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:

Engineered matrices reveal stiffness-mediated chemoresistance in patient-derived pancreatic cancer organoids

Abstract

Pancreatic ductal adenocarcinoma (PDAC) is characterized by its fibrotic and stiff extracellular matrix. However, how the altered cell/extracellular-matrix signalling contributes to the PDAC tumour phenotype has been difficult to dissect. Here we design and engineer matrices that recapitulate the key hallmarks of the PDAC tumour extracellular matrix to address this knowledge gap. We show that patient-derived PDAC organoids from three patients develop resistance to several clinically relevant chemotherapies when cultured within high-stiffness matrices mechanically matched to in vivo tumours. Using genetic barcoding, we find that while matrix-specific clonal selection occurs, cellular heterogeneity is not the main driver of chemoresistance. Instead, matrix-induced chemoresistance occurs within a stiff environment due to the increased expression of drug efflux transporters mediated by CD44 receptor interactions with hyaluronan. Moreover, PDAC chemoresistance is reversible following transfer from high- to low-stiffness matrices, suggesting that targeting the fibrotic extracellular matrix may sensitize chemoresistant tumours. Overall, our findings support the potential of engineered matrices and patient-derived organoids for elucidating extracellular matrix contributions to human disease pathophysiology.

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: ECM stiffness drives PDAC chemoresistance.
Fig. 2: Long-term culture in stiff matrices drives PDAC chemoresistance.
Fig. 3: Drug efflux transporter expression and activity mediates PDAC organoid chemoresistance.
Fig. 4: Influence of HA on PDAC chemoresistance.
Fig. 5: HA-mediated CD44 signalling mediates PDAC organoid chemoresistance.
Fig. 6: Stiffness-mediated PDAC chemoresistance is reversible.

Similar content being viewed by others

Data availability

All data needed to evaluate the conclusions in the paper are present in the paper and/or the Supplementary Information. Raw data related to this paper are uploaded to the Stanford Digital Repository, which can be accessed through the persistent URL (https://purl.stanford.edu/nw595bg6402) and the DOI (https://doi.org/10.25740/nw595bg6402). RNA-seq data from the TCGA and GTEx databases were accessed and plotted using the GEPIA online tool (http://gepia.cancer-pku.cn). Source data are provided with this paper.

References

  1. Kleeff, J. et al. Pancreatic cancer. Nat. Rev. Dis. Primers 2, 16022 (2016).

    PubMed  Google Scholar 

  2. Siegel, R. L., Miller, K. D. & Jemal, A. Cancer statistics, 2015. CA Cancer J. Clin. 65, 5–29 (2015).

    PubMed  Google Scholar 

  3. Amrutkar, M. & Gladhaug, I. P. Pancreatic cancer chemoresistance to gemcitabine. Cancers 9, 157 (2017).

    PubMed  PubMed Central  Google Scholar 

  4. Burris, H. A. et al. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J. Clin. Oncol. 15, 2403–2413 (1997).

    CAS  PubMed  Google Scholar 

  5. Rahib, L., Fleshman, J. M., Matrisian, L. M. & Berlin, J. D. Evaluation of pancreatic cancer clinical trials and benchmarks for clinically meaningful future trials: a systematic review. JAMA Oncol. 2, 1209–1216 (2016).

    PubMed  Google Scholar 

  6. Hosein, A. N., Brekken, R. A. & Maitra, A. Pancreatic cancer stroma: an update on therapeutic targeting strategies. Nat. Rev. Gastroenterol. Hepatol. 17, 487–505 (2020).

    PubMed  PubMed Central  Google Scholar 

  7. Feig, C. et al. The pancreas cancer microenvironment. Clin. Cancer Res. 18, 4266–4276 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Tian, C. et al. Proteomic analyses of ECM during pancreatic ductal adenocarcinoma progression reveal different contributions by tumor and stromal cells. Proc. Natl Acad. Sci. USA 116, 19609–19618 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Provenzano, P. P. et al. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell 21, 418–429 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Olive, K. P. et al. Inhibition of hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science 324, 1457–1461 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Hakim, N., Patel, R., Devoe, C. & Saif, M. W. Why HALO 301 failed and implications for treatment of pancreatic cancer. Pancreas Open J. 3, e1–e4 (2019).

    Google Scholar 

  12. Catenacci, D. V. T. et al. Randomized phase Ib/II study of gemcitabine plus placebo or vismodegib, a hedgehog pathway inhibitor, in patients with metastatic pancreatic cancer. J. Clin. Oncol. 33, 4284–4292 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Henke, E., Nandigama, R. & Ergün, S. Extracellular matrix in the tumor microenvironment and its impact on cancer therapy. Front. Mol. Biosci. 6, 160 (2020).

    PubMed  PubMed Central  Google Scholar 

  14. Paszek, M. J. et al. Tensional homeostasis and the malignant phenotype. Cancer Cell 8, 241–254 (2005).

    CAS  PubMed  Google Scholar 

  15. Chaudhuri, O. et al. Extracellular matrix stiffness and composition jointly regulate the induction of malignant phenotypes in mammary epithelium. Nat. Mater. 13, 970–978 (2014).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  17. Drost, J. & Clevers, H. Organoids in cancer research. Nat. Rev. Cancer 18, 407–418 (2018).

    CAS  PubMed  Google Scholar 

  18. Boj, S. F. et al. Organoid models of human and mouse ductal pancreatic cancer. Cell 160, 324–338 (2015).

    CAS  PubMed  Google Scholar 

  19. Tiriac, H. et al. Organoid profiling identifies common responders to chemotherapy in pancreatic cancer. Cancer Discov. 8, 1112–1129 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Hughes, C. S., Postovit, L. M. & Lajoie, G. A. Matrigel: a complex protein mixture required for optimal growth of cell culture. Proteomics 10, 1886–1890 (2010).

    CAS  PubMed  Google Scholar 

  21. Gjorevski, N. et al. Designer matrices for intestinal stem cell and organoid culture. Nature 539, 560–564 (2016).

    CAS  PubMed  Google Scholar 

  22. Hunt, D. R. et al. Engineered matrices enable the culture of human patient-derived intestinal organoids. Adv. Sci. 8, 2004705 (2021).

    CAS  Google Scholar 

  23. Hernandez-Gordillo, V. et al. Fully synthetic matrices for in vitro culture of primary human intestinal enteroids and endometrial organoids. Biomaterials 254, 120125 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Below, C. R. et al. A microenvironment-inspired synthetic three-dimensional model for pancreatic ductal adenocarcinoma organoids. Nat. Mater. 21, 110–119 (2022).

    CAS  PubMed  Google Scholar 

  25. Xiao, W. et al. Matrix stiffness mediates pancreatic cancer chemoresistance through induction of exosome hypersecretion in a cancer associated fibroblasts-tumor organoid biomimetic model. Matrix Biol. Plus 14, 100111 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Tang, Z. et al. GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 45, W98–W102 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. LeSavage, B. L., Suhar, N. A., Madl, C. M. & Heilshorn, S. C. Production of elastin-like protein hydrogels for encapsulation and immunostaining of cells in 3D. J. Vis. Exp. 135, e57739 (2018).

    Google Scholar 

  28. Connor, A. A. & Gallinger, S. Pancreatic cancer evolution and heterogeneity: integrating omics and clinical data. Nat. Rev. Cancer 22, 131–142 (2021).

    PubMed  Google Scholar 

  29. Roerink, S. F. et al. Intra-tumour diversification in colorectal cancer at the single-cell level. Nature 556, 437–462 (2018).

    Google Scholar 

  30. Krieger, T. G. et al. Single-cell analysis of patient-derived PDAC organoids reveals cell state heterogeneity and a conserved developmental hierarchy. Nat. Commun. 12, 5826 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Karlsson, K. et al. Deterministic evolution and stringent selection during preneoplasia. Nature 618, 383–393 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Dean, M., Fojo, T. & Bates, S. Tumour stem cells and drug resistance. Nat. Rev. Cancer 5, 275–284 (2005).

    CAS  PubMed  Google Scholar 

  33. Fletcher, J. I., Williams, R. T., Henderson, M. J., Norris, M. D. & Haber, M. ABC transporters as mediators of drug resistance and contributors to cancer cell biology. Drug Resist. Updates 26, 1–9 (2016).

    Google Scholar 

  34. Marzac, C. et al. ATP binding cassette transporters associated with chemoresistance: transcriptional profiling in extreme cohorts and their prognostic impact in a cohort of 281 acute myeloid leukemia patients. Haematologica 96, 1293–1301 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Robey, R. W. et al. Revisiting the role of ABC transporters in multidrug-resistant cancer. Nat. Rev. Cancer 18, 452–464 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Adamska, A. & Falasca, M. ATP-binding cassette transporters in progression and clinical outcome of pancreatic cancer: what is the way forward? World J. Gastroenterol. 24, 3222–3238 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. De Wolf, C. et al. Contribution of the drug transporter ABCG2 (breast cancer resistance protein) to resistance against anticancer nucleosides. Mol. Cancer Ther. 7, 3092–3102 (2008).

    PubMed  Google Scholar 

  38. Rudin, D., Li, L., Niu, N., Kalari, R. K. & Gilbert, A. J. Gemcitabine cytotoxicity: interaction of efflux and deamination. J. Drug Metab. Toxicol. 2, 1000107 (2011).

    CAS  Google Scholar 

  39. Zhou, J. et al. Persistence of side population cells with high drug efflux capacity in pancreatic cancer. World J. Gastroenterol. 14, 925–930 (2008).

    PubMed  PubMed Central  Google Scholar 

  40. Zhang, Z., Wang, J., Shen, B., Peng, C. & Zheng, M. The ABCC4 gene is a promising target for pancreatic cancer therapy. Gene 491, 194–199 (2012).

    CAS  PubMed  Google Scholar 

  41. Hsu, M. C. et al. Protein arginine methyltransferase 3 enhances chemoresistance in pancreatic cancer by methylating hnRNPA1 to increase ABCG2 expression. Cancers 11, 8 (2019).

    CAS  Google Scholar 

  42. Golebiewska, A., Brons, N. H. C., Bjerkvig, R. & Niclou, S. P. Critical appraisal of the side population assay in stem cell and cancer stem cell research. Cell Stem Cell 8, 136–147 (2011).

    CAS  PubMed  Google Scholar 

  43. Zhou, S. et al. The ABC transporter Bcrp1/ABCG2 is expressed in a wide variety of stem cells and is a molecular determinant of the side-population phenotype. Nat. Med. 7, 1028–1034 (2001).

    CAS  PubMed  Google Scholar 

  44. Toyoda, Y., Takada, T. & Suzuki, H. Inhibitors of human ABCG2: from technical background to recent updates with clinical implications. Front. Pharmacol. 10, 208 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Kim, Y. & Kumar, S. CD44-mediated adhesion to hyaluronic acid contributes to mechanosensing and invasive motility. Mol. Cancer Res. 12, 1416–1429 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Geiger, B., Spatz, J. P. & Bershadsky, A. D. Environmental sensing through focal adhesions. Nat. Rev. Mol. Cell Biol. 10, 21–33 (2009).

    CAS  PubMed  Google Scholar 

  47. Nieberler, M. et al. Exploring the role of RGD-recognizing integrins in cancer. Cancers 9, 116 (2017).

    PubMed  PubMed Central  Google Scholar 

  48. Chen, C., Zhao, S., Karnad, A. & Freeman, J. W. The biology and role of CD44 in cancer progression: therapeutic implications. J. Hematol. Oncol. 11, 64 (2018).

    PubMed  PubMed Central  Google Scholar 

  49. Chung, W.-M. et al. Increase paclitaxel sensitivity to better suppress serous epithelial ovarian cancer via ablating androgen receptor/aryl hydrocarbon receptor-ABCG2 axis. Cancers 11, 463 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Sheridan, C. Pancreatic cancer provides testbed for first mechanotherapeutics. Nat. Biotechnol. 37, 829–831 (2019).

    CAS  PubMed  Google Scholar 

  51. Neal, J. T. et al. Organoid modeling of the tumor immune microenvironment. Cell 175, 1972–1988.e16 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Goldman, M. J. et al. Visualizing and interpreting cancer genomics data via the Xena platform. Nat. Biotechnol. 38, 675–678 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Vivian, J. et al. Toil enables reproducible, open source, big biomedical data analyses. Nat. Biotechnol. 35, 314–316 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank T. Lozanoski, E. Rankin, S. Natarajan and J. Roth for insightful conversations and editing of the paper. We are grateful to the patients who donated tissue samples used for this research. We thank the Stanford Tissue Bank for their assistance in procuring patient tissue samples. We thank Z. Ma for help with barcode DNA sequencing. We thank P. Chu at the Stanford Human Pathology/Histology Service Center for assistance with tissue and organoid paraffin embedding and sectioning. B.L.L. acknowledges financial support from the Siebel Scholars Program and Stanford Bio-X Bowes Graduate Fellowship. D.Z. acknowledges financial support from the Stanford Graduate Fellowship and the National Science Foundation Graduate Research Fellowship Program. C.H.-L. acknowledges financial support from Fundación Alfonso Martín Escudero. B.A.K. acknowledges financial support from the Stanford Bio-X Bowes Graduate Fellowship. K. Karlsson acknowledges financial support from the Swedish Research Council (2018-00454). K.C.K. acknowledges financial support from the National Science Foundation Graduate Research Fellowship Program. M.S.H. acknowledges financial support from the National Institutes of Health F31 Pre-Doctoral Fellowship (NS132505), the Stanford ChEM-H O’Leary-Thiry Graduate Fellowship and the Gerald J. Lieberman Fellowship. This work was supported by funding from the National Institutes of Health (R01 EB027171 to S.C.H.; R01CA2515143, U01CA217851 and U54CA224081 to C.J.K.; U01CA217851 and DP1CA238296 to C.C.), Stand Up to Cancer and Cancer Research UK (C.J.K.), and the National Science Foundation (CBET 2033302 to S.C.H.). Flow cytometry data were collected on BD FACSymphony A5 in the Shared FACS Facility, obtained using a NIH S10 Shared Instrument Grant (1S10OD026831-01). Part of this work was performed at the Stanford Nano Shared Facilities (SNSF), supported by the National Science Foundation under award ECCS-2026822.

Author information

Authors and Affiliations

Authors

Contributions

B.L.L., D.Z., C.H.-L., A.E.G., B.A.K., A.R.S. and S.C.H. designed the research. B.L.L., D.Z., C.H.-L., A.E.G., B.A.K., K. Karagyozova, K.C.K., M.S.H., C.L., G.K. and C.M.M. conducted experiments. B.L.L., D.Z., C.H.-L., A.E.G., K. Karlsson, C.L., G.K. and C.M.M. analysed data. K. Karlsson, A.R.S., C.C. and C.J.K. provided organoids and reagents. B.L.L., D.Z., C.H.-L. and A.E.G. wrote the paper. B.L.L., D.Z., C.H.-L., A.E.G., B.A.K., K. Karlsson, A.R.S., K. Karagyozova, K.C.K., M.S.H., C.L., G.K., C.M.M., P.L.B., C.C., C.J.K. and S.C.H. edited and approved the final paper.

Corresponding author

Correspondence to Sarah C. Heilshorn.

Ethics declarations

Competing interests

S.C.H. has a patent application pending related to the hydrogel formulations used in this paper, as described in an international patent application PCT/US/2021/057925 with an international filing date of 3 November 2021. This application has been submitted to the U.S. Patent Office and published on 16 November 2023 as US 2023-0365940 A1, and this same application has entered the national phase in China (publication number 116829126), Singapore (application number 11202303493U), Europe (publication number 4240327), Japan (serial number 2023-526661), Canada (serial number 3,196,621), Israel (serial number 302453), South Korea (serial number 10-2023-7018686) and Australia (serial number 2021376143). The named inventors are S. Heilshorn, R. Suhar and D. Hunt, and the named applicant is the Board of Trustees of the Leland Stanford Junior University. C.C. is an advisor and holds equity in Grail, Ravel and Deepcell, and is an advisor to Genentech and NanoString. C.J.K. is an advisor and holds equity in Surrozen, Mozart and NextVivo. The other authors declare no competing interests.

Peer review

Peer review information

Nature Materials thanks Toshiro Sato 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 Matrix stiffness mediates PDAC organoid clonal heterogeneity.

a, Schematic summarizing the protocol and potential outcomes of genetically barcoded PDAC organoid expansion within Cultrex and HELP matrices. b, Number of barcodes present within PDAC organoid populations expanded within Cultrex and HELP matrices for one to three passages (N = 3 independent biological replicates per matrix type, mean ± 95% confidence interval, center line represents linear fit for each matrix over three passages, shaded region represents 95% confidence interval of linear fit). Statistical analysis was performed using an ordinary one-way ANOVA with Tukey multiple comparisons correction between data points at passage three (****P < 0.0001 for comparison between HELP High and Cultrex/HELP Low, comparison between HELP Low and Cultrex was not significantly different). The initial Parent population was the same for all matrices (N = 1). c, Slope of linear fit from b for each matrix (N = 3 independent biological replicates per matrix type, mean ± 95% confidence interval, ordinary one-way ANOVA with Tukey multiple comparisons correction, ****P < 0.0001, NS, not significant). d, Frequencies of barcodes present in the indicated matrix at passage three. Barcodes are listed in rank order and colored bars correspond to barcodes unique to only the indicated matrix. The number of these unique barcodes and their cumulative frequency are reported for each matrix. e, Venn diagram of barcodes at passage three across indicated matrices. f, Correlation plots comparing barcode frequencies across Cultrex and HELP at passage three. Pearson r values are reported for each pairing. Dashed line represents perfect correlation. In d-f, only barcodes with a frequency >0.01% across all three biological replicates at passage three were included.

Source Data

Extended Data Fig. 2 Long-term drug efflux inhibitor treatment exacerbates PDAC organoid chemoresistance.

a, PDAC viability following treatment with DMSO (control, normalized to 1), 66 nM gemcitabine (Gem), or 66 nM gemcitabine + 20 μM Ko143 (N = 4-5 independent experimental replicate hydrogels, mean ± s.d.). b, PDAC toxicity following treatment with DMSO (control, normalized to 0), 100 nM gemcitabine, 100 nM gemcitabine + 20 μM Ko143, or 3,333 nM gemcitabine (positive control, normalized to 1) (N = 4-5 independent experimental replicate hydrogels, mean ± s.d.). In a and b, PDAC organoids were expanded for four passages in HELP Low (left) or HELP High (right) prior to gemcitabine (+ Ko143) treatment for six days on single cells during log-phase growth (a) or for three days following formation of ~75-μm diameter multicellular organoids (b). Statistical analysis comparing experimental gemcitabine treatment and gemcitabine + Ko143 treatment was performed using an unpaired two-tailed t-test (*P < 0.05, ****P < 0.0001), and the fold change between these two conditions is reported for each comparison. c, Illustrated summary of results from a and b. d,e, qPCR (d) and Western blot (e) quantification of mRNA and protein-level ABC-family drug efflux transporter expression in PDAC organoids expanded within HELP Low or HELP High for four passages and treated with either DMSO or 20 μM Ko143 (N = 4 independent experimental replicates, qPCR: mean ± 95% confidence interval, Western: mean ± s.d.). Statistical analysis comparing DMSO vs. Ko143 treatment for each matrix was performed using an unpaired two-tailed t-test (*P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001). qPCR data are normalized to GAPDH gene expression and respective marker expression of Cultrex DMSO samples. Western data are normalized to β-actin expression and DMSO samples for each marker and matrix. PDAC cells were treated with Ko143 throughout single-cell log-phase growth for six days. f, Illustrated summary of results from d and e.

Source Data

Extended Data Fig. 3 Influence of RGD ligand on PDAC organoid chemoresistance.

a, Schematic of recombinant elastin-like protein (ELP) component of HELP, which can be engineered to display a non-interactive, scrambled RDG sequence, resulting in a HELP matrix without RGD, but with identical mechanical properties and HA concentration. b, Stiffness measurements of HELP RDG matrices stiffness-matched to HELP Low and HELP High (N = 3 independent experimental replicate hydrogels, mean ± s.d., unpaired two-tailed t-test, ****P < 0.0001). c, Representative bright-field images of PDAC organoids expanded within HELP RDG Low (top) or HELP RDG High (bottom) for four passages. Scale bar, 250 μm. d, qPCR quantification of mRNA-level ABCG2 expression in PDAC organoids expanded within HELP RDG Low or High for four passages (N = 4 independent experimental replicate hydrogels, mean ± 95% confidence interval). Statistical analysis comparing marker expression in Low vs. High matrices was performed using an unpaired two-tailed t-test (**P < 0.01). All data are normalized to GAPDH gene expression and respective marker expression in the PDAC organoid parent population cultured within Cultrex prior to expansion in HELP RDG (that is, Cultrex P0). e, Single-cell-level (left) and organoid-level (right) gemcitabine dose-response curves for PDAC organoids expanded within HELP RDG Low or High for four passages. Each data point represents the mean ± s.e.m. (N = 4 independent experimental replicate hydrogels, solid center line is nonlinear least squares regression of data; shaded region represents 95% confidence bands of nonlinear fit; data are normalized to positive controls (DMSO for single cells, 3,333 nM gemcitabine for organoids). f, Gemcitabine IC50 values calculated from nonlinear fit of dose-response curves shown in e for single-cell (left) and organoid (right) drug treatment in HELP RDG compared to HELP containing RGD. Each bar represents the mean ± s.e.m. (N = 4 independent experimental replicate hydrogels, unpaired two-tailed t-test between Low and High for each matrix variation, **P < 0.01, ***P < 0.001, ****P < 0.0001).

Source Data

Supplementary information

Supplementary Information

Supplementary Figs. 1–31, Tables 1–6 and Methods.

Reporting Summary

Supplementary Data 1

Statistical source data for Supplementary Figs. 1–31.

Source data

Source Data Figs. 1–6 and Extended Data Figs. 1–3

Statistical source data for Figs. 1–6 and Extended Data Figs. 1–3.

Source Data Fig. 1

Western blots.

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

LeSavage, B.L., Zhang, D., Huerta-López, C. et al. Engineered matrices reveal stiffness-mediated chemoresistance in patient-derived pancreatic cancer organoids. Nat. Mater. 23, 1138–1149 (2024). https://doi.org/10.1038/s41563-024-01908-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41563-024-01908-x

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