Skip to main content
Top
Published in: BMC Cancer 1/2023

Open Access 01-12-2023 | Gastric Cancer | Review

Promising preclinical patient-derived organoid (PDO) and xenograft (PDX) models in upper gastrointestinal cancers: progress and challenges

Authors: Jing Gao, Jianqiang Lan, Haiyan Liao, Fang Yang, Pei Qiu, Feng Jin, Shubin Wang, Lin Shen, Tengfei Chao, Cheng Zhang, Yu Zhu

Published in: BMC Cancer | Issue 1/2023

Login to get access

Abstract

Gastrointestinal (GI) cancers (gastric cancer, oesophageal cancer, liver cancer, colorectal cancer, etc.) are the most common cancers with the highest morbidity and mortality in the world. The therapy for most GI cancers is difficult and is associated with a poor prognosis. In China, upper GI cancers, mainly gastric cancer (GC) and oesophageal cancer (EC), are very common due to Chinese people’s characteristics, and more than half of patients are diagnosed with distant metastatic or locally advanced disease. Compared to other solid cancers, such as lung cancer and breast cancer, personalized therapies, especially targeted therapy and immunotherapy, in GC and EC are relatively lacking, leading to poor prognosis. For a long time, most studies were carried out by using in vitro cancer cell lines or in vivo cell line-derived xenograft models, which are unable to reproduce the characteristics of tumours derived from patients, leading to the possible misguidance of subsequent clinical validation. The patient-derived models represented by patient-derived organoid (PDO) and xenograft (PDX) models, known for their high preservation of patient tumour features, have emerged as a very popular platform that has been widely used in numerous studies, especially in the research and development of antitumour drugs and personalized medicine. Herein, based on some of the available published literature, we review the research and application status of PDO and PDX models in GC and EC, as well as detail their future challenges and prospects, to promote their use in basic and translational studies or personalized therapy.
Literature
1.
go back to reference Dizdar Ö, Kılıçkap S. Global epidemiology of gastrointestinal cancers. Textbook of Gastrointestinal Oncology, 2019: p. 1–12. Dizdar Ö, Kılıçkap S. Global epidemiology of gastrointestinal cancers. Textbook of Gastrointestinal Oncology, 2019: p. 1–12.
2.
go back to reference Arnold M, et al. Global burden of 5 major types of gastrointestinal cancer. Gastroenterology. 2020;159(1):335–49. e15.PubMedCrossRef Arnold M, et al. Global burden of 5 major types of gastrointestinal cancer. Gastroenterology. 2020;159(1):335–49. e15.PubMedCrossRef
3.
go back to reference Sung H, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. Cancer J Clin. 2021;71(3):209–49.CrossRef Sung H, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. Cancer J Clin. 2021;71(3):209–49.CrossRef
4.
go back to reference Li M, et al. Time trends of esophageal and gastric cancer mortality in China, 1991–2009: an age-period-cohort analysis. Sci Rep. 2017;7(1):1–8. Li M, et al. Time trends of esophageal and gastric cancer mortality in China, 1991–2009: an age-period-cohort analysis. Sci Rep. 2017;7(1):1–8.
5.
go back to reference Cunningham D, et al. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med. 2006;355(1):11–20.PubMedCrossRef Cunningham D, et al. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med. 2006;355(1):11–20.PubMedCrossRef
6.
go back to reference Al-Batran S-E, et al. Perioperative chemotherapy with fluorouracil plus leucovorin, oxaliplatin, and docetaxel versus fluorouracil or capecitabine plus cisplatin and epirubicin for locally advanced, resectable gastric or gastro-oesophageal junction adenocarcinoma (FLOT4): a randomised, phase 2/3 trial. The Lancet. 2019;393(10184):1948–57.CrossRef Al-Batran S-E, et al. Perioperative chemotherapy with fluorouracil plus leucovorin, oxaliplatin, and docetaxel versus fluorouracil or capecitabine plus cisplatin and epirubicin for locally advanced, resectable gastric or gastro-oesophageal junction adenocarcinoma (FLOT4): a randomised, phase 2/3 trial. The Lancet. 2019;393(10184):1948–57.CrossRef
7.
go back to reference Reynolds J, et al. ICORG 10–14: NEOadjuvant trial in Adenocarcinoma of the oEsophagus and oesophagoGastric junction International Study (Neo-AEGIS). BMC Cancer. 2017;17(1):1–10.CrossRef Reynolds J, et al. ICORG 10–14: NEOadjuvant trial in Adenocarcinoma of the oEsophagus and oesophagoGastric junction International Study (Neo-AEGIS). BMC Cancer. 2017;17(1):1–10.CrossRef
8.
go back to reference Shapiro J, et al. Neoadjuvant chemoradiotherapy plus surgery versus surgery alone for oesophageal or junctional cancer (CROSS): long-term results of a randomised controlled trial. Lancet Oncol. 2015;16(9):1090–8.PubMedCrossRef Shapiro J, et al. Neoadjuvant chemoradiotherapy plus surgery versus surgery alone for oesophageal or junctional cancer (CROSS): long-term results of a randomised controlled trial. Lancet Oncol. 2015;16(9):1090–8.PubMedCrossRef
9.
go back to reference Sitarz R, et al. Gastric cancer: epidemiology, prevention, classification, and treatment. Cancer Manage Res. 2018;10:239.CrossRef Sitarz R, et al. Gastric cancer: epidemiology, prevention, classification, and treatment. Cancer Manage Res. 2018;10:239.CrossRef
11.
go back to reference Yang Y-M, et al. Advances in targeted therapy for esophageal cancer. Signal Transduct Target Therapy. 2020;5(1):1–11. Yang Y-M, et al. Advances in targeted therapy for esophageal cancer. Signal Transduct Target Therapy. 2020;5(1):1–11.
12.
go back to reference Matsuoka T, Yashiro M. Precision medicine for gastrointestinal cancer: recent progress and future perspective. World J Gastrointest Oncol. 2020;12(1):1.PubMedPubMedCentralCrossRef Matsuoka T, Yashiro M. Precision medicine for gastrointestinal cancer: recent progress and future perspective. World J Gastrointest Oncol. 2020;12(1):1.PubMedPubMedCentralCrossRef
13.
go back to reference Ranga A, Gjorevski N, Lutolf MP. Drug discovery through stem cell-based organoid models. Adv Drug Deliv Rev. 2014;69:19–28.PubMedCrossRef Ranga A, Gjorevski N, Lutolf MP. Drug discovery through stem cell-based organoid models. Adv Drug Deliv Rev. 2014;69:19–28.PubMedCrossRef
14.
go back to reference Li A, et al. Genomic changes and gene expression profiles reveal that established glioma cell lines are poorly Representative of Primary Human Gliomas. Mol Cancer Res. 2008;6(1):21–30.PubMedCrossRef Li A, et al. Genomic changes and gene expression profiles reveal that established glioma cell lines are poorly Representative of Primary Human Gliomas. Mol Cancer Res. 2008;6(1):21–30.PubMedCrossRef
15.
go back to reference Gillet JP, et al. Redefining the relevance of established cancer cell lines to the study of mechanisms of clinical anti-cancer drug resistance. Proc Natl Acad Sci U S A. 2011;108(46):18708–13.PubMedPubMedCentralCrossRef Gillet JP, et al. Redefining the relevance of established cancer cell lines to the study of mechanisms of clinical anti-cancer drug resistance. Proc Natl Acad Sci U S A. 2011;108(46):18708–13.PubMedPubMedCentralCrossRef
17.
go back to reference Sharpless NE, Depinho RA. The mighty mouse: genetically engineered mouse models in cancer drug development. Nat Rev Drug Discov. 2006;5(9):741–54.PubMedCrossRef Sharpless NE, Depinho RA. The mighty mouse: genetically engineered mouse models in cancer drug development. Nat Rev Drug Discov. 2006;5(9):741–54.PubMedCrossRef
18.
go back to reference Tentler JJ, et al. Patient-derived tumour xenografts as models for oncology drug development. Nat Reviews Clin Oncol. 2012;9(6):338–50.CrossRef Tentler JJ, et al. Patient-derived tumour xenografts as models for oncology drug development. Nat Reviews Clin Oncol. 2012;9(6):338–50.CrossRef
20.
go back to reference Yoshida GJ. Applications of patient-derived tumor xenograft models and tumor organoids. J Hematol Oncol. 2020;13(1):1–16.CrossRef Yoshida GJ. Applications of patient-derived tumor xenograft models and tumor organoids. J Hematol Oncol. 2020;13(1):1–16.CrossRef
21.
22.
go back to reference Pauli C, et al. Vitro and in vivo Cancer models to Guide Precision MedicinePersonalized Cancer Models to Guide Precision Medicine. Cancer Discov. 2017;7(5):462–77.PubMedPubMedCentralCrossRef Pauli C, et al. Vitro and in vivo Cancer models to Guide Precision MedicinePersonalized Cancer Models to Guide Precision Medicine. Cancer Discov. 2017;7(5):462–77.PubMedPubMedCentralCrossRef
23.
go back to reference Sato T, 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, et al. Long-term expansion of epithelial organoids from human Colon, Adenoma, Adenocarcinoma, and Barrett’s epithelium. Gastroenterology. 2011;141(5):1762–72.PubMedCrossRef
24.
go back to reference Whelan KA, Muir AB, Nakagawa H. Esophageal 3D Culture Systems as modeling tools in esophageal epithelial pathobiology and Personalized Medicine. Cell Mol Gastroenterol Hepatol. 2018;5(4):461–78.PubMedPubMedCentralCrossRef Whelan KA, Muir AB, Nakagawa H. Esophageal 3D Culture Systems as modeling tools in esophageal epithelial pathobiology and Personalized Medicine. Cell Mol Gastroenterol Hepatol. 2018;5(4):461–78.PubMedPubMedCentralCrossRef
25.
go back to reference Zheng B, et al. A new murine esophageal organoid culture method and organoid-based model of esophageal squamous cell neoplasia. Iscience. 2021;24(12):103440.PubMedPubMedCentralCrossRef Zheng B, et al. A new murine esophageal organoid culture method and organoid-based model of esophageal squamous cell neoplasia. Iscience. 2021;24(12):103440.PubMedPubMedCentralCrossRef
26.
go back to reference Vega ME, et al. Inhibition of notch signaling enhances transdifferentiation of the esophageal squamous epithelium towards a Barrett’s-like metaplasia via KLF4. Cell Cycle. 2014;13(24):3857–66.PubMedCrossRef Vega ME, et al. Inhibition of notch signaling enhances transdifferentiation of the esophageal squamous epithelium towards a Barrett’s-like metaplasia via KLF4. Cell Cycle. 2014;13(24):3857–66.PubMedCrossRef
27.
go back to reference Karakasheva TA, et al. Generation and characterization of patient-derived head and neck, oral, and esophageal cancer organoids. Curr Protoc Stem Cell Biol. 2020;53(1):e109.PubMedPubMedCentralCrossRef Karakasheva TA, et al. Generation and characterization of patient-derived head and neck, oral, and esophageal cancer organoids. Curr Protoc Stem Cell Biol. 2020;53(1):e109.PubMedPubMedCentralCrossRef
28.
go back to reference Kijima T, et al. Three-Dimensional Organoids reveal Therapy Resistance of esophageal and oropharyngeal squamous cell carcinoma cells. Cell Mol Gastroenterol Hepatol. 2019;7(1):73–91.PubMedCrossRef Kijima T, et al. Three-Dimensional Organoids reveal Therapy Resistance of esophageal and oropharyngeal squamous cell carcinoma cells. Cell Mol Gastroenterol Hepatol. 2019;7(1):73–91.PubMedCrossRef
29.
30.
go back to reference Li X, et al. Organoid cultures recapitulate esophageal adenocarcinoma heterogeneity providing a model for clonality studies and precision therapeutics. Nat Commun. 2018;9(1):1–13. Li X, et al. Organoid cultures recapitulate esophageal adenocarcinoma heterogeneity providing a model for clonality studies and precision therapeutics. Nat Commun. 2018;9(1):1–13.
31.
go back to reference Derouet MF, et al. Towards personalized induction therapy for esophageal adenocarcinoma: organoids derived from endoscopic biopsy recapitulate the pre-treatment tumor. Sci Rep. 2020;10(1):1–11.CrossRef Derouet MF, et al. Towards personalized induction therapy for esophageal adenocarcinoma: organoids derived from endoscopic biopsy recapitulate the pre-treatment tumor. Sci Rep. 2020;10(1):1–11.CrossRef
32.
go back to reference Dodbiba L, et al. Primary esophageal and gastro-esophageal junction cancer xenograft models: clinicopathological features and engraftment. Lab Invest. 2013;93(4):397–407.CrossRef Dodbiba L, et al. Primary esophageal and gastro-esophageal junction cancer xenograft models: clinicopathological features and engraftment. Lab Invest. 2013;93(4):397–407.CrossRef
33.
go back to reference Damhofer H, et al. Establishment of patient-derived xenograft models and cell lines for malignancies of the upper gastrointestinal tract. J Translational Med. 2015;13(1):1–14.CrossRef Damhofer H, et al. Establishment of patient-derived xenograft models and cell lines for malignancies of the upper gastrointestinal tract. J Translational Med. 2015;13(1):1–14.CrossRef
34.
go back to reference Zhu H, et al. A subset of esophageal squamous cell carcinoma patient-derived xenografts respond to cetuximab, which is predicted by high EGFR expression and amplification. J Thorac Disease. 2018;10(9):5328.CrossRef Zhu H, et al. A subset of esophageal squamous cell carcinoma patient-derived xenografts respond to cetuximab, which is predicted by high EGFR expression and amplification. J Thorac Disease. 2018;10(9):5328.CrossRef
35.
go back to reference Dodbiba L, et al. Appropriateness of using patient-derived xenograft models for pharmacologic evaluation of novel therapies for esophageal/gastro-esophageal junction cancers. PLoS ONE. 2015;10(3):e0121872.PubMedPubMedCentralCrossRef Dodbiba L, et al. Appropriateness of using patient-derived xenograft models for pharmacologic evaluation of novel therapies for esophageal/gastro-esophageal junction cancers. PLoS ONE. 2015;10(3):e0121872.PubMedPubMedCentralCrossRef
36.
go back to reference Zou J, et al. Establishment and genomic characterizations of patient-derived esophageal squamous cell carcinoma xenograft models using biopsies for treatment optimization. J Translational Med. 2018;16(1):1–11.CrossRef Zou J, et al. Establishment and genomic characterizations of patient-derived esophageal squamous cell carcinoma xenograft models using biopsies for treatment optimization. J Translational Med. 2018;16(1):1–11.CrossRef
37.
go back to reference Zhang J, et al. Establishment and characterization of esophageal squamous cell carcinoma patient-derived xenograft mouse models for preclinical drug discovery. Lab Invest. 2014;94(8):917–26.PubMedCrossRef Zhang J, et al. Establishment and characterization of esophageal squamous cell carcinoma patient-derived xenograft mouse models for preclinical drug discovery. Lab Invest. 2014;94(8):917–26.PubMedCrossRef
38.
go back to reference Liu F, et al. Targeting integrin αvβ3 with indomethacin inhibits patient-derived xenograft tumour growth and recurrence in oesophageal squamous cell carcinoma. Clin Translational Med. 2021;11(10):e548.CrossRef Liu F, et al. Targeting integrin αvβ3 with indomethacin inhibits patient-derived xenograft tumour growth and recurrence in oesophageal squamous cell carcinoma. Clin Translational Med. 2021;11(10):e548.CrossRef
39.
go back to reference Ma F, et al. Heterogeneity analysis of esophageal squamous cell carcinoma in cell lines, tumor tissues and patient-derived xenografts. J Cancer. 2021;12(13):3930.PubMedPubMedCentralCrossRef Ma F, et al. Heterogeneity analysis of esophageal squamous cell carcinoma in cell lines, tumor tissues and patient-derived xenografts. J Cancer. 2021;12(13):3930.PubMedPubMedCentralCrossRef
40.
go back to reference Li J, et al. Malignant ascites-derived organoid (MADO) cultures for gastric cancer in vitro modelling and drug screening. J Cancer Res Clin Oncol. 2019;145(11):2637–47.PubMedCrossRef Li J, et al. Malignant ascites-derived organoid (MADO) cultures for gastric cancer in vitro modelling and drug screening. J Cancer Res Clin Oncol. 2019;145(11):2637–47.PubMedCrossRef
41.
go back to reference Gao M, et al. Development of patient-derived gastric cancer organoids from endoscopic biopsies and surgical tissues. Ann Surg Oncol. 2018;25(9):2767–75.PubMedCrossRef Gao M, et al. Development of patient-derived gastric cancer organoids from endoscopic biopsies and surgical tissues. Ann Surg Oncol. 2018;25(9):2767–75.PubMedCrossRef
42.
go back to reference Steele NG, et al. An organoid-based preclinical model of human gastric cancer. Cell Mol Gastroenterol Hepatol. 2019;7(1):161–84.PubMedCrossRef Steele NG, et al. An organoid-based preclinical model of human gastric cancer. Cell Mol Gastroenterol Hepatol. 2019;7(1):161–84.PubMedCrossRef
44.
go back to reference Beckers J, et al. Identification and validation of novel ERBB2 (HER2, NEU) targets including genes involved in angiogenesis. Int J Cancer. 2005;114(4):590–7.PubMedCrossRef Beckers J, et al. Identification and validation of novel ERBB2 (HER2, NEU) targets including genes involved in angiogenesis. Int J Cancer. 2005;114(4):590–7.PubMedCrossRef
45.
46.
go back to reference Yan HH, et al. A comprehensive human gastric cancer organoid biobank captures tumor subtype heterogeneity and enables therapeutic screening. Cell Stem Cell. 2018;23(6):882–97. e11.PubMedCrossRef Yan HH, et al. A comprehensive human gastric cancer organoid biobank captures tumor subtype heterogeneity and enables therapeutic screening. Cell Stem Cell. 2018;23(6):882–97. e11.PubMedCrossRef
47.
go back to reference Li X, Ootani A, Kuo C. An air–liquid interface culture system for 3D organoid culture of diverse primary gastrointestinal tissues, in Gastrointestinal Physiology and Diseases. 2016, Springer. p. 33–40. Li X, Ootani A, Kuo C. An air–liquid interface culture system for 3D organoid culture of diverse primary gastrointestinal tissues, in Gastrointestinal Physiology and Diseases. 2016, Springer. p. 33–40.
49.
go back to reference Chakrabarti J et al. Mouse-derived gastric organoid and immune cell co-culture for the study of the tumor microenvironment, in Epithelial Cell Culture. 2018, Springer. p. 157–168. Chakrabarti J et al. Mouse-derived gastric organoid and immune cell co-culture for the study of the tumor microenvironment, in Epithelial Cell Culture. 2018, Springer. p. 157–168.
51.
go back to reference Chakrabarti J, et al. Disruption of Her2-induced PD-L1 inhibits tumor cell immune evasion in patient-derived gastric cancer organoids. Cancers. 2021;13(24):6158.PubMedPubMedCentralCrossRef Chakrabarti J, et al. Disruption of Her2-induced PD-L1 inhibits tumor cell immune evasion in patient-derived gastric cancer organoids. Cancers. 2021;13(24):6158.PubMedPubMedCentralCrossRef
52.
go back to reference Li X. Calvin Kuo < an air–liquid Interface Culture System for 3D Organoid Culture of Diverse primary gastrointestinal Tissues.pdf>. Methods Mol Biol. 2016;1422:33–40.PubMedCrossRef Li X. Calvin Kuo < an air–liquid Interface Culture System for 3D Organoid Culture of Diverse primary gastrointestinal Tissues.pdf>. Methods Mol Biol. 2016;1422:33–40.PubMedCrossRef
53.
go back to reference Kawasaki K, et al. An organoid biobank of neuroendocrine neoplasms enables genotype-phenotype mapping. Cell. 2020;183(5):1420–35. e21.PubMedCrossRef Kawasaki K, et al. An organoid biobank of neuroendocrine neoplasms enables genotype-phenotype mapping. Cell. 2020;183(5):1420–35. e21.PubMedCrossRef
54.
go back to reference Wang H, et al. Establishment of patient-derived gastric cancer xenografts: a useful tool for preclinical evaluation of targeted therapies involving alterations in HER-2, MET and FGFR2 signaling pathways. BMC Cancer. 2017;17(1):1–11. Wang H, et al. Establishment of patient-derived gastric cancer xenografts: a useful tool for preclinical evaluation of targeted therapies involving alterations in HER-2, MET and FGFR2 signaling pathways. BMC Cancer. 2017;17(1):1–11.
55.
go back to reference Zhu Y, et al. Establishment and characterization of patient-derived tumor xenograft using gastroscopic biopsies in gastric cancer. Sci Rep. 2015;5(1):1–8. Zhu Y, et al. Establishment and characterization of patient-derived tumor xenograft using gastroscopic biopsies in gastric cancer. Sci Rep. 2015;5(1):1–8.
56.
go back to reference Choi YY, et al. Establishment and characterisation of patient-derived xenografts as paraclinical models for gastric cancer. Sci Rep. 2016;6(1):1–12. Choi YY, et al. Establishment and characterisation of patient-derived xenografts as paraclinical models for gastric cancer. Sci Rep. 2016;6(1):1–12.
57.
go back to reference Zhang T, et al. Patient-derived gastric carcinoma xenograft mouse models faithfully represent human tumor molecular diversity. PLoS ONE. 2015;10(7):e0134493.PubMedPubMedCentralCrossRef Zhang T, et al. Patient-derived gastric carcinoma xenograft mouse models faithfully represent human tumor molecular diversity. PLoS ONE. 2015;10(7):e0134493.PubMedPubMedCentralCrossRef
58.
go back to reference Wang J et al. Personalized Treatment of Advanced Gastric Cancer Guided by the MiniPDX Model. Journal of Oncology, 2022. 2022. Wang J et al. Personalized Treatment of Advanced Gastric Cancer Guided by the MiniPDX Model. Journal of Oncology, 2022. 2022.
59.
60.
go back to reference Song S, et al. Patient-derived cell lines and orthotopic mouse model of peritoneal carcinomatosis recapitulate molecular and phenotypic features of human gastric adenocarcinoma. J Experimental Clin Cancer Res. 2021;40(1):1–15.CrossRef Song S, et al. Patient-derived cell lines and orthotopic mouse model of peritoneal carcinomatosis recapitulate molecular and phenotypic features of human gastric adenocarcinoma. J Experimental Clin Cancer Res. 2021;40(1):1–15.CrossRef
61.
go back to reference Kuwata T, et al. Establishment of novel gastric cancer patient-derived xenografts and cell lines: pathological comparison between primary tumor, patient-derived, and cell-line derived xenografts. Cells. 2019;8(6):585.PubMedPubMedCentralCrossRef Kuwata T, et al. Establishment of novel gastric cancer patient-derived xenografts and cell lines: pathological comparison between primary tumor, patient-derived, and cell-line derived xenografts. Cells. 2019;8(6):585.PubMedPubMedCentralCrossRef
62.
63.
go back to reference Corso S, et al. A Comprehensive PDX Gastric Cancer Collection captures Cancer cell–intrinsic transcriptional MSI TraitsMSI signature derived from a gastric Cancer PDX platform. Cancer Res. 2019;79(22):5884–96.PubMedCrossRef Corso S, et al. A Comprehensive PDX Gastric Cancer Collection captures Cancer cell–intrinsic transcriptional MSI TraitsMSI signature derived from a gastric Cancer PDX platform. Cancer Res. 2019;79(22):5884–96.PubMedCrossRef
64.
go back to reference Chen Z, et al. Characterization and validation of potential therapeutic targets based on the molecular signature of patient-derived xenografts in gastric cancer. J Hematol Oncol. 2018;11(1):1–12.CrossRef Chen Z, et al. Characterization and validation of potential therapeutic targets based on the molecular signature of patient-derived xenografts in gastric cancer. J Hematol Oncol. 2018;11(1):1–12.CrossRef
65.
go back to reference Dugger SA, Platt A, Goldstein DB. Drug development in the era of precision medicine. Nat Rev Drug Discovery. 2018;17(3):183–96.PubMedCrossRef Dugger SA, Platt A, Goldstein DB. Drug development in the era of precision medicine. Nat Rev Drug Discovery. 2018;17(3):183–96.PubMedCrossRef
66.
go back to reference Wensink GE, et al. Patient-derived organoids as a predictive biomarker for treatment response in cancer patients. NPJ Precision Oncology. 2021;5(1):1–13.CrossRef Wensink GE, et al. Patient-derived organoids as a predictive biomarker for treatment response in cancer patients. NPJ Precision Oncology. 2021;5(1):1–13.CrossRef
67.
go back to reference Bar-Ephraim YE, Kretzschmar K, Clevers H. Organoids in immunological research. Nat Rev Immunol. 2020;20(5):279–93.CrossRef Bar-Ephraim YE, Kretzschmar K, Clevers H. Organoids in immunological research. Nat Rev Immunol. 2020;20(5):279–93.CrossRef
69.
go back to reference Schutgens F, Clevers H. Human organoids: tools for understanding biology and treating diseases. Annu Rev Pathol. 2020;15:211–34.PubMedCrossRef Schutgens F, Clevers H. Human organoids: tools for understanding biology and treating diseases. Annu Rev Pathol. 2020;15:211–34.PubMedCrossRef
70.
go back to reference Hu Y, et al. Lung cancer organoids analyzed on microwell arrays predict drug responses of patients within a week. Nat Commun. 2021;12(1):1–14. Hu Y, et al. Lung cancer organoids analyzed on microwell arrays predict drug responses of patients within a week. Nat Commun. 2021;12(1):1–14.
71.
go back to reference Shi R, et al. Organoid cultures as Preclinical Models of non–small cell lung CancerNon–Small cell Lung Cancer Organoids. Clin Cancer Res. 2020;26(5):1162–74.PubMedCrossRef Shi R, et al. Organoid cultures as Preclinical Models of non–small cell lung CancerNon–Small cell Lung Cancer Organoids. Clin Cancer Res. 2020;26(5):1162–74.PubMedCrossRef
72.
go back to reference Kim S-Y, et al. Modeling clinical responses to targeted therapies by patient-derived organoids of Advanced Lung AdenocarcinomaClinical Relevance of Lung Cancer patient-derived Organoids. Clin Cancer Res. 2021;27(15):4397–409.PubMedPubMedCentralCrossRef Kim S-Y, et al. Modeling clinical responses to targeted therapies by patient-derived organoids of Advanced Lung AdenocarcinomaClinical Relevance of Lung Cancer patient-derived Organoids. Clin Cancer Res. 2021;27(15):4397–409.PubMedPubMedCentralCrossRef
73.
go back to reference Ooft SN, et al. Patient-derived organoids can predict response to chemotherapy in metastatic colorectal cancer patients. Sci Transl Med. 2019;11(513):eaay2574.PubMedCrossRef Ooft SN, et al. Patient-derived organoids can predict response to chemotherapy in metastatic colorectal cancer patients. Sci Transl Med. 2019;11(513):eaay2574.PubMedCrossRef
74.
go back to reference Yao Y, 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, et al. Patient-derived organoids predict chemoradiation responses of locally advanced rectal cancer. Cell Stem Cell. 2020;26(1):17–26. e6.PubMedCrossRef
75.
go back to reference Bruun J, et al. Patient-derived organoids from multiple colorectal Cancer Liver Metastases Reveal Moderate Intra-patient Pharmacotranscriptomic HeterogeneityPharmacological Heterogeneity of Colorectal Liver Metastases. Clin Cancer Res. 2020;26(15):4107–19.PubMedCrossRef Bruun J, et al. Patient-derived organoids from multiple colorectal Cancer Liver Metastases Reveal Moderate Intra-patient Pharmacotranscriptomic HeterogeneityPharmacological Heterogeneity of Colorectal Liver Metastases. Clin Cancer Res. 2020;26(15):4107–19.PubMedCrossRef
76.
go back to reference Sachs N, et al. A living biobank of breast cancer organoids captures disease heterogeneity. Cell. 2018;172(1–2):373–86. e10.PubMedCrossRef Sachs N, et al. A living biobank of breast cancer organoids captures disease heterogeneity. Cell. 2018;172(1–2):373–86. e10.PubMedCrossRef
77.
go back to reference Chen P, et al. Patient-derived Organoids can Guide Personalized‐Therapies for patients with advanced breast Cancer. Adv Sci. 2021;8(22):2101176.CrossRef Chen P, et al. Patient-derived Organoids can Guide Personalized‐Therapies for patients with advanced breast Cancer. Adv Sci. 2021;8(22):2101176.CrossRef
78.
go back to reference de Witte CJ, 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, 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
79.
go back to reference Kopper O, et al. An organoid platform for ovarian cancer captures intra-and interpatient heterogeneity. Nat Med. 2019;25(5):838–49.PubMedCrossRef Kopper O, et al. An organoid platform for ovarian cancer captures intra-and interpatient heterogeneity. Nat Med. 2019;25(5):838–49.PubMedCrossRef
80.
go back to reference Driehuis E, et al. Pancreatic cancer organoids recapitulate disease and allow personalized drug screening. Proc Natl Acad Sci. 2019;116(52):26580–90.PubMedPubMedCentralCrossRef Driehuis E, et al. Pancreatic cancer organoids recapitulate disease and allow personalized drug screening. Proc Natl Acad Sci. 2019;116(52):26580–90.PubMedPubMedCentralCrossRef
81.
go back to reference Tiriac H, et al. Organoid Profiling identifies common responders to Chemotherapy in Pancreatic CancerPancreatic Cancer Organoids parallel patient response. Cancer Discov. 2018;8(9):1112–29.PubMedPubMedCentralCrossRef Tiriac H, et al. Organoid Profiling identifies common responders to Chemotherapy in Pancreatic CancerPancreatic Cancer Organoids parallel patient response. Cancer Discov. 2018;8(9):1112–29.PubMedPubMedCentralCrossRef
82.
83.
go back to reference Cao W, et al. Modeling liver cancer and therapy responsiveness using organoids derived from primary mouse liver tumors. Carcinogenesis. 2019;40(1):145–54.PubMedCrossRef Cao W, et al. Modeling liver cancer and therapy responsiveness using organoids derived from primary mouse liver tumors. Carcinogenesis. 2019;40(1):145–54.PubMedCrossRef
84.
go back to reference Junttila MR, de Sauvage FJ. Influence of tumour micro-environment heterogeneity on therapeutic response. Nature. 2013;501(7467):346–54.PubMedCrossRef Junttila MR, de Sauvage FJ. Influence of tumour micro-environment heterogeneity on therapeutic response. Nature. 2013;501(7467):346–54.PubMedCrossRef
85.
go back to reference Chuprin J, et al. Humanized mouse models for immuno-oncology research. Nat Reviews Clin Oncol. 2023;20(3):192–206.CrossRef Chuprin J, et al. Humanized mouse models for immuno-oncology research. Nat Reviews Clin Oncol. 2023;20(3):192–206.CrossRef
Metadata
Title
Promising preclinical patient-derived organoid (PDO) and xenograft (PDX) models in upper gastrointestinal cancers: progress and challenges
Authors
Jing Gao
Jianqiang Lan
Haiyan Liao
Fang Yang
Pei Qiu
Feng Jin
Shubin Wang
Lin Shen
Tengfei Chao
Cheng Zhang
Yu Zhu
Publication date
01-12-2023
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2023
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-023-11434-9

Other articles of this Issue 1/2023

BMC Cancer 1/2023 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