Skip to main content
Top
Published in: Cancer Cell International 1/2021

Open Access 01-12-2021 | Primary research

Establishment and drug screening of patient-derived extrahepatic biliary tract carcinoma organoids

Authors: Zhiwei Wang, Yinghao Guo, Yun Jin, Xiaoxiao Zhang, Hao Geng, Guangyuan Xie, Dan Ye, Yuanquan Yu, Daren Liu, Donger Zhou, Baizhou Li, Yan Luo, Shuyou Peng, Jiangtao Li

Published in: Cancer Cell International | Issue 1/2021

Login to get access

Abstract

Background

Patient-derived organoids (PDO) have been proposed as a novel in vitro method of drug screening for different types of cancer. However, to date, extrahepatic biliary tract carcinoma (eBTC) PDOs have not yet been fully established.

Methods

We collected six samples of gallbladder carcinoma (GBC) and one sample of extrahepatic cholangiocarcinoma (eCCA) from seven patients to attempt to establish eBTC PDOs for drug screening. We successfully established five GBC and one eCCA PDOs. Histological staining was used to compare structural features between the original tissues and cancer PDOs. Whole exome sequencing (WES) was performed to analyze the genetic profiles of original tissues and cancer PDOs. Drug screening, including gemcitabine, 5-fluorouracil, cisplatin, paclitaxel, infigratinib, and ivosidenib, was measured and verified by clinical effects in certain cases.

Results

Different PDOs exhibited diverse growth rates during in vitro culture. Hematoxylin and eosin staining demonstrated that the structures of most cancer PDOs retained the original structures of adenocarcinoma. Immunohistological and periodic acid-schiff staining revealed that marker expression in cancer PDOs was similar to that of the original specimens. Genetic profiles from the four original specimens, as well as paired cancer PDOs, were analyzed using whole exome sequencing. Three of the four PDOs exhibited a high degree of similarity when compared to the original specimens, except for GBC2 PDO, which only had a concordance of 74% in the proportion of single nucleotide polymorphisms in the coding sequence. In general, gemcitabine was found to be the most efficient drug for eBTC treatment, as it showed moderate or significant inhibitory impact on cancer growth. Results from drug screening were confirmed to a certain extent by three clinical cases.

Conclusions

Our study successfully established a series of eBTC PDOs, which contributed to the field of eBTC PDOs. Additional enhancements should be explored to improve the growth rate of PDOs and to preserve their immune microenvironment.
Appendix
Available only for authorised users
Literature
1.
go back to reference Okano K, Yoshizawa T, Miura T, Ishido K, Kudo D, Kimura N, et al. Impact of the histological phenotype of extrahepatic bile duct carcinoma. Mol Clin Oncol. 2018;8(1):54–60.PubMed Okano K, Yoshizawa T, Miura T, Ishido K, Kudo D, Kimura N, et al. Impact of the histological phenotype of extrahepatic bile duct carcinoma. Mol Clin Oncol. 2018;8(1):54–60.PubMed
2.
go back to reference Zhou Z, Nie SD, Jiang B, Wang J, Lv P. Risk factors for extrahepatic cholangiocarcinoma: a case-control study in China. Eur J Cancer Prev. 2019;28(4):254–7.CrossRef Zhou Z, Nie SD, Jiang B, Wang J, Lv P. Risk factors for extrahepatic cholangiocarcinoma: a case-control study in China. Eur J Cancer Prev. 2019;28(4):254–7.CrossRef
3.
go back to reference Zhou Y, Zhou Q, Lin Q, Chen R, Gong Y, Liu Y, et al. Evaluation of risk factors for extrahepatic cholangiocarcinoma: ABO blood group, hepatitis B virus and their synergism. Int J Cancer. 2013;133(8):1867–75.CrossRef Zhou Y, Zhou Q, Lin Q, Chen R, Gong Y, Liu Y, et al. Evaluation of risk factors for extrahepatic cholangiocarcinoma: ABO blood group, hepatitis B virus and their synergism. Int J Cancer. 2013;133(8):1867–75.CrossRef
4.
go back to reference Sun LJ, Guan A, Xu WY, Liu MX, Yin HH, Jin B, et al. γ-glutamyl transferase-to-platelet ratio based nomogram predicting overall survival of gallbladder carcinoma. World J Gastrointest Oncol. 2020;12(9):1014–30.CrossRef Sun LJ, Guan A, Xu WY, Liu MX, Yin HH, Jin B, et al. γ-glutamyl transferase-to-platelet ratio based nomogram predicting overall survival of gallbladder carcinoma. World J Gastrointest Oncol. 2020;12(9):1014–30.CrossRef
5.
go back to reference Lu K, Feng F, Yang Y, Liu K, Duan J, Liu H, et al. High-throughput screening identified miR-7-2-3p and miR-29c-3p as metastasis suppressors in gallbladder carcinoma. J Gastroenterol. 2020;55(1):51–66.CrossRef Lu K, Feng F, Yang Y, Liu K, Duan J, Liu H, et al. High-throughput screening identified miR-7-2-3p and miR-29c-3p as metastasis suppressors in gallbladder carcinoma. J Gastroenterol. 2020;55(1):51–66.CrossRef
6.
go back to reference Aloia TA, Vauthey JN, Tzeng CD, Herman JM, Koong AC, Krishnan SX, et al. Benefits and limitations of middle bile duct segmental resection for extrahepatic cholangiocarcinoma. Cancer Med. 2020;19(2):147–52. Aloia TA, Vauthey JN, Tzeng CD, Herman JM, Koong AC, Krishnan SX, et al. Benefits and limitations of middle bile duct segmental resection for extrahepatic cholangiocarcinoma. Cancer Med. 2020;19(2):147–52.
7.
go back to reference Tantau AI, Mandrutiu A, Pop A, Zaharie RD, Crisan D, Preda CM, et al. Extrahepatic cholangiocarcinoma: current status of endoscopic approach and additional therapies. World J Hepatol. 2021;13(2):166–86.CrossRef Tantau AI, Mandrutiu A, Pop A, Zaharie RD, Crisan D, Preda CM, et al. Extrahepatic cholangiocarcinoma: current status of endoscopic approach and additional therapies. World J Hepatol. 2021;13(2):166–86.CrossRef
8.
go back to reference Motoyama H, Kubota K, Notake T, Fukushima K, Ikehara T, Hayashi H, et al. Feasibility and efficacy evaluation of metallic biliary stents eluting gemcitabine and cisplatin for extrahepatic cholangiocarcinoma. Ann Surg Oncol. 2020;26(31):4589–606. Motoyama H, Kubota K, Notake T, Fukushima K, Ikehara T, Hayashi H, et al. Feasibility and efficacy evaluation of metallic biliary stents eluting gemcitabine and cisplatin for extrahepatic cholangiocarcinoma. Ann Surg Oncol. 2020;26(31):4589–606.
9.
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;26:4107–19.CrossRef 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;26:4107–19.CrossRef
10.
go back to reference Seppala TT, Zimmerman JW, Sereni E, Plenker D, Suri R, Rozich N, et al. Patient-derived organoid pharmacotyping is a clinically tractable strategy for precision medicine in pancreatic cancer. Ann Surg. 2020;272(3):427–35.CrossRef Seppala TT, Zimmerman JW, Sereni E, Plenker D, Suri R, Rozich N, et al. Patient-derived organoid pharmacotyping is a clinically tractable strategy for precision medicine in pancreatic cancer. Ann Surg. 2020;272(3):427–35.CrossRef
11.
go back to reference Saito Y, Muramatsu T, Kanai Y, Ojima H, Sukeda A, Hiraoka N, et al. Establishment of patient-derived organoids and drug screening for biliary tract carcinoma. Cell Rep. 2019;27(4):1265–76.CrossRef Saito Y, Muramatsu T, Kanai Y, Ojima H, Sukeda A, Hiraoka N, et al. Establishment of patient-derived organoids and drug screening for biliary tract carcinoma. Cell Rep. 2019;27(4):1265–76.CrossRef
12.
go back to reference Engel RM, Chan WH, Nickless D, Hlavca S, Richards E, Kerr G, et al. Patient-derived colorectal cancer organoids upregulate revival stem cell marker genes following chemotherapeutic treatment. J Clin Med. 2020;9(1):128.CrossRef Engel RM, Chan WH, Nickless D, Hlavca S, Richards E, Kerr G, et al. Patient-derived colorectal cancer organoids upregulate revival stem cell marker genes following chemotherapeutic treatment. J Clin Med. 2020;9(1):128.CrossRef
13.
go back to reference Vlachogiannis G, Hedayat S, Vatsiou A, Jamin Y, Fernandez-Mateos J, Khan K, et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science. 2018;359(6378):920–6.CrossRef Vlachogiannis G, Hedayat S, Vatsiou A, Jamin Y, Fernandez-Mateos J, Khan K, et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science. 2018;359(6378):920–6.CrossRef
14.
go back to reference Broutier L, Mastrogiovanni G, Verstegen MM, Francies HE, Gavarro LM, Bradshaw CR, et al. Human primary liver cancer-derived organoid cultures for disease modeling and drug screening. Nat Med. 2017;23(12):1424–35.CrossRef Broutier L, Mastrogiovanni G, Verstegen MM, Francies HE, Gavarro LM, Bradshaw CR, et al. Human primary liver cancer-derived organoid cultures for disease modeling and drug screening. Nat Med. 2017;23(12):1424–35.CrossRef
15.
go back to reference Ganesh K, Wu C, O’Rourke KP, Szeglin BC, Zheng Y, Sauve CG, et al. A rectal cancer organoid platform to study individual responses to chemoradiation. Nat Med. 2019;25:1607–14.CrossRef Ganesh K, Wu C, O’Rourke KP, Szeglin BC, Zheng Y, Sauve CG, et al. A rectal cancer organoid platform to study individual responses to chemoradiation. Nat Med. 2019;25:1607–14.CrossRef
16.
go back to reference Wang Z, Jin Y, Guo Y, Tan Z, Zhang X, Ye D, et al. Conversion therapy of intrahepatic cholangiocarcinoma is associated with improved prognosis and verified by a case of patient-derived organoid. Cancers. 2021;13(5):1179.CrossRef Wang Z, Jin Y, Guo Y, Tan Z, Zhang X, Ye D, et al. Conversion therapy of intrahepatic cholangiocarcinoma is associated with improved prognosis and verified by a case of patient-derived organoid. Cancers. 2021;13(5):1179.CrossRef
17.
go back to reference Soroka CJ, Assis DN, Alrabadi LS, Roberts S, Cusack L, Jaffe AB, et al. Bile-derived organoids from patients with primary sclerosing cholangitis recapitulate their inflammatory immune profile. Hepatology. 2019;70(3):871–82.CrossRef Soroka CJ, Assis DN, Alrabadi LS, Roberts S, Cusack L, Jaffe AB, et al. Bile-derived organoids from patients with primary sclerosing cholangitis recapitulate their inflammatory immune profile. Hepatology. 2019;70(3):871–82.CrossRef
18.
go back to reference Usui T, Sakurai M, Nishikawa S, Umata K, Nemoto Y, Haraguchi T, et al. Establishment of a dog primary prostate cancer organoid using the urine cancer stem cells. Cancer Sci. 2017;108(12):2383–92.CrossRef Usui T, Sakurai M, Nishikawa S, Umata K, Nemoto Y, Haraguchi T, et al. Establishment of a dog primary prostate cancer organoid using the urine cancer stem cells. Cancer Sci. 2017;108(12):2383–92.CrossRef
19.
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.CrossRef 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.CrossRef
20.
go back to reference Phifer CJ, Bergdorf KN, Bechard ME, Vilgelm A, Baregamian N, McDonald OG, et al. Obtaining patient-derived cancer organoid cultures via fine-needle aspiration. STAR Protoc. 2021;2(1):100220.CrossRef Phifer CJ, Bergdorf KN, Bechard ME, Vilgelm A, Baregamian N, McDonald OG, et al. Obtaining patient-derived cancer organoid cultures via fine-needle aspiration. STAR Protoc. 2021;2(1):100220.CrossRef
21.
go back to reference Nuciforo S, Fofana I, Matter MS, Blumer T, Calabrese D, Boldanova T, et al. Organoid models of human liver cancers derived from tumor needle biopsies. Cell Rep. 2018;24(5):1363–76.CrossRef Nuciforo S, Fofana I, Matter MS, Blumer T, Calabrese D, Boldanova T, et al. Organoid models of human liver cancers derived from tumor needle biopsies. Cell Rep. 2018;24(5):1363–76.CrossRef
22.
go back to reference Xu H, Jiao Y, Qin S, Zhao W, Chu Q, Wu K. Organoid technology in disease modelling, drug development, personalized treatment and regeneration medicine. Exp Hematol Oncol. 2018;7:30.CrossRef Xu H, Jiao Y, Qin S, Zhao W, Chu Q, Wu K. Organoid technology in disease modelling, drug development, personalized treatment and regeneration medicine. Exp Hematol Oncol. 2018;7:30.CrossRef
23.
go back to reference Seino T, Kawasaki S, Shimokawa M, Tamagawa H, Toshimitsu K, Fujii M, et al. Human pancreatic tumor organoids reveal loss of stem cell niche factor dependence during disease progression. Cell Stem Cell. 2018;22(3):454–67 e6.CrossRef Seino T, Kawasaki S, Shimokawa M, Tamagawa H, Toshimitsu K, Fujii M, et al. Human pancreatic tumor organoids reveal loss of stem cell niche factor dependence during disease progression. Cell Stem Cell. 2018;22(3):454–67 e6.CrossRef
24.
go back to reference Zhu J, Zhao Y, Liu M, Gonzalez-Rivas D, Xu X, Cai W, et al. Developing a new qPCR-based system for screening mutation. Small. 2019;15(9):e1805285.CrossRef Zhu J, Zhao Y, Liu M, Gonzalez-Rivas D, Xu X, Cai W, et al. Developing a new qPCR-based system for screening mutation. Small. 2019;15(9):e1805285.CrossRef
25.
go back to reference Stevenson M, Pagnamenta AT, Reichart S, Philpott C, Lines KE, OxClinWgs, et al. Whole genome sequence analysis identifies a PAX2 mutation to establish a correct diagnosis for a syndromic form of hyperuricemia. Am J Med Genet A. 2020;182(11):2521–8.CrossRef Stevenson M, Pagnamenta AT, Reichart S, Philpott C, Lines KE, OxClinWgs, et al. Whole genome sequence analysis identifies a PAX2 mutation to establish a correct diagnosis for a syndromic form of hyperuricemia. Am J Med Genet A. 2020;182(11):2521–8.CrossRef
26.
go back to reference Lee KH, Lee TH, Choi MK, Kwon IS, Bae GE, Yeo MK. Identification of a clinical cutoff value for multiplex KRAS(G12/G13) mutation detection in colorectal adenocarcinoma patients using digital droplet PCR, and comparison with sanger sequencing and PNA clamping assay. J Clin Med. 2020;9(7):2283.CrossRef Lee KH, Lee TH, Choi MK, Kwon IS, Bae GE, Yeo MK. Identification of a clinical cutoff value for multiplex KRAS(G12/G13) mutation detection in colorectal adenocarcinoma patients using digital droplet PCR, and comparison with sanger sequencing and PNA clamping assay. J Clin Med. 2020;9(7):2283.CrossRef
27.
go back to reference Zhao H, Yan C, Hu Y, Mu L, Liu S, Huang K, et al. Differentiated cancer cell-originated lactate promotes the self-renewal of cancer stem cells in patient-derived colorectal cancer organoids. Cancer Lett. 2020;493:236–44.CrossRef Zhao H, Yan C, Hu Y, Mu L, Liu S, Huang K, et al. Differentiated cancer cell-originated lactate promotes the self-renewal of cancer stem cells in patient-derived colorectal cancer organoids. Cancer Lett. 2020;493:236–44.CrossRef
28.
go back to reference Jacob F, Salinas RD, Zhang DY, Nguyen PTT, Schnoll JG, Wong SZH, et al. A patient-derived glioblastoma organoid model and biobank recapitulates inter- and intra-tumoral heterogeneity. Cell. 2020;180(1):188-204 e22.CrossRef Jacob F, Salinas RD, Zhang DY, Nguyen PTT, Schnoll JG, Wong SZH, et al. A patient-derived glioblastoma organoid model and biobank recapitulates inter- and intra-tumoral heterogeneity. Cell. 2020;180(1):188-204 e22.CrossRef
29.
go back to reference de Witte CJ, Espejo Valle-Inclan J, Hami N, Lohmussaar K, Kopper O, Vreuls CPH, et al. Patient-derived ovarian cancer organoids mimic clinical response and exhibit heterogeneous inter- and intrapatient drug responses. Cell Rep. 2020;31(11):107762.CrossRef de Witte CJ, Espejo Valle-Inclan J, Hami N, Lohmussaar K, Kopper O, Vreuls CPH, et al. Patient-derived ovarian cancer organoids mimic clinical response and exhibit heterogeneous inter- and intrapatient drug responses. Cell Rep. 2020;31(11):107762.CrossRef
30.
go back to reference Frappart PO, Walter K, Gout J, Beutel AK, Morawe M, Arnold F, et al. Pancreatic cancer-derived organoids—a disease modeling tool to predict drug response. United Eur Gastroenterol J. 2020;8:594–606.CrossRef Frappart PO, Walter K, Gout J, Beutel AK, Morawe M, Arnold F, et al. Pancreatic cancer-derived organoids—a disease modeling tool to predict drug response. United Eur Gastroenterol J. 2020;8:594–606.CrossRef
31.
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;25:5376–87.CrossRef 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;25:5376–87.CrossRef
32.
go back to reference Tuveson D, Clevers H. Cancer modeling meets human organoid technology. Science. 2019;364(6444):952–5.CrossRef Tuveson D, Clevers H. Cancer modeling meets human organoid technology. Science. 2019;364(6444):952–5.CrossRef
33.
go back to reference Lee SH, Hu W, Matulay JT, Silva MV, Owczarek TB, Kim K, et al. Tumor evolution and drug response in patient-derived organoid models of bladder cancer. Cell. 2018;173(2):515–28 e17.CrossRef Lee SH, Hu W, Matulay JT, Silva MV, Owczarek TB, Kim K, et al. Tumor evolution and drug response in patient-derived organoid models of bladder cancer. Cell. 2018;173(2):515–28 e17.CrossRef
34.
go back to reference Pape J, Magdeldin T, Stamati K, Nyga A, Loizidou M, Emberton M, et al. Cancer-associated fibroblasts mediate cancer progression and remodel the tumouroid stroma. Br J Cancer. 2020;123:1178–90.CrossRef Pape J, Magdeldin T, Stamati K, Nyga A, Loizidou M, Emberton M, et al. Cancer-associated fibroblasts mediate cancer progression and remodel the tumouroid stroma. Br J Cancer. 2020;123:1178–90.CrossRef
35.
go back to reference Nakamura H, Sugano M, Miyashita T, Hashimoto H, Ochiai A, Suzuki K, et al. Organoid culture containing cancer cells and stromal cells reveals that podoplanin-positive cancer-associated fibroblasts enhance proliferation of lung cancer cells. Lung Cancer. 2019;134:100–7.CrossRef Nakamura H, Sugano M, Miyashita T, Hashimoto H, Ochiai A, Suzuki K, et al. Organoid culture containing cancer cells and stromal cells reveals that podoplanin-positive cancer-associated fibroblasts enhance proliferation of lung cancer cells. Lung Cancer. 2019;134:100–7.CrossRef
36.
go back to reference Liu J, Li P, Wang L, Li M, Ge Z, Noordam L, et al. Cancer-associated fibroblasts provide a stromal niche for liver cancer organoids that confers trophic effects and therapy resistance. Cell Mol Gastroenterol Hepatol. 2020;11:407–31.CrossRef Liu J, Li P, Wang L, Li M, Ge Z, Noordam L, et al. Cancer-associated fibroblasts provide a stromal niche for liver cancer organoids that confers trophic effects and therapy resistance. Cell Mol Gastroenterol Hepatol. 2020;11:407–31.CrossRef
37.
go back to reference Montal R, Sia D, Montironi C, Leow WQ, Esteban-Fabro R, Pinyol R, et al. Molecular classification and therapeutic targets in extrahepatic cholangiocarcinoma. J Hepatol. 2020;73:315–27.CrossRef Montal R, Sia D, Montironi C, Leow WQ, Esteban-Fabro R, Pinyol R, et al. Molecular classification and therapeutic targets in extrahepatic cholangiocarcinoma. J Hepatol. 2020;73:315–27.CrossRef
38.
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.CrossRef 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.CrossRef
39.
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.CrossRef 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.CrossRef
Metadata
Title
Establishment and drug screening of patient-derived extrahepatic biliary tract carcinoma organoids
Authors
Zhiwei Wang
Yinghao Guo
Yun Jin
Xiaoxiao Zhang
Hao Geng
Guangyuan Xie
Dan Ye
Yuanquan Yu
Daren Liu
Donger Zhou
Baizhou Li
Yan Luo
Shuyou Peng
Jiangtao Li
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Cancer Cell International / Issue 1/2021
Electronic ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-021-02219-w

Other articles of this Issue 1/2021

Cancer Cell International 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