Skip to main content
Erschienen in: Cellular Oncology 5/2022

29.08.2022 | Original Article

Heterogeneity, inherent and acquired drug resistance in patient-derived organoid models of primary liver cancer

verfasst von: Linfeng Xian, Pei Zhao, Xi Chen, Zhimin Wei, Hongxiang Ji, Jun Zhao, Wenbin Liu, Zishuai Li, Donghong Liu, Xue Han, Youwen Qian, Hui Dong, Xiong Zhou, Junyan Fan, Xiaoqiong Zhu, Jianhua Yin, Xiaojie Tan, Dongming Jiang, Hongping Yu, Guangwen Cao

Erschienen in: Cellular Oncology | Ausgabe 5/2022

Einloggen, um Zugang zu erhalten

Abstract

Purpose

We aimed to elucidate the applicability of tumor organoids for inherent drug resistance of primary liver cancer (PLC) and mechanisms of acquired drug resistance.

Methods

PLC tissues were used to establish organoids, organoid-derived xenograft (ODX) and patient-derived xenograft (PDX) models. Acquired drug resistance was induced in hepatocellular carcinoma (HCC) organoids. Gene expression profiling was performed by RNA-sequencing.

Results

Fifty-two organoids were established from 153 PLC patients. Compared with establishing PDX models, establishing organoids of HCC showed a trend toward a higher success rate (29.0% vs. 23.7%) and took less time (13.0 ± 4.7 vs. 25.1 ± 5.4 days, p = 2.28 × 10−13). Larger tumors, vascular invasion, higher serum AFP levels, advanced stages and upregulation of stemness- and proliferation-related genes were significantly associated with the successful establishment of HCC organoids and PDX. Organoids and ODX recapitulated PLC histopathological features, but were enriched in more aggressive cell types. PLC organoids were mostly resistant to lenvatinib in vitro but sensitive to lenvatinib in ODX models. Stemness– and epithelial–mesenchymal transition (EMT)–related gene sets were found to be upregulated, whereas liver development– and liver specific molecule–related gene sets were downregulated in acquired sorafenib-resistant organoids. Targeting the mTOR signaling pathway was effective in treating acquired sorafenib-resistant HCC organoids, possibly via inducing phosphorylated S6 kinase. Genes upregulated in acquired sorafenib-resistant HCC organoids were associated with an unfavorable prognosis.

Conclusions

HCC organoids perform better than PDX for drug screening. Acquired sorafenib resistance in organoids promotes HCC aggressiveness via facilitating stemness, retro-differentiation and EMT. Phosphorylated S6 kinase may be predictive for drug resistance in HCC.
Anhänge
Nur mit Berechtigung zugänglich
Literatur
1.
Zurück zum Zitat H. Sung, J. Ferlay, R.L. Siegel, M. Laversanne, I. Soerjomataram, A. Jemal, F. Bray, Global Cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 71, 209–249 (2021)PubMedCrossRef H. Sung, J. Ferlay, R.L. Siegel, M. Laversanne, I. Soerjomataram, A. Jemal, F. Bray, Global Cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 71, 209–249 (2021)PubMedCrossRef
2.
Zurück zum Zitat D. Jiang, L. Zhang, W. Liu, Y. Ding, J. Yin, R. Ren, Q. Li, Y. Chen, J. Shen, X. Tan, H. Zhang, G. Cao, Trends in cancer mortality in China from 2004 to 2018: A nationwide longitudinal study. Cancer Commun. (Lond.) 41, 1024–1036 (2021)CrossRef D. Jiang, L. Zhang, W. Liu, Y. Ding, J. Yin, R. Ren, Q. Li, Y. Chen, J. Shen, X. Tan, H. Zhang, G. Cao, Trends in cancer mortality in China from 2004 to 2018: A nationwide longitudinal study. Cancer Commun. (Lond.) 41, 1024–1036 (2021)CrossRef
3.
Zurück zum Zitat F. Yang, L. Ma, Y. Yang, W. Liu, J. Zhao, X. Chen, M. Wang, H. Zhang, S. Cheng, F. Shen, H. Wang, W. Zhou, G. Cao, Contribution of hepatitis B virus infection to the aggressiveness of primary liver cancer: A clinical epidemiological study in eastern China. Front. Oncol. 9, 370 (2019)PubMedPubMedCentralCrossRef F. Yang, L. Ma, Y. Yang, W. Liu, J. Zhao, X. Chen, M. Wang, H. Zhang, S. Cheng, F. Shen, H. Wang, W. Zhou, G. Cao, Contribution of hepatitis B virus infection to the aggressiveness of primary liver cancer: A clinical epidemiological study in eastern China. Front. Oncol. 9, 370 (2019)PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat G. Marasco, F. Ravaioli, A. Vestito, B. Rossini, E. Dajti, L. Colecchia, K. Gjini, M. Renzulli, R. Golfieri, D. Festi, A. Colecchia, Predictive factors for hepatocellular carcinoma recurrence after curative treatments. Hepatoma Res. 6, 33 (2020) G. Marasco, F. Ravaioli, A. Vestito, B. Rossini, E. Dajti, L. Colecchia, K. Gjini, M. Renzulli, R. Golfieri, D. Festi, A. Colecchia, Predictive factors for hepatocellular carcinoma recurrence after curative treatments. Hepatoma Res. 6, 33 (2020)
5.
Zurück zum Zitat A. Rammohan, M. Rela, Risk factors and management of post-liver transplant recurrence of hepatocellular carcinoma. Hepatoma Res. 7, 49 (2021) A. Rammohan, M. Rela, Risk factors and management of post-liver transplant recurrence of hepatocellular carcinoma. Hepatoma Res. 7, 49 (2021)
6.
Zurück zum Zitat Y. Lai, X. Wei, S. Lin, L. Qin, L. Cheng, P. Li, Current status and perspectives of patient-derived xenograft models in cancer research. J. Hematol. Oncol. 10, 106 (2017)PubMedPubMedCentralCrossRef Y. Lai, X. Wei, S. Lin, L. Qin, L. Cheng, P. Li, Current status and perspectives of patient-derived xenograft models in cancer research. J. Hematol. Oncol. 10, 106 (2017)PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat M.R. Kuracha, P. Thomas, B.W. Loggie, V. Govindarajan, Patient-derived xenograft mouse models of pseudomyxoma peritonei recapitulate the human inflammatory tumor microenvironment. Cancer Med. 5, 711–719 (2016)PubMedPubMedCentralCrossRef M.R. Kuracha, P. Thomas, B.W. Loggie, V. Govindarajan, Patient-derived xenograft mouse models of pseudomyxoma peritonei recapitulate the human inflammatory tumor microenvironment. Cancer Med. 5, 711–719 (2016)PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat G.J. Yoshida, Applications of patient-derived tumor xenograft models and tumor organoids. J. Hematol. Oncol. 7, 13:4 (2020) G.J. Yoshida, Applications of patient-derived tumor xenograft models and tumor organoids. J. Hematol. Oncol. 7, 13:4 (2020)
9.
Zurück zum Zitat S. Shi, M.M.A. Verstegen, H.P. Roest, A.I. Ardisasmita, W. Cao, F.J.M. Roos, P.E. de Ruiter, M. Niemeijer, Q. Pan, J.N.M. IJzermans, L.J.W. van der Laan, Recapitulating cholangiopathy-associated necroptotic cell death in vitro using human cholangiocyte organoids. Cell Mol. Gastroenterol. Hepatol. 13, 541–564 (2021)PubMedPubMedCentralCrossRef S. Shi, M.M.A. Verstegen, H.P. Roest, A.I. Ardisasmita, W. Cao, F.J.M. Roos, P.E. de Ruiter, M. Niemeijer, Q. Pan, J.N.M. IJzermans, L.J.W. van der Laan, Recapitulating cholangiopathy-associated necroptotic cell death in vitro using human cholangiocyte organoids. Cell Mol. Gastroenterol. Hepatol. 13, 541–564 (2021)PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat L. Broutier, G. Mastrogiovanni, M.M. Verstegen, H.E. Francies, L.M. Gavarró, C.R. Bradshaw, G.E. Allen, R. Arnes-Benito, O. Sidorova, M.P. Gaspersz, N. Georgakopoulos, B.K. Koo, S. Dietmann, S.E. Davies, R.K. Praseedom, R. Lieshout, J.N.M. Ijzermans, S.J. Wigmore, K. Saeb-Parsy, et al., Human primary liver cancer-derived organoid cultures for disease modeling and drug screening. Nat. Med. 23, 1424–1435 (2017)PubMedPubMedCentralCrossRef L. Broutier, G. Mastrogiovanni, M.M. Verstegen, H.E. Francies, L.M. Gavarró, C.R. Bradshaw, G.E. Allen, R. Arnes-Benito, O. Sidorova, M.P. Gaspersz, N. Georgakopoulos, B.K. Koo, S. Dietmann, S.E. Davies, R.K. Praseedom, R. Lieshout, J.N.M. Ijzermans, S.J. Wigmore, K. Saeb-Parsy, et al., Human primary liver cancer-derived organoid cultures for disease modeling and drug screening. Nat. Med. 23, 1424–1435 (2017)PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat S. Nuciforo, I. Fofana, M.S. Matter, T. Blumer, D. Calabrese, T. Boldanova, S. Piscuoglio, S. Wieland, F. Ringnalda, G. Schwank, L.M. Terracciano, C.K.Y. Ng, M.H. Heim, Organoid models of human liver cancers derived from tumor needle biopsies. Cell Rep. 24, 1363–1376 (2018)PubMedPubMedCentralCrossRef S. Nuciforo, I. Fofana, M.S. Matter, T. Blumer, D. Calabrese, T. Boldanova, S. Piscuoglio, S. Wieland, F. Ringnalda, G. Schwank, L.M. Terracciano, C.K.Y. Ng, M.H. Heim, Organoid models of human liver cancers derived from tumor needle biopsies. Cell Rep. 24, 1363–1376 (2018)PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat L. Li, H. Knutsdottir, K. Hui, M.J. Weiss, J. He, B. Philosophe, A.M. Cameron, C.L. Wolfgang, T.M. Pawlik, G. Ghiaur, A.J. Ewald, E. Mezey, J.S. Bader, F.M. Selaru, Human primary liver cancer organoids reveal intratumor and interpatient drug response heterogeneity. JCI Insight 4, e121490 (2019)PubMedCentralCrossRef L. Li, H. Knutsdottir, K. Hui, M.J. Weiss, J. He, B. Philosophe, A.M. Cameron, C.L. Wolfgang, T.M. Pawlik, G. Ghiaur, A.J. Ewald, E. Mezey, J.S. Bader, F.M. Selaru, Human primary liver cancer organoids reveal intratumor and interpatient drug response heterogeneity. JCI Insight 4, e121490 (2019)PubMedCentralCrossRef
13.
Zurück zum Zitat S. Wang, Y. Wang, X. Xun, C. Zhang, X. Xiang, Q. Cheng, S. Hu, Z. Li, J. Zhu, Hedgehog signaling promotes sorafenib resistance in hepatocellular carcinoma patient-derived organoids. J. Exp. Clin. Cancer Res. 39, 22 (2020)PubMedPubMedCentralCrossRef S. Wang, Y. Wang, X. Xun, C. Zhang, X. Xiang, Q. Cheng, S. Hu, Z. Li, J. Zhu, Hedgehog signaling promotes sorafenib resistance in hepatocellular carcinoma patient-derived organoids. J. Exp. Clin. Cancer Res. 39, 22 (2020)PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Y. Zhao, Z.X. Li, Y.J. Zhu, J. Fu, X.F. Zhao, Y.N. Zhang, S. Wang, J.M. Wu, K.T. Wang, S.C.J. Wu, S.Y. Shen, X. Wu, H.Y. Wang, D. Gao, L. Chen, Single cell transcriptome analysis uncovers intratumoral heterogeneity and underlying mechanisms for drug resistance in hepatobiliary tumor organoids. Adv. Sci, (Weinh.) 8, e2003897 (2021) Y. Zhao, Z.X. Li, Y.J. Zhu, J. Fu, X.F. Zhao, Y.N. Zhang, S. Wang, J.M. Wu, K.T. Wang, S.C.J. Wu, S.Y. Shen, X. Wu, H.Y. Wang, D. Gao, L. Chen, Single cell transcriptome analysis uncovers intratumoral heterogeneity and underlying mechanisms for drug resistance in hepatobiliary tumor organoids. Adv. Sci, (Weinh.) 8, e2003897 (2021)
15.
Zurück zum Zitat C.O.N. Leung, M. Tong, K.P.S. Chung, L. Zhou, N. Che, K.H. Tang, J. Ding, E.Y.T. Lau, I.O.L. Ng, S. Ma, T.K.W. Lee, Overriding adaptive resistance to sorafenib through combination therapy with Src homology 2 domain-containing phosphatase 2 blockade in hepatocellular carcinoma. Hepatology 72, 155–168 (2020)PubMedCrossRef C.O.N. Leung, M. Tong, K.P.S. Chung, L. Zhou, N. Che, K.H. Tang, J. Ding, E.Y.T. Lau, I.O.L. Ng, S. Ma, T.K.W. Lee, Overriding adaptive resistance to sorafenib through combination therapy with Src homology 2 domain-containing phosphatase 2 blockade in hepatocellular carcinoma. Hepatology 72, 155–168 (2020)PubMedCrossRef
16.
Zurück zum Zitat S.R. Veiga, X. Ge, C.A. Mercer, M.I. Hernández-Álvarez, H.E. Thomas, J. Hernandez-Losa, R.Y. Cajal, S, Zorzano A, Thomas G, Kozma SC., Phenformin-induced mitochondrial dysfunction sensitizes hepatocellular carcinoma for dual inhibition of mTOR. Clin. Cancer Res. 24, 3767–3780 (2018)PubMedCrossRef S.R. Veiga, X. Ge, C.A. Mercer, M.I. Hernández-Álvarez, H.E. Thomas, J. Hernandez-Losa, R.Y. Cajal, S, Zorzano A, Thomas G, Kozma SC., Phenformin-induced mitochondrial dysfunction sensitizes hepatocellular carcinoma for dual inhibition of mTOR. Clin. Cancer Res. 24, 3767–3780 (2018)PubMedCrossRef
17.
Zurück zum Zitat W. Chang, X. Gao, Y. Han, Y. Du, Q. Liu, L. Wang, X. Tan, Q. Zhang, Y. Liu, Y. Zhu, Y. Yu, X. Fan, H. Zhang, W. Zhou, J. Wang, C. Fu, G. Cao, Gene expression profiling-derived immunohistochemistry signature with high prognostic value in colorectal carcinoma. Gut 63, 1457–1467 (2014)PubMedCrossRef W. Chang, X. Gao, Y. Han, Y. Du, Q. Liu, L. Wang, X. Tan, Q. Zhang, Y. Liu, Y. Zhu, Y. Yu, X. Fan, H. Zhang, W. Zhou, J. Wang, C. Fu, G. Cao, Gene expression profiling-derived immunohistochemistry signature with high prognostic value in colorectal carcinoma. Gut 63, 1457–1467 (2014)PubMedCrossRef
18.
Zurück zum Zitat M.A. Rodríguez-Hernández, R. Chapresto-Garzón, M. Cadenas, E. Navarro-Villarán, M. Negrete, M.A. Gómez-Bravo, V.M. Victor, F.J. Padillo, J. Muntané, Differential effectiveness of tyrosine kinase inhibitors in 2D/3D culture according to cell differentiation, p53 status and mitochondrial respiration in liver cancer cells. Cell Death Dis. 11, 339 (2020)PubMedPubMedCentralCrossRef M.A. Rodríguez-Hernández, R. Chapresto-Garzón, M. Cadenas, E. Navarro-Villarán, M. Negrete, M.A. Gómez-Bravo, V.M. Victor, F.J. Padillo, J. Muntané, Differential effectiveness of tyrosine kinase inhibitors in 2D/3D culture according to cell differentiation, p53 status and mitochondrial respiration in liver cancer cells. Cell Death Dis. 11, 339 (2020)PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat M. Pertea, G.M. Pertea, C.M. Antonescu, T.C. Chang, J.T. Mendell, S.L. Salzberg, StringTie enables improved reconstruction of a transcriptome from RNA-seq reads. Nat. Biotechnol. 33, 290–295 (2015)PubMedPubMedCentralCrossRef M. Pertea, G.M. Pertea, C.M. Antonescu, T.C. Chang, J.T. Mendell, S.L. Salzberg, StringTie enables improved reconstruction of a transcriptome from RNA-seq reads. Nat. Biotechnol. 33, 290–295 (2015)PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat C. Trapnell, A. Roberts, L. Goff, G. Pertea, D. Kim, D.R. Kelley, H. Pimentel, S.L. Salzberg, J.L. Rinn, L. Pachter, Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and cufflinks. Nat. Protoc. 7, 562–578 (2012)PubMedPubMedCentralCrossRef C. Trapnell, A. Roberts, L. Goff, G. Pertea, D. Kim, D.R. Kelley, H. Pimentel, S.L. Salzberg, J.L. Rinn, L. Pachter, Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and cufflinks. Nat. Protoc. 7, 562–578 (2012)PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat A. Roberts, L. Pachter, Streaming fragment assignment for real-time analysis of sequencing experiments. Nat. Methods 10, 71–73 (2013)PubMedCrossRef A. Roberts, L. Pachter, Streaming fragment assignment for real-time analysis of sequencing experiments. Nat. Methods 10, 71–73 (2013)PubMedCrossRef
26.
Zurück zum Zitat D.J. McCarthy, Y. Chen, G.K. Smyth, Differential expression analysis of multifactor RNA-Seq experiments with respect to biological variation. Nucleic Acids Res. 40, 4288–4297 (2012)PubMedPubMedCentralCrossRef D.J. McCarthy, Y. Chen, G.K. Smyth, Differential expression analysis of multifactor RNA-Seq experiments with respect to biological variation. Nucleic Acids Res. 40, 4288–4297 (2012)PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat A. Subramanian, P. Tamayo, V.K. Mootha, S. Mukherjee, B.L. Ebert, M.A. Gillette, A. Paulovich, S.L. Pomeroy, T.R. Golub, E.S. Lander, J.P. Mesirov, Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. U. S. A. 102, 15545–15550 (2005)PubMedPubMedCentralCrossRef A. Subramanian, P. Tamayo, V.K. Mootha, S. Mukherjee, B.L. Ebert, M.A. Gillette, A. Paulovich, S.L. Pomeroy, T.R. Golub, E.S. Lander, J.P. Mesirov, Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. U. S. A. 102, 15545–15550 (2005)PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat F.M. Ippen, C.A. Alvarez-Breckenridge, B.M. Kuter, A.L. Fink, I.V. Bihun, M. Lastrapes, T. Penson, S.P. Schmidt, G.R. Wojtkiewicz, J. Ning, M. Subramanian, A. Giobbie-Hurder, M. Martinez-Lage, S.L. Carter, D.P. Cahill, H. Wakimoto, P.K. Brastianos, The dual PI3K/mTOR pathway inhibitor GDC-0084 achieves antitumor activity in PIK3CA-mutant breast cancer brain metastases. Clin. Cancer Res. 25, 3374–3383 (2019)PubMedPubMedCentralCrossRef F.M. Ippen, C.A. Alvarez-Breckenridge, B.M. Kuter, A.L. Fink, I.V. Bihun, M. Lastrapes, T. Penson, S.P. Schmidt, G.R. Wojtkiewicz, J. Ning, M. Subramanian, A. Giobbie-Hurder, M. Martinez-Lage, S.L. Carter, D.P. Cahill, H. Wakimoto, P.K. Brastianos, The dual PI3K/mTOR pathway inhibitor GDC-0084 achieves antitumor activity in PIK3CA-mutant breast cancer brain metastases. Clin. Cancer Res. 25, 3374–3383 (2019)PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat N.Y. Kim, S. Lee, J. Yu, N. Kim, S.S. Won, H. Park, W.D. Heo, Optogenetic control of mRNA localization and translation in live cells. Nat. Cell Biol. 22, 341–352 (2020)PubMedCrossRef N.Y. Kim, S. Lee, J. Yu, N. Kim, S.S. Won, H. Park, W.D. Heo, Optogenetic control of mRNA localization and translation in live cells. Nat. Cell Biol. 22, 341–352 (2020)PubMedCrossRef
30.
Zurück zum Zitat J. Yin, N. Li, Y. Han, J. Xue, Y. Deng, J. Shi, W. Guo, H. Zhang, H. Wang, S. Cheng, G. Cao, Effect of antiviral treatment with nucleotide/nucleoside analogs on postoperative prognosis of hepatitis B virus-related hepatocellular carcinoma: A two-stage longitudinal clinical study. J. Clin. Oncol. 31, 3647–3655 (2013)PubMedCrossRef J. Yin, N. Li, Y. Han, J. Xue, Y. Deng, J. Shi, W. Guo, H. Zhang, H. Wang, S. Cheng, G. Cao, Effect of antiviral treatment with nucleotide/nucleoside analogs on postoperative prognosis of hepatitis B virus-related hepatocellular carcinoma: A two-stage longitudinal clinical study. J. Clin. Oncol. 31, 3647–3655 (2013)PubMedCrossRef
31.
Zurück zum Zitat T. Kawai, K. Yasuchika, T. Ishii, H. Katayama, E.Y. Yoshitoshi, S. Ogiso, S. Kita, K. Yasuda, K. Fukumitsu, M. Mizumoto, E. Hatano, S. Uemoto, Keratin 19, a cancer stem cell marker in human epatocellular carcinoma. Clin. Cancer Res. 21, 3081–3091 (2015)PubMedCrossRef T. Kawai, K. Yasuchika, T. Ishii, H. Katayama, E.Y. Yoshitoshi, S. Ogiso, S. Kita, K. Yasuda, K. Fukumitsu, M. Mizumoto, E. Hatano, S. Uemoto, Keratin 19, a cancer stem cell marker in human epatocellular carcinoma. Clin. Cancer Res. 21, 3081–3091 (2015)PubMedCrossRef
32.
Zurück zum Zitat G. Chen, Y. Wang, X. Zhao, X.Z. Xie, J.G. Zhao, T. Deng, Z.Y. Chen, H.B. Chen, Y.F. Tong, Z. Yang, X.W. Ding, P.Y. Guo, H.T. Yu, L.J. Wu, S.N. Zhang, Q.D. Zhu, J.J. Li, Y.F. Shan, F.X. Yu, et al., A positive feedback loop between Periostin and TGFβ1 induces and maintains the stemness of hepatocellular carcinoma cells via AP-2α activation. J. Exp. Clin. Cancer Res. 40, 218 (2021)PubMedPubMedCentralCrossRef G. Chen, Y. Wang, X. Zhao, X.Z. Xie, J.G. Zhao, T. Deng, Z.Y. Chen, H.B. Chen, Y.F. Tong, Z. Yang, X.W. Ding, P.Y. Guo, H.T. Yu, L.J. Wu, S.N. Zhang, Q.D. Zhu, J.J. Li, Y.F. Shan, F.X. Yu, et al., A positive feedback loop between Periostin and TGFβ1 induces and maintains the stemness of hepatocellular carcinoma cells via AP-2α activation. J. Exp. Clin. Cancer Res. 40, 218 (2021)PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Y. Pan, M. Han, X. Zhang, Y. He, C. Yuan, Y. Xiong, X. Li, C. Zeng, K. Lu, H. Zhu, X. Lu, Q. Liu, H. Liang, Z. Liao, Z. Ding, Z. Zhang, X. Chen, W. Zhang, B. Zhang, Discoidin domain receptor 1 promotes hepatocellular carcinoma progression through modulation of SLC1A5 and the mTORC1 signaling pathway. Cell. Oncol. 45, 163–178 (2022)CrossRef Y. Pan, M. Han, X. Zhang, Y. He, C. Yuan, Y. Xiong, X. Li, C. Zeng, K. Lu, H. Zhu, X. Lu, Q. Liu, H. Liang, Z. Liao, Z. Ding, Z. Zhang, X. Chen, W. Zhang, B. Zhang, Discoidin domain receptor 1 promotes hepatocellular carcinoma progression through modulation of SLC1A5 and the mTORC1 signaling pathway. Cell. Oncol. 45, 163–178 (2022)CrossRef
34.
Zurück zum Zitat D.J. Marchant, C.L. Bellac, T.J. Moraes, S.J. Wadsworth, A. Dufour, G.S. Butler, L.M. Bilawchuk, R.G. Hendry, A.G. Robertson, C.T. Cheung, J. Ng, L. Ang, Z. Luo, K. Heilbron, M.J. Norris, W. Duan, T. Bucyk, A. Karpov, L. Devel, et al., A new transcriptional role for matrix metalloproteinase-12 in antiviral immunity. Nat. Med. 20, 493–502 (2014)PubMedCrossRef D.J. Marchant, C.L. Bellac, T.J. Moraes, S.J. Wadsworth, A. Dufour, G.S. Butler, L.M. Bilawchuk, R.G. Hendry, A.G. Robertson, C.T. Cheung, J. Ng, L. Ang, Z. Luo, K. Heilbron, M.J. Norris, W. Duan, T. Bucyk, A. Karpov, L. Devel, et al., A new transcriptional role for matrix metalloproteinase-12 in antiviral immunity. Nat. Med. 20, 493–502 (2014)PubMedCrossRef
35.
Zurück zum Zitat Q. Wu, W. Zhou, S. Yin, Y. Zhou, T. Chen, J. Qian, R. Su, L. Hong, H. Lu, F. Zhang, H. Xie, L. Zhou, S. Zheng, Blocking triggering receptor expressed on myeloid cells-1-positive tumor-associated macrophages induced by hypoxia reverses immunosuppression and anti-programmed cell death ligand 1 resistance in liver cancer. Hepatology 70, 198–214 (2019)PubMedCrossRef Q. Wu, W. Zhou, S. Yin, Y. Zhou, T. Chen, J. Qian, R. Su, L. Hong, H. Lu, F. Zhang, H. Xie, L. Zhou, S. Zheng, Blocking triggering receptor expressed on myeloid cells-1-positive tumor-associated macrophages induced by hypoxia reverses immunosuppression and anti-programmed cell death ligand 1 resistance in liver cancer. Hepatology 70, 198–214 (2019)PubMedCrossRef
36.
Zurück zum Zitat J. Shen, T. Liu, J. Lv, S. Xu, Identification of an immune-related prognostic gene CLEC5A based on immune microenvironment and risk modeling of ovarian cancer. Front Cell Dev. Biol. 9, 746932 (2021)PubMedPubMedCentralCrossRef J. Shen, T. Liu, J. Lv, S. Xu, Identification of an immune-related prognostic gene CLEC5A based on immune microenvironment and risk modeling of ovarian cancer. Front Cell Dev. Biol. 9, 746932 (2021)PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Y.H. Kuo, Y.H. Yen, Y.Y. Chen, K.M. Kee, C.H. Hung, S.N. Lu, T.H. Hu, C.H. Chen, J.H. Wang, Nivolumab versus regorafenib in patients with hepatocellular carcinoma after sorafenib failure. Front. Oncol. 11, 683341 (2021)PubMedPubMedCentralCrossRef Y.H. Kuo, Y.H. Yen, Y.Y. Chen, K.M. Kee, C.H. Hung, S.N. Lu, T.H. Hu, C.H. Chen, J.H. Wang, Nivolumab versus regorafenib in patients with hepatocellular carcinoma after sorafenib failure. Front. Oncol. 11, 683341 (2021)PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat M. Zhu, L. Li, T. Lu, H. Yoo, J. Zhu, P. Gopal, S.C. Wang, M.R. Porembka, N.E. Rich, S. Kagan, M. Odewole, V. Renteria, A.K. Waljee, T. Wang, A.G. Singal, A.C. Yopp, H. Zhu, Uncovering biological factors that regulate hepatocellular carcinoma growth using patient-derived xenograft assays. Hepatology 72, 1085–1101 (2020)PubMedCrossRef M. Zhu, L. Li, T. Lu, H. Yoo, J. Zhu, P. Gopal, S.C. Wang, M.R. Porembka, N.E. Rich, S. Kagan, M. Odewole, V. Renteria, A.K. Waljee, T. Wang, A.G. Singal, A.C. Yopp, H. Zhu, Uncovering biological factors that regulate hepatocellular carcinoma growth using patient-derived xenograft assays. Hepatology 72, 1085–1101 (2020)PubMedCrossRef
39.
Zurück zum Zitat J.L. Carroll, L.L. Nielsen, S.B. Pruett, J.M. Mathis, The role of natural killer cells in adenovirus-mediated p53 gene therapy. Mol. Cancer Ther. 1, 49–60 (2001)PubMed J.L. Carroll, L.L. Nielsen, S.B. Pruett, J.M. Mathis, The role of natural killer cells in adenovirus-mediated p53 gene therapy. Mol. Cancer Ther. 1, 49–60 (2001)PubMed
40.
Zurück zum Zitat Q. Zhang, H. Liu, H. Wang, M. Lu, Y. Miao, J. Ding, H. Li, X. Gao, S. Sun, J. Zheng, Lenvatinib promotes antitumor immunity by enhancing the tumor infiltration and activation of NK cells. Am. J. Cancer Res. 9, 1382–1395 (2019)PubMedPubMedCentral Q. Zhang, H. Liu, H. Wang, M. Lu, Y. Miao, J. Ding, H. Li, X. Gao, S. Sun, J. Zheng, Lenvatinib promotes antitumor immunity by enhancing the tumor infiltration and activation of NK cells. Am. J. Cancer Res. 9, 1382–1395 (2019)PubMedPubMedCentral
41.
Zurück zum Zitat J.M. Llovet, S. Ricci, V. Mazzaferro, P. Hilgard, E. Gane, J.F. Blanc, A.C. de Oliveira, A. Santoro, J.L. Raoul, A. Forner, M. Schwartz, C. Porta, S. Zeuzem, L. Bolondi, T.F. Greten, P.R. Galle, J.F. Seitz, I. Borbath, D. Häussinger, et al., SHARP investigators study group. Sorafenib in advanced hepatocellular carcinoma. N. Engl. J. Med. 359, 378–390 (2008)PubMedCrossRef J.M. Llovet, S. Ricci, V. Mazzaferro, P. Hilgard, E. Gane, J.F. Blanc, A.C. de Oliveira, A. Santoro, J.L. Raoul, A. Forner, M. Schwartz, C. Porta, S. Zeuzem, L. Bolondi, T.F. Greten, P.R. Galle, J.F. Seitz, I. Borbath, D. Häussinger, et al., SHARP investigators study group. Sorafenib in advanced hepatocellular carcinoma. N. Engl. J. Med. 359, 378–390 (2008)PubMedCrossRef
42.
Zurück zum Zitat Q. Zhu, H. Ren, X. Li, B. Qian, S. Fan, F. Hu, L. Xu, B. Zhai, Silencing KIF14 reverses acquired resistance to sorafenib in hepatocellular carcinoma. Aging (Albany NY) 12, 22975–23003 (2020) Q. Zhu, H. Ren, X. Li, B. Qian, S. Fan, F. Hu, L. Xu, B. Zhai, Silencing KIF14 reverses acquired resistance to sorafenib in hepatocellular carcinoma. Aging (Albany NY) 12, 22975–23003 (2020)
43.
Zurück zum Zitat V. Tovar, H. Cornella, A. Moeini, S. Vidal, Y. Hoshida, D. Sia, J. Peix, L. Cabellos, C. Alsinet, S. Torrecilla, I. Martinez-Quetglas, J.J. Lozano, C. Desbois-Mouthon, M. Solé, J. Domingo-Domenech, A. Villanueva, J.M. Llovet, Tumour initiating cells and IGF/FGF signalling contribute to sorafenib resistance in hepatocellular carcinoma. Gut 66, 530–540 (2017)PubMedCrossRef V. Tovar, H. Cornella, A. Moeini, S. Vidal, Y. Hoshida, D. Sia, J. Peix, L. Cabellos, C. Alsinet, S. Torrecilla, I. Martinez-Quetglas, J.J. Lozano, C. Desbois-Mouthon, M. Solé, J. Domingo-Domenech, A. Villanueva, J.M. Llovet, Tumour initiating cells and IGF/FGF signalling contribute to sorafenib resistance in hepatocellular carcinoma. Gut 66, 530–540 (2017)PubMedCrossRef
44.
Zurück zum Zitat M. Liu, Q. Hu, M. Tu, X. Wang, Z. Yang, G. Yang, R. Luo, MCM6 promotes metastasis of hepatocellular carcinoma via MEK/ERK pathway and serves as a novel serum biomarker for early recurrence. J. Exp. Clin. Cancer Res. 37, 10 (2018)PubMedPubMedCentralCrossRef M. Liu, Q. Hu, M. Tu, X. Wang, Z. Yang, G. Yang, R. Luo, MCM6 promotes metastasis of hepatocellular carcinoma via MEK/ERK pathway and serves as a novel serum biomarker for early recurrence. J. Exp. Clin. Cancer Res. 37, 10 (2018)PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat J. Wang, Z. Li, C. Zuo, Q. Xie, H. Li, J. Jia, Z. Zhen, R. Qi, Z. Li, D. Liu, B. Sun, Knockdown of RRS1 by lentiviral-mediated RNAi promotes apoptosis and suppresses proliferation of human hepatocellular carcinoma cells. Oncol. Rep. 38, 2166–2172 (2017)PubMedPubMedCentralCrossRef J. Wang, Z. Li, C. Zuo, Q. Xie, H. Li, J. Jia, Z. Zhen, R. Qi, Z. Li, D. Liu, B. Sun, Knockdown of RRS1 by lentiviral-mediated RNAi promotes apoptosis and suppresses proliferation of human hepatocellular carcinoma cells. Oncol. Rep. 38, 2166–2172 (2017)PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Z. Zhou, X. Liu, Y. Li, J. Li, W. Deng, J. Zhong, L. Chen, Y. Li, X. Zeng, G. Wang, J. Zhu, B. Fu, TP53INP2 modulates epithelial-to-mesenchymal transition via the GSK-3β/β-catenin/Snail1 pathway in bladder cancer cells. Onco Targets Ther 13, 9587–9597 (2020)PubMedPubMedCentralCrossRef Z. Zhou, X. Liu, Y. Li, J. Li, W. Deng, J. Zhong, L. Chen, Y. Li, X. Zeng, G. Wang, J. Zhu, B. Fu, TP53INP2 modulates epithelial-to-mesenchymal transition via the GSK-3β/β-catenin/Snail1 pathway in bladder cancer cells. Onco Targets Ther 13, 9587–9597 (2020)PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat A. Surcel, E.S. Schiffhauer, D.G. Thomas, Q. Zhu, K.T. DiNapoli, M. Herbig, O. Otto, H. West-Foyle, A. Jacobi, M. Kräter, K. Plak, J. Guck, E.M. Jaffee, P.A. Iglesias, R.A. Anders, D.N. Robinson, Targeting mechanoresponsive proteins in pancreatic cancer: 4-hydroxyacetophenone blocks dissemination and invasion by activating MYH14. Cancer Res. 79, 4665–4678 (2019)PubMedPubMedCentralCrossRef A. Surcel, E.S. Schiffhauer, D.G. Thomas, Q. Zhu, K.T. DiNapoli, M. Herbig, O. Otto, H. West-Foyle, A. Jacobi, M. Kräter, K. Plak, J. Guck, E.M. Jaffee, P.A. Iglesias, R.A. Anders, D.N. Robinson, Targeting mechanoresponsive proteins in pancreatic cancer: 4-hydroxyacetophenone blocks dissemination and invasion by activating MYH14. Cancer Res. 79, 4665–4678 (2019)PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat B. Zhang, H.Y. Wang, D.X. Zhao, D.X. Wang, Q. Zeng, J.F. Xi, X. Nan, L.J. He, J.N. Zhou, X.T. Pei, W. Yue, The splicing regulatory factor hnRNPU is a novel transcriptional target of c-Myc in hepatocellular carcinoma. FEBS Lett. 595, 68–84 (2021)PubMedCrossRef B. Zhang, H.Y. Wang, D.X. Zhao, D.X. Wang, Q. Zeng, J.F. Xi, X. Nan, L.J. He, J.N. Zhou, X.T. Pei, W. Yue, The splicing regulatory factor hnRNPU is a novel transcriptional target of c-Myc in hepatocellular carcinoma. FEBS Lett. 595, 68–84 (2021)PubMedCrossRef
49.
Zurück zum Zitat S. Talukdar, L. Emdad, S.K. Das, P.B. Fisher, EGFR: An essential receptor tyrosine kinase-regulator of cancer stem cells. Adv. Cancer Res. 147, 161–188 (2020)PubMedCrossRef S. Talukdar, L. Emdad, S.K. Das, P.B. Fisher, EGFR: An essential receptor tyrosine kinase-regulator of cancer stem cells. Adv. Cancer Res. 147, 161–188 (2020)PubMedCrossRef
50.
Zurück zum Zitat J. Chen, H. Zhu, Q. Liu, D. Ning, Z. Zhang, L. Zhang, J. Mo, P. Du, X. Liu, S. Song, Y. Fan, H. Liang, J. Liu, B. Zhang, X. Chen, DEPTOR induces a partial epithelial-to-mesenchymal transition and metastasis via autocrine TGFβ1 signaling and is associated with poor prognosis in hepatocellular carcinoma. J. Exp. Clin. Cancer Res. 38, 273 (2019)PubMedPubMedCentralCrossRef J. Chen, H. Zhu, Q. Liu, D. Ning, Z. Zhang, L. Zhang, J. Mo, P. Du, X. Liu, S. Song, Y. Fan, H. Liang, J. Liu, B. Zhang, X. Chen, DEPTOR induces a partial epithelial-to-mesenchymal transition and metastasis via autocrine TGFβ1 signaling and is associated with poor prognosis in hepatocellular carcinoma. J. Exp. Clin. Cancer Res. 38, 273 (2019)PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat J. Xing, V. Bhuria, K.C. Bui, M.L.T. Nguyen, Z. Hu, C.J. Hsieh, K. Wittstein, M. Stadler, L. Wilkens, J. Li, M. Kalesse, P. Bozko, R.R. Plentz, Haprolid inhibits tumor growth of hepatocellular carcinoma through Rb/E2F and Akt/mTOR inhibition. Cancers (Basel) 12, 615 (2020)CrossRef J. Xing, V. Bhuria, K.C. Bui, M.L.T. Nguyen, Z. Hu, C.J. Hsieh, K. Wittstein, M. Stadler, L. Wilkens, J. Li, M. Kalesse, P. Bozko, R.R. Plentz, Haprolid inhibits tumor growth of hepatocellular carcinoma through Rb/E2F and Akt/mTOR inhibition. Cancers (Basel) 12, 615 (2020)CrossRef
52.
Zurück zum Zitat L. Zhu, N. Yang, G. Du, C. Li, G. Liu, S. Liu, Y. Xu, Y. Di, W. Pan, X. Li, LncRNA CRNDE promotes the epithelial-mesenchymal transition of hepatocellular carcinoma cells via enhancing the Wnt/β-catenin signaling pathway. J. Cell. Biochem. 120, 1156–1164 (2018)PubMedCentralCrossRef L. Zhu, N. Yang, G. Du, C. Li, G. Liu, S. Liu, Y. Xu, Y. Di, W. Pan, X. Li, LncRNA CRNDE promotes the epithelial-mesenchymal transition of hepatocellular carcinoma cells via enhancing the Wnt/β-catenin signaling pathway. J. Cell. Biochem. 120, 1156–1164 (2018)PubMedCentralCrossRef
53.
Zurück zum Zitat J. Stebbing, A. Filipović, G. Giamas, Claudin-1 as a promoter of EMT in hepatocellular carcinoma. Oncogene 32, 4871–4872 (2013)PubMedCrossRef J. Stebbing, A. Filipović, G. Giamas, Claudin-1 as a promoter of EMT in hepatocellular carcinoma. Oncogene 32, 4871–4872 (2013)PubMedCrossRef
54.
Zurück zum Zitat M. Hu, M. Li, H. Huang, C. Lu, Isolated cancer stem cells from human liver cancer: Morphological and functional characteristics in primary culture. Clin. Transl. Oncol. 24, 48–56 (2022)PubMedCrossRef M. Hu, M. Li, H. Huang, C. Lu, Isolated cancer stem cells from human liver cancer: Morphological and functional characteristics in primary culture. Clin. Transl. Oncol. 24, 48–56 (2022)PubMedCrossRef
55.
Zurück zum Zitat X. Sang, F. Wu, D. Wu, S. Lin, J. Li, N. Zhao, X. Chen, A. Xu, Human hepatic cancer stem cells (HCSCs) markers correlated with immune infiltrates reveal prognostic significance of hepatocellular carcinoma. Front. Genet. 11, 112 (2020)PubMedPubMedCentralCrossRef X. Sang, F. Wu, D. Wu, S. Lin, J. Li, N. Zhao, X. Chen, A. Xu, Human hepatic cancer stem cells (HCSCs) markers correlated with immune infiltrates reveal prognostic significance of hepatocellular carcinoma. Front. Genet. 11, 112 (2020)PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat I. Pastushenko, C. Blanpain, EMT transition states during tumor progression and metastasis. Trends Cell Biol. 29, 212–226 (2019)PubMedCrossRef I. Pastushenko, C. Blanpain, EMT transition states during tumor progression and metastasis. Trends Cell Biol. 29, 212–226 (2019)PubMedCrossRef
57.
Zurück zum Zitat I. Pastushenko, A. Brisebarre, A. Sifrim, M. Fioramonti, T. Revenco, S. Boumahdi, A. Van Keymeulen, D. Brown, V. Moers, S. Lemaire, S. De Clercq, E. Minguijón, C. Balsat, Y. Sokolow, C. Dubois, F. De Cock, S. Scozzaro, F. Sopena, A. Lanas, et al., Identification of the tumour transition states occurring during EMT. Nature 556, 463–468 (2018)PubMedCrossRef I. Pastushenko, A. Brisebarre, A. Sifrim, M. Fioramonti, T. Revenco, S. Boumahdi, A. Van Keymeulen, D. Brown, V. Moers, S. Lemaire, S. De Clercq, E. Minguijón, C. Balsat, Y. Sokolow, C. Dubois, F. De Cock, S. Scozzaro, F. Sopena, A. Lanas, et al., Identification of the tumour transition states occurring during EMT. Nature 556, 463–468 (2018)PubMedCrossRef
58.
Zurück zum Zitat M. Masuda, W.Y. Chen, A. Miyanaga, Y. Nakamura, K. Kawasaki, T. Sakuma, M. Ono, C.L. Chen, K. Honda, T. Yamada, Alternative mammalian target of rapamycin (mTOR) signal activation in sorafenib-resistant hepatocellular carcinoma cells revealed by array-based pathway profiling. Mol. Cell. Proteomics 13, 1429–1438 (2014)PubMedPubMedCentralCrossRef M. Masuda, W.Y. Chen, A. Miyanaga, Y. Nakamura, K. Kawasaki, T. Sakuma, M. Ono, C.L. Chen, K. Honda, T. Yamada, Alternative mammalian target of rapamycin (mTOR) signal activation in sorafenib-resistant hepatocellular carcinoma cells revealed by array-based pathway profiling. Mol. Cell. Proteomics 13, 1429–1438 (2014)PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat I. Eliseeva, M. Vasilieva, L.P. Ovchinnikov, Translation of human β-actin mRNA is regulated by mTOR pathway. Genes (Basel) 10, 96 (2019)CrossRef I. Eliseeva, M. Vasilieva, L.P. Ovchinnikov, Translation of human β-actin mRNA is regulated by mTOR pathway. Genes (Basel) 10, 96 (2019)CrossRef
Metadaten
Titel
Heterogeneity, inherent and acquired drug resistance in patient-derived organoid models of primary liver cancer
verfasst von
Linfeng Xian
Pei Zhao
Xi Chen
Zhimin Wei
Hongxiang Ji
Jun Zhao
Wenbin Liu
Zishuai Li
Donghong Liu
Xue Han
Youwen Qian
Hui Dong
Xiong Zhou
Junyan Fan
Xiaoqiong Zhu
Jianhua Yin
Xiaojie Tan
Dongming Jiang
Hongping Yu
Guangwen Cao
Publikationsdatum
29.08.2022
Verlag
Springer Netherlands
Erschienen in
Cellular Oncology / Ausgabe 5/2022
Print ISSN: 2211-3428
Elektronische ISSN: 2211-3436
DOI
https://doi.org/10.1007/s13402-022-00707-3

Weitere Artikel der Ausgabe 5/2022

Cellular Oncology 5/2022 Zur Ausgabe

Neu im Fachgebiet Pathologie

Molekularpathologische Untersuchungen im Wandel der Zeit

Open Access Biomarker Leitthema

Um auch an kleinen Gewebeproben zuverlässige und reproduzierbare Ergebnisse zu gewährleisten ist eine strenge Qualitätskontrolle in jedem Schritt des Arbeitsablaufs erforderlich. Eine nicht ordnungsgemäße Prüfung oder Behandlung des …

Vergleichende Pathologie in der onkologischen Forschung

Pathologie Leitthema

Die vergleichende experimentelle Pathologie („comparative experimental pathology“) ist ein Fachbereich an der Schnittstelle von Human- und Veterinärmedizin. Sie widmet sich der vergleichenden Erforschung von Gemeinsamkeiten und Unterschieden von …

Gastrointestinale Stromatumoren

Open Access GIST CME-Artikel

Gastrointestinale Stromatumoren (GIST) stellen seit über 20 Jahren ein Paradigma für die zielgerichtete Therapie mit Tyrosinkinaseinhibitoren dar. Eine elementare Voraussetzung für eine mögliche neoadjuvante oder adjuvante Behandlung bei …

Personalisierte Medizin in der Onkologie

Aufgrund des erheblichen technologischen Fortschritts in der molekularen und genetischen Diagnostik sowie zunehmender Erkenntnisse über die molekulare Pathogenese von Krankheiten hat in den letzten zwei Jahrzehnten ein grundlegender …