Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2024

Open Access 01-12-2024 | Colorectal Cancer | Research

Organoids as a biomarker for personalized treatment in metastatic colorectal cancer: drug screen optimization and correlation with patient response

Authors: Lidwien P. Smabers, Emerens Wensink, Carla S. Verissimo, Esmee Koedoot, Katerina-Chara Pitsa, Maarten A. Huismans, Celia Higuera Barón, Mayke Doorn, Liselot B. Valkenburg-van Iersel, Geert A. Cirkel, Anneta Brousali, René Overmeer, Miriam Koopman, Manon N. Braat, Bas Penning de Vries, Sjoerd G. Elias, Robert G. Vries, Onno Kranenburg, Sylvia F. Boj, Jeanine M. Roodhart

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2024

Login to get access

Abstract

Background

The inability to predict treatment response of colorectal cancer patients results in unnecessary toxicity, decreased efficacy and survival. Response testing on patient-derived organoids (PDOs) is a promising biomarker for treatment efficacy. The aim of this study is to optimize PDO drug screening methods for correlation with patient response and explore the potential to predict responses to standard chemotherapies.

Methods

We optimized drug screen methods on 5–11 PDOs per condition of the complete set of 23 PDOs from patients treated for metastatic colorectal cancer (mCRC). PDOs were exposed to 5-fluorouracil (5-FU), irinotecan- and oxaliplatin-based chemotherapy. We compared medium with and without N-acetylcysteine (NAC), different readouts and different combination treatment set-ups to capture the strongest association with patient response. We expanded the screens using the optimized methods for all PDOs. Organoid sensitivity was correlated to the patient’s response, determined by % change in the size of target lesions. We assessed organoid sensitivity in relation to prior exposure to chemotherapy, mutational status and sidedness.

Results

Drug screen optimization involved excluding N-acetylcysteine from the medium and biphasic curve fitting for 5-FU & oxaliplatin combination screens. CellTiter-Glo measurements were comparable with CyQUANT and did not affect the correlation with patient response. Furthermore, the correlation improved with application of growth rate metrics, when 5-FU & oxaliplatin was screened in a ratio, and 5-FU & SN-38 using a fixed dose of SN-38. Area under the curve was the most robust drug response curve metric. After optimization, organoid and patient response showed a correlation coefficient of 0.58 for 5-FU (n = 6, 95% CI -0.44,0.95), 0.61 for irinotecan- (n = 10, 95% CI -0.03,0.90) and 0.60 for oxaliplatin-based chemotherapy (n = 11, 95% CI -0.01,0.88). Median progression-free survival of patients with resistant PDOs to oxaliplatin-based chemotherapy was significantly shorter than sensitive PDOs (3.3 vs 10.9 months, p = 0.007). Increased resistance to 5-FU in patients with prior exposure to 5-FU/capecitabine was adequately reflected in PDOs (p = 0.003).

Conclusions

Our study emphasizes the critical impact of the screening methods for determining correlation between PDO drug screens and mCRC patient outcomes. Our 5-step optimization strategy provides a basis for future research on the clinical utility of PDO screens.
Appendix
Available only for authorised users
Literature
1.
go back to reference Yamazaki K, Nagase M, Tamagawa H, Ueda S, Tamura T, Murata K, et al. Randomized phase III study of bevacizumab plus FOLFIRI and bevacizumab plus mFOLFOX6 as first-line treatment for patients with metastatic colorectal cancer (WJOG4407G). Ann Oncol. 2016;27(8):1539–46.PubMedCrossRef Yamazaki K, Nagase M, Tamagawa H, Ueda S, Tamura T, Murata K, et al. Randomized phase III study of bevacizumab plus FOLFIRI and bevacizumab plus mFOLFOX6 as first-line treatment for patients with metastatic colorectal cancer (WJOG4407G). Ann Oncol. 2016;27(8):1539–46.PubMedCrossRef
2.
go back to reference Venook AP, Niedzwiecki D, Lenz HJ, Innocenti F, Fruth B, Meyerhardt JA, et al. Effect of first-line chemotherapy combined with Cetuximab or bevacizumab on overall survival in patients with KRAS wild-type advanced or metastatic colorectal Cancer: a randomized clinical trial. JAMA. 2017;317(23):2392–401.PubMedPubMedCentralCrossRef Venook AP, Niedzwiecki D, Lenz HJ, Innocenti F, Fruth B, Meyerhardt JA, et al. Effect of first-line chemotherapy combined with Cetuximab or bevacizumab on overall survival in patients with KRAS wild-type advanced or metastatic colorectal Cancer: a randomized clinical trial. JAMA. 2017;317(23):2392–401.PubMedPubMedCentralCrossRef
3.
go back to reference Kwakman JJM, Simkens LHJ, van Rooijen JM, van de Wouw AJ, Ten Tije AJ, Creemers GJM, et al. Randomized phase III trial of S-1 versus capecitabine in the first-line treatment of metastatic colorectal cancer: SALTO study by the Dutch colorectal Cancer group. Ann Oncol. 2017;28(6):1288–93.PubMedCrossRef Kwakman JJM, Simkens LHJ, van Rooijen JM, van de Wouw AJ, Ten Tije AJ, Creemers GJM, et al. Randomized phase III trial of S-1 versus capecitabine in the first-line treatment of metastatic colorectal cancer: SALTO study by the Dutch colorectal Cancer group. Ann Oncol. 2017;28(6):1288–93.PubMedCrossRef
4.
go back to reference Overman MJ, Lonardi S, Wong KYM, Lenz HJ, Gelsomino F, Aglietta M, et al. Durable clinical benefit with Nivolumab plus Ipilimumab in DNA mismatch repair-deficient/microsatellite instability-high metastatic colorectal Cancer. J Clin Oncol. 2018;36(8):773–9.PubMedCrossRef Overman MJ, Lonardi S, Wong KYM, Lenz HJ, Gelsomino F, Aglietta M, et al. Durable clinical benefit with Nivolumab plus Ipilimumab in DNA mismatch repair-deficient/microsatellite instability-high metastatic colorectal Cancer. J Clin Oncol. 2018;36(8):773–9.PubMedCrossRef
6.
7.
go back to reference Wensink GE, Elias SG, Mullenders J, Koopman M, Boj SF, Kranenburg OW, Roodhart JML. Patient-derived organoids as a predictive biomarker for treatment response in cancer patients. Npj Precision Oncol. 2021;5(1):30.CrossRef Wensink GE, Elias SG, Mullenders J, Koopman M, Boj SF, Kranenburg OW, Roodhart JML. Patient-derived organoids as a predictive biomarker for treatment response in cancer patients. Npj Precision Oncol. 2021;5(1):30.CrossRef
9.
go back to reference Li M, Izpisua Belmonte JC. Organoids - preclinical models of human disease. Reply N Engl J Med. 2019;380(20):1982.PubMed Li M, Izpisua Belmonte JC. Organoids - preclinical models of human disease. Reply N Engl J Med. 2019;380(20):1982.PubMed
10.
go back to reference van de Wetering M, Francies HE, Francis JM, Bounova G, Iorio F, Pronk A, et al. Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell. 2015;161(4):933–45.PubMedPubMedCentralCrossRef van de Wetering M, Francies HE, Francis JM, Bounova G, Iorio F, Pronk A, et al. Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell. 2015;161(4):933–45.PubMedPubMedCentralCrossRef
11.
go back to reference Sato T, Stange DE, Ferrante M, Vries RG, Van Es JH, Van den Brink S, et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett’s epithelium. Gastroenterology. 2011;141(5):1762–72.PubMedCrossRef Sato T, Stange DE, Ferrante M, Vries RG, Van Es JH, Van den Brink S, et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett’s epithelium. Gastroenterology. 2011;141(5):1762–72.PubMedCrossRef
13.
go back to reference Mertens S, Huismans MA, Verissimo CS, Ponsioen B, Overmeer R, Proost N, et al. Drug-repurposing screen on patient-derived organoids identifies therapy-induced vulnerability in KRAS-mutant colon cancer. Cell Rep. 2023;42(4):112324.PubMedCrossRef Mertens S, Huismans MA, Verissimo CS, Ponsioen B, Overmeer R, Proost N, et al. Drug-repurposing screen on patient-derived organoids identifies therapy-induced vulnerability in KRAS-mutant colon cancer. Cell Rep. 2023;42(4):112324.PubMedCrossRef
14.
go back to reference Ganesh K, Wu C, O’Rourke KP, Szeglin BC, Zheng Y, Sauvé C-EG, et al. A rectal cancer organoid platform to study individual responses to chemoradiation. Nat Med. 2019;25(10):1607–14.PubMedPubMedCentralCrossRef Ganesh K, Wu C, O’Rourke KP, Szeglin BC, Zheng Y, Sauvé C-EG, et al. A rectal cancer organoid platform to study individual responses to chemoradiation. Nat Med. 2019;25(10):1607–14.PubMedPubMedCentralCrossRef
15.
go back to reference Cho YW, Min DW, Kim HP, An Y, Kim S, Youk J, et al. Patient-derived organoids as a preclinical platform for precision medicine in colorectal cancer. Mol Oncol. 2022;16(12):2396–412.PubMedPubMedCentralCrossRef Cho YW, Min DW, Kim HP, An Y, Kim S, Youk J, et al. Patient-derived organoids as a preclinical platform for precision medicine in colorectal cancer. Mol Oncol. 2022;16(12):2396–412.PubMedPubMedCentralCrossRef
16.
go back to reference Wang T, Pan W, Zheng H, Zheng H, Wang Z, Li JJ, et al. Accuracy of using a patient-derived tumor organoid culture model to predict the response to chemotherapy regimens in stage IV colorectal Cancer: a blinded study. Dis Colon Rectum. 2021;64(7):833–50.PubMedCrossRef Wang T, Pan W, Zheng H, Zheng H, Wang Z, Li JJ, et al. Accuracy of using a patient-derived tumor organoid culture model to predict the response to chemotherapy regimens in stage IV colorectal Cancer: a blinded study. Dis Colon Rectum. 2021;64(7):833–50.PubMedCrossRef
17.
go back to reference Vlachogiannis G, Hedayat S, Vatsiou A, Jamin Y, Fernández-Mateos J, Khan K, et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science. 2018;359(6378):920–6.ADSPubMedPubMedCentralCrossRef Vlachogiannis G, Hedayat S, Vatsiou A, Jamin Y, Fernández-Mateos J, Khan K, et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science. 2018;359(6378):920–6.ADSPubMedPubMedCentralCrossRef
18.
go back to reference Yao L, Zao XL, Pan XF, Zhang HG, Wang FJ, Qiao PF. Application of tumoroids derived from advanced colorectal cancer patients to predict individual response to chemotherapy. J Chemother. 2022;1–13. Yao L, Zao XL, Pan XF, Zhang HG, Wang FJ, Qiao PF. Application of tumoroids derived from advanced colorectal cancer patients to predict individual response to chemotherapy. J Chemother. 2022;1–13.
19.
go back to reference Yao Y, Xu X, Yang L, Zhu J, Wan J, Shen L, et al. Patient-derived organoids predict Chemoradiation responses of locally advanced rectal Cancer. Cell Stem Cell. 2020;26(1):17–26.e6.PubMedCrossRef Yao Y, Xu X, Yang L, Zhu J, Wan J, Shen L, et al. Patient-derived organoids predict Chemoradiation responses of locally advanced rectal Cancer. Cell Stem Cell. 2020;26(1):17–26.e6.PubMedCrossRef
20.
go back to reference Narasimhan V, Wright JA, Churchill M, Wang T, Rosati R, Lannagan TRM, et al. Medium-throughput drug screening of patient-derived organoids from colorectal peritoneal metastases to direct personalized therapy. Clin Cancer Res. 2020;26(14):3662–70.PubMedPubMedCentralCrossRef Narasimhan V, Wright JA, Churchill M, Wang T, Rosati R, Lannagan TRM, et al. Medium-throughput drug screening of patient-derived organoids from colorectal peritoneal metastases to direct personalized therapy. Clin Cancer Res. 2020;26(14):3662–70.PubMedPubMedCentralCrossRef
21.
go back to reference Mo S, Tang P, Luo W, Zhang L, Li Y, Hu X, et al. Patient-derived organoids from colorectal Cancer with paired liver metastasis reveal tumor heterogeneity and predict response to chemotherapy. Adv Sci (Weinh). 2022;9(31):e2204097.PubMedCrossRef Mo S, Tang P, Luo W, Zhang L, Li Y, Hu X, et al. Patient-derived organoids from colorectal Cancer with paired liver metastasis reveal tumor heterogeneity and predict response to chemotherapy. Adv Sci (Weinh). 2022;9(31):e2204097.PubMedCrossRef
22.
go back to reference Tang Y, Wang T, Hu Y, Ji H, Yan B, Hu X, et al. Cutoff value of IC(50) for drug sensitivity in patient-derived tumor organoids in colorectal cancer. iScience. 2023;26(7):107116.ADSPubMedPubMedCentralCrossRef Tang Y, Wang T, Hu Y, Ji H, Yan B, Hu X, et al. Cutoff value of IC(50) for drug sensitivity in patient-derived tumor organoids in colorectal cancer. iScience. 2023;26(7):107116.ADSPubMedPubMedCentralCrossRef
23.
go back to reference Ooft SN, Weeber F, Dijkstra KK, McLean CM, Kaing S, van Werkhoven E, et al. Patient-derived organoids can predict response to chemotherapy in metastatic colorectal cancer patients. Sci Transl Med. 2019;11(513). Ooft SN, Weeber F, Dijkstra KK, McLean CM, Kaing S, van Werkhoven E, et al. Patient-derived organoids can predict response to chemotherapy in metastatic colorectal cancer patients. Sci Transl Med. 2019;11(513).
24.
go back to reference Driehuis E, Kretzschmar K, Clevers H. Establishment of patient-derived cancer organoids for drug-screening applications. Nat Protoc. 2020;15(10):3380–409.PubMedCrossRef Driehuis E, Kretzschmar K, Clevers H. Establishment of patient-derived cancer organoids for drug-screening applications. Nat Protoc. 2020;15(10):3380–409.PubMedCrossRef
25.
go back to reference Shin YJ, Jo EH, Oh Y, Kim DS, Hyun S, Yu A, et al. Improved drug-response prediction model of APC mutant Colon Cancer patient-derived organoids for precision medicine. Cancers (Basel). 2023;15(23). Shin YJ, Jo EH, Oh Y, Kim DS, Hyun S, Yu A, et al. Improved drug-response prediction model of APC mutant Colon Cancer patient-derived organoids for precision medicine. Cancers (Basel). 2023;15(23).
26.
go back to reference Li X, Francies HE, Secrier M, Perner J, Miremadi A, Galeano-Dalmau N, et al. Organoid cultures recapitulate esophageal adenocarcinoma heterogeneity providing a model for clonality studies and precision therapeutics. Nat Commun. 2018;9(1):2983.ADSPubMedPubMedCentralCrossRef Li X, Francies HE, Secrier M, Perner J, Miremadi A, Galeano-Dalmau N, et al. Organoid cultures recapitulate esophageal adenocarcinoma heterogeneity providing a model for clonality studies and precision therapeutics. Nat Commun. 2018;9(1):2983.ADSPubMedPubMedCentralCrossRef
27.
go back to reference Mohn C, Hacker HG, Hilger RA, Gutschow M, Jaehde U. Defining the role of MRP-mediated efflux and glutathione in detoxification of oxaliplatin. Pharmazie. 2013;68(7):622–7.PubMed Mohn C, Hacker HG, Hilger RA, Gutschow M, Jaehde U. Defining the role of MRP-mediated efflux and glutathione in detoxification of oxaliplatin. Pharmazie. 2013;68(7):622–7.PubMed
28.
go back to reference Laoukili J, Constantinides A, Wassenaar ECE, Elias SG, Raats DAE, van Schelven SJ, et al. Peritoneal metastases from colorectal cancer belong to consensus molecular subtype 4 and are sensitised to oxaliplatin by inhibiting reducing capacity. Br J Cancer. 2022;126(12):1824–33.PubMedPubMedCentralCrossRef Laoukili J, Constantinides A, Wassenaar ECE, Elias SG, Raats DAE, van Schelven SJ, et al. Peritoneal metastases from colorectal cancer belong to consensus molecular subtype 4 and are sensitised to oxaliplatin by inhibiting reducing capacity. Br J Cancer. 2022;126(12):1824–33.PubMedPubMedCentralCrossRef
30.
go back to reference Sahin IH, Klostergaard J. BRAF mutations as actionable targets: a paradigm shift in the Management of Colorectal Cancer and Novel Avenues. JCO Oncol Pract. 2021;17(12):723–30.PubMedCrossRef Sahin IH, Klostergaard J. BRAF mutations as actionable targets: a paradigm shift in the Management of Colorectal Cancer and Novel Avenues. JCO Oncol Pract. 2021;17(12):723–30.PubMedCrossRef
31.
32.
go back to reference Hafner M, Niepel M, Chung M, Sorger PK. Growth rate inhibition metrics correct for confounders in measuring sensitivity to cancer drugs. Nat Methods. 2016;13(6):521–7.PubMedPubMedCentralCrossRef Hafner M, Niepel M, Chung M, Sorger PK. Growth rate inhibition metrics correct for confounders in measuring sensitivity to cancer drugs. Nat Methods. 2016;13(6):521–7.PubMedPubMedCentralCrossRef
34.
go back to reference Song SL, Li B, Carvalho MR, Wang HJ, Mao DL, Wei JT, et al. Complex in vitro 3D models of digestive system tumors to advance precision medicine and drug testing: Progress, challenges, and trends. Pharmacol Ther. 2022;239:108276.PubMedCrossRef Song SL, Li B, Carvalho MR, Wang HJ, Mao DL, Wei JT, et al. Complex in vitro 3D models of digestive system tumors to advance precision medicine and drug testing: Progress, challenges, and trends. Pharmacol Ther. 2022;239:108276.PubMedCrossRef
35.
go back to reference Sui Y, Wu Z. Alternative statistical parameter for high-throughput screening assay quality assessment. J Biomol Screen. 2007;12(2):229–34.PubMedCrossRef Sui Y, Wu Z. Alternative statistical parameter for high-throughput screening assay quality assessment. J Biomol Screen. 2007;12(2):229–34.PubMedCrossRef
36.
go back to reference Chan GK, Kleinheinz TL, Peterson D, Moffat JG. A simple high-content cell cycle assay reveals frequent discrepancies between cell number and ATP and MTS proliferation assays. PLoS One. 2013;8(5):e63583.ADSPubMedPubMedCentralCrossRef Chan GK, Kleinheinz TL, Peterson D, Moffat JG. A simple high-content cell cycle assay reveals frequent discrepancies between cell number and ATP and MTS proliferation assays. PLoS One. 2013;8(5):e63583.ADSPubMedPubMedCentralCrossRef
37.
go back to reference Jones LJ, Gray M, Yue ST, Haugland RP, Singer VL. Sensitive determination of cell number using the CyQUANT cell proliferation assay. J Immunol Methods. 2001;254(1–2):85–98.PubMedCrossRef Jones LJ, Gray M, Yue ST, Haugland RP, Singer VL. Sensitive determination of cell number using the CyQUANT cell proliferation assay. J Immunol Methods. 2001;254(1–2):85–98.PubMedCrossRef
38.
go back to reference Alvarez-Varela A, Novellasdemunt L, Barriga FM, Hernando-Momblona X, Canellas-Socias A, Cano-Crespo S, et al. Mex3a marks drug-tolerant persister colorectal cancer cells that mediate relapse after chemotherapy. Nat Cancer. 2022;3(9):1052–70.PubMedCrossRef Alvarez-Varela A, Novellasdemunt L, Barriga FM, Hernando-Momblona X, Canellas-Socias A, Cano-Crespo S, et al. Mex3a marks drug-tolerant persister colorectal cancer cells that mediate relapse after chemotherapy. Nat Cancer. 2022;3(9):1052–70.PubMedCrossRef
39.
go back to reference Luo Z, Wang B, Luo F, Guo Y, Jiang N, Wei J, et al. Establishment of a large-scale patient-derived high-risk colorectal adenoma organoid biobank for high-throughput and high-content drug screening. BMC Med. 2023;21(1):336.PubMedPubMedCentralCrossRef Luo Z, Wang B, Luo F, Guo Y, Jiang N, Wei J, et al. Establishment of a large-scale patient-derived high-risk colorectal adenoma organoid biobank for high-throughput and high-content drug screening. BMC Med. 2023;21(1):336.PubMedPubMedCentralCrossRef
40.
go back to reference Jensen LH, Rogatto SR, Lindebjerg J, Havelund B, Abildgaard C, do Canto LM, et al. Precision medicine applied to metastatic colorectal cancer using tumor-derived organoids and in-vitro sensitivity testing: a phase 2, single-center, open-label, and non-comparative study. J Exp Clin Cancer Res. 2023;42(1):115.PubMedPubMedCentralCrossRef Jensen LH, Rogatto SR, Lindebjerg J, Havelund B, Abildgaard C, do Canto LM, et al. Precision medicine applied to metastatic colorectal cancer using tumor-derived organoids and in-vitro sensitivity testing: a phase 2, single-center, open-label, and non-comparative study. J Exp Clin Cancer Res. 2023;42(1):115.PubMedPubMedCentralCrossRef
41.
go back to reference Zoetemelk M, Ramzy GM, Rausch M, Nowak-Sliwinska P. Drug-drug interactions of Irinotecan, 5-fluorouracil, Folinic acid and Oxaliplatin and its activity in colorectal carcinoma treatment. Molecules. 2020;25(11). Zoetemelk M, Ramzy GM, Rausch M, Nowak-Sliwinska P. Drug-drug interactions of Irinotecan, 5-fluorouracil, Folinic acid and Oxaliplatin and its activity in colorectal carcinoma treatment. Molecules. 2020;25(11).
42.
go back to reference Tejpar S, Stintzing S, Ciardiello F, Tabernero J, Van Cutsem E, Beier F, et al. Prognostic and predictive relevance of primary tumor location in patients with RAS wild-type metastatic colorectal Cancer: retrospective analyses of the CRYSTAL and FIRE-3 trials. JAMA Oncol. 2017;3(2):194–201.PubMedPubMedCentralCrossRef Tejpar S, Stintzing S, Ciardiello F, Tabernero J, Van Cutsem E, Beier F, et al. Prognostic and predictive relevance of primary tumor location in patients with RAS wild-type metastatic colorectal Cancer: retrospective analyses of the CRYSTAL and FIRE-3 trials. JAMA Oncol. 2017;3(2):194–201.PubMedPubMedCentralCrossRef
43.
go back to reference Holch JW, Ricard I, Stintzing S, Modest DP, Heinemann V. The relevance of primary tumour location in patients with metastatic colorectal cancer: a meta-analysis of first-line clinical trials. Eur J Cancer. 2017;70:87–98.PubMedCrossRef Holch JW, Ricard I, Stintzing S, Modest DP, Heinemann V. The relevance of primary tumour location in patients with metastatic colorectal cancer: a meta-analysis of first-line clinical trials. Eur J Cancer. 2017;70:87–98.PubMedCrossRef
44.
go back to reference Martini G, Belli V, Napolitano S, Ciaramella V, Ciardiello D, Belli A, et al. Establishment of patient-derived tumor organoids to functionally inform treatment decisions in metastatic colorectal cancer. ESMO Open. 2023;8(3):101198.PubMedPubMedCentralCrossRef Martini G, Belli V, Napolitano S, Ciaramella V, Ciardiello D, Belli A, et al. Establishment of patient-derived tumor organoids to functionally inform treatment decisions in metastatic colorectal cancer. ESMO Open. 2023;8(3):101198.PubMedPubMedCentralCrossRef
45.
go back to reference Tan T, Mouradov D, Lee M, Gard G, Hirokawa Y, Li S, et al. Unified framework for patient-derived, tumor-organoid-based predictive testing of standard-of-care therapies in metastatic colorectal cancer. Cell Rep Med. 2023;4(12):101335.PubMedPubMedCentralCrossRef Tan T, Mouradov D, Lee M, Gard G, Hirokawa Y, Li S, et al. Unified framework for patient-derived, tumor-organoid-based predictive testing of standard-of-care therapies in metastatic colorectal cancer. Cell Rep Med. 2023;4(12):101335.PubMedPubMedCentralCrossRef
46.
go back to reference Sachs N, de Ligt J, Kopper O, Gogola E, Bounova G, Weeber F, et al. A living biobank of breast Cancer organoids captures disease heterogeneity. Cell. 2018;172(1–2):373–86 e10.PubMedCrossRef Sachs N, de Ligt J, Kopper O, Gogola E, Bounova G, Weeber F, et al. A living biobank of breast Cancer organoids captures disease heterogeneity. Cell. 2018;172(1–2):373–86 e10.PubMedCrossRef
47.
go back to reference de Witte CJ, Espejo Valle-Inclan J, Hami N, Lohmussaar K, Kopper O, Vreuls CPH, et al. Patient-derived ovarian Cancer organoids mimic clinical response and exhibit heterogeneous inter- and Intrapatient drug responses. Cell Rep. 2020;31(11):107762.PubMedCrossRef de Witte CJ, Espejo Valle-Inclan J, Hami N, Lohmussaar K, Kopper O, Vreuls CPH, et al. Patient-derived ovarian Cancer organoids mimic clinical response and exhibit heterogeneous inter- and Intrapatient drug responses. Cell Rep. 2020;31(11):107762.PubMedCrossRef
48.
go back to reference Tiriac H, Belleau P, Engle DD, Plenker D, Deschenes A, Somerville TDD, et al. Organoid profiling identifies common responders to chemotherapy in pancreatic Cancer. Cancer Discov. 2018;8(9):1112–29.PubMedPubMedCentralCrossRef Tiriac H, Belleau P, Engle DD, Plenker D, Deschenes A, Somerville TDD, et al. Organoid profiling identifies common responders to chemotherapy in pancreatic Cancer. Cancer Discov. 2018;8(9):1112–29.PubMedPubMedCentralCrossRef
49.
go back to reference Hsu KS, Adileh M, Martin ML, Makarov V, Chen J, Wu C, et al. Colorectal Cancer develops inherent Radiosensitivity that can be predicted using patient-derived organoids. Cancer Res. 2022;82(12):2298–312.PubMedPubMedCentralCrossRef Hsu KS, Adileh M, Martin ML, Makarov V, Chen J, Wu C, et al. Colorectal Cancer develops inherent Radiosensitivity that can be predicted using patient-derived organoids. Cancer Res. 2022;82(12):2298–312.PubMedPubMedCentralCrossRef
50.
go back to reference Xue W, Wang T, Yao J, Wu W, Chen D, Yan B, et al. Use of patient-derived tumor organoid platform to predict the benefit of postoperative adjuvant chemotherapy for poor responders to neoadjuvant chemoradiotherapy in locally advanced rectal cancer. Bioeng Transl Med. 2023;8(6):e10586.PubMedPubMedCentralCrossRef Xue W, Wang T, Yao J, Wu W, Chen D, Yan B, et al. Use of patient-derived tumor organoid platform to predict the benefit of postoperative adjuvant chemotherapy for poor responders to neoadjuvant chemoradiotherapy in locally advanced rectal cancer. Bioeng Transl Med. 2023;8(6):e10586.PubMedPubMedCentralCrossRef
51.
go back to reference Ooft SN, Weeber F, Schipper L, Dijkstra KK, McLean CM, Kaing S, et al. Prospective experimental treatment of colorectal cancer patients based on organoid drug responses. ESMO Open. 2021;6(3):100103.PubMedPubMedCentralCrossRef Ooft SN, Weeber F, Schipper L, Dijkstra KK, McLean CM, Kaing S, et al. Prospective experimental treatment of colorectal cancer patients based on organoid drug responses. ESMO Open. 2021;6(3):100103.PubMedPubMedCentralCrossRef
Metadata
Title
Organoids as a biomarker for personalized treatment in metastatic colorectal cancer: drug screen optimization and correlation with patient response
Authors
Lidwien P. Smabers
Emerens Wensink
Carla S. Verissimo
Esmee Koedoot
Katerina-Chara Pitsa
Maarten A. Huismans
Celia Higuera Barón
Mayke Doorn
Liselot B. Valkenburg-van Iersel
Geert A. Cirkel
Anneta Brousali
René Overmeer
Miriam Koopman
Manon N. Braat
Bas Penning de Vries
Sjoerd G. Elias
Robert G. Vries
Onno Kranenburg
Sylvia F. Boj
Jeanine M. Roodhart
Publication date
01-12-2024
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2024
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-024-02980-6

Other articles of this Issue 1/2024

Journal of Experimental & Clinical Cancer Research 1/2024 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine