Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2019

Open Access 01-12-2019 | Gastric Cancer | Research

A subset of diffuse-type gastric cancer is susceptible to mTOR inhibitors and checkpoint inhibitors

Authors: Hiroshi Fukamachi, Seon-Kyu Kim, Jiwon Koh, Hye Seung Lee, Yasushi Sasaki, Kentaro Yamashita, Taketo Nishikawaji, Shu Shimada, Yoshimitsu Akiyama, Sun-ju Byeon, Dong-Hyuck Bae, Keisuke Okuno, Masatoshi Nakagawa, Toshiro Tanioka, Mikito Inokuchi, Hiroshi Kawachi, Kiichiro Tsuchiya, Kazuyuki Kojima, Takashi Tokino, Yoshinobu Eishi, Yong Sung Kim, Woo Ho Kim, Yasuhito Yuasa, Shinji Tanaka

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2019

Login to get access

Abstract

Background

Mechanistic target of rapamycin (mTOR) pathway is essential for the growth of gastric cancer (GC), but mTOR inhibitor everolimus was not effective for the treatment of GCs. The Cancer Genome Atlas (TCGA) researchers reported that most diffuse-type GCs were genomically stable (GS). Pathological analysis suggested that some diffuse-type GCs developed from intestinal-type GCs.

Methods

We established patient-derived xenograft (PDX) lines from diffuse-type GCs, and searched for drugs that suppressed their growth. Diffuse-type GCs were classified into subtypes by their gene expression profiles.

Results

mTOR inhibitor temsirolimus strongly suppressed the growth of PDX-derived diffuse-type GC-initiating cells, which was regulated via Wnt-mTOR axis. These cells were microsatellite unstable (MSI) or chromosomally unstable (CIN), inconsistent with TCGA report. Diffuse-type GCs in TCGA cohort could be classified into two clusters, and GS subtype was major in cluster I while CIN and MSI subtypes were predominant in cluster II where PDX-derived diffuse-type GC cells were included. We estimated that about 9 and 55% of the diffuse-type GCs in cluster II were responders to mTOR inhibitors and checkpoint inhibitors, respectively, by identifying PIK3CA mutations and MSI condition in TCGA cohort. These ratios were far greater than those of diffuse-type GCs in cluster I or intestinal-type GCs. Further analysis suggested that diffuse-type GCs in cluster II developed from intestinal-type GCs while those in cluster I from normal gastric epithelial cells.

Conclusion

mTOR inhibitors and checkpoint inhibitors might be useful for the treatment of a subset of diffuse-type GCs which may develop from intestinal-type GCs.
Appendix
Available only for authorised users
Literature
1.
go back to reference Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136:E359–86.CrossRef Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136:E359–86.CrossRef
2.
go back to reference Lauren P. The two histological main types of gastric carcinoma: diffuse and so-called intestinal-type carcinoma. An attempt at a histo-clinical classification. Acta Pathol Microbiol Scand. 1965;64:31–49.CrossRef Lauren P. The two histological main types of gastric carcinoma: diffuse and so-called intestinal-type carcinoma. An attempt at a histo-clinical classification. Acta Pathol Microbiol Scand. 1965;64:31–49.CrossRef
3.
go back to reference Vauhkonen M, Vauhkonen H, Sipponen P. Pathology and molecular biology of gastric cancer. Best Pract Res Clin Gastroenterol. 2006;20:651–74.CrossRef Vauhkonen M, Vauhkonen H, Sipponen P. Pathology and molecular biology of gastric cancer. Best Pract Res Clin Gastroenterol. 2006;20:651–74.CrossRef
4.
go back to reference Ikeda Y, Mori M, Kamakura T, Haraguchi Y, Saku M, Sugimachi K. Increased incidence of undifferentiated type of gastric cancer with tumor progression in 912 patients with early gastric cancer and 1245 with advanced gastric cancer. Cancer. 1994;73:2459–63.CrossRef Ikeda Y, Mori M, Kamakura T, Haraguchi Y, Saku M, Sugimachi K. Increased incidence of undifferentiated type of gastric cancer with tumor progression in 912 patients with early gastric cancer and 1245 with advanced gastric cancer. Cancer. 1994;73:2459–63.CrossRef
5.
go back to reference Arai T, Sakurai U, Sawabe M, Honma N, Aida J, Ushio Y, et al. Frequent microsatellite instability in papillary and solid-type, poorly differentiated adenocarcinomas of the stomach. Gastric Cancer. 2013;16:505–12.CrossRef Arai T, Sakurai U, Sawabe M, Honma N, Aida J, Ushio Y, et al. Frequent microsatellite instability in papillary and solid-type, poorly differentiated adenocarcinomas of the stomach. Gastric Cancer. 2013;16:505–12.CrossRef
6.
go back to reference Zheng HC, Li XH, Hara T, Masuda S, Yang XH, Guan YF, et al. Mixed-type gastric carcinomas exhibit more aggressive features and indicate the histogenesis of carcinomas. Virchows Arch. 2008;452:525–34.CrossRef Zheng HC, Li XH, Hara T, Masuda S, Yang XH, Guan YF, et al. Mixed-type gastric carcinomas exhibit more aggressive features and indicate the histogenesis of carcinomas. Virchows Arch. 2008;452:525–34.CrossRef
7.
go back to reference Komatsu S, Ichikawa D, Miyamae M, Shimizu H, Konishi H, Shiozaki A, et al. Histological mixed-type as an independent prognostic factor in stage I gastric carcinoma. World J Gastroenterol. 2015;21:549–55.CrossRef Komatsu S, Ichikawa D, Miyamae M, Shimizu H, Konishi H, Shiozaki A, et al. Histological mixed-type as an independent prognostic factor in stage I gastric carcinoma. World J Gastroenterol. 2015;21:549–55.CrossRef
8.
go back to reference Ikeguchi M, Miyake T, Matsunaga T, Yamamoto M, Fukumoto Y, Yamada Y, et al. Recent results of therapy for scirrhous gastric cancer. Surg Today. 2009;39:290–4.CrossRef Ikeguchi M, Miyake T, Matsunaga T, Yamamoto M, Fukumoto Y, Yamada Y, et al. Recent results of therapy for scirrhous gastric cancer. Surg Today. 2009;39:290–4.CrossRef
9.
go back to reference Lordick F, Janjigian YY. Clinical impact of tumour biology in the management of gastroesophageal cancer. Nat Rev Clin Oncol. 2016;13:348–60.CrossRef Lordick F, Janjigian YY. Clinical impact of tumour biology in the management of gastroesophageal cancer. Nat Rev Clin Oncol. 2016;13:348–60.CrossRef
10.
go back to reference Clarke MF, Dick JE, Dirks PB, Eaves CJ, Jamieson CH, Jones DL, et al. Cancer stem cells-perspectives on current status and future directions: AACR workshop on cancer stem cells. Cancer Res. 2006;66:9339–44.CrossRef Clarke MF, Dick JE, Dirks PB, Eaves CJ, Jamieson CH, Jones DL, et al. Cancer stem cells-perspectives on current status and future directions: AACR workshop on cancer stem cells. Cancer Res. 2006;66:9339–44.CrossRef
11.
go back to reference Batlle E, Clevers H. Cancer stem cells revisited. Nat Med. 2017;23:1124–34.CrossRef Batlle E, Clevers H. Cancer stem cells revisited. Nat Med. 2017;23:1124–34.CrossRef
12.
go back to reference Dashzeveg NK, Taftaf R, Ramos EK, Torre-Healy L, Chumakova A, Silver DJ, et al. New advances and challenges of targeting cancer stem cells. Cancer Res. 2017;77:5222–7.CrossRef Dashzeveg NK, Taftaf R, Ramos EK, Torre-Healy L, Chumakova A, Silver DJ, et al. New advances and challenges of targeting cancer stem cells. Cancer Res. 2017;77:5222–7.CrossRef
13.
go back to reference Bekaii-Saab T, El-Rayes B. Identifying and targeting cancer stem cells in the treatment of gastric cancer. Cancer. 2017;123:1303–12.CrossRef Bekaii-Saab T, El-Rayes B. Identifying and targeting cancer stem cells in the treatment of gastric cancer. Cancer. 2017;123:1303–12.CrossRef
14.
go back to reference Apicella M, Corso S, Giordano S. Targeted therapies for gastric cancer: failures and hopes from clinical trials. Oncotarget. 2017;8:57654–69.CrossRef Apicella M, Corso S, Giordano S. Targeted therapies for gastric cancer: failures and hopes from clinical trials. Oncotarget. 2017;8:57654–69.CrossRef
15.
go back to reference Krebsbach PH, Villa-Diaz LG. The role of integrin alpha6 (CD49f) in stem cells: more than a conserved biomarker. Stem Cells Dev. 2017;26:1090–9.CrossRef Krebsbach PH, Villa-Diaz LG. The role of integrin alpha6 (CD49f) in stem cells: more than a conserved biomarker. Stem Cells Dev. 2017;26:1090–9.CrossRef
16.
go back to reference Fukamachi H, Seol HS, Shimada S, Funasaka C, Baba K, Kim JH, et al. CD49fhigh cells retain sphere-forming and tumor-initiating activities in human gastric tumors. PLoS One. 2013;8:e72438.CrossRef Fukamachi H, Seol HS, Shimada S, Funasaka C, Baba K, Kim JH, et al. CD49fhigh cells retain sphere-forming and tumor-initiating activities in human gastric tumors. PLoS One. 2013;8:e72438.CrossRef
17.
go back to reference Bartfeld S, Bayram T, van de Wetering M, Huch M, Begthel H, Kujala P, et al. In vitro expansion of human gastric epithelial stem cells and their responses to bacterial infection. Gastroenterology. 2015;148:126–36 e6.CrossRef Bartfeld S, Bayram T, van de Wetering M, Huch M, Begthel H, Kujala P, et al. In vitro expansion of human gastric epithelial stem cells and their responses to bacterial infection. Gastroenterology. 2015;148:126–36 e6.CrossRef
18.
go back to reference Broutier L, Andersson-Rolf A, Hindley CJ, Boj SF, Clevers H, Koo BK, et al. Culture and establishment of self-renewing human and mouse adult liver and pancreas 3D organoids and their genetic manipulation. Nat Protoc. 2016;11:1724–43.CrossRef Broutier L, Andersson-Rolf A, Hindley CJ, Boj SF, Clevers H, Koo BK, et al. Culture and establishment of self-renewing human and mouse adult liver and pancreas 3D organoids and their genetic manipulation. Nat Protoc. 2016;11:1724–43.CrossRef
19.
go back to reference Kim SK, Kim SY, Kim JH, Roh SA, Cho DH, Kim YS, et al. A nineteen gene-based risk score classifier predicts prognosis of colorectal cancer patients. Mol Oncol. 2014;8:1653–66.CrossRef Kim SK, Kim SY, Kim JH, Roh SA, Cho DH, Kim YS, et al. A nineteen gene-based risk score classifier predicts prognosis of colorectal cancer patients. Mol Oncol. 2014;8:1653–66.CrossRef
20.
go back to reference Sohn BH, Hwang JE, Jang HJ, Lee HS, Oh SC, Shim JJ, et al. Clinical significance of four molecular subtypes of gastric cancer identified by the cancer genome atlas project. Clin Cancer Res. 2017;23:1–9.CrossRef Sohn BH, Hwang JE, Jang HJ, Lee HS, Oh SC, Shim JJ, et al. Clinical significance of four molecular subtypes of gastric cancer identified by the cancer genome atlas project. Clin Cancer Res. 2017;23:1–9.CrossRef
21.
go back to reference Koh J, Ock CY, Kim JW, Nam SK, Kwak Y, Yun S, et al. Clinicopathologic implications of immune classification by PD-L1 expression and CD8-positive tumor-infiltrating lymphocytes in stage II and III gastric cancer patients. Oncotarget. 2017;8:26356–67. Koh J, Ock CY, Kim JW, Nam SK, Kwak Y, Yun S, et al. Clinicopathologic implications of immune classification by PD-L1 expression and CD8-positive tumor-infiltrating lymphocytes in stage II and III gastric cancer patients. Oncotarget. 2017;8:26356–67.
22.
go back to reference Byeon SJ, Han N, Choi J, Kim MA, Kim WH. Prognostic implication of TSC1 and mTOR expression in gastric carcinoma. J Surg Oncol. 2017;109:812–7.CrossRef Byeon SJ, Han N, Choi J, Kim MA, Kim WH. Prognostic implication of TSC1 and mTOR expression in gastric carcinoma. J Surg Oncol. 2017;109:812–7.CrossRef
23.
go back to reference Ikeda H, Ishiguro K, Igarashi T, Aoki Y, Hayashi T, Ishida T, et al. Molecular diagnostics of a single drug-resistant multiple myeloma case using targeted next-generation sequencing. Onco Targets Ther. 2015;8:2805–15.CrossRef Ikeda H, Ishiguro K, Igarashi T, Aoki Y, Hayashi T, Ishida T, et al. Molecular diagnostics of a single drug-resistant multiple myeloma case using targeted next-generation sequencing. Onco Targets Ther. 2015;8:2805–15.CrossRef
24.
go back to reference Wanajo A, Sasaki A, Nagasaki H, Shimada S, Otsubo T, Owaki S, et al. Methylation of the calcium channel-related gene, CACNA2D3, is frequent and a poor prognostic factor in gastric cancer. Gastroenterology. 2008;135:580–90.CrossRef Wanajo A, Sasaki A, Nagasaki H, Shimada S, Otsubo T, Owaki S, et al. Methylation of the calcium channel-related gene, CACNA2D3, is frequent and a poor prognostic factor in gastric cancer. Gastroenterology. 2008;135:580–90.CrossRef
25.
go back to reference Herman JG, Graff JR, Myöhänen S, Nelkin BD, Baylin SB. Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands. Proc Natl Acad Sci U S A. 1996;93:9821–6.CrossRef Herman JG, Graff JR, Myöhänen S, Nelkin BD, Baylin SB. Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands. Proc Natl Acad Sci U S A. 1996;93:9821–6.CrossRef
26.
go back to reference Yamashita K, Arimura Y, Kurokawa S, Itoh F, Endo T, Hirata K, et al. Microsatellite instability in patients with multiple primary cancers of the gastrointestinal tract. Gut. 2000;46:790–4.CrossRef Yamashita K, Arimura Y, Kurokawa S, Itoh F, Endo T, Hirata K, et al. Microsatellite instability in patients with multiple primary cancers of the gastrointestinal tract. Gut. 2000;46:790–4.CrossRef
27.
go back to reference Fletcher J. Immortalized cell lines: chromosome preparation and banding. In: Gosden R, editor. Chromosome analysis protocols. Totowa: Hamana Press; 1994. p. 51–7.CrossRef Fletcher J. Immortalized cell lines: chromosome preparation and banding. In: Gosden R, editor. Chromosome analysis protocols. Totowa: Hamana Press; 1994. p. 51–7.CrossRef
28.
go back to reference The Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513:202–9.CrossRef The Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513:202–9.CrossRef
29.
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 modeling of anticancer drug sensitivity. Nature. 2012;483:603–7.CrossRef Barretina J, Caponigro G, Stransky N, Venkatesan K, Margolin AA, Kim S, et al. The cancer cell line encyclopedia enables predictive modeling of anticancer drug sensitivity. Nature. 2012;483:603–7.CrossRef
30.
go back to reference Johnson JI, Decker S, Zaharevitz D, Rubinstein LV, Venditti JM, Schepartz S, et al. Relationships between drug activity in NCI preclinical in vitro and in vivo models and early clinical trials. Br J Cancer. 2001;84:1424–31.CrossRef Johnson JI, Decker S, Zaharevitz D, Rubinstein LV, Venditti JM, Schepartz S, et al. Relationships between drug activity in NCI preclinical in vitro and in vivo models and early clinical trials. Br J Cancer. 2001;84:1424–31.CrossRef
31.
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;108:18708–13.CrossRef 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;108:18708–13.CrossRef
32.
go back to reference Weeber F, Ooft SN, Dijkstra KK, Voest EE. Tumor organoids as a pre-clinical cancer model for drug discovery. Cell Chem Biol. 2017;24:1092–100.CrossRef Weeber F, Ooft SN, Dijkstra KK, Voest EE. Tumor organoids as a pre-clinical cancer model for drug discovery. Cell Chem Biol. 2017;24:1092–100.CrossRef
33.
go back to reference Liu X, Meltzer SJ. Gastric cancer in the era of precision medicine. Cell Mol Gastroenterol Hepatol. 2017;3:348–58.CrossRef Liu X, Meltzer SJ. Gastric cancer in the era of precision medicine. Cell Mol Gastroenterol Hepatol. 2017;3:348–58.CrossRef
34.
go back to reference Seidlitz T, Merker SR, Rothe A, Zakrzewski F, von Neubeck C, Grutzmann K, et al. Human gastric cancer modelling using organoids. Gut. 2019;68:207–17.CrossRef Seidlitz T, Merker SR, Rothe A, Zakrzewski F, von Neubeck C, Grutzmann K, et al. Human gastric cancer modelling using organoids. Gut. 2019;68:207–17.CrossRef
35.
go back to reference Tiriac H, Belleau P, Engle DD, Plenker D, Deschenes A, Somerville T, et al. Organoid profiling identifies common responders to chemotherapy in pancreatic cancer. Cancer Discov. 2018;8:1112–29.CrossRef Tiriac H, Belleau P, Engle DD, Plenker D, Deschenes A, Somerville T, et al. Organoid profiling identifies common responders to chemotherapy in pancreatic cancer. Cancer Discov. 2018;8:1112–29.CrossRef
36.
go back to reference Nanki K, Toshimitsu K, Takano A, Fujii M, Shimokawa M, Ohta Y, et al. Divergent routes toward Wnt and R-spondin niche independency during human gastric carcinogenesis. Cell. 2018;174:856–69 e17.CrossRef Nanki K, Toshimitsu K, Takano A, Fujii M, Shimokawa M, Ohta Y, et al. Divergent routes toward Wnt and R-spondin niche independency during human gastric carcinogenesis. Cell. 2018;174:856–69 e17.CrossRef
37.
go back to reference Yan HHN, Siu HC, Law S, Ho SL, Yue SSK, Tsui WY, et al. A comprehensive human gastric cancer organoid biobank captures tumor subtype heterogeneity and enables therapeutic screening. Cell Stem Cell. 2018;23:882–97 e811.CrossRef Yan HHN, Siu HC, Law S, Ho SL, Yue SSK, Tsui WY, et al. A comprehensive human gastric cancer organoid biobank captures tumor subtype heterogeneity and enables therapeutic screening. Cell Stem Cell. 2018;23:882–97 e811.CrossRef
38.
go back to reference Rini B, Kar S, Kirkpatrick P. Temsirolimus. Nat Rev Drug Discov. 2007;6:599–600.CrossRef Rini B, Kar S, Kirkpatrick P. Temsirolimus. Nat Rev Drug Discov. 2007;6:599–600.CrossRef
39.
40.
go back to reference Al-Batran SE, Ducreux M, Ohtsu A. mTOR as a therapeutic target in patients with gastric cancer. Int J Cancer. 2012;130:491–6.CrossRef Al-Batran SE, Ducreux M, Ohtsu A. mTOR as a therapeutic target in patients with gastric cancer. Int J Cancer. 2012;130:491–6.CrossRef
41.
go back to reference Ohtsu A, Ajani JA, Bai YX, Bang YJ, Chung HC, Pan HM, et al. Everolimus for previously treated advanced gastric cancer: results of the randomized, double-blind, phase III GRANITE-1 study. J Clin Oncol. 2013;31:3935–43.CrossRef Ohtsu A, Ajani JA, Bai YX, Bang YJ, Chung HC, Pan HM, et al. Everolimus for previously treated advanced gastric cancer: results of the randomized, double-blind, phase III GRANITE-1 study. J Clin Oncol. 2013;31:3935–43.CrossRef
42.
go back to reference Shen YC, Li CP, Yen CJ, Hsu C, Lin YL, Lin ZZ, et al. Phase II multicentered study of low-dose everolimus plus cisplatin and weekly 24-hour infusion of high-dose 5-fluorouracil and leucovorin as first-line treatment for patients with advanced gastric cancer. Oncology. 2014;87:104–13.CrossRef Shen YC, Li CP, Yen CJ, Hsu C, Lin YL, Lin ZZ, et al. Phase II multicentered study of low-dose everolimus plus cisplatin and weekly 24-hour infusion of high-dose 5-fluorouracil and leucovorin as first-line treatment for patients with advanced gastric cancer. Oncology. 2014;87:104–13.CrossRef
43.
go back to reference Park JH, Ryu MH, Park YS, Park SR, Na YS, Rhoo BY, et al. Successful control of heavily pretreated metastatic gastric cancer with the mTOR inhibitor everolimus (RAD001) in a patient with PIK3CA mutation and pS6 overexpression. BMC Cancer. 2015;15:119.CrossRef Park JH, Ryu MH, Park YS, Park SR, Na YS, Rhoo BY, et al. Successful control of heavily pretreated metastatic gastric cancer with the mTOR inhibitor everolimus (RAD001) in a patient with PIK3CA mutation and pS6 overexpression. BMC Cancer. 2015;15:119.CrossRef
44.
go back to reference Zhou J, Tan SH, Nicolas V, Bauvy C, Yang ND, Zhang J, et al. Activation of lysosomal function in the course of autophagy via mTORC1 suppression and autophagosome-lysosome fusion. Cell Res. 2013;23:508–23.CrossRef Zhou J, Tan SH, Nicolas V, Bauvy C, Yang ND, Zhang J, et al. Activation of lysosomal function in the course of autophagy via mTORC1 suppression and autophagosome-lysosome fusion. Cell Res. 2013;23:508–23.CrossRef
45.
go back to reference Chiang GG, Abraham RT. Phosphorylation of mammalian target of rapamycin (mTOR) at Ser-2448 is mediated by p70S6 kinase. J Biol Chem. 2005;280:25485–90.CrossRef Chiang GG, Abraham RT. Phosphorylation of mammalian target of rapamycin (mTOR) at Ser-2448 is mediated by p70S6 kinase. J Biol Chem. 2005;280:25485–90.CrossRef
46.
go back to reference Bhavanasi D, Klein PS. Wnt signaling in normal and malignant stem cells. Curr Stem Cell Rep. 2016;2:379–87.CrossRef Bhavanasi D, Klein PS. Wnt signaling in normal and malignant stem cells. Curr Stem Cell Rep. 2016;2:379–87.CrossRef
47.
go back to reference Acebron SP, Niehrs C. beta-catenin-independent roles of Wnt/LRP6 signaling. Trends Cell Biol. 2016;26:956–67.CrossRef Acebron SP, Niehrs C. beta-catenin-independent roles of Wnt/LRP6 signaling. Trends Cell Biol. 2016;26:956–67.CrossRef
48.
go back to reference Cho SY, Park JW, Liu Y, Park YS, Kim JH, Yang H, et al. Sporadic early-onset diffuse gastric cancers have high frequency of somatic CDH1 alterations, but low frequency of somatic RHOA mutations compared with late-onset cancers. Gastroenterology. 2017;153:536–49 e26.CrossRef Cho SY, Park JW, Liu Y, Park YS, Kim JH, Yang H, et al. Sporadic early-onset diffuse gastric cancers have high frequency of somatic CDH1 alterations, but low frequency of somatic RHOA mutations compared with late-onset cancers. Gastroenterology. 2017;153:536–49 e26.CrossRef
49.
go back to reference Shen J, Ju Z, Zhao W, Wang L, Peng Y, Ge Z, et al. ARID1A deficiency promotes mutability and potentiates therapeutic antitumor immunity unleashed by immune checkpoint blockade. Nat Med. 2018;24:556–62.CrossRef Shen J, Ju Z, Zhao W, Wang L, Peng Y, Ge Z, et al. ARID1A deficiency promotes mutability and potentiates therapeutic antitumor immunity unleashed by immune checkpoint blockade. Nat Med. 2018;24:556–62.CrossRef
50.
go back to reference Jakubowska A, Nej K, Huzarski T, Scott RJ, Lubiński J. BRCA2 gene mutations in families with aggregations of breast and stomach cancers. Br J Cancer. 2002;87:888–91.CrossRef Jakubowska A, Nej K, Huzarski T, Scott RJ, Lubiński J. BRCA2 gene mutations in families with aggregations of breast and stomach cancers. Br J Cancer. 2002;87:888–91.CrossRef
51.
go back to reference Janku F, Tsimberidou AM, Garrido-Laguna I, Wang X, Luthra R, Hong DS, et al. PIK3CA mutations in patients with advanced cancers treated with PI3K/AKT/mTOR axis inhibitors. Mol Cancer Ther. 2011;10:558–65.CrossRef Janku F, Tsimberidou AM, Garrido-Laguna I, Wang X, Luthra R, Hong DS, et al. PIK3CA mutations in patients with advanced cancers treated with PI3K/AKT/mTOR axis inhibitors. Mol Cancer Ther. 2011;10:558–65.CrossRef
52.
go back to reference Chalmers ZR, Connelly CF, Fabrizio D, Gay L, Ali SM, Ennis R, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017;9:34.CrossRef Chalmers ZR, Connelly CF, Fabrizio D, Gay L, Ali SM, Ennis R, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017;9:34.CrossRef
53.
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;49:1567–75.CrossRef 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;49:1567–75.CrossRef
54.
go back to reference Ma C, Patel K, Singhi AD, Ren B, Zhu B, Shaikh F, et al. Programmed death-ligand 1 expression is common in gastric cancer associated with Epstein-Barr virus or microsatellite instability. Am J Surg Pathol. 2016;40:1496–506.CrossRef Ma C, Patel K, Singhi AD, Ren B, Zhu B, Shaikh F, et al. Programmed death-ligand 1 expression is common in gastric cancer associated with Epstein-Barr virus or microsatellite instability. Am J Surg Pathol. 2016;40:1496–506.CrossRef
55.
go back to reference Gu L, Chen M, Guo D, Zhu H, Zhang W, Pan J, et al. PD-L1 and gastric cancer prognosis: a systematic review and meta-analysis. PLoS One. 2017;12:e0182692.CrossRef Gu L, Chen M, Guo D, Zhu H, Zhang W, Pan J, et al. PD-L1 and gastric cancer prognosis: a systematic review and meta-analysis. PLoS One. 2017;12:e0182692.CrossRef
56.
go back to reference Taieb J, Moehler M, Boku N, Ajani JA, Yanez Ruiz E, Ryu MH, et al. Evolution of checkpoint inhibitors for the treatment of metastatic gastric cancers: current status and future perspectives. Cancer Treat Rev. 2018;66:104–13.CrossRef Taieb J, Moehler M, Boku N, Ajani JA, Yanez Ruiz E, Ryu MH, et al. Evolution of checkpoint inhibitors for the treatment of metastatic gastric cancers: current status and future perspectives. Cancer Treat Rev. 2018;66:104–13.CrossRef
57.
go back to reference Lastwika KJ, Wilson W 3rd, Li QK, Norris J, Xu H, Ghazarian SR, et al. Control of PD-L1 expression by oncogenic activation of the AKT-mTOR pathway in non-small cell lung cancer. Cancer Res. 2016;76:227–38.CrossRef Lastwika KJ, Wilson W 3rd, Li QK, Norris J, Xu H, Ghazarian SR, et al. Control of PD-L1 expression by oncogenic activation of the AKT-mTOR pathway in non-small cell lung cancer. Cancer Res. 2016;76:227–38.CrossRef
58.
go back to reference Liu J, Quan L, Zhang C, Liu A, Tong D, Wang J. Over-activated PD-1/PD-L1 axis facilitates the chemoresistance of diffuse large B-cell lymphoma cells to the CHOP regimen. Oncol Lett. 2018;15:3321–8. Liu J, Quan L, Zhang C, Liu A, Tong D, Wang J. Over-activated PD-1/PD-L1 axis facilitates the chemoresistance of diffuse large B-cell lymphoma cells to the CHOP regimen. Oncol Lett. 2018;15:3321–8.
59.
go back to reference Wu X, Li Y, Liu X, Chen C, Harrington SM, Cao S, et al. Targeting B7-H1 (PD-L1) sensitizes cancer cells to chemotherapy. Heliyon. 2018;4:e01039.CrossRef Wu X, Li Y, Liu X, Chen C, Harrington SM, Cao S, et al. Targeting B7-H1 (PD-L1) sensitizes cancer cells to chemotherapy. Heliyon. 2018;4:e01039.CrossRef
60.
go back to reference Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357:409–13.CrossRef Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357:409–13.CrossRef
61.
go back to reference Kim ST, Cristescu R, Bass AJ, Kim KM, Odegaard JI, Kim K, et al. Comprehensive molecular characterization of clinical responses to PD-1 inhibition in metastatic gastric cancer. Nat Med. 2018;24:1449–58.CrossRef Kim ST, Cristescu R, Bass AJ, Kim KM, Odegaard JI, Kim K, et al. Comprehensive molecular characterization of clinical responses to PD-1 inhibition in metastatic gastric cancer. Nat Med. 2018;24:1449–58.CrossRef
62.
go back to reference Velho S, Oliveira C, Ferreira A, Ferreira AC, Suriano G, Schwartz S Jr, et al. The prevalence of PIK3CA mutations in gastric and colon cancer. Eur J Cancer. 2005;41:1649–54.CrossRef Velho S, Oliveira C, Ferreira A, Ferreira AC, Suriano G, Schwartz S Jr, et al. The prevalence of PIK3CA mutations in gastric and colon cancer. Eur J Cancer. 2005;41:1649–54.CrossRef
63.
go back to reference Polom K, Marrelli D, Roviello G, Pascale V, Voglino C, Vindigni C, et al. PIK3CA mutation in gastric cancer and the role of microsatellite instability status in mutations of exons 9 and 20 of the PIK3CA gene. Adv Clin Exp Med. 2018;27:963–9.CrossRef Polom K, Marrelli D, Roviello G, Pascale V, Voglino C, Vindigni C, et al. PIK3CA mutation in gastric cancer and the role of microsatellite instability status in mutations of exons 9 and 20 of the PIK3CA gene. Adv Clin Exp Med. 2018;27:963–9.CrossRef
64.
go back to reference Böger C, Kruger S, Behrens HM, Bock S, Haag J, Kalthoff H, et al. Epstein-Barr virus-associated gastric cancer reveals intratumoral heterogeneity of PIK3CA mutations. Ann Oncol. 2017;28:1005–14.CrossRef Böger C, Kruger S, Behrens HM, Bock S, Haag J, Kalthoff H, et al. Epstein-Barr virus-associated gastric cancer reveals intratumoral heterogeneity of PIK3CA mutations. Ann Oncol. 2017;28:1005–14.CrossRef
65.
go back to reference Katsuno Y, Ehata S, Yashiro M, Yanagihara K, Hirakawa K, Miyazono K. Coordinated expression of REG4 and aldehyde dehydrogenase 1 regulating tumourigenic capacity of diffuse-type gastric carcinoma-initiating cells is inhibited by TGF-beta. J Pathol. 2012;228:391–404.CrossRef Katsuno Y, Ehata S, Yashiro M, Yanagihara K, Hirakawa K, Miyazono K. Coordinated expression of REG4 and aldehyde dehydrogenase 1 regulating tumourigenic capacity of diffuse-type gastric carcinoma-initiating cells is inhibited by TGF-beta. J Pathol. 2012;228:391–404.CrossRef
66.
go back to reference Rocco A, Liguori E, Pirozzi G, Tirino V, Compare D, Franco R, et al. CD133 and CD44 cell surface markers do not identify cancer stem cells in primary human gastric tumors. J Cell Physiol. 2012;227:2686–93.CrossRef Rocco A, Liguori E, Pirozzi G, Tirino V, Compare D, Franco R, et al. CD133 and CD44 cell surface markers do not identify cancer stem cells in primary human gastric tumors. J Cell Physiol. 2012;227:2686–93.CrossRef
67.
go back to reference Lau WM, Teng E, Chong HS, Lopez KA, Tay AY, Salto-Tellez M, et al. CD44v8-10 is a cancer-specific marker for gastric cancer stem cells. Cancer Res. 2014;74:2630–41.CrossRef Lau WM, Teng E, Chong HS, Lopez KA, Tay AY, Salto-Tellez M, et al. CD44v8-10 is a cancer-specific marker for gastric cancer stem cells. Cancer Res. 2014;74:2630–41.CrossRef
68.
go back to reference Nguyen PH, Giraud J, Chambonnier L, Dubus P, Wittkop L, Belleannee G, et al. Characterization of biomarkers of tumorigenic and chemoresistant cancer stem cells in human gastric carcinoma. Clin Cancer Res. 2017;23:1586–97.CrossRef Nguyen PH, Giraud J, Chambonnier L, Dubus P, Wittkop L, Belleannee G, et al. Characterization of biomarkers of tumorigenic and chemoresistant cancer stem cells in human gastric carcinoma. Clin Cancer Res. 2017;23:1586–97.CrossRef
69.
go back to reference Cristescu R, Lee J, Nebozhyn M, Kim KM, Ting JC, Wong SS, et al. Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat Med. 2015;21:449–56.CrossRef Cristescu R, Lee J, Nebozhyn M, Kim KM, Ting JC, Wong SS, et al. Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat Med. 2015;21:449–56.CrossRef
70.
go back to reference Wong SS, Kim KM, Ting JC, Yu K, Fu J, Liu S, et al. Genomic landscape and genetic heterogeneity in gastric adenocarcinoma revealed by whole-genome sequencing. Nat Commun. 2014;5:5477.CrossRef Wong SS, Kim KM, Ting JC, Yu K, Fu J, Liu S, et al. Genomic landscape and genetic heterogeneity in gastric adenocarcinoma revealed by whole-genome sequencing. Nat Commun. 2014;5:5477.CrossRef
71.
go back to reference Ge S, Xia X, Ding C, Zhen B, Zhou Q, Feng J, et al. A proteomic landscape of diffuse-type gastric cancer. Nat Commun. 2018;9:1012.CrossRef Ge S, Xia X, Ding C, Zhen B, Zhou Q, Feng J, et al. A proteomic landscape of diffuse-type gastric cancer. Nat Commun. 2018;9:1012.CrossRef
72.
go back to reference Klumpen HJ, Beijnen JH, Gurney H, Schellens JH. Inhibitors of mTOR. Oncologist. 2010;15:1262–9.CrossRef Klumpen HJ, Beijnen JH, Gurney H, Schellens JH. Inhibitors of mTOR. Oncologist. 2010;15:1262–9.CrossRef
73.
go back to reference Murayama T, Inokuchi M, Takagi Y, Yamada H, Kojima K, Kumagai J, et al. Relation between outcomes and localisation of p-mTOR expression in gastric cancer. Br J Cancer. 2009;100:782–8.CrossRef Murayama T, Inokuchi M, Takagi Y, Yamada H, Kojima K, Kumagai J, et al. Relation between outcomes and localisation of p-mTOR expression in gastric cancer. Br J Cancer. 2009;100:782–8.CrossRef
74.
go back to reference Carneiro F, Huntsman DG, Smyrk TC, Owen DA, Seruca R, Pharoah P, et al. Model of the early development of diffuse gastric cancer in E-cadherin mutation carriers and its implications for patient screening. J Pathol. 2004;203:681–7.CrossRef Carneiro F, Huntsman DG, Smyrk TC, Owen DA, Seruca R, Pharoah P, et al. Model of the early development of diffuse gastric cancer in E-cadherin mutation carriers and its implications for patient screening. J Pathol. 2004;203:681–7.CrossRef
75.
go back to reference Shimada S, Mimata A, Sekine M, Mogushi K, Akiyama Y, Fukamachi H, et al. Synergistic tumour suppressor activity of E-cadherin and p53 in a conditional mouse model for metastatic diffuse-type gastric cancer. Gut. 2012;61:344–53.CrossRef Shimada S, Mimata A, Sekine M, Mogushi K, Akiyama Y, Fukamachi H, et al. Synergistic tumour suppressor activity of E-cadherin and p53 in a conditional mouse model for metastatic diffuse-type gastric cancer. Gut. 2012;61:344–53.CrossRef
76.
go back to reference Miyazawa K, Iwaya K, Kuroda M, Harada M, Serizawa H, Koyanagi Y, et al. Nuclear accumulation of beta-catenin in intestinal-type gastric carcinoma: correlation with early tumor invasion. Virchows Arch. 2000;437:508–13.CrossRef Miyazawa K, Iwaya K, Kuroda M, Harada M, Serizawa H, Koyanagi Y, et al. Nuclear accumulation of beta-catenin in intestinal-type gastric carcinoma: correlation with early tumor invasion. Virchows Arch. 2000;437:508–13.CrossRef
77.
go back to reference Nakatsuru S, Yanagisawa A, Ichii S, Tahara E, Kato Y, Nakamura Y, et al. Somatic mutation of the APC gene in gastric cancer: frequent mutations in very well differentiated adenocarcinoma and signet-ring cell carcinoma. Hum Mol Genet. 1992;1:559–63.CrossRef Nakatsuru S, Yanagisawa A, Ichii S, Tahara E, Kato Y, Nakamura Y, et al. Somatic mutation of the APC gene in gastric cancer: frequent mutations in very well differentiated adenocarcinoma and signet-ring cell carcinoma. Hum Mol Genet. 1992;1:559–63.CrossRef
78.
go back to reference Sano T, Tsujino T, Yoshida K, Nakayama H, Haruma K, Ito H, et al. Frequent loss of heterozygosity on chromosomes 1q, 5q, and 17p in human gastric carcinomas. Cancer Res. 1991;51:2926–31. Sano T, Tsujino T, Yoshida K, Nakayama H, Haruma K, Ito H, et al. Frequent loss of heterozygosity on chromosomes 1q, 5q, and 17p in human gastric carcinomas. Cancer Res. 1991;51:2926–31.
79.
go back to reference Park WS, Oh RR, Park JY, Lee SH, Shin MS, Kim YS, et al. Frequent somatic mutations of the beta-catenin gene in intestinal-type gastric cancer. Cancer Res. 1999;59:4257–60. Park WS, Oh RR, Park JY, Lee SH, Shin MS, Kim YS, et al. Frequent somatic mutations of the beta-catenin gene in intestinal-type gastric cancer. Cancer Res. 1999;59:4257–60.
Metadata
Title
A subset of diffuse-type gastric cancer is susceptible to mTOR inhibitors and checkpoint inhibitors
Authors
Hiroshi Fukamachi
Seon-Kyu Kim
Jiwon Koh
Hye Seung Lee
Yasushi Sasaki
Kentaro Yamashita
Taketo Nishikawaji
Shu Shimada
Yoshimitsu Akiyama
Sun-ju Byeon
Dong-Hyuck Bae
Keisuke Okuno
Masatoshi Nakagawa
Toshiro Tanioka
Mikito Inokuchi
Hiroshi Kawachi
Kiichiro Tsuchiya
Kazuyuki Kojima
Takashi Tokino
Yoshinobu Eishi
Yong Sung Kim
Woo Ho Kim
Yasuhito Yuasa
Shinji Tanaka
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2019
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-019-1121-3

Other articles of this Issue 1/2019

Journal of Experimental & Clinical Cancer Research 1/2019 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