Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2021

Open Access 01-12-2021 | Colorectal Cancer | Review

Preclinical models as patients’ avatars for precision medicine in colorectal cancer: past and future challenges

Authors: Erika Durinikova, Kristi Buzo, Sabrina Arena

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2021

Login to get access

Abstract

Colorectal cancer (CRC) is a complex and heterogeneous disease, characterized by dismal prognosis and low survival rate in the advanced (metastatic) stage. During the last decade, the establishment of novel preclinical models, leading to the generation of translational discovery and validation platforms, has opened up a new scenario for the clinical practice of CRC patients. To bridge the results developed at the bench with the medical decision process, the ideal model should be easily scalable, reliable to predict treatment responses, and flexibly adapted for various applications in the research. As such, the improved benefit of novel therapies being tested initially on valuable and reproducible preclinical models would lie in personalized treatment recommendations based on the biology and genomics of the patient’s tumor with the overall aim to avoid overtreatment and unnecessary toxicity. In this review, we summarize different in vitro and in vivo models, which proved efficacy in detection of novel CRC culprits and shed light into the biology and therapy of this complex disease. Even though cell lines and patient-derived xenografts remain the mainstay of colorectal cancer research, the field has been confidently shifting to the use of organoids as the most relevant preclinical model. Prioritization of organoids is supported by increasing body of evidence that these represent excellent tools worth further therapeutic explorations. In addition, novel preclinical models such as zebrafish avatars are emerging as useful tools for pharmacological interrogation. Finally, all available models represent complementary tools that can be utilized for precision medicine applications.
Literature
1.
go back to reference Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424.PubMedCrossRef Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424.PubMedCrossRef
2.
4.
go back to reference Van Cutsem E, Cervantes A, Adam R, Sobrero A, Van Krieken JH, Aderka D, et al. ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann Oncol. 2016;27(8):1386–422.PubMedCrossRef Van Cutsem E, Cervantes A, Adam R, Sobrero A, Van Krieken JH, Aderka D, et al. ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann Oncol. 2016;27(8):1386–422.PubMedCrossRef
5.
go back to reference Martini G, Troiani T, Cardone C, Vitiello P, Sforza V, Ciardiello D, et al. Present and future of metastatic colorectal cancer treatment: a review of new candidate targets. World J Gastroenterol. 2017;23(26):4675–88.PubMedPubMedCentralCrossRef Martini G, Troiani T, Cardone C, Vitiello P, Sforza V, Ciardiello D, et al. Present and future of metastatic colorectal cancer treatment: a review of new candidate targets. World J Gastroenterol. 2017;23(26):4675–88.PubMedPubMedCentralCrossRef
6.
go back to reference Argiles G, Tabernero J, Labianca R, Hochhauser D, Salazar R, Iveson T, et al. Localised Colon Cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2020;31(10):1291–305. Argiles G, Tabernero J, Labianca R, Hochhauser D, Salazar R, Iveson T, et al. Localised Colon Cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2020;31(10):1291–305.
7.
go back to reference Rimassa L, Bozzarelli S, Pietrantonio F, Cordio S, Lonardi S, Toppo L, et al. Phase II study of Tivantinib and Cetuximab in patients with KRAS wild-type metastatic colorectal Cancer with acquired resistance to EGFR inhibitors and emergence of MET overexpression: lesson learned for future trials with EGFR/MET dual inhibition. Clin Colorectal Cancer. 2019;18(2):125–32 e2.PubMedCrossRef Rimassa L, Bozzarelli S, Pietrantonio F, Cordio S, Lonardi S, Toppo L, et al. Phase II study of Tivantinib and Cetuximab in patients with KRAS wild-type metastatic colorectal Cancer with acquired resistance to EGFR inhibitors and emergence of MET overexpression: lesson learned for future trials with EGFR/MET dual inhibition. Clin Colorectal Cancer. 2019;18(2):125–32 e2.PubMedCrossRef
8.
go back to reference Cremolini C, Pietrantonio F, Tomasello G, Dadduzio V, Moretto R, Morano F, et al. Vinorelbine in BRAF V600E mutated metastatic colorectal cancer: a prospective multicentre phase II clinical study. ESMO Open. 2017;2(3):e000241.PubMedPubMedCentralCrossRef Cremolini C, Pietrantonio F, Tomasello G, Dadduzio V, Moretto R, Morano F, et al. Vinorelbine in BRAF V600E mutated metastatic colorectal cancer: a prospective multicentre phase II clinical study. ESMO Open. 2017;2(3):e000241.PubMedPubMedCentralCrossRef
9.
go back to reference Satoh T, Xu RH, Chung HC, Sun GP, Doi T, Xu JM, et al. Lapatinib plus paclitaxel versus paclitaxel alone in the second-line treatment of HER2-amplified advanced gastric cancer in Asian populations: TyTAN--a randomized, phase III study. J Clin Oncol. 2014;32(19):2039–49.PubMedCrossRef Satoh T, Xu RH, Chung HC, Sun GP, Doi T, Xu JM, et al. Lapatinib plus paclitaxel versus paclitaxel alone in the second-line treatment of HER2-amplified advanced gastric cancer in Asian populations: TyTAN--a randomized, phase III study. J Clin Oncol. 2014;32(19):2039–49.PubMedCrossRef
10.
go back to reference Catenacci DVT, Tebbutt NC, Davidenko I, Murad AM, Al-Batran SE, Ilson DH, et al. Rilotumumab plus epirubicin, cisplatin, and capecitabine as first-line therapy in advanced MET-positive gastric or gastro-oesophageal junction cancer (RILOMET-1): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2017;18(11):1467–82.PubMedPubMedCentralCrossRef Catenacci DVT, Tebbutt NC, Davidenko I, Murad AM, Al-Batran SE, Ilson DH, et al. Rilotumumab plus epirubicin, cisplatin, and capecitabine as first-line therapy in advanced MET-positive gastric or gastro-oesophageal junction cancer (RILOMET-1): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2017;18(11):1467–82.PubMedPubMedCentralCrossRef
11.
go back to reference Sharma SV, Haber DA, Settleman J. Cell line-based platforms to evaluate the therapeutic efficacy of candidate anticancer agents. Nat Rev Cancer. 2010;10(4):241–53.PubMedCrossRef Sharma SV, Haber DA, Settleman J. Cell line-based platforms to evaluate the therapeutic efficacy of candidate anticancer agents. Nat Rev Cancer. 2010;10(4):241–53.PubMedCrossRef
12.
go back to reference Mullins CS, Micheel B, Matschos S, Leuchter M, Bürtin F, Krohn M, et al. Integrated biobanking and tumor model establishment of human colorectal carcinoma provides excellent tools for preclinical research. Cancers (Basel). 2019;11(10):1520. Mullins CS, Micheel B, Matschos S, Leuchter M, Bürtin F, Krohn M, et al. Integrated biobanking and tumor model establishment of human colorectal carcinoma provides excellent tools for preclinical research. Cancers (Basel). 2019;11(10):1520.
14.
go back to reference Fazio M, Ablain J, Chuan Y, Langenau DM, Zon LI. Zebrafish patient avatars in cancer biology and precision cancer therapy. Nat Rev Cancer. 2020;20(5):263–73.PubMedPubMedCentralCrossRef Fazio M, Ablain J, Chuan Y, Langenau DM, Zon LI. Zebrafish patient avatars in cancer biology and precision cancer therapy. Nat Rev Cancer. 2020;20(5):263–73.PubMedPubMedCentralCrossRef
15.
go back to reference Linnebacher M, Maletzki C, Ostwald C, Klier U, Krohn M, Klar E, et al. Cryopreservation of human colorectal carcinomas prior to xenografting. BMC Cancer. 2010;10:362.PubMedPubMedCentralCrossRef Linnebacher M, Maletzki C, Ostwald C, Klier U, Krohn M, Klar E, et al. Cryopreservation of human colorectal carcinomas prior to xenografting. BMC Cancer. 2010;10:362.PubMedPubMedCentralCrossRef
16.
go back to reference Bürtin F, Mullins CS, Linnebacher M. Mouse models of colorectal cancer: past, present and future perspectives. World J Gastroenterol. 2020;26(13):1394–426.PubMedPubMedCentralCrossRef Bürtin F, Mullins CS, Linnebacher M. Mouse models of colorectal cancer: past, present and future perspectives. World J Gastroenterol. 2020;26(13):1394–426.PubMedPubMedCentralCrossRef
17.
go back to reference Maletzki C, Bock S, Fruh P, Macius K, Witt A, Prall F, et al. NSG mice as hosts for oncological precision medicine. Lab Investig. 2020;100(1):27–37.PubMedCrossRef Maletzki C, Bock S, Fruh P, Macius K, Witt A, Prall F, et al. NSG mice as hosts for oncological precision medicine. Lab Investig. 2020;100(1):27–37.PubMedCrossRef
18.
go back to reference Xiao X, Zhou X, Ming H, Zhang J, Huang G, Zhang Z, et al. Chick Chorioallantoic membrane assay: a 3D animal model for study of human nasopharyngeal carcinoma. PLoS One. 2015;10(6):e0130935.PubMedPubMedCentralCrossRef Xiao X, Zhou X, Ming H, Zhang J, Huang G, Zhang Z, et al. Chick Chorioallantoic membrane assay: a 3D animal model for study of human nasopharyngeal carcinoma. PLoS One. 2015;10(6):e0130935.PubMedPubMedCentralCrossRef
19.
go back to reference Debreova M, Csaderova L, Burikova M, Lukacikova L, Kajanova I, Sedlakova O, et al. CAIX regulates Invadopodia formation through both a pH-dependent mechanism and interplay with actin regulatory proteins. Int J Mol Sci. 2019;20(11):2745. Debreova M, Csaderova L, Burikova M, Lukacikova L, Kajanova I, Sedlakova O, et al. CAIX regulates Invadopodia formation through both a pH-dependent mechanism and interplay with actin regulatory proteins. Int J Mol Sci. 2019;20(11):2745.
20.
go back to reference DeBord LC, Pathak RR, Villaneuva M, Liu HC, Harrington DA, Yu W, et al. The chick chorioallantoic membrane (CAM) as a versatile patient-derived xenograft (PDX) platform for precision medicine and preclinical research. Am J Cancer Res. 2018;8(8):1642–60.PubMedPubMedCentral DeBord LC, Pathak RR, Villaneuva M, Liu HC, Harrington DA, Yu W, et al. The chick chorioallantoic membrane (CAM) as a versatile patient-derived xenograft (PDX) platform for precision medicine and preclinical research. Am J Cancer Res. 2018;8(8):1642–60.PubMedPubMedCentral
21.
go back to reference Langenau DM, Traver D, Ferrando AA, Kutok JL, Aster JC, Kanki JP, et al. Myc-induced T cell leukemia in transgenic zebrafish. Science. 2003;299(5608):887–90.PubMedCrossRef Langenau DM, Traver D, Ferrando AA, Kutok JL, Aster JC, Kanki JP, et al. Myc-induced T cell leukemia in transgenic zebrafish. Science. 2003;299(5608):887–90.PubMedCrossRef
22.
go back to reference Nicoli S, Ribatti D, Cotelli F, Presta M. Mammalian tumor xenografts induce neovascularization in zebrafish embryos. Cancer Res. 2007;67(7):2927–31.PubMedCrossRef Nicoli S, Ribatti D, Cotelli F, Presta M. Mammalian tumor xenografts induce neovascularization in zebrafish embryos. Cancer Res. 2007;67(7):2927–31.PubMedCrossRef
23.
go back to reference Haldi M, Ton C, Seng WL, McGrath P. Human melanoma cells transplanted into zebrafish proliferate, migrate, produce melanin, form masses and stimulate angiogenesis in zebrafish. Angiogenesis. 2006;9(3):139–51.PubMedCrossRef Haldi M, Ton C, Seng WL, McGrath P. Human melanoma cells transplanted into zebrafish proliferate, migrate, produce melanin, form masses and stimulate angiogenesis in zebrafish. Angiogenesis. 2006;9(3):139–51.PubMedCrossRef
24.
go back to reference Yan C, Brunson DC, Tang Q, Do D, Iftimia NA, Moore JC, et al. Visualizing engrafted human Cancer and therapy responses in Immunodeficient Zebrafish. Cell. 2019;177(7):1903–14 e14.PubMedPubMedCentralCrossRef Yan C, Brunson DC, Tang Q, Do D, Iftimia NA, Moore JC, et al. Visualizing engrafted human Cancer and therapy responses in Immunodeficient Zebrafish. Cell. 2019;177(7):1903–14 e14.PubMedPubMedCentralCrossRef
25.
go back to reference Lin J, Zhang W, Zhao JJ, Kwart AH, Yang C, Ma D, et al. A clinically relevant in vivo zebrafish model of human multiple myeloma to study preclinical therapeutic efficacy. Blood. 2016;128(2):249–52.PubMedPubMedCentralCrossRef Lin J, Zhang W, Zhao JJ, Kwart AH, Yang C, Ma D, et al. A clinically relevant in vivo zebrafish model of human multiple myeloma to study preclinical therapeutic efficacy. Blood. 2016;128(2):249–52.PubMedPubMedCentralCrossRef
26.
go back to reference Mercatali L, La Manna F, Groenewoud A, Casadei R, Recine F, Miserocchi G, et al. Development of a patient-derived Xenograft (PDX) of breast Cancer bone metastasis in a Zebrafish model. Int J Mol Sci. 2016;17(8):1375. Mercatali L, La Manna F, Groenewoud A, Casadei R, Recine F, Miserocchi G, et al. Development of a patient-derived Xenograft (PDX) of breast Cancer bone metastasis in a Zebrafish model. Int J Mol Sci. 2016;17(8):1375.
28.
go back to reference Hason M, Bartůněk P. Zebrafish models of Cancer-new insights on modeling human Cancer in a non-mammalian vertebrate. Genes (Basel). 2019;10(11):935. Hason M, Bartůněk P. Zebrafish models of Cancer-new insights on modeling human Cancer in a non-mammalian vertebrate. Genes (Basel). 2019;10(11):935.
29.
go back to reference White RM, Cech J, Ratanasirintrawoot S, Lin CY, Rahl PB, Burke CJ, et al. DHODH modulates transcriptional elongation in the neural crest and melanoma. Nature. 2011;471(7339):518–22.PubMedPubMedCentralCrossRef White RM, Cech J, Ratanasirintrawoot S, Lin CY, Rahl PB, Burke CJ, et al. DHODH modulates transcriptional elongation in the neural crest and melanoma. Nature. 2011;471(7339):518–22.PubMedPubMedCentralCrossRef
30.
go back to reference Clements WK, Traver D. Fish pharming: zebrafish antileukemia screening. Blood. 2012;119(24):5614–5.PubMedCrossRef Clements WK, Traver D. Fish pharming: zebrafish antileukemia screening. Blood. 2012;119(24):5614–5.PubMedCrossRef
31.
go back to reference Camus S, Quevedo C, Menéndez S, Paramonov I, Stouten PF, Janssen RA, et al. Identification of phosphorylase kinase as a novel therapeutic target through high-throughput screening for anti-angiogenesis compounds in zebrafish. Oncogene. 2012;31(39):4333–42.PubMedCrossRef Camus S, Quevedo C, Menéndez S, Paramonov I, Stouten PF, Janssen RA, et al. Identification of phosphorylase kinase as a novel therapeutic target through high-throughput screening for anti-angiogenesis compounds in zebrafish. Oncogene. 2012;31(39):4333–42.PubMedCrossRef
32.
go back to reference Astin JW, Jamieson SM, Eng TC, Flores MV, Misa JP, Chien A, et al. An in vivo antilymphatic screen in zebrafish identifies novel inhibitors of mammalian lymphangiogenesis and lymphatic-mediated metastasis. Mol Cancer Ther. 2014;13(10):2450–62.PubMedCrossRef Astin JW, Jamieson SM, Eng TC, Flores MV, Misa JP, Chien A, et al. An in vivo antilymphatic screen in zebrafish identifies novel inhibitors of mammalian lymphangiogenesis and lymphatic-mediated metastasis. Mol Cancer Ther. 2014;13(10):2450–62.PubMedCrossRef
33.
go back to reference Fior R, Póvoa V, Mendes RV, Carvalho T, Gomes A, Figueiredo N, et al. Single-cell functional and chemosensitive profiling of combinatorial colorectal therapy in zebrafish xenografts. Proc Natl Acad Sci U S A. 2017;114(39):E8234–E43.PubMedPubMedCentralCrossRef Fior R, Póvoa V, Mendes RV, Carvalho T, Gomes A, Figueiredo N, et al. Single-cell functional and chemosensitive profiling of combinatorial colorectal therapy in zebrafish xenografts. Proc Natl Acad Sci U S A. 2017;114(39):E8234–E43.PubMedPubMedCentralCrossRef
34.
go back to reference Costa B, Ferreira S, Póvoa V, Cardoso MJ, Vieira S, Stroom J, et al. Developments in zebrafish avatars as radiotherapy sensitivity reporters - towards personalized medicine. EBioMedicine. 2020;51:102578.PubMedCrossRef Costa B, Ferreira S, Póvoa V, Cardoso MJ, Vieira S, Stroom J, et al. Developments in zebrafish avatars as radiotherapy sensitivity reporters - towards personalized medicine. EBioMedicine. 2020;51:102578.PubMedCrossRef
35.
go back to reference Póvoa V, Rebelo de Almeida C, Maia-Gil M, Sobral D, Domingues M, Martinez-Lopez M, et al. Innate immune evasion revealed in a colorectal zebrafish xenograft model. Nat Commun. 2021;12(1):1156.PubMedPubMedCentralCrossRef Póvoa V, Rebelo de Almeida C, Maia-Gil M, Sobral D, Domingues M, Martinez-Lopez M, et al. Innate immune evasion revealed in a colorectal zebrafish xenograft model. Nat Commun. 2021;12(1):1156.PubMedPubMedCentralCrossRef
36.
go back to reference Kemper K, Krijgsman O, Cornelissen-Steijger P, Shahrabi A, Weeber F, Song JY, et al. Intra- and inter-tumor heterogeneity in a vemurafenib-resistant melanoma patient and derived xenografts. EMBO Mol Med. 2015;7(9):1104–18.PubMedPubMedCentralCrossRef Kemper K, Krijgsman O, Cornelissen-Steijger P, Shahrabi A, Weeber F, Song JY, et al. Intra- and inter-tumor heterogeneity in a vemurafenib-resistant melanoma patient and derived xenografts. EMBO Mol Med. 2015;7(9):1104–18.PubMedPubMedCentralCrossRef
37.
go back to reference Siravegna G, Lazzari L, Crisafulli G, Sartore-Bianchi A, Mussolin B, Cassingena A, et al. Radiologic and Genomic Evolution of Individual Metastases during HER2 Blockade in Colorectal Cancer. Cancer Cell. 2018;34(1):148–62 e7.CrossRefPubMed Siravegna G, Lazzari L, Crisafulli G, Sartore-Bianchi A, Mussolin B, Cassingena A, et al. Radiologic and Genomic Evolution of Individual Metastases during HER2 Blockade in Colorectal Cancer. Cancer Cell. 2018;34(1):148–62 e7.CrossRefPubMed
38.
go back to reference Puig I, Chicote I, Tenbaum SP, Arqués O, Herance JR, Gispert JD, et al. A personalized preclinical model to evaluate the metastatic potential of patient-derived colon cancer initiating cells. Clin Cancer Res. 2013;19(24):6787–801.PubMedCrossRef Puig I, Chicote I, Tenbaum SP, Arqués O, Herance JR, Gispert JD, et al. A personalized preclinical model to evaluate the metastatic potential of patient-derived colon cancer initiating cells. Clin Cancer Res. 2013;19(24):6787–801.PubMedCrossRef
39.
go back to reference Janakiraman H, Zhu Y, Becker SA, Wang C, Cross A, Curl E, et al. Modeling rectal cancer to advance neoadjuvant precision therapy. Int J Cancer. 2020;147(5):1405–18. Janakiraman H, Zhu Y, Becker SA, Wang C, Cross A, Curl E, et al. Modeling rectal cancer to advance neoadjuvant precision therapy. Int J Cancer. 2020;147(5):1405–18.
40.
go back to reference Rajaram S, Roth MA, Malato J, VandenBerg S, Hann B, Atreya CE, et al. A multi-modal data resource for investigating topographic heterogeneity in patient-derived xenograft tumors. Sci Data. 2019;6(1):253.PubMedPubMedCentralCrossRef Rajaram S, Roth MA, Malato J, VandenBerg S, Hann B, Atreya CE, et al. A multi-modal data resource for investigating topographic heterogeneity in patient-derived xenograft tumors. Sci Data. 2019;6(1):253.PubMedPubMedCentralCrossRef
41.
go back to reference Guinney J, Dienstmann R, Wang X, de Reyniès A, Schlicker A, Soneson C, et al. The consensus molecular subtypes of colorectal cancer. Nat Med. 2015;21(11):1350–6.PubMedPubMedCentralCrossRef Guinney J, Dienstmann R, Wang X, de Reyniès A, Schlicker A, Soneson C, et al. The consensus molecular subtypes of colorectal cancer. Nat Med. 2015;21(11):1350–6.PubMedPubMedCentralCrossRef
42.
go back to reference Dienstmann R, Vermeulen L, Guinney J, Kopetz S, Tejpar S, Tabernero J. Consensus molecular subtypes and the evolution of precision medicine in colorectal cancer. Nat Rev Cancer. 2017;17(2):79–92.PubMedCrossRef Dienstmann R, Vermeulen L, Guinney J, Kopetz S, Tejpar S, Tabernero J. Consensus molecular subtypes and the evolution of precision medicine in colorectal cancer. Nat Rev Cancer. 2017;17(2):79–92.PubMedCrossRef
43.
go back to reference Sveen A, Bruun J, Eide PW, Eilertsen IA, Ramirez L, Murumägi A, et al. Colorectal Cancer consensus molecular subtypes translated to preclinical models uncover potentially targetable Cancer cell dependencies. Clin Cancer Res. 2018;24(4):794–806.PubMedCrossRef Sveen A, Bruun J, Eide PW, Eilertsen IA, Ramirez L, Murumägi A, et al. Colorectal Cancer consensus molecular subtypes translated to preclinical models uncover potentially targetable Cancer cell dependencies. Clin Cancer Res. 2018;24(4):794–806.PubMedCrossRef
44.
go back to reference Prasetyanti PR, van Hooff SR, van Herwaarden T, de Vries N, Kalloe K, Rodermond H, et al. Capturing colorectal cancer inter-tumor heterogeneity in patient-derived xenograft (PDX) models. Int J Cancer. 2019;144(2):366–71.PubMedCrossRef Prasetyanti PR, van Hooff SR, van Herwaarden T, de Vries N, Kalloe K, Rodermond H, et al. Capturing colorectal cancer inter-tumor heterogeneity in patient-derived xenograft (PDX) models. Int J Cancer. 2019;144(2):366–71.PubMedCrossRef
45.
go back to reference Oh BY, Lee WY, Jung S, Hong HK, Nam DH, Park YA, et al. Correlation between tumor engraftment in patient-derived xenograft models and clinical outcomes in colorectal cancer patients. Oncotarget. 2015;6(18):16059–68.PubMedPubMedCentralCrossRef Oh BY, Lee WY, Jung S, Hong HK, Nam DH, Park YA, et al. Correlation between tumor engraftment in patient-derived xenograft models and clinical outcomes in colorectal cancer patients. Oncotarget. 2015;6(18):16059–68.PubMedPubMedCentralCrossRef
46.
go back to reference Isella C, Brundu F, Bellomo SE, Galimi F, Zanella E, Porporato R, et al. Selective analysis of cancer-cell intrinsic transcriptional traits defines novel clinically relevant subtypes of colorectal cancer. Nat Commun. 2017;8:15107.PubMedPubMedCentralCrossRef Isella C, Brundu F, Bellomo SE, Galimi F, Zanella E, Porporato R, et al. Selective analysis of cancer-cell intrinsic transcriptional traits defines novel clinically relevant subtypes of colorectal cancer. Nat Commun. 2017;8:15107.PubMedPubMedCentralCrossRef
47.
go back to reference Morgan KM, Riedlinger GM, Rosenfeld J, Ganesan S, Pine SR. Patient-derived Xenograft models of non-small cell lung Cancer and their potential utility in personalized medicine. Front Oncol. 2017;7:2.PubMedPubMedCentralCrossRef Morgan KM, Riedlinger GM, Rosenfeld J, Ganesan S, Pine SR. Patient-derived Xenograft models of non-small cell lung Cancer and their potential utility in personalized medicine. Front Oncol. 2017;7:2.PubMedPubMedCentralCrossRef
48.
go back to reference Ben-David U, Ha G, Tseng YY, Greenwald NF, Oh C, Shih J, et al. Patient-derived xenografts undergo mouse-specific tumor evolution. Nat Genet. 2017;49(11):1567–75.PubMedPubMedCentralCrossRef Ben-David U, Ha G, Tseng YY, Greenwald NF, Oh C, Shih J, et al. Patient-derived xenografts undergo mouse-specific tumor evolution. Nat Genet. 2017;49(11):1567–75.PubMedPubMedCentralCrossRef
49.
go back to reference Hidalgo M, Amant F, Biankin AV, Budinská E, Byrne AT, Caldas C, et al. Patient-derived xenograft models: an emerging platform for translational cancer research. Cancer Discov. 2014;4(9):998–1013.PubMedPubMedCentralCrossRef Hidalgo M, Amant F, Biankin AV, Budinská E, Byrne AT, Caldas C, et al. Patient-derived xenograft models: an emerging platform for translational cancer research. Cancer Discov. 2014;4(9):998–1013.PubMedPubMedCentralCrossRef
50.
go back to reference Woo XY, Giordano J, Srivastava A, Zhao ZM, Lloyd MW, de Bruijn R, et al. Conservation of copy number profiles during engraftment and passaging of patient-derived cancer xenografts. Nat Genet. 2021;53(1):86–99.PubMedPubMedCentralCrossRef Woo XY, Giordano J, Srivastava A, Zhao ZM, Lloyd MW, de Bruijn R, et al. Conservation of copy number profiles during engraftment and passaging of patient-derived cancer xenografts. Nat Genet. 2021;53(1):86–99.PubMedPubMedCentralCrossRef
51.
go back to reference Byrne AT, Alférez DG, Amant F, Annibali D, Arribas J, Biankin AV, et al. Interrogating open issues in cancer precision medicine with patient-derived xenografts. Nat Rev Cancer. 2017;17(4):254–68.PubMedCrossRef Byrne AT, Alférez DG, Amant F, Annibali D, Arribas J, Biankin AV, et al. Interrogating open issues in cancer precision medicine with patient-derived xenografts. Nat Rev Cancer. 2017;17(4):254–68.PubMedCrossRef
52.
go back to reference Evrard YA, Srivastava A, Randjelovic J, Consortium NP, Doroshow JH, Dean DA, et al. Systematic Establishment of Robustness and Standards in Patient-Derived Xenograft Experiments and Analysis. Cancer Res. 2020;80(11):2286–97. Evrard YA, Srivastava A, Randjelovic J, Consortium NP, Doroshow JH, Dean DA, et al. Systematic Establishment of Robustness and Standards in Patient-Derived Xenograft Experiments and Analysis. Cancer Res. 2020;80(11):2286–97.
53.
go back to reference Meehan TF, Conte N, Goldstein T, Inghirami G, Murakami MA, Brabetz S, et al. PDX-MI: minimal information for patient-derived tumor Xenograft models. Cancer Res. 2017;77(21):e62–e6.PubMedPubMedCentralCrossRef Meehan TF, Conte N, Goldstein T, Inghirami G, Murakami MA, Brabetz S, et al. PDX-MI: minimal information for patient-derived tumor Xenograft models. Cancer Res. 2017;77(21):e62–e6.PubMedPubMedCentralCrossRef
54.
go back to reference Gao H, Korn JM, Ferretti S, Monahan JE, Wang Y, Singh M, et al. High-throughput screening using patient-derived tumor xenografts to predict clinical trial drug response. Nat Med. 2015;21(11):1318–25.PubMedCrossRef Gao H, Korn JM, Ferretti S, Monahan JE, Wang Y, Singh M, et al. High-throughput screening using patient-derived tumor xenografts to predict clinical trial drug response. Nat Med. 2015;21(11):1318–25.PubMedCrossRef
55.
go back to reference Bertotti A, Migliardi G, Galimi F, Sassi F, Torti D, Isella C, et al. A molecularly annotated platform of patient-derived xenografts ("xenopatients") identifies HER2 as an effective therapeutic target in cetuximab-resistant colorectal cancer. Cancer Discov. 2011;1(6):508–23.PubMedCrossRef Bertotti A, Migliardi G, Galimi F, Sassi F, Torti D, Isella C, et al. A molecularly annotated platform of patient-derived xenografts ("xenopatients") identifies HER2 as an effective therapeutic target in cetuximab-resistant colorectal cancer. Cancer Discov. 2011;1(6):508–23.PubMedCrossRef
56.
go back to reference Kavuri SM, Jain N, Galimi F, Cottino F, Leto SM, Migliardi G, et al. HER2 activating mutations are targets for colorectal cancer treatment. Cancer Discov. 2015;5(8):832–41.PubMedPubMedCentralCrossRef Kavuri SM, Jain N, Galimi F, Cottino F, Leto SM, Migliardi G, et al. HER2 activating mutations are targets for colorectal cancer treatment. Cancer Discov. 2015;5(8):832–41.PubMedPubMedCentralCrossRef
57.
go back to reference Bertotti A, Papp E, Jones S, Adleff V, Anagnostou V, Lupo B, et al. The genomic landscape of response to EGFR blockade in colorectal cancer. Nature. 2015;526(7572):263–7.PubMedPubMedCentralCrossRef Bertotti A, Papp E, Jones S, Adleff V, Anagnostou V, Lupo B, et al. The genomic landscape of response to EGFR blockade in colorectal cancer. Nature. 2015;526(7572):263–7.PubMedPubMedCentralCrossRef
58.
go back to reference Sartore-Bianchi A, Trusolino L, Martino C, Bencardino K, Lonardi S, Bergamo F, et al. Dual-targeted therapy with trastuzumab and lapatinib in treatment-refractory, KRAS codon 12/13 wild-type, HER2-positive metastatic colorectal cancer (HERACLES): a proof-of-concept, multicentre, open-label, phase 2 trial. Lancet Oncol. 2016;17(6):738–46.PubMedCrossRef Sartore-Bianchi A, Trusolino L, Martino C, Bencardino K, Lonardi S, Bergamo F, et al. Dual-targeted therapy with trastuzumab and lapatinib in treatment-refractory, KRAS codon 12/13 wild-type, HER2-positive metastatic colorectal cancer (HERACLES): a proof-of-concept, multicentre, open-label, phase 2 trial. Lancet Oncol. 2016;17(6):738–46.PubMedCrossRef
59.
go back to reference Sartore-Bianchi A, Lonardi S, Aglietta M, Martino C, Ciardiello F, Marsoni S, et al. Central nervous system as possible site of relapse in ERBB2-positive metastatic colorectal Cancer: long-term results of treatment with Trastuzumab and Lapatinib. JAMA Oncol. 2020;6(6):927–9.PubMedPubMedCentralCrossRef Sartore-Bianchi A, Lonardi S, Aglietta M, Martino C, Ciardiello F, Marsoni S, et al. Central nervous system as possible site of relapse in ERBB2-positive metastatic colorectal Cancer: long-term results of treatment with Trastuzumab and Lapatinib. JAMA Oncol. 2020;6(6):927–9.PubMedPubMedCentralCrossRef
60.
go back to reference Tosi F, Sartore-Bianchi A, Lonardi S, Amatu A, Leone F, Ghezzi S, et al. Long-term clinical outcome of Trastuzumab and Lapatinib for HER2-positive metastatic colorectal Cancer. Clin Colorectal Cancer. 2020;19(4):256–62.e2. Tosi F, Sartore-Bianchi A, Lonardi S, Amatu A, Leone F, Ghezzi S, et al. Long-term clinical outcome of Trastuzumab and Lapatinib for HER2-positive metastatic colorectal Cancer. Clin Colorectal Cancer. 2020;19(4):256–62.e2.
61.
go back to reference Sartore-Bianchi A, Lonardi S, Martino C, Fenocchio E, Tosi F, Ghezzi S, et al. Pertuzumab and trastuzumab emtansine in patients with HER2-amplified metastatic colorectal cancer: the phase II HERACLES-B trial. ESMO Open. 2020;5(5):e000911. Sartore-Bianchi A, Lonardi S, Martino C, Fenocchio E, Tosi F, Ghezzi S, et al. Pertuzumab and trastuzumab emtansine in patients with HER2-amplified metastatic colorectal cancer: the phase II HERACLES-B trial. ESMO Open. 2020;5(5):e000911.
62.
go back to reference Lazzari L, Corti G, Picco G, Isella C, Montone M, Arcella P, et al. Patient-derived Xenografts and matched cell lines identify Pharmacogenomic vulnerabilities in colorectal Cancer. Clin Cancer Res. 2019;25(20):6243–59.PubMedCrossRefPubMedCentral Lazzari L, Corti G, Picco G, Isella C, Montone M, Arcella P, et al. Patient-derived Xenografts and matched cell lines identify Pharmacogenomic vulnerabilities in colorectal Cancer. Clin Cancer Res. 2019;25(20):6243–59.PubMedCrossRefPubMedCentral
63.
go back to reference Sartore-Bianchi A, Amatu A, Porcu L, Ghezzi S, Lonardi S, Leone F, et al. HER2 positivity predicts unresponsiveness to EGFR-targeted treatment in metastatic colorectal Cancer. Oncologist. 2019;24(10):1395–402.PubMedPubMedCentralCrossRef Sartore-Bianchi A, Amatu A, Porcu L, Ghezzi S, Lonardi S, Leone F, et al. HER2 positivity predicts unresponsiveness to EGFR-targeted treatment in metastatic colorectal Cancer. Oncologist. 2019;24(10):1395–402.PubMedPubMedCentralCrossRef
64.
go back to reference Martinelli E, Troiani T, Sforza V, Martini G, Cardone C, Vitiello PP, et al. Sequential HER2 blockade as effective therapy in chemorefractory, HER2 gene-amplified, RAS wild-type, metastatic colorectal cancer: learning from a clinical case. ESMO Open. 2018;3(1):e000299.PubMedPubMedCentralCrossRef Martinelli E, Troiani T, Sforza V, Martini G, Cardone C, Vitiello PP, et al. Sequential HER2 blockade as effective therapy in chemorefractory, HER2 gene-amplified, RAS wild-type, metastatic colorectal cancer: learning from a clinical case. ESMO Open. 2018;3(1):e000299.PubMedPubMedCentralCrossRef
65.
go back to reference Li BT, Michelini F, Misale S, Cocco E, Baldino L, Cai Y, et al. HER2-mediated internalization of cytotoxic agents in. Cancer Discov. 2020;10(5):674–87.PubMedPubMedCentralCrossRef Li BT, Michelini F, Misale S, Cocco E, Baldino L, Cai Y, et al. HER2-mediated internalization of cytotoxic agents in. Cancer Discov. 2020;10(5):674–87.PubMedPubMedCentralCrossRef
66.
go back to reference Roque-Lima B, Roque CCTA, Begnami MD, Peresi P, Lima ENP, Mello CAL, et al. Development of patient-derived orthotopic xenografts from metastatic colorectal cancer in nude mice. J Drug Target. 2019;27(9):943–9.PubMedCrossRef Roque-Lima B, Roque CCTA, Begnami MD, Peresi P, Lima ENP, Mello CAL, et al. Development of patient-derived orthotopic xenografts from metastatic colorectal cancer in nude mice. J Drug Target. 2019;27(9):943–9.PubMedCrossRef
67.
go back to reference Galimi F, Torti D, Sassi F, Isella C, Corà D, Gastaldi S, et al. Genetic and expression analysis of MET, MACC1, and HGF in metastatic colorectal cancer: response to met inhibition in patient xenografts and pathologic correlations. Clin Cancer Res. 2011;17(10):3146–56.PubMedCrossRef Galimi F, Torti D, Sassi F, Isella C, Corà D, Gastaldi S, et al. Genetic and expression analysis of MET, MACC1, and HGF in metastatic colorectal cancer: response to met inhibition in patient xenografts and pathologic correlations. Clin Cancer Res. 2011;17(10):3146–56.PubMedCrossRef
68.
go back to reference Picco G, Petti C, Sassi F, Grillone K, Migliardi G, Rossi T, et al. Efficacy of NEDD8 Pathway Inhibition in Preclinical Models of Poorly Differentiated, Clinically Aggressive Colorectal Cancer. J Natl Cancer Inst. 2017;109(2):djw209. Picco G, Petti C, Sassi F, Grillone K, Migliardi G, Rossi T, et al. Efficacy of NEDD8 Pathway Inhibition in Preclinical Models of Poorly Differentiated, Clinically Aggressive Colorectal Cancer. J Natl Cancer Inst. 2017;109(2):djw209.
69.
go back to reference André T, Shiu KK, Kim TW, Jensen BV, Jensen LH, Punt C, et al. Pembrolizumab in microsatellite-instability-high advanced colorectal Cancer. N Engl J Med. 2020;383(23):2207–18.PubMedCrossRef André T, Shiu KK, Kim TW, Jensen BV, Jensen LH, Punt C, et al. Pembrolizumab in microsatellite-instability-high advanced colorectal Cancer. N Engl J Med. 2020;383(23):2207–18.PubMedCrossRef
70.
go back to reference Ganesh K, Stadler ZK, Cercek A, Mendelsohn RB, Shia J, Segal NH, et al. Immunotherapy in colorectal cancer: rationale, challenges and potential. Nat Rev Gastroenterol Hepatol. 2019;16(6):361–75.PubMedPubMedCentralCrossRef Ganesh K, Stadler ZK, Cercek A, Mendelsohn RB, Shia J, Segal NH, et al. Immunotherapy in colorectal cancer: rationale, challenges and potential. Nat Rev Gastroenterol Hepatol. 2019;16(6):361–75.PubMedPubMedCentralCrossRef
71.
go back to reference Golshani G, Zhang Y. Advances in immunotherapy for colorectal cancer: a review. Ther Adv Gastroenterol. 2020;13:1756284820917527.CrossRef Golshani G, Zhang Y. Advances in immunotherapy for colorectal cancer: a review. Ther Adv Gastroenterol. 2020;13:1756284820917527.CrossRef
72.
go back to reference Tintelnot J, Stein A. Immunotherapy in colorectal cancer: available clinical evidence, challenges and novel approaches. World J Gastroenterol. 2019;25(29):3920–8.PubMedPubMedCentralCrossRef Tintelnot J, Stein A. Immunotherapy in colorectal cancer: available clinical evidence, challenges and novel approaches. World J Gastroenterol. 2019;25(29):3920–8.PubMedPubMedCentralCrossRef
73.
go back to reference Choi Y, Lee S, Kim K, Kim SH, Chung YJ, Lee C. Studying cancer immunotherapy using patient-derived xenografts (PDXs) in humanized mice. Exp Mol Med. 2018;50(8):99.PubMedCentral Choi Y, Lee S, Kim K, Kim SH, Chung YJ, Lee C. Studying cancer immunotherapy using patient-derived xenografts (PDXs) in humanized mice. Exp Mol Med. 2018;50(8):99.PubMedCentral
74.
go back to reference Teng R, Zhao J, Zhao Y, Gao J, Li H, Zhou S, et al. Chimeric antigen receptor-modified T cells repressed solid tumors and their relapse in an established patient-derived Colon carcinoma Xenograft model. J Immunother. 2019;42(2):33–42.PubMedCrossRef Teng R, Zhao J, Zhao Y, Gao J, Li H, Zhou S, et al. Chimeric antigen receptor-modified T cells repressed solid tumors and their relapse in an established patient-derived Colon carcinoma Xenograft model. J Immunother. 2019;42(2):33–42.PubMedCrossRef
75.
go back to reference Janssen E, Subtil B, de la Jara OF, Verheul HMW, Tauriello DVF. Combinatorial Immunotherapies for Metastatic Colorectal Cancer. Cancers (Basel). 2020;12(7):1875. Janssen E, Subtil B, de la Jara OF, Verheul HMW, Tauriello DVF. Combinatorial Immunotherapies for Metastatic Colorectal Cancer. Cancers (Basel). 2020;12(7):1875.
76.
go back to reference Hidalgo M, Bruckheimer E, Rajeshkumar NV, Garrido-Laguna I, De Oliveira E, Rubio-Viqueira B, et al. A pilot clinical study of treatment guided by personalized tumorgrafts in patients with advanced cancer. Mol Cancer Ther. 2011;10(8):1311–6.PubMedPubMedCentralCrossRef Hidalgo M, Bruckheimer E, Rajeshkumar NV, Garrido-Laguna I, De Oliveira E, Rubio-Viqueira B, et al. A pilot clinical study of treatment guided by personalized tumorgrafts in patients with advanced cancer. Mol Cancer Ther. 2011;10(8):1311–6.PubMedPubMedCentralCrossRef
77.
go back to reference Morelli MP, Calvo E, Ordoñez E, Wick MJ, Viqueira BR, Lopez-Casas PP, et al. Prioritizing phase I treatment options through preclinical testing on personalized tumorgraft. J Clin Oncol. 2012;30(4):e45–8.PubMedCrossRef Morelli MP, Calvo E, Ordoñez E, Wick MJ, Viqueira BR, Lopez-Casas PP, et al. Prioritizing phase I treatment options through preclinical testing on personalized tumorgraft. J Clin Oncol. 2012;30(4):e45–8.PubMedCrossRef
78.
go back to reference Clohessy JG, Pandolfi PP. Mouse hospital and co-clinical trial project--from bench to bedside. Nat Rev Clin Oncol. 2015;12(8):491–8.PubMedCrossRef Clohessy JG, Pandolfi PP. Mouse hospital and co-clinical trial project--from bench to bedside. Nat Rev Clin Oncol. 2015;12(8):491–8.PubMedCrossRef
79.
go back to reference Julien S, Merino-Trigo A, Lacroix L, Pocard M, Goéré D, Mariani P, et al. Characterization of a large panel of patient-derived tumor xenografts representing the clinical heterogeneity of human colorectal cancer. Clin Cancer Res. 2012;18(19):5314–28.PubMedCrossRef Julien S, Merino-Trigo A, Lacroix L, Pocard M, Goéré D, Mariani P, et al. Characterization of a large panel of patient-derived tumor xenografts representing the clinical heterogeneity of human colorectal cancer. Clin Cancer Res. 2012;18(19):5314–28.PubMedCrossRef
80.
go back to reference Peng S, Creighton CJ, Zhang Y, Sen B, Mazumdar T, Myers JN, et al. Tumor grafts derived from patients with head and neck squamous carcinoma authentically maintain the molecular and histologic characteristics of human cancers. J Transl Med. 2013;11:198.PubMedPubMedCentralCrossRef Peng S, Creighton CJ, Zhang Y, Sen B, Mazumdar T, Myers JN, et al. Tumor grafts derived from patients with head and neck squamous carcinoma authentically maintain the molecular and histologic characteristics of human cancers. J Transl Med. 2013;11:198.PubMedPubMedCentralCrossRef
81.
go back to reference DeRose YS, Wang G, Lin YC, Bernard PS, Buys SS, Ebbert MT, et al. Tumor grafts derived from women with breast cancer authentically reflect tumor pathology, growth, metastasis and disease outcomes. Nat Med. 2011;17(11):1514–20.PubMedPubMedCentralCrossRef DeRose YS, Wang G, Lin YC, Bernard PS, Buys SS, Ebbert MT, et al. Tumor grafts derived from women with breast cancer authentically reflect tumor pathology, growth, metastasis and disease outcomes. Nat Med. 2011;17(11):1514–20.PubMedPubMedCentralCrossRef
82.
go back to reference Chao C, Widen SG, Wood TG, Zatarain JR, Johnson P, Gajjar A, et al. Patient-derived Xenografts from colorectal carcinoma: a temporal and hierarchical study of murine stromal cell replacement. Anticancer Res. 2017;37(7):3405–12.PubMedPubMedCentral Chao C, Widen SG, Wood TG, Zatarain JR, Johnson P, Gajjar A, et al. Patient-derived Xenografts from colorectal carcinoma: a temporal and hierarchical study of murine stromal cell replacement. Anticancer Res. 2017;37(7):3405–12.PubMedPubMedCentral
83.
go back to reference Dieter SM, Giessler KM, Kriegsmann M, Dubash TD, Möhrmann L, Schulz ER, et al. Patient-derived xenografts of gastrointestinal cancers are susceptible to rapid and delayed B-lymphoproliferation. Int J Cancer. 2017;140(6):1356–63.PubMedCrossRef Dieter SM, Giessler KM, Kriegsmann M, Dubash TD, Möhrmann L, Schulz ER, et al. Patient-derived xenografts of gastrointestinal cancers are susceptible to rapid and delayed B-lymphoproliferation. Int J Cancer. 2017;140(6):1356–63.PubMedCrossRef
84.
go back to reference Weinstein JN, Myers TG, O'Connor PM, Friend SH, Fornace AJ, Kohn KW, et al. An information-intensive approach to the molecular pharmacology of cancer. Science. 1997;275(5298):343–9.PubMedCrossRef Weinstein JN, Myers TG, O'Connor PM, Friend SH, Fornace AJ, Kohn KW, et al. An information-intensive approach to the molecular pharmacology of cancer. Science. 1997;275(5298):343–9.PubMedCrossRef
85.
go back to reference Shoemaker RH. The NCI60 human tumour cell line anticancer drug screen. Nat Rev Cancer. 2006;6(10):813–23.PubMedCrossRef Shoemaker RH. The NCI60 human tumour cell line anticancer drug screen. Nat Rev Cancer. 2006;6(10):813–23.PubMedCrossRef
86.
go back to reference Garnett MJ, Edelman EJ, Heidorn SJ, Greenman CD, Dastur A, Lau KW, et al. Systematic identification of genomic markers of drug sensitivity in cancer cells. Nature. 2012;483(7391):570–5.PubMedPubMedCentralCrossRef Garnett MJ, Edelman EJ, Heidorn SJ, Greenman CD, Dastur A, Lau KW, et al. Systematic identification of genomic markers of drug sensitivity in cancer cells. Nature. 2012;483(7391):570–5.PubMedPubMedCentralCrossRef
87.
go back to reference Seashore-Ludlow B, Rees MG, Cheah JH, Cokol M, Price EV, Coletti ME, et al. Harnessing connectivity in a large-scale small-molecule sensitivity dataset. Cancer Discov. 2015;5(11):1210–23.PubMedPubMedCentralCrossRef Seashore-Ludlow B, Rees MG, Cheah JH, Cokol M, Price EV, Coletti ME, et al. Harnessing connectivity in a large-scale small-molecule sensitivity dataset. Cancer Discov. 2015;5(11):1210–23.PubMedPubMedCentralCrossRef
88.
go back to reference Barretina J, Caponigro G, Stransky N, Venkatesan K, Margolin AA, Kim S, et al. The Cancer cell line encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature. 2012;483(7391):603–7.PubMedPubMedCentralCrossRef Barretina J, Caponigro G, Stransky N, Venkatesan K, Margolin AA, Kim S, et al. The Cancer cell line encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature. 2012;483(7391):603–7.PubMedPubMedCentralCrossRef
89.
go back to reference Ghandi M, Huang FW, Jané-Valbuena J, Kryukov GV, Lo CC, McDonald ER, et al. Next-generation characterization of the Cancer cell line encyclopedia. Nature. 2019;569(7757):503–8.PubMedPubMedCentralCrossRef Ghandi M, Huang FW, Jané-Valbuena J, Kryukov GV, Lo CC, McDonald ER, et al. Next-generation characterization of the Cancer cell line encyclopedia. Nature. 2019;569(7757):503–8.PubMedPubMedCentralCrossRef
90.
go back to reference Behan FM, Iorio F, Picco G, Gonçalves E, Beaver CM, Migliardi G, et al. Prioritization of cancer therapeutic targets using CRISPR-Cas9 screens. Nature. 2019;568(7753):511–6.PubMedCrossRef Behan FM, Iorio F, Picco G, Gonçalves E, Beaver CM, Migliardi G, et al. Prioritization of cancer therapeutic targets using CRISPR-Cas9 screens. Nature. 2019;568(7753):511–6.PubMedCrossRef
91.
go back to reference Picco G, Chen ED, Alonso LG, Behan FM, Gonçalves E, Bignell G, et al. Functional linkage of gene fusions to cancer cell fitness assessed by pharmacological and CRISPR-Cas9 screening. Nat Commun. 2019;10(1):2198.PubMedPubMedCentralCrossRef Picco G, Chen ED, Alonso LG, Behan FM, Gonçalves E, Bignell G, et al. Functional linkage of gene fusions to cancer cell fitness assessed by pharmacological and CRISPR-Cas9 screening. Nat Commun. 2019;10(1):2198.PubMedPubMedCentralCrossRef
92.
go back to reference Petljak M, Alexandrov LB, Brammeld JS, Price S, Wedge DC, Grossmann S, et al. Characterizing Mutational Signatures in Human Cancer Cell Lines Reveals Episodic APOBEC Mutagenesis. Cell. 2019;176(6):1282–94 e20.PubMedPubMedCentralCrossRef Petljak M, Alexandrov LB, Brammeld JS, Price S, Wedge DC, Grossmann S, et al. Characterizing Mutational Signatures in Human Cancer Cell Lines Reveals Episodic APOBEC Mutagenesis. Cell. 2019;176(6):1282–94 e20.PubMedPubMedCentralCrossRef
93.
go back to reference Medico E, Russo M, Picco G, Cancelliere C, Valtorta E, Corti G, et al. The molecular landscape of colorectal cancer cell lines unveils clinically actionable kinase targets. Nat Commun. 2015;6:7002.PubMedCrossRef Medico E, Russo M, Picco G, Cancelliere C, Valtorta E, Corti G, et al. The molecular landscape of colorectal cancer cell lines unveils clinically actionable kinase targets. Nat Commun. 2015;6:7002.PubMedCrossRef
94.
go back to reference Arena S, Corti G, Durinikova E, Montone M, Reilly NM, Russo M, et al. A subset of colorectal cancers with Cross-sensitivity to Olaparib and Oxaliplatin. Clin Cancer Res. 2020;26(6):1372–84.PubMedCrossRef Arena S, Corti G, Durinikova E, Montone M, Reilly NM, Russo M, et al. A subset of colorectal cancers with Cross-sensitivity to Olaparib and Oxaliplatin. Clin Cancer Res. 2020;26(6):1372–84.PubMedCrossRef
95.
go back to reference Rospo G, Lorenzato A, Amirouchene-Angelozzi N, Magrì A, Cancelliere C, Corti G, et al. Evolving neoantigen profiles in colorectal cancers with DNA repair defects. Genome Med. 2019;11(1):42.PubMedPubMedCentralCrossRef Rospo G, Lorenzato A, Amirouchene-Angelozzi N, Magrì A, Cancelliere C, Corti G, et al. Evolving neoantigen profiles in colorectal cancers with DNA repair defects. Genome Med. 2019;11(1):42.PubMedPubMedCentralCrossRef
96.
go back to reference Catalano I, Grassi E, Bertotti A, Trusolino L. Immunogenomics of colorectal tumors: facts and hypotheses on an evolving Saga. Trends Cancer. 2019;5(12):779–88.PubMedCrossRef Catalano I, Grassi E, Bertotti A, Trusolino L. Immunogenomics of colorectal tumors: facts and hypotheses on an evolving Saga. Trends Cancer. 2019;5(12):779–88.PubMedCrossRef
97.
98.
go back to reference Wang J, Mouradov D, Wang X, Jorissen RN, Chambers MC, Zimmerman LJ, et al. Colorectal Cancer cell line proteomes are representative of primary tumors and predict drug sensitivity. Gastroenterology. 2017;153(4):1082–95.PubMedCrossRef Wang J, Mouradov D, Wang X, Jorissen RN, Chambers MC, Zimmerman LJ, et al. Colorectal Cancer cell line proteomes are representative of primary tumors and predict drug sensitivity. Gastroenterology. 2017;153(4):1082–95.PubMedCrossRef
99.
go back to reference Gillet JP, Calcagno AM, Varma S, Marino M, Green LJ, Vora MI, 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, Calcagno AM, Varma S, Marino M, Green LJ, Vora MI, 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
101.
go back to reference Hughes P, Marshall D, Reid Y, Parkes H, Gelber C. The costs of using unauthenticated, over-passaged cell lines: how much more data do we need? Biotechniques. 2007;43(5):575 7–8, 81–2 passim.PubMedCrossRef Hughes P, Marshall D, Reid Y, Parkes H, Gelber C. The costs of using unauthenticated, over-passaged cell lines: how much more data do we need? Biotechniques. 2007;43(5):575 7–8, 81–2 passim.PubMedCrossRef
102.
go back to reference Maletzki C, Stier S, Gruenert U, Gock M, Ostwald C, Prall F, et al. Establishment, characterization and chemosensitivity of three mismatch repair deficient cell lines from sporadic and inherited colorectal carcinomas. PLoS One. 2012;7(12):e52485.PubMedPubMedCentralCrossRef Maletzki C, Stier S, Gruenert U, Gock M, Ostwald C, Prall F, et al. Establishment, characterization and chemosensitivity of three mismatch repair deficient cell lines from sporadic and inherited colorectal carcinomas. PLoS One. 2012;7(12):e52485.PubMedPubMedCentralCrossRef
103.
go back to reference Rowehl RA, Burke S, Bialkowska AB, Pettet DW, Rowehl L, Li E, et al. Establishment of highly tumorigenic human colorectal cancer cell line (CR4) with properties of putative cancer stem cells. PLoS One. 2014;9(6):e99091.PubMedPubMedCentralCrossRef Rowehl RA, Burke S, Bialkowska AB, Pettet DW, Rowehl L, Li E, et al. Establishment of highly tumorigenic human colorectal cancer cell line (CR4) with properties of putative cancer stem cells. PLoS One. 2014;9(6):e99091.PubMedPubMedCentralCrossRef
104.
go back to reference Maletzki C, Gock M, Randow M, Klar E, Huehns M, Prall F, et al. Establishment and characterization of cell lines from chromosomal instable colorectal cancer. World J Gastroenterol. 2015;21(1):164–76.PubMedPubMedCentralCrossRef Maletzki C, Gock M, Randow M, Klar E, Huehns M, Prall F, et al. Establishment and characterization of cell lines from chromosomal instable colorectal cancer. World J Gastroenterol. 2015;21(1):164–76.PubMedPubMedCentralCrossRef
105.
go back to reference Song HN, Lee C, Kim ST, Kim SY, Kim NK, Jang J, et al. Molecular characterization of colorectal cancer patients and concomitant patient-derived tumor cell establishment. Oncotarget. 2016;7(15):19610–9.PubMedPubMedCentralCrossRef Song HN, Lee C, Kim ST, Kim SY, Kim NK, Jang J, et al. Molecular characterization of colorectal cancer patients and concomitant patient-derived tumor cell establishment. Oncotarget. 2016;7(15):19610–9.PubMedPubMedCentralCrossRef
106.
go back to reference Ku JL, Shin YK, Kim DW, Kim KH, Choi JS, Hong SH, et al. Establishment and characterization of 13 human colorectal carcinoma cell lines: mutations of genes and expressions of drug-sensitivity genes and cancer stem cell markers. Carcinogenesis. 2010;31(6):1003–9.PubMedCrossRef Ku JL, Shin YK, Kim DW, Kim KH, Choi JS, Hong SH, et al. Establishment and characterization of 13 human colorectal carcinoma cell lines: mutations of genes and expressions of drug-sensitivity genes and cancer stem cell markers. Carcinogenesis. 2010;31(6):1003–9.PubMedCrossRef
107.
go back to reference Boot A, van Eendenburg J, Crobach S, Ruano D, Speetjens F, Calame J, et al. Characterization of novel low passage primary and metastatic colorectal cancer cell lines. Oncotarget. 2016;7(12):14499–509.PubMedPubMedCentralCrossRef Boot A, van Eendenburg J, Crobach S, Ruano D, Speetjens F, Calame J, et al. Characterization of novel low passage primary and metastatic colorectal cancer cell lines. Oncotarget. 2016;7(12):14499–509.PubMedPubMedCentralCrossRef
108.
go back to reference Kim SC, Hong CW, Jang SG, Kim YA, Yoo BC, Shin YK, et al. Establishment and characterization of paired primary and peritoneal seeding human colorectal Cancer cell lines: identification of genes that mediate metastatic potential. Transl Oncol. 2018;11(5):1232–43.PubMedPubMedCentralCrossRef Kim SC, Hong CW, Jang SG, Kim YA, Yoo BC, Shin YK, et al. Establishment and characterization of paired primary and peritoneal seeding human colorectal Cancer cell lines: identification of genes that mediate metastatic potential. Transl Oncol. 2018;11(5):1232–43.PubMedPubMedCentralCrossRef
109.
go back to reference Gock M, Mullins CS, Bergner C, Prall F, Ramer R, Göder A, et al. Establishment, functional and genetic characterization of three novel patient-derived rectal cancer cell lines. World J Gastroenterol. 2018;24(43):4880–92.PubMedPubMedCentralCrossRef Gock M, Mullins CS, Bergner C, Prall F, Ramer R, Göder A, et al. Establishment, functional and genetic characterization of three novel patient-derived rectal cancer cell lines. World J Gastroenterol. 2018;24(43):4880–92.PubMedPubMedCentralCrossRef
110.
go back to reference Francipane MG, Bulanin D, Lagasse E. Establishment and Characterization of 5-Fluorouracil-Resistant Human Colorectal Cancer Stem-Like Cells: Tumor Dynamics under Selection Pressure. Int J Mol Sci. 2019;20(8):1817. Francipane MG, Bulanin D, Lagasse E. Establishment and Characterization of 5-Fluorouracil-Resistant Human Colorectal Cancer Stem-Like Cells: Tumor Dynamics under Selection Pressure. Int J Mol Sci. 2019;20(8):1817.
111.
go back to reference Kim SC, Shin R, Seo HY, Kim M, Park JW, Jeong SY, et al. Identification of a novel fusion gene, FAM174A-WWC1, in early-onset colorectal Cancer: establishment and characterization of four human Cancer cell lines from early-onset colorectal cancers. Transl Oncol. 2019;12(9):1185–95.PubMedPubMedCentralCrossRef Kim SC, Shin R, Seo HY, Kim M, Park JW, Jeong SY, et al. Identification of a novel fusion gene, FAM174A-WWC1, in early-onset colorectal Cancer: establishment and characterization of four human Cancer cell lines from early-onset colorectal cancers. Transl Oncol. 2019;12(9):1185–95.PubMedPubMedCentralCrossRef
112.
go back to reference Dangles-Marie V, Pocard M, Richon S, Weiswald LB, Assayag F, Saulnier P, et al. Establishment of human colon cancer cell lines from fresh tumors versus xenografts: comparison of success rate and cell line features. Cancer Res. 2007;67(1):398–407.PubMedCrossRef Dangles-Marie V, Pocard M, Richon S, Weiswald LB, Assayag F, Saulnier P, et al. Establishment of human colon cancer cell lines from fresh tumors versus xenografts: comparison of success rate and cell line features. Cancer Res. 2007;67(1):398–407.PubMedCrossRef
113.
go back to reference Lerescu L, Tucureanu C, Caraş I, Neagu S, Melinceanu L, Sălăgeanu A. Primary cell culture of human adenocarcinomas--practical considerations. Roum Arch Microbiol Immunol. 2008;67(3–4):55–66.PubMed Lerescu L, Tucureanu C, Caraş I, Neagu S, Melinceanu L, Sălăgeanu A. Primary cell culture of human adenocarcinomas--practical considerations. Roum Arch Microbiol Immunol. 2008;67(3–4):55–66.PubMed
114.
go back to reference Bruna A, Rueda OM, Greenwood W, Batra AS, Callari M, Batra RN, et al. A Biobank of Breast Cancer Explants with Preserved Intra-tumor Heterogeneity to Screen Anticancer Compounds. Cell. 2016;167(1):260–74 e22.PubMedPubMedCentralCrossRef Bruna A, Rueda OM, Greenwood W, Batra AS, Callari M, Batra RN, et al. A Biobank of Breast Cancer Explants with Preserved Intra-tumor Heterogeneity to Screen Anticancer Compounds. Cell. 2016;167(1):260–74 e22.PubMedPubMedCentralCrossRef
115.
go back to reference Damhofer H, Ebbing EA, Steins A, Welling L, Tol JA, Krishnadath KK, et al. Establishment of patient-derived xenograft models and cell lines for malignancies of the upper gastrointestinal tract. J Transl Med. 2015;13:115.PubMedPubMedCentralCrossRef Damhofer H, Ebbing EA, Steins A, Welling L, Tol JA, Krishnadath KK, et al. Establishment of patient-derived xenograft models and cell lines for malignancies of the upper gastrointestinal tract. J Transl Med. 2015;13:115.PubMedPubMedCentralCrossRef
116.
go back to reference Knudsen ES, Balaji U, Mannakee B, Vail P, Eslinger C, Moxom C, et al. Pancreatic cancer cell lines as patient-derived avatars: genetic characterisation and functional utility. Gut. 2018;67(3):508–20.PubMedCrossRef Knudsen ES, Balaji U, Mannakee B, Vail P, Eslinger C, Moxom C, et al. Pancreatic cancer cell lines as patient-derived avatars: genetic characterisation and functional utility. Gut. 2018;67(3):508–20.PubMedCrossRef
117.
go back to reference Fan F, Bellister S, Lu J, Ye X, Boulbes DR, Tozzi F, et al. The requirement for freshly isolated human colorectal cancer (CRC) cells in isolating CRC stem cells. Br J Cancer. 2015;112(3):539–46.PubMedCrossRef Fan F, Bellister S, Lu J, Ye X, Boulbes DR, Tozzi F, et al. The requirement for freshly isolated human colorectal cancer (CRC) cells in isolating CRC stem cells. Br J Cancer. 2015;112(3):539–46.PubMedCrossRef
118.
go back to reference Leto SM, Sassi F, Catalano I, Torri V, Migliardi G, Zanella ER, et al. Sustained inhibition of HER3 and EGFR is necessary to induce regression of HER2-amplified gastrointestinal carcinomas. Clin Cancer Res. 2015;21(24):5519–31.PubMedCrossRef Leto SM, Sassi F, Catalano I, Torri V, Migliardi G, Zanella ER, et al. Sustained inhibition of HER3 and EGFR is necessary to induce regression of HER2-amplified gastrointestinal carcinomas. Clin Cancer Res. 2015;21(24):5519–31.PubMedCrossRef
119.
go back to reference Sato T, Vries RG, Snippert HJ, van de Wetering M, Barker N, Stange DE, et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 2009;459(7244):262–5.PubMedCrossRef Sato T, Vries RG, Snippert HJ, van de Wetering M, Barker N, Stange DE, et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 2009;459(7244):262–5.PubMedCrossRef
120.
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
121.
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):eaay2574. 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):eaay2574.
122.
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
123.
go back to reference Fujii M, Shimokawa M, Date S, Takano A, Matano M, Nanki K, et al. A colorectal tumor Organoid library demonstrates progressive loss of niche factor requirements during tumorigenesis. Cell Stem Cell. 2016;18(6):827–38.PubMedCrossRef Fujii M, Shimokawa M, Date S, Takano A, Matano M, Nanki K, et al. A colorectal tumor Organoid library demonstrates progressive loss of niche factor requirements during tumorigenesis. Cell Stem Cell. 2016;18(6):827–38.PubMedCrossRef
124.
go back to reference Fujii M, Matano M, Toshimitsu K, Takano A, Mikami Y, Nishikori S, et al. Human Intestinal Organoids Maintain Self-Renewal Capacity and Cellular Diversity in Niche-Inspired Culture Condition. Cell Stem Cell. 2018;23(6):787–93 e6.PubMedCrossRef Fujii M, Matano M, Toshimitsu K, Takano A, Mikami Y, Nishikori S, et al. Human Intestinal Organoids Maintain Self-Renewal Capacity and Cellular Diversity in Niche-Inspired Culture Condition. Cell Stem Cell. 2018;23(6):787–93 e6.PubMedCrossRef
125.
go back to reference Pleguezuelos-Manzano C, Puschhof J, van den Brink S, Geurts V, Beumer J, Clevers H. Establishment and culture of human intestinal Organoids derived from adult stem cells. Curr Protoc Immunol. 2020;130(1):e106.PubMed Pleguezuelos-Manzano C, Puschhof J, van den Brink S, Geurts V, Beumer J, Clevers H. Establishment and culture of human intestinal Organoids derived from adult stem cells. Curr Protoc Immunol. 2020;130(1):e106.PubMed
126.
go back to reference Crespo M, Vilar E, Tsai SY, Chang K, Amin S, Srinivasan T, et al. Colonic organoids derived from human induced pluripotent stem cells for modeling colorectal cancer and drug testing. Nat Med. 2017;23(7):878–84.PubMedPubMedCentralCrossRef Crespo M, Vilar E, Tsai SY, Chang K, Amin S, Srinivasan T, et al. Colonic organoids derived from human induced pluripotent stem cells for modeling colorectal cancer and drug testing. Nat Med. 2017;23(7):878–84.PubMedPubMedCentralCrossRef
127.
go back to reference Kondo J, Endo H, Okuyama H, Ishikawa O, Iishi H, Tsujii M, et al. Retaining cell-cell contact enables preparation and culture of spheroids composed of pure primary cancer cells from colorectal cancer. Proc Natl Acad Sci U S A. 2011;108(15):6235–40.PubMedPubMedCentralCrossRef Kondo J, Endo H, Okuyama H, Ishikawa O, Iishi H, Tsujii M, et al. Retaining cell-cell contact enables preparation and culture of spheroids composed of pure primary cancer cells from colorectal cancer. Proc Natl Acad Sci U S A. 2011;108(15):6235–40.PubMedPubMedCentralCrossRef
128.
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
129.
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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
130.
go back to reference Pasch CA, Favreau PF, Yueh AE, Babiarz CP, Gillette AA, Sharick JT, et al. Patient-derived Cancer Organoid cultures to predict sensitivity to chemotherapy and radiation. Clin Cancer Res. 2019;25(17):5376–87.PubMedPubMedCentralCrossRef Pasch CA, Favreau PF, Yueh AE, Babiarz CP, Gillette AA, Sharick JT, et al. Patient-derived Cancer Organoid cultures to predict sensitivity to chemotherapy and radiation. Clin Cancer Res. 2019;25(17):5376–87.PubMedPubMedCentralCrossRef
131.
go back to reference Roerink SF, Sasaki N, Lee-Six H, Young MD, Alexandrov LB, Behjati S, et al. Intra-tumour diversification in colorectal cancer at the single-cell level. Nature. 2018;556(7702):457–62.PubMedCrossRef Roerink SF, Sasaki N, Lee-Six H, Young MD, Alexandrov LB, Behjati S, et al. Intra-tumour diversification in colorectal cancer at the single-cell level. Nature. 2018;556(7702):457–62.PubMedCrossRef
132.
go back to reference Yan HHN, Siu HC, Ho SL, Yue SSK, Gao Y, Tsui WY, et al. Organoid cultures of early-onset colorectal cancers reveal distinct and rare genetic profiles. Gut. 2020;69(12):2165–79. Yan HHN, Siu HC, Ho SL, Yue SSK, Gao Y, Tsui WY, et al. Organoid cultures of early-onset colorectal cancers reveal distinct and rare genetic profiles. Gut. 2020;69(12):2165–79.
133.
go back to reference Bruun J, Kryeziu K, Eide PW, Moosavi SH, Eilertsen IA, Langerud J, et al. Patient-derived organoids from multiple colorectal cancer liver metastases reveal moderate intra-patient pharmacotranscriptomic heterogeneity. Clin Cancer Res. 2020;26(15):4107–19. Bruun J, Kryeziu K, Eide PW, Moosavi SH, Eilertsen IA, Langerud J, et al. Patient-derived organoids from multiple colorectal cancer liver metastases reveal moderate intra-patient pharmacotranscriptomic heterogeneity. Clin Cancer Res. 2020;26(15):4107–19.
134.
go back to reference Narasimhan V, Wright JA, Churchill M, Wang T, Rosati R, Lannagan TR, 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. Narasimhan V, Wright JA, Churchill M, Wang T, Rosati R, Lannagan TR, 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.
135.
go back to reference Li X, Larsson P, Ljuslinder I, Öhlund D, Myte R, Löfgren-Burström A, et al. Ex Vivo Organoid Cultures Reveal the Importance of the Tumor Microenvironment for Maintenance of Colorectal Cancer Stem Cells. Cancers (Basel). 2020;12(4):923. Li X, Larsson P, Ljuslinder I, Öhlund D, Myte R, Löfgren-Burström A, et al. Ex Vivo Organoid Cultures Reveal the Importance of the Tumor Microenvironment for Maintenance of Colorectal Cancer Stem Cells. Cancers (Basel). 2020;12(4):923.
136.
go back to reference Nikolaev M, Mitrofanova O, Broguiere N, Geraldo S, Dutta D, Tabata Y, et al. Homeostatic mini-intestines through scaffold-guided organoid morphogenesis. Nature. 2020;585(7826):574–8.PubMedCrossRef Nikolaev M, Mitrofanova O, Broguiere N, Geraldo S, Dutta D, Tabata Y, et al. Homeostatic mini-intestines through scaffold-guided organoid morphogenesis. Nature. 2020;585(7826):574–8.PubMedCrossRef
137.
go back to reference Verissimo CS, Overmeer RM, Ponsioen B, Drost J, Mertens S, Verlaan-Klink I, et al. Targeting mutant RAS in patient-derived colorectal cancer organoids by combinatorial drug screening. Elife. 2016;5:e18489. Verissimo CS, Overmeer RM, Ponsioen B, Drost J, Mertens S, Verlaan-Klink I, et al. Targeting mutant RAS in patient-derived colorectal cancer organoids by combinatorial drug screening. Elife. 2016;5:e18489.
138.
go back to reference Smyth E, Khan K, Valeri N. Translational research and application of basic biology to clinical trial development in GI cancers. Ann Transl Med. 2018;6(9):164.PubMedPubMedCentralCrossRef Smyth E, Khan K, Valeri N. Translational research and application of basic biology to clinical trial development in GI cancers. Ann Transl Med. 2018;6(9):164.PubMedPubMedCentralCrossRef
139.
go back to reference Lindeboom RG, van Voorthuijsen L, Oost KC, Rodríguez-Colman MJ, Luna-Velez MV, Furlan C, et al. Integrative multi-omics analysis of intestinal organoid differentiation. Mol Syst Biol. 2018;14(6):e8227.PubMedPubMedCentralCrossRef Lindeboom RG, van Voorthuijsen L, Oost KC, Rodríguez-Colman MJ, Luna-Velez MV, Furlan C, et al. Integrative multi-omics analysis of intestinal organoid differentiation. Mol Syst Biol. 2018;14(6):e8227.PubMedPubMedCentralCrossRef
140.
go back to reference Brazovskaja A, Treutlein B, Camp JG. High-throughput single-cell transcriptomics on organoids. Curr Opin Biotechnol. 2019;55:167–71.PubMedCrossRef Brazovskaja A, Treutlein B, Camp JG. High-throughput single-cell transcriptomics on organoids. Curr Opin Biotechnol. 2019;55:167–71.PubMedCrossRef
141.
go back to reference Schumacher D, Andrieux G, Boehnke K, Keil M, Silvestri A, Silvestrov M, et al. Heterogeneous pathway activation and drug response modelled in colorectal-tumor-derived 3D cultures. PLoS Genet. 2019;15(3):e1008076.PubMedPubMedCentralCrossRef Schumacher D, Andrieux G, Boehnke K, Keil M, Silvestri A, Silvestrov M, et al. Heterogeneous pathway activation and drug response modelled in colorectal-tumor-derived 3D cultures. PLoS Genet. 2019;15(3):e1008076.PubMedPubMedCentralCrossRef
142.
go back to reference Picco G, Petti C, Centonze A, Torchiaro E, Crisafulli G, Novara L, et al. Loss of AXIN1 drives acquired resistance to WNT pathway blockade in colorectal cancer cells carrying RSPO3 fusions. EMBO Mol Med. 2017;9(3):293–303.PubMedPubMedCentralCrossRef Picco G, Petti C, Centonze A, Torchiaro E, Crisafulli G, Novara L, et al. Loss of AXIN1 drives acquired resistance to WNT pathway blockade in colorectal cancer cells carrying RSPO3 fusions. EMBO Mol Med. 2017;9(3):293–303.PubMedPubMedCentralCrossRef
143.
go back to reference Seshagiri S, Stawiski EW, Durinck S, Modrusan Z, Storm EE, Conboy CB, et al. Recurrent R-spondin fusions in colon cancer. Nature. 2012;488(7413):660–4.PubMedPubMedCentralCrossRef Seshagiri S, Stawiski EW, Durinck S, Modrusan Z, Storm EE, Conboy CB, et al. Recurrent R-spondin fusions in colon cancer. Nature. 2012;488(7413):660–4.PubMedPubMedCentralCrossRef
144.
go back to reference Bolhaqueiro ACF, Ponsioen B, Bakker B, Klaasen SJ, Kucukkose E, van Jaarsveld RH, et al. Ongoing chromosomal instability and karyotype evolution in human colorectal cancer organoids. Nat Genet. 2019;51(5):824–34.PubMedCrossRef Bolhaqueiro ACF, Ponsioen B, Bakker B, Klaasen SJ, Kucukkose E, van Jaarsveld RH, et al. Ongoing chromosomal instability and karyotype evolution in human colorectal cancer organoids. Nat Genet. 2019;51(5):824–34.PubMedCrossRef
145.
go back to reference Moore K, Colombo N, Scambia G, Kim BG, Oaknin A, Friedlander M, et al. Maintenance Olaparib in patients with newly diagnosed advanced ovarian Cancer. N Engl J Med. 2018;379(26):2495–505.CrossRefPubMed Moore K, Colombo N, Scambia G, Kim BG, Oaknin A, Friedlander M, et al. Maintenance Olaparib in patients with newly diagnosed advanced ovarian Cancer. N Engl J Med. 2018;379(26):2495–505.CrossRefPubMed
146.
go back to reference Golan T, Hammel P, Reni M, Van Cutsem E, Macarulla T, Hall MJ, et al. Maintenance Olaparib for Germline BRCA-mutated metastatic pancreatic Cancer. N Engl J Med. 2019;381(4):317–27.PubMedPubMedCentralCrossRef Golan T, Hammel P, Reni M, Van Cutsem E, Macarulla T, Hall MJ, et al. Maintenance Olaparib for Germline BRCA-mutated metastatic pancreatic Cancer. N Engl J Med. 2019;381(4):317–27.PubMedPubMedCentralCrossRef
147.
go back to reference Amodio V, Yaeger R, Arcella P, Cancelliere C, Lamba S, Lorenzato A, et al. EGFR blockade reverts resistance to KRAS G12C inhibition in colorectal cancer. Cancer Discov. 2020;10(8):1129–39. Amodio V, Yaeger R, Arcella P, Cancelliere C, Lamba S, Lorenzato A, et al. EGFR blockade reverts resistance to KRAS G12C inhibition in colorectal cancer. Cancer Discov. 2020;10(8):1129–39.
148.
go back to reference Lorenzato A, Magrì A, Matafora V, Audrito V, Arcella P, Lazzari L, et al. Vitamin C Restricts the Emergence of Acquired Resistance to EGFR-Targeted Therapies in Colorectal Cancer. Cancers (Basel). 2020;12(3). Lorenzato A, Magrì A, Matafora V, Audrito V, Arcella P, Lazzari L, et al. Vitamin C Restricts the Emergence of Acquired Resistance to EGFR-Targeted Therapies in Colorectal Cancer. Cancers (Basel). 2020;12(3).
149.
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
150.
go back to reference Neal JT, Li X, Zhu J, Giangarra V, Grzeskowiak CL, Ju J, et al. Organoid Modeling of the Tumor Immune Microenvironment. Cell. 2018;175(7):1972–88 e16.PubMedPubMedCentralCrossRef Neal JT, Li X, Zhu J, Giangarra V, Grzeskowiak CL, Ju J, et al. Organoid Modeling of the Tumor Immune Microenvironment. Cell. 2018;175(7):1972–88 e16.PubMedPubMedCentralCrossRef
151.
go back to reference Cattaneo CM, Dijkstra KK, Fanchi LF, Kelderman S, Kaing S, van Rooij N, et al. Tumor organoid-T-cell coculture systems. Nat Protoc. 2020;15(1):15–39.PubMedCrossRef Cattaneo CM, Dijkstra KK, Fanchi LF, Kelderman S, Kaing S, van Rooij N, et al. Tumor organoid-T-cell coculture systems. Nat Protoc. 2020;15(1):15–39.PubMedCrossRef
152.
go back to reference Dijkstra KK, Cattaneo CM, Weeber F, Chalabi M, van de Haar J, Fanchi LF, et al. Generation of Tumor-Reactive T Cells by Co-culture of Peripheral Blood Lymphocytes and Tumor Organoids. Cell. 2018;174(6):1586–98 e12.PubMedPubMedCentralCrossRef Dijkstra KK, Cattaneo CM, Weeber F, Chalabi M, van de Haar J, Fanchi LF, et al. Generation of Tumor-Reactive T Cells by Co-culture of Peripheral Blood Lymphocytes and Tumor Organoids. Cell. 2018;174(6):1586–98 e12.PubMedPubMedCentralCrossRef
153.
go back to reference Fujii M, Clevers H, Sato T. Modeling human digestive diseases with CRISPR-Cas9-modified Organoids. Gastroenterology. 2019;156(3):562–76.PubMedCrossRef Fujii M, Clevers H, Sato T. Modeling human digestive diseases with CRISPR-Cas9-modified Organoids. Gastroenterology. 2019;156(3):562–76.PubMedCrossRef
154.
go back to reference Matano M, Date S, Shimokawa M, Takano A, Fujii M, Ohta Y, et al. Modeling colorectal cancer using CRISPR-Cas9-mediated engineering of human intestinal organoids. Nat Med. 2015;21(3):256–62.PubMedCrossRef Matano M, Date S, Shimokawa M, Takano A, Fujii M, Ohta Y, et al. Modeling colorectal cancer using CRISPR-Cas9-mediated engineering of human intestinal organoids. Nat Med. 2015;21(3):256–62.PubMedCrossRef
155.
go back to reference O'Rourke KP, Loizou E, Livshits G, Schatoff EM, Baslan T, Manchado E, et al. Transplantation of engineered organoids enables rapid generation of metastatic mouse models of colorectal cancer. Nat Biotechnol. 2017;35(6):577–82.PubMedPubMedCentralCrossRef O'Rourke KP, Loizou E, Livshits G, Schatoff EM, Baslan T, Manchado E, et al. Transplantation of engineered organoids enables rapid generation of metastatic mouse models of colorectal cancer. Nat Biotechnol. 2017;35(6):577–82.PubMedPubMedCentralCrossRef
156.
go back to reference Cortina C, Turon G, Stork D, Hernando-Momblona X, Sevillano M, Aguilera M, et al. A genome editing approach to study cancer stem cells in human tumors. EMBO Mol Med. 2017;9(7):869–79.PubMedPubMedCentralCrossRef Cortina C, Turon G, Stork D, Hernando-Momblona X, Sevillano M, Aguilera M, et al. A genome editing approach to study cancer stem cells in human tumors. EMBO Mol Med. 2017;9(7):869–79.PubMedPubMedCentralCrossRef
157.
go back to reference Drost J, van Boxtel R, Blokzijl F, Mizutani T, Sasaki N, Sasselli V, et al. Use of CRISPR-modified human stem cell organoids to study the origin of mutational signatures in cancer. Science. 2017;358(6360):234–8.PubMedPubMedCentralCrossRef Drost J, van Boxtel R, Blokzijl F, Mizutani T, Sasaki N, Sasselli V, et al. Use of CRISPR-modified human stem cell organoids to study the origin of mutational signatures in cancer. Science. 2017;358(6360):234–8.PubMedPubMedCentralCrossRef
158.
go back to reference Michels BE, Mosa MH, Streibl BI, Zhan T, Menche C, Abou-El-Ardat K, et al. Pooled In Vitro and In Vivo CRISPR-Cas9 Screening Identifies Tumor Suppressors in Human Colon Organoids. Cell Stem Cell. 2020;26(5):782–92 e7.PubMedCrossRef Michels BE, Mosa MH, Streibl BI, Zhan T, Menche C, Abou-El-Ardat K, et al. Pooled In Vitro and In Vivo CRISPR-Cas9 Screening Identifies Tumor Suppressors in Human Colon Organoids. Cell Stem Cell. 2020;26(5):782–92 e7.PubMedCrossRef
159.
go back to reference Ringel T, Frey N, Ringnalda F, Janjuha S, Cherkaoui S, Butz S, et al. Genome-Scale CRISPR Screening in Human Intestinal Organoids Identifies Drivers of TGF-β Resistance. Cell Stem Cell. 2020;26(3):431–40 e8.PubMedCrossRef Ringel T, Frey N, Ringnalda F, Janjuha S, Cherkaoui S, Butz S, et al. Genome-Scale CRISPR Screening in Human Intestinal Organoids Identifies Drivers of TGF-β Resistance. Cell Stem Cell. 2020;26(3):431–40 e8.PubMedCrossRef
160.
go back to reference Lannagan TRM, Lee YK, Wang T, Roper J, Bettington ML, Fennell L, et al. Genetic editing of colonic organoids provides a molecularly distinct and orthotopic preclinical model of serrated carcinogenesis. Gut. 2019;68(4):684–92.PubMedCrossRef Lannagan TRM, Lee YK, Wang T, Roper J, Bettington ML, Fennell L, et al. Genetic editing of colonic organoids provides a molecularly distinct and orthotopic preclinical model of serrated carcinogenesis. Gut. 2019;68(4):684–92.PubMedCrossRef
161.
go back to reference Kawasaki K, Fujii M, Sugimoto S, Ishikawa K, Matano M, Ohta Y, et al. Chromosome Engineering of Human Colon-Derived Organoids to Develop a Model of Traditional Serrated Adenoma. Gastroenterology. 2020;158(3):638–51 e8.PubMedCrossRef Kawasaki K, Fujii M, Sugimoto S, Ishikawa K, Matano M, Ohta Y, et al. Chromosome Engineering of Human Colon-Derived Organoids to Develop a Model of Traditional Serrated Adenoma. Gastroenterology. 2020;158(3):638–51 e8.PubMedCrossRef
162.
go back to reference Stangl C, Post JB, van Roosmalen MJ, Hami N, Verlaan-Klink I, Vos HR, et al. Diverse BRAF gene fusions confer resistance to EGFR-targeted therapy via differential modulation of BRAF activity. Mol Cancer Res. 2020;18(4):537–48.PubMedCrossRef Stangl C, Post JB, van Roosmalen MJ, Hami N, Verlaan-Klink I, Vos HR, et al. Diverse BRAF gene fusions confer resistance to EGFR-targeted therapy via differential modulation of BRAF activity. Mol Cancer Res. 2020;18(4):537–48.PubMedCrossRef
163.
go back to reference Tsai YH, Czerwinski M, Wu A, Dame MK, Attili D, Hill E, et al. A Method for Cryogenic Preservation of Human Biopsy Specimens and Subsequent Organoid Culture. Cell Mol Gastroenterol Hepatol. 2018;6(2):218–22 e7.PubMedPubMedCentralCrossRef Tsai YH, Czerwinski M, Wu A, Dame MK, Attili D, Hill E, et al. A Method for Cryogenic Preservation of Human Biopsy Specimens and Subsequent Organoid Culture. Cell Mol Gastroenterol Hepatol. 2018;6(2):218–22 e7.PubMedPubMedCentralCrossRef
164.
go back to reference Bui BN, Boretto M, Kobayashi H, van Hoesel M, Steba GS, van Hoogenhuijze N, et al. Organoids can be established reliably from cryopreserved biopsy catheter-derived endometrial tissue of infertile women. Reprod BioMed Online. 2020;41(3):465–73.PubMedCrossRef Bui BN, Boretto M, Kobayashi H, van Hoesel M, Steba GS, van Hoogenhuijze N, et al. Organoids can be established reliably from cryopreserved biopsy catheter-derived endometrial tissue of infertile women. Reprod BioMed Online. 2020;41(3):465–73.PubMedCrossRef
165.
go back to reference Di Nicolantonio F, Vitiello PP, Marsoni S, Siena S, Tabernero J, Trusolino L, et al. Precision oncology in metastatic colorectal cancer - from biology to medicine. Nat Rev Clin Oncol. 2021. Online ahead of print. Di Nicolantonio F, Vitiello PP, Marsoni S, Siena S, Tabernero J, Trusolino L, et al. Precision oncology in metastatic colorectal cancer - from biology to medicine. Nat Rev Clin Oncol. 2021. Online ahead of print.
166.
167.
go back to reference Meric-Bernstam F, Hurwitz H, Raghav KPS, McWilliams RR, Fakih M, VanderWalde A, et al. Pertuzumab plus trastuzumab for HER2-amplified metastatic colorectal cancer (MyPathway): an updated report from a multicentre, open-label, phase 2a, multiple basket study. Lancet Oncol. 2019;20(4):518–30.PubMedPubMedCentralCrossRef Meric-Bernstam F, Hurwitz H, Raghav KPS, McWilliams RR, Fakih M, VanderWalde A, et al. Pertuzumab plus trastuzumab for HER2-amplified metastatic colorectal cancer (MyPathway): an updated report from a multicentre, open-label, phase 2a, multiple basket study. Lancet Oncol. 2019;20(4):518–30.PubMedPubMedCentralCrossRef
168.
go back to reference Tol J, Nagtegaal ID, Punt CJ. BRAF mutation in metastatic colorectal cancer. N Engl J Med. 2009;361(1):98–9.PubMedCrossRef Tol J, Nagtegaal ID, Punt CJ. BRAF mutation in metastatic colorectal cancer. N Engl J Med. 2009;361(1):98–9.PubMedCrossRef
169.
go back to reference Kopetz S, Grothey A, Yaeger R, Van Cutsem E, Desai J, Yoshino T, et al. Encorafenib, Binimetinib, and Cetuximab in BRAF V600E-mutated colorectal Cancer. N Engl J Med. 2019;381(17):1632–43.PubMedCrossRef Kopetz S, Grothey A, Yaeger R, Van Cutsem E, Desai J, Yoshino T, et al. Encorafenib, Binimetinib, and Cetuximab in BRAF V600E-mutated colorectal Cancer. N Engl J Med. 2019;381(17):1632–43.PubMedCrossRef
170.
go back to reference Corcoran RB, Atreya CE, Falchook GS, Kwak EL, Ryan DP, Bendell JC, et al. Combined BRAF and MEK inhibition with Dabrafenib and Trametinib in BRAF V600-mutant colorectal Cancer. J Clin Oncol. 2015;33(34):4023–31.PubMedPubMedCentralCrossRef Corcoran RB, Atreya CE, Falchook GS, Kwak EL, Ryan DP, Bendell JC, et al. Combined BRAF and MEK inhibition with Dabrafenib and Trametinib in BRAF V600-mutant colorectal Cancer. J Clin Oncol. 2015;33(34):4023–31.PubMedPubMedCentralCrossRef
172.
173.
go back to reference Sartore-Bianchi A, Ardini E, Bosotti R, Amatu A, Valtorta E, Somaschini A, et al. Sensitivity to Entrectinib Associated With a Novel LMNA-NTRK1 Gene Fusion in Metastatic Colorectal Cancer. J Natl Cancer Inst. 2016;108(1). Sartore-Bianchi A, Ardini E, Bosotti R, Amatu A, Valtorta E, Somaschini A, et al. Sensitivity to Entrectinib Associated With a Novel LMNA-NTRK1 Gene Fusion in Metastatic Colorectal Cancer. J Natl Cancer Inst. 2016;108(1).
174.
go back to reference Créancier L, Vandenberghe I, Gomes B, Dejean C, Blanchet JC, Meilleroux J, et al. Chromosomal rearrangements involving the NTRK1 gene in colorectal carcinoma. Cancer Lett. 2015;365(1):107–11.PubMedCrossRef Créancier L, Vandenberghe I, Gomes B, Dejean C, Blanchet JC, Meilleroux J, et al. Chromosomal rearrangements involving the NTRK1 gene in colorectal carcinoma. Cancer Lett. 2015;365(1):107–11.PubMedCrossRef
175.
go back to reference Drilon A, Laetsch TW, Kummar S, DuBois SG, Lassen UN, Demetri GD, et al. Efficacy of Larotrectinib in TRK fusion-positive cancers in adults and children. N Engl J Med. 2018;378(8):731–9.PubMedPubMedCentralCrossRef Drilon A, Laetsch TW, Kummar S, DuBois SG, Lassen UN, Demetri GD, et al. Efficacy of Larotrectinib in TRK fusion-positive cancers in adults and children. N Engl J Med. 2018;378(8):731–9.PubMedPubMedCentralCrossRef
176.
go back to reference Russo M, Misale S, Wei G, Siravegna G, Crisafulli G, Lazzari L, et al. Acquired resistance to the TRK inhibitor Entrectinib in colorectal Cancer. Cancer Discov. 2016;6(1):36–44.PubMedCrossRef Russo M, Misale S, Wei G, Siravegna G, Crisafulli G, Lazzari L, et al. Acquired resistance to the TRK inhibitor Entrectinib in colorectal Cancer. Cancer Discov. 2016;6(1):36–44.PubMedCrossRef
177.
go back to reference Maekawa H, Miyoshi H, Yamaura T, Itatani Y, Kawada K, Sakai Y, et al. A Chemosensitivity study of colorectal Cancer using Xenografts of patient-derived tumor-initiating cells. Mol Cancer Ther. 2018;17(10):2187–96.PubMedCrossRef Maekawa H, Miyoshi H, Yamaura T, Itatani Y, Kawada K, Sakai Y, et al. A Chemosensitivity study of colorectal Cancer using Xenografts of patient-derived tumor-initiating cells. Mol Cancer Ther. 2018;17(10):2187–96.PubMedCrossRef
178.
go back to reference Ganesh K, Wu C, O'Rourke KP, Szeglin BC, Zheng Y, Sauvé CG, 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 CG, et al. A rectal cancer organoid platform to study individual responses to chemoradiation. Nat Med. 2019;25(10):1607–14.PubMedPubMedCentralCrossRef
Metadata
Title
Preclinical models as patients’ avatars for precision medicine in colorectal cancer: past and future challenges
Authors
Erika Durinikova
Kristi Buzo
Sabrina Arena
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2021
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-021-01981-z

Other articles of this Issue 1/2021

Journal of Experimental & Clinical Cancer Research 1/2021 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