigel. Commun. Biol. 4, 1387 (2021).
Oxford, J. T., Reeck, J. C. & Hardy, M. J. Extracellular matrix in development and disease. Int. J. Mol. Sci. https://doi.org/10.3390/ijms20010205 (2019).
Feng B, Y. H. et al. Collagen-based biomaterials in organoid technology for reproductive medicine: composition, characteristics, and applications. Collagen Leather 5, 35 (2023).
Barnes, C. et al. From single cells to tissues: interactions between the matrix and human breast cells in real time. PLoS ONE 9, e93325 (2014).
Vörsmann, H. et al. Development of a human three-dimensional organotypic skin-melanoma spheroid model for in vitro drug testing. Cell Death Dis. 4, e719 (2013).
Wang, H. K., Broker, T. R. & Chow, L. T. Robust HPV-18 production in organotypic cultures of primary human keratinocytes. Methods Mol. Biol. 1249, 93–109 (2015).
Maione, A. G. et al. Three-dimensional human tissue models that incorporate diabetic foot ulcer-derived fibroblasts mimic in vivo features of chronic wounds. Tissue Eng. Part C Methods 21, 499–508 (2015).
Parry, D. A. D. The molecular fibrillar structure of collagen and its relationship to the mechanical properties of connective tissue. Biophys. Chem. 29, 195–209 (1988).
Abbas, Y. et al. Generation of a three-dimensional collagen scaffold-based model of the human endometrium. Interface Focus 10, 20190079 (2020).
Krause, S., Maffini, M. V., Soto, A. M. & Sonnenschein, C. A novel 3D in vitro culture model to study stromal-epithelial interactions in the mammary gland. Tissue Eng. Part C Methods 14, 261–271 (2008).
Yu, W. et al. β1-integrin orients epithelial polarity via Rac1 and laminin. Mol. Biol. Cell 16, 433–445 (2005).
Manninen, A. Epithelial polarity–generating and integrating signals from the ECM with integrins. Exp. Cell. Res. 334, 337–349 (2015).
Gjorevski, N. et al. Designer matrices for intestinal stem cell and organoid culture. Nature 539, 560–564 (2016).
Damljanović, V., Lagerholm, B. C. & Jacobson, K. Bulk and micropatterned conjugation of extracellular matrix proteins to characterized polyacrylamide substrates for cell mechanotransduction assays. Biotechniques 39, 847–851 (2005).
Tse, J. R. & Engler, A. J. Preparation of hydrogel substrates with tunable mechanical properties. Curr. Protoc. Cell Biol. 47, 10.16.11–10.16.16 (2010).
Lin, C. -C. & Anseth, K. S. PEG hydrogels for the controlled release of biomolecules in regenerative medicine. Pharm. Res. 26, 631–643 (2009).
Phelps, E. A. et al. Maleimide cross-linked bioactive PEG hydrogel exhibits improved reaction kinetics and cross-linking for cell encapsulation and in-situ delivery. Adv. Mater. 24, 64–70 (2011).
Kumar, V. B., Tiwari, O. S., Finkelstein-Zuta, G., Rencus-Lazar, S. & Gazit, E. Design of functional RGD peptide-based biomaterials for tissue engineering. Pharmaceutics 15, 345 (2023).
Caliari, S. R. & Burdick, J. A. A practical guide to hydrogels for cell culture. Nat. Methods 13, 405–414 (2016).
Urciuolo, A. et al. Hydrogel-in-hydrogel live bioprinting for guidance and control of organoids and organotypic cultures. Nat. Commun. 14, 3128 (2023).
Lee, J. M. et al. Conductive hydrogel/nanowire micropattern-based sensor for neural stem cell differentiation. Sens. Actuators B Chem. 258, 1042–1050 (2018).
Sokol, E. S. et al. Growth of human breast tissues from patient cells in 3D hydrogel scaffolds. Breast Cancer Res. 18, 19 (2016).
Rauner, G. et al. Next-generation breast organoids capture tissue organogenesis with high-resolution live imaging. Preprint at bioRxiv https://doi.org/10.1101/2023.10.02.560364 (2025).
Rauner, G. et al. Breast tissue regeneration is driven by cell-matrix interactions coordinating multi-lineage stem cell differentiation through DDR1. Nat. Commun. 12, 7116 (2021).
Guenther, C. Stiffness regulates dendritic cell and macrophage subtype development and increased stiffness induces a tumor–associated macrophage phenotype in cancer co–cultures. Front. Immunol. 15, 1434030 (2024).
Wang, C., Tong, X. & Yang, F. Bioengineered 3D brain tumor model to elucidate the effects of matrix stiffness on glioblastoma cell behavior using PEG-based hydrogels. Mol. Pharm. 11, 2115–2125 (2014).
Wang, Z. et al. Spatial cell fate manipulation of human pluripotent stem cells by controlling the microenvironment using photocurable hydrogel. Development 151, dev201621 (2024).
Guimarães, C. F., Gasperini, L., Marques, A. P. & Reis, R. L. The stiffness of living tissues and its implications for tissue engineering. Nat. Rev. Mater. 5, 351–370 (2020).
Linnemann, J. R. et al. Quantification of regenerative potential in primary human mammary epithelial cells. Development 142, 3239–3251 (2015).
Buchmann, B. et al. Mechanical plasticity of collagen directs branch elongation in human mammary gland organoids. Nat. Commun. 12, 2759 (2021).
Trepicchio, C. et al. DDR1 regulates RUNX1-CBFβ to control breast stem cell differentiation. Stem Cell Rep. https://doi.org/10.1016/j.stemcr.2025.102576 (2025).
Bankhead, P. et al. QuPath: open source software for digital pathology image analysis. Sci. Rep. 7, 16878 (2017).
Ershov, D. et al. TrackMate 7: integrating state-of-the-art segmentation algorithms into tracking pipelines. Nat. Methods 19, 829–832 (2022).
Moen, E. et al. Deep learning for cellular image analysis. Nat. Methods 16, 1233–1246 (2019).
Choo, N. et al. High-throughput imaging assay for drug screening of 3D prostate cancer organoids. SLAS Discov. 26, 1107–1124 (2021).
Li, X. et al. Assay establishment and validation of a high-throughput organoid-based drug screening platform. Stem Cell Res. Ther. 13, 219 (2022).
Lee, B. H. et al. Control of lumen geometry and topology by the interplay between pressure and cell proliferation rate in pancreatic organoids. Preprint at bioRxiv https://doi.org/10.1101/2024.05.29.596462 (2024).
Lukonin, I., Zinner, M. & Liberali, P. Organoids in image-based phenotypic chemical screens. Exp. Mol. Med. 53, 1495–1502 (2021).
Barroso, M., Monaghan, M. G., Niesner, R. & Dmitriev, R. I. Probing organoid metabolism using fluorescence lifetime imaging microscopy (FLIM): the next frontier of drug discovery and disease understanding. Adv. Drug Deliv. Rev. 201, 115081 (2023).
Kelm, J. M., Timmins, N. E., Brown, C. J., Fussenegger, M. & Nielsen, L. K. Method for generation of homogeneous multicellular tumor spheroids applicable to a wide variety of cell types. Biotechnol. Bioeng. 83, 173–180 (2003).
Friedrich, J., Seidel, C., Ebner, R. & Kunz-Schughart, L. A. Spheroid-based drug screen: considerations and practical approach. Nat. Protoc. 4, 309–324 (2009).
Abbas, Z. N., Al-Saffar, A. Z., Jasim, S. M. & Sulaiman, G. M. Comparative analysis between 2D and 3D colorectal cancer culture models for insights into cellular morphological and transcriptomic variations. Sci. Rep. 13, 18380 (2023).
Ma, X. -L. et al. Sphere-forming culture enriches liver cancer stem cells and reveals Stearoyl-CoA desaturase 1 as a potential therapeutic target. BMC Cancer 19, 760 (2019).
Ponti, D. et al. Isolation and in vitro propagation of tumorigenic breast cancer cells with stem/progenitor cell properties. Cancer Res. 65, 5506–5511 (2005).
Yu, S. -c et al. Isolation and characterization of cancer stem cells from a human glioblastoma cell line U87. Cancer Lett. 265, 124–134 (2008).
Takasato, M. et al. Kidney organoids from human iPS cells contain multiple lineages and model human nephrogenesis. Nature 526, 564–568 (2015).
Quadrato, G. et al. Cell diversity and network dynamics in photosensitive human brain organoids. Nature 545, 48–53 (2017).
Bersini, S. et al. A microfluidic 3D in vitro model for specificity of breast cancer metastasis to bone. Biomaterials 35, 2454–2461 (2014).
Skardal, A., Devarasetty, M., Forsythe, S., Atala, A. & Soker, S. A reductionist metastasis-on-a-chip platform for in vitro tumor progression modeling and drug screening. Biotechnol. Bioeng. 113, 2020–2032 (2016).
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).
Acknowledgements
This research was supported by the National Institute of General Medical Sciences (7R01GM124491, to C.K.), DOD-BCRP (W81XWH2010018, to G.R.), Breast Cancer Research Foundation (to C.K.) and FTC Breast Cancer Foundation (to C.K.).
Author information
Authors and Affiliations
Contributions
G.R., P.B.G. and C.K. jointly conceived the perspective, and wrote the manuscript with contributions, editorial review and approval from all authors.
Corresponding author
Ethics declarations
Competing interests
C.K. is a cofounder and consultant of Naveris. P.B.G. is a cofounder, Chief Science & Technology Officer and Executive Chairman of Naveris. The remaining authors declare no competing interests.
Peer review
Peer review information
Nature Methods thanks Andrea McClatchey and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Madhura Mukhopadhyay, in collaboration with the Nature Methods team.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
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
Rauner, G., Gupta, P.B. & Kuperwasser, C. From 2D to 3D and beyond: the evolution and impact of in vitro tumor models in cancer research. Nat Methods 22, 1776–1787 (2025). https://doi.org/10.1038/s41592-025-02769-1
Received:
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1038/s41592-025-02769-1
This article is cited by
-
A tale of tumors
Nature Methods (2025)