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

01-12-2021 | Metastasis | Research article

Mesothelin blockage by Amatuximab suppresses cell invasiveness, enhances gemcitabine sensitivity and regulates cancer cell stemness in mesothelin-positive pancreatic cancer cells

Authors: Fumihiko Matsuzawa, Hirofumi Kamachi, Tatsuzo Mizukami, Takahiro Einama, Futoshi Kawamata, Yuki Fujii, Moto Fukai, Nozomi Kobayashi, Yutaka Hatanaka, Akinobu Taketomi

Published in: BMC Cancer | Issue 1/2021

Login to get access

Abstract

Background

Mesothelin is a 40-kDa glycoprotein that is highly overexpressed in various types of cancers, however molecular mechanism of mesothelin has not been well-known. Amatuximab is a chimeric monoclonal IgG1/k antibody targeting mesothelin. We recently demonstrated that the combine therapy of Amatuximab and gemcitabine was effective for peritonitis of pancreatic cancer in mouse model.

Methods

We discover the role and potential mechanism of mesothelin blockage by Amatuximab in human pancreatic cells both expressing high or low level of mesothelin in vitro experiment and peritonitis mouse model of pancreatic cancer.

Results

Mesothelin blockage by Amatuximab lead to suppression of invasiveness and migration capacity in AsPC-1 and Capan-2 (high mesothelin expression) and reduce levels of pMET expression. The combination of Amatuximab and gemcitabine suppressed proliferation of AsPC-1 and Capan-2 more strongly than gemcitabine alone. These phenomena were not observed in Panc-1 and MIA Paca-2 (Mesothelin low expression). We previously demonstrated that Amatuximab reduced the peritoneal mass in mouse AsPC-1 peritonitis model and induced sherbet-like cancer cell aggregates, which were vanished by gemcitabine. In this study, we showed that the cancer stem cell related molecule such as ALDH1, CD44, c-MET, as well as proliferation related molecules, were suppressed in sherbet-like aggregates, but once sherbet-like aggregates attached to peritoneum, they expressed these molecules strongly without the morphological changes.

Conclusions

Our work suggested that Amatuximab inhibits the adhesion of cancer cells to peritoneum and suppresses the stemness and viability of those, that lead to enhance the sensitivity for gemcitabine.
Appendix
Available only for authorised users
Literature
1.
go back to reference Kleeff J, Korc M, Apte M, La Vecchia C, Johnson CD, Biankin AV, Neale RE, Tempero M, Tuveson DA, Hruban RH, et al. Pancreatic cancer. Nat Rev Dis Primers. 2016;2:16022.PubMedCrossRef Kleeff J, Korc M, Apte M, La Vecchia C, Johnson CD, Biankin AV, Neale RE, Tempero M, Tuveson DA, Hruban RH, et al. Pancreatic cancer. Nat Rev Dis Primers. 2016;2:16022.PubMedCrossRef
2.
go back to reference Tempero MA, Malafa MP, Al-Hawary M, Asbun H, Bain A, Behrman SW, Benson AB 3rd, Binder E, Cardin DB, Cha C, et al. Pancreatic adenocarcinoma, version 2.2017, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2017;15(8):1028–61.PubMedCrossRef Tempero MA, Malafa MP, Al-Hawary M, Asbun H, Bain A, Behrman SW, Benson AB 3rd, Binder E, Cardin DB, Cha C, et al. Pancreatic adenocarcinoma, version 2.2017, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2017;15(8):1028–61.PubMedCrossRef
3.
go back to reference Vauthey JN, Dixon E. AHPBA/SSO/SSAT consensus conference on Resectable and borderline Resectable pancreatic Cancer: rationale and overview of the conference. Ann Surg Oncol. 2009;16(7):1725–6.PubMedCrossRef Vauthey JN, Dixon E. AHPBA/SSO/SSAT consensus conference on Resectable and borderline Resectable pancreatic Cancer: rationale and overview of the conference. Ann Surg Oncol. 2009;16(7):1725–6.PubMedCrossRef
4.
go back to reference Hattangadi JA, Hong TS, Yeap BY, Mamon HJ. Results and patterns of failure in patients treated with adjuvant combined chemoradiation therapy for resected pancreatic adenocarcinoma. Cancer. 2009;115(16):3640–50.PubMedCrossRef Hattangadi JA, Hong TS, Yeap BY, Mamon HJ. Results and patterns of failure in patients treated with adjuvant combined chemoradiation therapy for resected pancreatic adenocarcinoma. Cancer. 2009;115(16):3640–50.PubMedCrossRef
5.
go back to reference Neoptolemos JP, Stocken DD, Bassi C, Ghaneh P, Cunningham D, Goldstein D, Padbury R, Moore MJ, Gallinger S, Mariette C, et al. Adjuvant chemotherapy with fluorouracil plus folinic acid vs gemcitabine following pancreatic cancer resection: a randomized controlled trial. JAMA. 2010;304(10):1073–81.PubMedCrossRef Neoptolemos JP, Stocken DD, Bassi C, Ghaneh P, Cunningham D, Goldstein D, Padbury R, Moore MJ, Gallinger S, Mariette C, et al. Adjuvant chemotherapy with fluorouracil plus folinic acid vs gemcitabine following pancreatic cancer resection: a randomized controlled trial. JAMA. 2010;304(10):1073–81.PubMedCrossRef
6.
go back to reference Uesaka K, Boku N, Fukutomi A, Okamura Y, Konishi M, Matsumoto I, Kaneoka Y, Shimizu Y, Nakamori S, Sakamoto H, et al. Adjuvant chemotherapy of S-1 versus gemcitabine for resected pancreatic cancer: a phase 3, open-label, randomised, non-inferiority trial (JASPAC 01). Lancet (London, England). 2016;388(10041):248–57.CrossRef Uesaka K, Boku N, Fukutomi A, Okamura Y, Konishi M, Matsumoto I, Kaneoka Y, Shimizu Y, Nakamori S, Sakamoto H, et al. Adjuvant chemotherapy of S-1 versus gemcitabine for resected pancreatic cancer: a phase 3, open-label, randomised, non-inferiority trial (JASPAC 01). Lancet (London, England). 2016;388(10041):248–57.CrossRef
7.
go back to reference Ducreux M, Boige V, Malka D. Treatment of advanced pancreatic cancer. Semin Oncol. 2007;34(2 Suppl 1):S25–30.PubMedCrossRef Ducreux M, Boige V, Malka D. Treatment of advanced pancreatic cancer. Semin Oncol. 2007;34(2 Suppl 1):S25–30.PubMedCrossRef
8.
go back to reference Chang K, Pastan I. Molecular cloning of mesothelin, a differentiation antigen present on mesothelium, mesotheliomas, and ovarian cancers. Proc Natl Acad Sci U S A. 1996;93(1):136–40.PubMedPubMedCentralCrossRef Chang K, Pastan I. Molecular cloning of mesothelin, a differentiation antigen present on mesothelium, mesotheliomas, and ovarian cancers. Proc Natl Acad Sci U S A. 1996;93(1):136–40.PubMedPubMedCentralCrossRef
9.
go back to reference Hassan R, Laszik ZG, Lerner M, Raffeld M, Postier R, Brackett D. Mesothelin is overexpressed in pancreaticobiliary adenocarcinomas but not in normal pancreas and chronic pancreatitis. Am J Clin Pathol. 2005;124(6):838–45.PubMedCrossRef Hassan R, Laszik ZG, Lerner M, Raffeld M, Postier R, Brackett D. Mesothelin is overexpressed in pancreaticobiliary adenocarcinomas but not in normal pancreas and chronic pancreatitis. Am J Clin Pathol. 2005;124(6):838–45.PubMedCrossRef
10.
go back to reference Li M, Bharadwaj U, Zhang R, Zhang S, Mu H, Fisher WE, Brunicardi FC, Chen C, Yao Q. Mesothelin is a malignant factor and therapeutic vaccine target for pancreatic cancer. Mol Cancer Ther. 2008;7(2):286–96.PubMedPubMedCentralCrossRef Li M, Bharadwaj U, Zhang R, Zhang S, Mu H, Fisher WE, Brunicardi FC, Chen C, Yao Q. Mesothelin is a malignant factor and therapeutic vaccine target for pancreatic cancer. Mol Cancer Ther. 2008;7(2):286–96.PubMedPubMedCentralCrossRef
11.
go back to reference He X, Wang L, Riedel H, Wang K, Yang Y, Dinu CZ, Rojanasakul Y. Mesothelin promotes epithelial-to-mesenchymal transition and tumorigenicity of human lung cancer and mesothelioma cells. Mol Cancer. 2017;16(1):63.PubMedPubMedCentralCrossRef He X, Wang L, Riedel H, Wang K, Yang Y, Dinu CZ, Rojanasakul Y. Mesothelin promotes epithelial-to-mesenchymal transition and tumorigenicity of human lung cancer and mesothelioma cells. Mol Cancer. 2017;16(1):63.PubMedPubMedCentralCrossRef
12.
go back to reference Einama T, Homma S, Kamachi H, Kawamata F, Takahashi K, Takahashi N, Taniguchi M, Kamiyama T, Furukawa H, Matsuno Y, et al. Luminal membrane expression of mesothelin is a prominent poor prognostic factor for gastric cancer. Br J Cancer. 2012;107(1):137–42.PubMedPubMedCentralCrossRef Einama T, Homma S, Kamachi H, Kawamata F, Takahashi K, Takahashi N, Taniguchi M, Kamiyama T, Furukawa H, Matsuno Y, et al. Luminal membrane expression of mesothelin is a prominent poor prognostic factor for gastric cancer. Br J Cancer. 2012;107(1):137–42.PubMedPubMedCentralCrossRef
13.
go back to reference Einama T, Kamachi H, Nishihara H, Homma S, Kanno H, Ishikawa M, Kawamata F, Konishi Y, Sato M, Tahara M, et al. Importance of luminal membrane mesothelin expression in intraductal papillary mucinous neoplasms. Oncol Lett. 2015;9(4):1583–9.PubMedPubMedCentralCrossRef Einama T, Kamachi H, Nishihara H, Homma S, Kanno H, Ishikawa M, Kawamata F, Konishi Y, Sato M, Tahara M, et al. Importance of luminal membrane mesothelin expression in intraductal papillary mucinous neoplasms. Oncol Lett. 2015;9(4):1583–9.PubMedPubMedCentralCrossRef
14.
go back to reference Einama T, Kamachi H, Nishihara H, Homma S, Kanno H, Takahashi K, Sasaki A, Tahara M, Okada K, Muraoka S, et al. Co-expression of mesothelin and CA125 correlates with unfavorable patient outcome in pancreatic ductal adenocarcinoma. Pancreas. 2011;40(8):1276–82.PubMedCrossRef Einama T, Kamachi H, Nishihara H, Homma S, Kanno H, Takahashi K, Sasaki A, Tahara M, Okada K, Muraoka S, et al. Co-expression of mesothelin and CA125 correlates with unfavorable patient outcome in pancreatic ductal adenocarcinoma. Pancreas. 2011;40(8):1276–82.PubMedCrossRef
15.
go back to reference Einama T, Kawamata F, Kamachi H, Nishihara H, Homma S, Matsuzawa F, Mizukami T, Konishi Y, Tahara M, Kamiyama T, et al. Clinical impacts of mesothelin expression in gastrointestinal carcinomas. World J Gastrointest Pathophysiol. 2016;7(2):218–22.PubMedPubMedCentralCrossRef Einama T, Kawamata F, Kamachi H, Nishihara H, Homma S, Matsuzawa F, Mizukami T, Konishi Y, Tahara M, Kamiyama T, et al. Clinical impacts of mesothelin expression in gastrointestinal carcinomas. World J Gastrointest Pathophysiol. 2016;7(2):218–22.PubMedPubMedCentralCrossRef
16.
go back to reference Kawamata F, Homma S, Kamachi H, Einama T, Kato Y, Tsuda M, Tanaka S, Maeda M, Kajino K, Hino O, et al. C-ERC/mesothelin provokes lymphatic invasion of colorectal adenocarcinoma. J Gastroenterol. 2014;49(1):81–92.PubMedCrossRef Kawamata F, Homma S, Kamachi H, Einama T, Kato Y, Tsuda M, Tanaka S, Maeda M, Kajino K, Hino O, et al. C-ERC/mesothelin provokes lymphatic invasion of colorectal adenocarcinoma. J Gastroenterol. 2014;49(1):81–92.PubMedCrossRef
17.
go back to reference Kawamata F, Kamachi H, Einama T, Homma S, Tahara M, Miyazaki M, Tanaka S, Kamiyama T, Nishihara H, Taketomi A, et al. Intracellular localization of mesothelin predicts patient prognosis of extrahepatic bile duct cancer. Int J Oncol. 2012;41(6):2109–18.PubMedCrossRef Kawamata F, Kamachi H, Einama T, Homma S, Tahara M, Miyazaki M, Tanaka S, Kamiyama T, Nishihara H, Taketomi A, et al. Intracellular localization of mesothelin predicts patient prognosis of extrahepatic bile duct cancer. Int J Oncol. 2012;41(6):2109–18.PubMedCrossRef
18.
go back to reference Morello A, Sadelain M, Adusumilli PS. Mesothelin-targeted CARs: driving T cells to solid tumors. Cancer Discov. 2016;6(2):133–46.PubMedCrossRef Morello A, Sadelain M, Adusumilli PS. Mesothelin-targeted CARs: driving T cells to solid tumors. Cancer Discov. 2016;6(2):133–46.PubMedCrossRef
19.
go back to reference Hassan R, Ebel W, Routhier EL, Patel R, Kline JB, Zhang J, Chao Q, Jacob S, Turchin H, Gibbs L, et al. Preclinical evaluation of MORAb-009, a chimeric antibody targeting tumor-associated mesothelin. Cancer Immun. 2007;7:20.PubMedPubMedCentral Hassan R, Ebel W, Routhier EL, Patel R, Kline JB, Zhang J, Chao Q, Jacob S, Turchin H, Gibbs L, et al. Preclinical evaluation of MORAb-009, a chimeric antibody targeting tumor-associated mesothelin. Cancer Immun. 2007;7:20.PubMedPubMedCentral
20.
go back to reference Fujisaka Y, Kurata T, Tanaka K, Kudo T, Okamoto K, Tsurutani J, Kaneda H, Okamoto I, Namiki M, Kitamura C, et al. Phase I study of amatuximab, a novel monoclonal antibody to mesothelin, in Japanese patients with advanced solid tumors. Investig New Drugs. 2015;33(2):380–8.CrossRef Fujisaka Y, Kurata T, Tanaka K, Kudo T, Okamoto K, Tsurutani J, Kaneda H, Okamoto I, Namiki M, Kitamura C, et al. Phase I study of amatuximab, a novel monoclonal antibody to mesothelin, in Japanese patients with advanced solid tumors. Investig New Drugs. 2015;33(2):380–8.CrossRef
21.
go back to reference Mizukami T, Kamachi H, Fujii Y, Matsuzawa F, Einama T, Kawamata F, Kobayashi N, Hatanaka Y, Taketomi A. The anti-mesothelin monoclonal antibody amatuximab enhances the anti-tumor effect of gemcitabine against mesothelin-high expressing pancreatic cancer cells in a peritoneal metastasis mouse model. Oncotarget. 2018;9(73):33844–52.PubMedPubMedCentralCrossRef Mizukami T, Kamachi H, Fujii Y, Matsuzawa F, Einama T, Kawamata F, Kobayashi N, Hatanaka Y, Taketomi A. The anti-mesothelin monoclonal antibody amatuximab enhances the anti-tumor effect of gemcitabine against mesothelin-high expressing pancreatic cancer cells in a peritoneal metastasis mouse model. Oncotarget. 2018;9(73):33844–52.PubMedPubMedCentralCrossRef
22.
go back to reference Awasthi N, Monahan S, Stefaniak A, Schwarz MA, Schwarz RE. Inhibition of the MEK/ERK pathway augments nab-paclitaxel-based chemotherapy effects in preclinical models of pancreatic cancer. Oncotarget. 2018;9(4):5274–86.PubMedCrossRef Awasthi N, Monahan S, Stefaniak A, Schwarz MA, Schwarz RE. Inhibition of the MEK/ERK pathway augments nab-paclitaxel-based chemotherapy effects in preclinical models of pancreatic cancer. Oncotarget. 2018;9(4):5274–86.PubMedCrossRef
24.
go back to reference Tang Z, Qian M, Ho M. The role of mesothelin in tumor progression and targeted therapy. Anti Cancer Agents Med Chem. 2013;13(2):276–80.CrossRef Tang Z, Qian M, Ho M. The role of mesothelin in tumor progression and targeted therapy. Anti Cancer Agents Med Chem. 2013;13(2):276–80.CrossRef
25.
go back to reference Scholz A, Heinze S, Detjen KM, Peters M, Welzel M, Hauff P, Schirner M, Wiedenmann B, Rosewicz S. Activated signal transducer and activator of transcription 3 (STAT3) supports the malignant phenotype of human pancreatic cancer. Gastroenterology. 2003;125(3):891–905.PubMedCrossRef Scholz A, Heinze S, Detjen KM, Peters M, Welzel M, Hauff P, Schirner M, Wiedenmann B, Rosewicz S. Activated signal transducer and activator of transcription 3 (STAT3) supports the malignant phenotype of human pancreatic cancer. Gastroenterology. 2003;125(3):891–905.PubMedCrossRef
26.
go back to reference DeArmond D, Brattain MG, Jessup JM, Kreisberg J, Malik S, Zhao S, Freeman JW. Autocrine-mediated ErbB-2 kinase activation of STAT3 is required for growth factor independence of pancreatic cancer cell lines. Oncogene. 2003;22(49):7781–95.PubMedCrossRef DeArmond D, Brattain MG, Jessup JM, Kreisberg J, Malik S, Zhao S, Freeman JW. Autocrine-mediated ErbB-2 kinase activation of STAT3 is required for growth factor independence of pancreatic cancer cell lines. Oncogene. 2003;22(49):7781–95.PubMedCrossRef
27.
go back to reference Toyonaga T, Nakano K, Nagano M, Zhao G, Yamaguchi K, Kuroki S, Eguchi T, Chijiiwa K, Tsuneyoshi M, Tanaka M. Blockade of constitutively activated Janus kinase/signal transducer and activator of transcription-3 pathway inhibits growth of human pancreatic cancer. Cancer Lett. 2003;201(1):107–16.PubMedCrossRef Toyonaga T, Nakano K, Nagano M, Zhao G, Yamaguchi K, Kuroki S, Eguchi T, Chijiiwa K, Tsuneyoshi M, Tanaka M. Blockade of constitutively activated Janus kinase/signal transducer and activator of transcription-3 pathway inhibits growth of human pancreatic cancer. Cancer Lett. 2003;201(1):107–16.PubMedCrossRef
28.
go back to reference Greten FR, Weber CK, Greten TF, Schneider G, Wagner M, Adler G, Schmid RM. Stat3 and NF-kappaB activation prevents apoptosis in pancreatic carcinogenesis. Gastroenterology. 2002;123(6):2052–63.PubMedCrossRef Greten FR, Weber CK, Greten TF, Schneider G, Wagner M, Adler G, Schmid RM. Stat3 and NF-kappaB activation prevents apoptosis in pancreatic carcinogenesis. Gastroenterology. 2002;123(6):2052–63.PubMedCrossRef
29.
go back to reference Bharadwaj U, Li M, Chen C, Yao Q. Mesothelin-induced pancreatic cancer cell proliferation involves alteration of cyclin E via activation of signal transducer and activator of transcription protein 3. Mol Cancer Res. 2008;6(11):1755–65.PubMedPubMedCentralCrossRef Bharadwaj U, Li M, Chen C, Yao Q. Mesothelin-induced pancreatic cancer cell proliferation involves alteration of cyclin E via activation of signal transducer and activator of transcription protein 3. Mol Cancer Res. 2008;6(11):1755–65.PubMedPubMedCentralCrossRef
30.
go back to reference Uehara N, Matsuoka Y, Tsubura A. Mesothelin promotes anchorage-independent growth and prevents anoikis via extracellular signal-regulated kinase signaling pathway in human breast cancer cells. Mol Cancer Res. 2008;6(2):186–93.PubMedCrossRef Uehara N, Matsuoka Y, Tsubura A. Mesothelin promotes anchorage-independent growth and prevents anoikis via extracellular signal-regulated kinase signaling pathway in human breast cancer cells. Mol Cancer Res. 2008;6(2):186–93.PubMedCrossRef
31.
go back to reference Chang MC, Chen CA, Hsieh CY, Lee CN, Su YN, Hu YH, Cheng WF. Mesothelin inhibits paclitaxel-induced apoptosis through the PI3K pathway. Biochem J. 2009;424(3):449–58.PubMedCrossRef Chang MC, Chen CA, Hsieh CY, Lee CN, Su YN, Hu YH, Cheng WF. Mesothelin inhibits paclitaxel-induced apoptosis through the PI3K pathway. Biochem J. 2009;424(3):449–58.PubMedCrossRef
32.
go back to reference Tan X, Egami H, Abe M, Nozawa F, Hirota M, Ogawa M. Involvement of MMP-7 in invasion of pancreatic cancer cells through activation of the EGFR mediated MEK-ERK signal transduction pathway. J Clin Pathol. 2005;58(12):1242–8.PubMedPubMedCentralCrossRef Tan X, Egami H, Abe M, Nozawa F, Hirota M, Ogawa M. Involvement of MMP-7 in invasion of pancreatic cancer cells through activation of the EGFR mediated MEK-ERK signal transduction pathway. J Clin Pathol. 2005;58(12):1242–8.PubMedPubMedCentralCrossRef
33.
go back to reference Zheng C, Jia W, Tang Y, Zhao H, Jiang Y, Sun S. Mesothelin regulates growth and apoptosis in pancreatic cancer cells through p53-dependent and -independent signal pathway. J Exp Clin Cancer Res. 2012;31(1):84.PubMedPubMedCentralCrossRef Zheng C, Jia W, Tang Y, Zhao H, Jiang Y, Sun S. Mesothelin regulates growth and apoptosis in pancreatic cancer cells through p53-dependent and -independent signal pathway. J Exp Clin Cancer Res. 2012;31(1):84.PubMedPubMedCentralCrossRef
34.
go back to reference Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414(6859):105–11.PubMedCrossRef Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414(6859):105–11.PubMedCrossRef
35.
go back to reference Dean M, Fojo T, Bates S. Tumour stem cells and drug resistance. Nat Rev Cancer. 2005;5(4):275–84.PubMedCrossRef Dean M, Fojo T, Bates S. Tumour stem cells and drug resistance. Nat Rev Cancer. 2005;5(4):275–84.PubMedCrossRef
37.
go back to reference Ouyang G, Wang Z, Fang X, Liu J, Yang CJ. Molecular signaling of the epithelial to mesenchymal transition in generating and maintaining cancer stem cells. Cell Mol Life Sci. 2010;67(15):2605–18.PubMedCrossRef Ouyang G, Wang Z, Fang X, Liu J, Yang CJ. Molecular signaling of the epithelial to mesenchymal transition in generating and maintaining cancer stem cells. Cell Mol Life Sci. 2010;67(15):2605–18.PubMedCrossRef
38.
go back to reference Awad O, Yustein JT, Shah P, Gul N, Katuri V, O'Neill A, Kong Y, Brown ML, Toretsky JA, Loeb DM. High ALDH activity identifies chemotherapy-resistant Ewing's sarcoma stem cells that retain sensitivity to EWS-FLI1 inhibition. PLoS One. 2010;5(11):e13r943.CrossRef Awad O, Yustein JT, Shah P, Gul N, Katuri V, O'Neill A, Kong Y, Brown ML, Toretsky JA, Loeb DM. High ALDH activity identifies chemotherapy-resistant Ewing's sarcoma stem cells that retain sensitivity to EWS-FLI1 inhibition. PLoS One. 2010;5(11):e13r943.CrossRef
39.
go back to reference Mueller MT, Hermann PC, Witthauer J, Rubio-Viqueira B, Leicht SF, Huber S, Ellwart JW, Mustafa M, Bartenstein P, D'Haese JG, et al. Combined targeted treatment to eliminate tumorigenic cancer stem cells in human pancreatic cancer. Gastroenterology. 2009;137(3):1102–13.PubMedCrossRef Mueller MT, Hermann PC, Witthauer J, Rubio-Viqueira B, Leicht SF, Huber S, Ellwart JW, Mustafa M, Bartenstein P, D'Haese JG, et al. Combined targeted treatment to eliminate tumorigenic cancer stem cells in human pancreatic cancer. Gastroenterology. 2009;137(3):1102–13.PubMedCrossRef
40.
go back to reference Dylla SJ, Beviglia L, Park IK, Chartier C, Raval J, Ngan L, Pickell K, Aguilar J, Lazetic S, Smith-Berdan S, et al. Colorectal cancer stem cells are enriched in xenogeneic tumors following chemotherapy. PLoS One. 2008;3(6):e2428.PubMedPubMedCentralCrossRef Dylla SJ, Beviglia L, Park IK, Chartier C, Raval J, Ngan L, Pickell K, Aguilar J, Lazetic S, Smith-Berdan S, et al. Colorectal cancer stem cells are enriched in xenogeneic tumors following chemotherapy. PLoS One. 2008;3(6):e2428.PubMedPubMedCentralCrossRef
42.
go back to reference Eramo A, Lotti F, Sette G, Pilozzi E, Biffoni M, Di Virgilio A, Conticello C, Ruco L, Peschle C, De Maria R. Identification and expansion of the tumorigenic lung cancer stem cell population. Cell Death Differ. 2008;15(3):504–14.PubMedCrossRef Eramo A, Lotti F, Sette G, Pilozzi E, Biffoni M, Di Virgilio A, Conticello C, Ruco L, Peschle C, De Maria R. Identification and expansion of the tumorigenic lung cancer stem cell population. Cell Death Differ. 2008;15(3):504–14.PubMedCrossRef
43.
go back to reference Michieli P, Mazzone M, Basilico C, Cavassa S, Sottile A, Naldini L, Comoglio PM. Targeting the tumor and its microenvironment by a dual-function decoy met receptor. Cancer Cell. 2004;6(1):61–73.PubMedCrossRef Michieli P, Mazzone M, Basilico C, Cavassa S, Sottile A, Naldini L, Comoglio PM. Targeting the tumor and its microenvironment by a dual-function decoy met receptor. Cancer Cell. 2004;6(1):61–73.PubMedCrossRef
44.
go back to reference Gherardi E, Birchmeier W, Birchmeier C, Vande Woude G. Targeting MET in cancer: rationale and progress. Nat Rev Cancer. 2012;12(2):89–103.PubMedCrossRef Gherardi E, Birchmeier W, Birchmeier C, Vande Woude G. Targeting MET in cancer: rationale and progress. Nat Rev Cancer. 2012;12(2):89–103.PubMedCrossRef
45.
go back to reference Marjanovic ND, Weinberg RA, Chaffer CL. Cell plasticity and heterogeneity in cancer. Clin Chem. 2013;59(1):168–79.PubMedCrossRef Marjanovic ND, Weinberg RA, Chaffer CL. Cell plasticity and heterogeneity in cancer. Clin Chem. 2013;59(1):168–79.PubMedCrossRef
46.
go back to reference Stankevicius V, Kunigenas L, Stankunas E, Kuodyte K, Strainiene E, Cicenas J, Samalavicius NE, Suziedelis K. The expression of cancer stem cell markers in human colorectal carcinoma cells in a microenvironment dependent manner. Biochem Biophys Res Commun. 2017;484(4):726–33.PubMedCrossRef Stankevicius V, Kunigenas L, Stankunas E, Kuodyte K, Strainiene E, Cicenas J, Samalavicius NE, Suziedelis K. The expression of cancer stem cell markers in human colorectal carcinoma cells in a microenvironment dependent manner. Biochem Biophys Res Commun. 2017;484(4):726–33.PubMedCrossRef
47.
go back to reference Shishido A, Mori S, Yokoyama Y, Hamada Y, Minami K, Qian Y, Wang J, Hirose H, Wu X, Kawaguchi N, et al. Mesothelial cells facilitate cancer stem-like properties in spheroids of ovarian cancer cells. Oncol Rep. 2018;40(4):2105–14.PubMed Shishido A, Mori S, Yokoyama Y, Hamada Y, Minami K, Qian Y, Wang J, Hirose H, Wu X, Kawaguchi N, et al. Mesothelial cells facilitate cancer stem-like properties in spheroids of ovarian cancer cells. Oncol Rep. 2018;40(4):2105–14.PubMed
48.
go back to reference Wang Y, Cheon DJ, Lu Z, Cunningham SL, Chen CM, Luo RZ, Xing D, Orsulic S, Bast RC Jr, Behringer RR. MUC16 expression during embryogenesis, in adult tissues, and ovarian cancer in the mouse. Differentiation. 2008;76(10):1081–92.PubMedPubMedCentralCrossRef Wang Y, Cheon DJ, Lu Z, Cunningham SL, Chen CM, Luo RZ, Xing D, Orsulic S, Bast RC Jr, Behringer RR. MUC16 expression during embryogenesis, in adult tissues, and ovarian cancer in the mouse. Differentiation. 2008;76(10):1081–92.PubMedPubMedCentralCrossRef
49.
go back to reference Hassan R, Schweizer C, Lu KF, Schuler B, Remaley AT, Weil SC, Pastan I. Inhibition of mesothelin-CA-125 interaction in patients with mesothelioma by the anti-mesothelin monoclonal antibody MORAb-009: Implications for cancer therapy. Lung Cancer. 2010;68(3):455–9.PubMedCrossRef Hassan R, Schweizer C, Lu KF, Schuler B, Remaley AT, Weil SC, Pastan I. Inhibition of mesothelin-CA-125 interaction in patients with mesothelioma by the anti-mesothelin monoclonal antibody MORAb-009: Implications for cancer therapy. Lung Cancer. 2010;68(3):455–9.PubMedCrossRef
Metadata
Title
Mesothelin blockage by Amatuximab suppresses cell invasiveness, enhances gemcitabine sensitivity and regulates cancer cell stemness in mesothelin-positive pancreatic cancer cells
Authors
Fumihiko Matsuzawa
Hirofumi Kamachi
Tatsuzo Mizukami
Takahiro Einama
Futoshi Kawamata
Yuki Fujii
Moto Fukai
Nozomi Kobayashi
Yutaka Hatanaka
Akinobu Taketomi
Publication date
01-12-2021
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2021
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-020-07722-3

Other articles of this Issue 1/2021

BMC Cancer 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