Skip to main content
Top
Published in: Cellular Oncology 6/2016

01-12-2016 | Original Paper

NFκB activation demarcates a subset of hepatocellular carcinoma patients for targeted therapy

Authors: Vignesh Ramesh, Karthikeyan Selvarasu, Jaishree Pandian, Soundarajan Myilsamy, Chidambaranathan Shanmugasundaram, Kumaresan Ganesan

Published in: Cellular Oncology | Issue 6/2016

Login to get access

Abstract

Background

Hepatocellular carcinoma (HCC) is the fifth most common cancer and the third leading cause of cancer death worldwide. It is a heterogeneous disorder and >80 % of the tumors develop in patients with liver cirrhosis, resulting from chronic inflammation and/or fibrosis. Here, we set out to identify novel targets for HCC therapy and to define a subgroup of patients that might benefit most from it.

Methods

Cellular pathway activation profiling of 45 transcription factors in a HCC-derived cell line (HEP3B), in vitro analysis of NFκB reporter activity in additional HCC-derived cell lines and pathway-focused integrative analyses of publicly available primary HCC-derived expression profiling data (GSE6764, GSE9843, E-TABM-36 and E-TABM-292) were employed to reveal a role of NFκB in HCC development. In order to identify potential targeting agents, a luciferase-based NFκB reporter screening assay was established in HEP3B cells. After screening of a drug library through this assay, a potent NFκB pathway inhibitor was identified and characterized using an array of additional in vitro assays.

Results

Using cellular pathway activation profiling, we found a high activation of NFκB-mediated signaling in HCC-derived cell lines and in primary HCC tumors. Through NFκB inhibitor screening we observed a highly efficacious NFκB pathway inhibitory potential of ornithogalum in HCC-derived HEP3B cells. Although its active component still remains to be defined, ornithogalum has been found to inhibit endoplasmic reticulum (ER) and oxidative stress responses. ER stress, oxidative stress and NFκB signaling were found to be enhanced in a subset of HCCs, as well as in (precancerous) liver cirrhosis tissues.

Conclusion

From our data we conclude that NFκB signaling is activated in precancerous cirrhosis tissues and in a subset of HCCs. We found that ornithogalum exhibits NFκB targeting and stress relieving activities. NFκB inhibitors, including the active component of ornithogalum, may serve as putative preventive and targeted therapeutic agents for at least a subset of HCCs in which the NFκB pathway is activated. These latter notions require further investigation in a translational context.
Appendix
Available only for authorised users
Literature
1.
go back to reference J. Ferlay, H. R. Shin, F. Bray, D. Forman, C. Mathers, D. M. Parkin, Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int. J. Cancer 127, 2893–2917 (2010)CrossRefPubMed J. Ferlay, H. R. Shin, F. Bray, D. Forman, C. Mathers, D. M. Parkin, Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int. J. Cancer 127, 2893–2917 (2010)CrossRefPubMed
2.
go back to reference L. Zender, M. S. Spector, W. Xue, P. Flemming, C. Cordon-Cardo, J. Silke, S. T. Fan, J. M. Luk, M. Wigler, G. J. Hannon, D. Mu, R. Lucito, S. Powers, S. W. Lowe, Identification and validation of oncogenes in liver cancer using an integrative oncogenomic approach. Cell 125, 1253–1267 (2006)CrossRefPubMedPubMedCentral L. Zender, M. S. Spector, W. Xue, P. Flemming, C. Cordon-Cardo, J. Silke, S. T. Fan, J. M. Luk, M. Wigler, G. J. Hannon, D. Mu, R. Lucito, S. Powers, S. W. Lowe, Identification and validation of oncogenes in liver cancer using an integrative oncogenomic approach. Cell 125, 1253–1267 (2006)CrossRefPubMedPubMedCentral
3.
4.
go back to reference R. Saeki, H. Nagai, S. Kaneko, M. Unoura, N. Yamanaka, E. Okamoto, K. Kobayashi, K. Matsubara, Intratumoral genomic heterogeneity in human hepatocellular carcinoma detected by restriction landmark genomic scanning. J. Hepatol. 33, 99–105 (2000)CrossRefPubMed R. Saeki, H. Nagai, S. Kaneko, M. Unoura, N. Yamanaka, E. Okamoto, K. Kobayashi, K. Matsubara, Intratumoral genomic heterogeneity in human hepatocellular carcinoma detected by restriction landmark genomic scanning. J. Hepatol. 33, 99–105 (2000)CrossRefPubMed
6.
go back to reference S. Fatima, N. P. Lee, J. M. Luk, Dickkopfs and Wnt/β-catenin signalling in liver cancer. World. J. Clin. Oncol. 2, 311–325 (2011) S. Fatima, N. P. Lee, J. M. Luk, Dickkopfs and Wnt/β-catenin signalling in liver cancer. World. J. Clin. Oncol. 2, 311–325 (2011)
8.
go back to reference Y. Totoki, K. Tatsuno, S. Yamamoto, Y. Arai, F. Hosoda, S. Ishikawa, S. Tsutsumi, K. Sonoda, H. Totsuka, T. Shirakihara, H. Sakamoto, L. Wang, H. Ojimo, K. Shimada, T. Kosuge, T. Okusaka, K. Kato, J. Kusuda, T. Yoshida, H. Aburatani, T. Shibata, High-resolution characterization of a hepatocellular carcinoma genome. Nat. Genet. 43, 464–469 (2011)CrossRefPubMed Y. Totoki, K. Tatsuno, S. Yamamoto, Y. Arai, F. Hosoda, S. Ishikawa, S. Tsutsumi, K. Sonoda, H. Totsuka, T. Shirakihara, H. Sakamoto, L. Wang, H. Ojimo, K. Shimada, T. Kosuge, T. Okusaka, K. Kato, J. Kusuda, T. Yoshida, H. Aburatani, T. Shibata, High-resolution characterization of a hepatocellular carcinoma genome. Nat. Genet. 43, 464–469 (2011)CrossRefPubMed
9.
go back to reference S. I. Grivennikov, M. Karin, Inflammatory cytokines in cancer: tumour necrosis factor and interleukin 6 take the stage. Ann. Rheum. Dis. 70, i104–i108 (2011)CrossRefPubMed S. I. Grivennikov, M. Karin, Inflammatory cytokines in cancer: tumour necrosis factor and interleukin 6 take the stage. Ann. Rheum. Dis. 70, i104–i108 (2011)CrossRefPubMed
10.
go back to reference G. Fattovich, T. Stroffolini, I. Zagni, F. Donato, Hepatocellular carcinoma in cirrhosis: incidence and risk factors. Gastroenterology 127, S35–S50 (2004)CrossRefPubMed G. Fattovich, T. Stroffolini, I. Zagni, F. Donato, Hepatocellular carcinoma in cirrhosis: incidence and risk factors. Gastroenterology 127, S35–S50 (2004)CrossRefPubMed
11.
12.
go back to reference K. Vazquez-Santillan, J. Melendez-Zajgla, L. Jimenez-Hernandez, G. Martinez-Ruiz, V. Maldonado, NF-κB signaling in cancer stem cells: a promising therapeutic target? Cell. Oncol. 38, 327–339 (2015)CrossRef K. Vazquez-Santillan, J. Melendez-Zajgla, L. Jimenez-Hernandez, G. Martinez-Ruiz, V. Maldonado, NF-κB signaling in cancer stem cells: a promising therapeutic target? Cell. Oncol. 38, 327–339 (2015)CrossRef
13.
go back to reference B. B. Aggarwal, A. Bhardwaj, R. S. Aggarwal, N. P. Seeram, S. Shishodia, Y. Takada, Role of resveratrol in prevention and therapy of cancer: preclinical and clinical studies. Anticancer Res. 24, 2783–2840 (2004)PubMed B. B. Aggarwal, A. Bhardwaj, R. S. Aggarwal, N. P. Seeram, S. Shishodia, Y. Takada, Role of resveratrol in prevention and therapy of cancer: preclinical and clinical studies. Anticancer Res. 24, 2783–2840 (2004)PubMed
14.
go back to reference N. Chainani-Wu, Safety and anti-inflammatory activity of curcumin: a component of tumeric (Curcuma longa. J. Altern. Complement. Med. 9, 161–168 (2003)CrossRefPubMed N. Chainani-Wu, Safety and anti-inflammatory activity of curcumin: a component of tumeric (Curcuma longa. J. Altern. Complement. Med. 9, 161–168 (2003)CrossRefPubMed
15.
go back to reference L. Fredriksson, B. Herpers, G. Benedetti, Q. Matadin, J. C. Puigvert, H. de Bont, S. Dragovic, N. P. Vermeulen, J. N. Commandeur, E. Danen, M. de Graauw, B. van de Water, Diclofenac inhibits tumor necrosis factor-α-induced nuclear factor-κB activation causing synergistic hepatocyte apoptosis. Hepatology 53, 2027–2041 (2011)CrossRefPubMed L. Fredriksson, B. Herpers, G. Benedetti, Q. Matadin, J. C. Puigvert, H. de Bont, S. Dragovic, N. P. Vermeulen, J. N. Commandeur, E. Danen, M. de Graauw, B. van de Water, Diclofenac inhibits tumor necrosis factor-α-induced nuclear factor-κB activation causing synergistic hepatocyte apoptosis. Hepatology 53, 2027–2041 (2011)CrossRefPubMed
16.
go back to reference S. C. Miller, R. Huang, S. Sakamuru, S. J. Shukla, M. S. Attene-Ramos, P. Shinn, D. Van Leer, W. Leister, C. P. Austin, M. Xia, Identification of known drugs that act as inhibitors of NF-kappaB signaling and their mechanism of action. Biochem. Pharmacol. 79, 1272–1280 (2010)CrossRefPubMedPubMedCentral S. C. Miller, R. Huang, S. Sakamuru, S. J. Shukla, M. S. Attene-Ramos, P. Shinn, D. Van Leer, W. Leister, C. P. Austin, M. Xia, Identification of known drugs that act as inhibitors of NF-kappaB signaling and their mechanism of action. Biochem. Pharmacol. 79, 1272–1280 (2010)CrossRefPubMedPubMedCentral
17.
go back to reference S. Shishodia, D. Koul, B. B. Aggarwal, Cyclooxygenase (COX)-2 inhibitor celecoxib abrogates TNF-induced NF-kappa B activation through inhibition of activation of I kappa B alpha kinase and Akt in human non-small cell lung carcinoma: correlation with suppression of COX-2 synthesis. J. Immunol. 173, 2011–2022 (2004)CrossRefPubMed S. Shishodia, D. Koul, B. B. Aggarwal, Cyclooxygenase (COX)-2 inhibitor celecoxib abrogates TNF-induced NF-kappa B activation through inhibition of activation of I kappa B alpha kinase and Akt in human non-small cell lung carcinoma: correlation with suppression of COX-2 synthesis. J. Immunol. 173, 2011–2022 (2004)CrossRefPubMed
18.
go back to reference X. Yao, J. Huang, H. Zhong, N. Shen, R. Faggioni, M. Fung, Y. Yao, Targeting interleukin-6 in inflammatory autoimmune diseases and cancers. Pharmacol. Ther. 141, 125–139 (2014)CrossRefPubMed X. Yao, J. Huang, H. Zhong, N. Shen, R. Faggioni, M. Fung, Y. Yao, Targeting interleukin-6 in inflammatory autoimmune diseases and cancers. Pharmacol. Ther. 141, 125–139 (2014)CrossRefPubMed
19.
go back to reference S. W. Tas, M. J. Vervoordeldonk, P. P. Tak, Gene therapy targeting nuclear factor-κB: towards clinical application in inflammatory diseases and cancer. Curr Gene Ther 9, 160–170 (2009)CrossRefPubMedPubMedCentral S. W. Tas, M. J. Vervoordeldonk, P. P. Tak, Gene therapy targeting nuclear factor-κB: towards clinical application in inflammatory diseases and cancer. Curr Gene Ther 9, 160–170 (2009)CrossRefPubMedPubMedCentral
20.
21.
go back to reference P. N. Verma, I. Vaid, Encyclopaedia of Homeopathic Pharmacopoeia, Vol. IB (Jain Publishers, New Delhi, India, 2002), p. 1864 P. N. Verma, I. Vaid, Encyclopaedia of Homeopathic Pharmacopoeia, Vol. IB (Jain Publishers, New Delhi, India, 2002), p. 1864
22.
go back to reference B. W. Dyer, F. A. Ferrer, D. K. Klinedinst, R. Rodriguez, A noncommercial dual luciferase enzyme assay system for reporter gene analysis. Anal. Biochem. 282, 158–161 (2000)CrossRefPubMed B. W. Dyer, F. A. Ferrer, D. K. Klinedinst, R. Rodriguez, A noncommercial dual luciferase enzyme assay system for reporter gene analysis. Anal. Biochem. 282, 158–161 (2000)CrossRefPubMed
23.
go back to reference T. Mosmann, Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J. Immunol. Methods 65, 55–63 (1983)CrossRefPubMed T. Mosmann, Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J. Immunol. Methods 65, 55–63 (1983)CrossRefPubMed
24.
go back to reference 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)CrossRefPubMedPubMedCentral 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)CrossRefPubMedPubMedCentral
25.
go back to reference J. Lamb, E. D. Crawford, D. Peck, J. W. Modell, I. C. Blat, M. J. Wrobel, J. Lerner, J. P. Brunet, A. Subramanian, K. N. Ross, M. Reich, H. Hieronymus, G. Wei, S. A. Armstrong, S. J. Haggarty, P. A. Clemons, R. Wei, S. A. Carr, E. S. Lander, T. R. Golub, The connectivity map: using gene-expression signatures to connect small molecules, genes, and disease. Science 313, 1929–1935 (2006)CrossRefPubMed J. Lamb, E. D. Crawford, D. Peck, J. W. Modell, I. C. Blat, M. J. Wrobel, J. Lerner, J. P. Brunet, A. Subramanian, K. N. Ross, M. Reich, H. Hieronymus, G. Wei, S. A. Armstrong, S. J. Haggarty, P. A. Clemons, R. Wei, S. A. Carr, E. S. Lander, T. R. Golub, The connectivity map: using gene-expression signatures to connect small molecules, genes, and disease. Science 313, 1929–1935 (2006)CrossRefPubMed
26.
go back to reference C. Li, W. H. Wong, Model-based analysis of oligonucleotide arrays: expression index computation and outlier detection. Proc. Natl. Acad. Sci. U. S. A. 98, 31–36 (2001)CrossRefPubMed C. Li, W. H. Wong, Model-based analysis of oligonucleotide arrays: expression index computation and outlier detection. Proc. Natl. Acad. Sci. U. S. A. 98, 31–36 (2001)CrossRefPubMed
27.
go back to reference J. Boelens, S. Lust, F. Offner, M. E. Bracke, B. W. Vanhoecke, The endoplasmic reticulum: a target for new anticancer drugs. In Vivo 21, 215–226 (2007)PubMed J. Boelens, S. Lust, F. Offner, M. E. Bracke, B. W. Vanhoecke, The endoplasmic reticulum: a target for new anticancer drugs. In Vivo 21, 215–226 (2007)PubMed
28.
go back to reference A. Shibata, T. Nagaya, T. Imai, H. Funahashi, A. Nakao, H. Seo, Inhibition of NF-κB activity decreases the VEGF mRNA expression in MDA-MB-231 breast cancer cells. Breast Cancer Res. Treat. 73, 237–243 (2002)CrossRefPubMed A. Shibata, T. Nagaya, T. Imai, H. Funahashi, A. Nakao, H. Seo, Inhibition of NF-κB activity decreases the VEGF mRNA expression in MDA-MB-231 breast cancer cells. Breast Cancer Res. Treat. 73, 237–243 (2002)CrossRefPubMed
30.
go back to reference A. Geurts van Kessel, The cancer genome: from structure to function. Cell. Oncol. 37, 155–165 (2014)CrossRef A. Geurts van Kessel, The cancer genome: from structure to function. Cell. Oncol. 37, 155–165 (2014)CrossRef
31.
go back to reference F. Colotta, P. Allavena, A. Sica, C. Garlanda, A. Mantovani, Cancer-related inflammation, the seventh hallmark of cancer: links to genetic instability. Carcinogenesis 30, 1073–1081 (2009)CrossRefPubMed F. Colotta, P. Allavena, A. Sica, C. Garlanda, A. Mantovani, Cancer-related inflammation, the seventh hallmark of cancer: links to genetic instability. Carcinogenesis 30, 1073–1081 (2009)CrossRefPubMed
32.
go back to reference S. Lüth, J. Schrader, S. Zander, A. Carambia, J. Buchkremer, S. Huber, K. Reifenberg, K. Yamamura, P. Schirmacher, A. W. Lohse, J. Herkel, Chronic inflammatory IFN-γ signaling suppresses hepatocarcinogenesis in mice by sensitizing hepatocytes for apoptosis. Cancer Res. 71, 3763–3771 (2011)CrossRefPubMed S. Lüth, J. Schrader, S. Zander, A. Carambia, J. Buchkremer, S. Huber, K. Reifenberg, K. Yamamura, P. Schirmacher, A. W. Lohse, J. Herkel, Chronic inflammatory IFN-γ signaling suppresses hepatocarcinogenesis in mice by sensitizing hepatocytes for apoptosis. Cancer Res. 71, 3763–3771 (2011)CrossRefPubMed
33.
go back to reference J. F. Rossi, S. Negrier, N. D. James, I. Kocak, R. Hawkins, H. Davis, U. Prabhakar, X. Qin, P. Mulders, B. Berns, A phase I/II study of siltuximab (CNTO 328), an anti-interleukin-6 monoclonal antibody, in metastatic renal cell cancer. Br. J. Cancer 103, 1154–1162 (2010)CrossRefPubMedPubMedCentral J. F. Rossi, S. Negrier, N. D. James, I. Kocak, R. Hawkins, H. Davis, U. Prabhakar, X. Qin, P. Mulders, B. Berns, A phase I/II study of siltuximab (CNTO 328), an anti-interleukin-6 monoclonal antibody, in metastatic renal cell cancer. Br. J. Cancer 103, 1154–1162 (2010)CrossRefPubMedPubMedCentral
34.
go back to reference F. Wang, P. Arun, J. Friedman, Z. Chen, C. Van Waes, Current and potential inflammation targeted therapies in head and neck cancer. Curr. Opin. Pharmacol. 9, 389–395 (2009)CrossRefPubMedPubMedCentral F. Wang, P. Arun, J. Friedman, Z. Chen, C. Van Waes, Current and potential inflammation targeted therapies in head and neck cancer. Curr. Opin. Pharmacol. 9, 389–395 (2009)CrossRefPubMedPubMedCentral
35.
go back to reference S. Maeda, H. Kamata, J. L. Luo, H. Leffert, M. Karin, IKKβ couples hepatocyte death to cytokine-driven compensatory proliferation that promotes chemical hepatocarcinogenesis. Cell 121, 977–990 (2005)CrossRefPubMed S. Maeda, H. Kamata, J. L. Luo, H. Leffert, M. Karin, IKKβ couples hepatocyte death to cytokine-driven compensatory proliferation that promotes chemical hepatocarcinogenesis. Cell 121, 977–990 (2005)CrossRefPubMed
36.
go back to reference P. J. Wysocki, Targeted therapy of hepatocellular cancer. Expert Opin Investig Drugs 19, 265–274 (2010)CrossRefPubMed P. J. Wysocki, Targeted therapy of hepatocellular cancer. Expert Opin Investig Drugs 19, 265–274 (2010)CrossRefPubMed
37.
go back to reference W. Yeo, T. S. Mok, B. Zee, T. W. Leung, P. B. Lai, W. Y. Lau, J. Koh, F. K. Mo, S. C. Yu, A. T. Chan, P. Hui, B. Ma, K. C. Lam, W. M. Ho, H. T. Wong, A. Tang, P. J. Johnson, A randomized phase III study of doxorubicin versus cisplatin/interferon α-2b/doxorubicin/fluorouracil (PIAF) combination chemotherapy for unresectable hepatocellular carcinoma. J. Natl. Cancer Inst. 97, 1532–1538 (2005)CrossRefPubMed W. Yeo, T. S. Mok, B. Zee, T. W. Leung, P. B. Lai, W. Y. Lau, J. Koh, F. K. Mo, S. C. Yu, A. T. Chan, P. Hui, B. Ma, K. C. Lam, W. M. Ho, H. T. Wong, A. Tang, P. J. Johnson, A randomized phase III study of doxorubicin versus cisplatin/interferon α-2b/doxorubicin/fluorouracil (PIAF) combination chemotherapy for unresectable hepatocellular carcinoma. J. Natl. Cancer Inst. 97, 1532–1538 (2005)CrossRefPubMed
38.
go back to reference A. R. Baudy, N. Saxena, H. Gordish, E. P. Hoffman, K. Nagaraju, A robust in vitro screening assay to identify NF-κB inhibitors for inflammatory muscle diseases. Int. Immunopharmacol. 9, 1209–1214 (2009)CrossRefPubMedPubMedCentral A. R. Baudy, N. Saxena, H. Gordish, E. P. Hoffman, K. Nagaraju, A robust in vitro screening assay to identify NF-κB inhibitors for inflammatory muscle diseases. Int. Immunopharmacol. 9, 1209–1214 (2009)CrossRefPubMedPubMedCentral
39.
go back to reference Y. Zhou, C. Garcia-Prieto, D. A. Carney, R. H. Xu, H. Pelicano, Y. Kang, W. Yu, C. Lou, S. Kondo, J. Liu, D. M. Harris, Z. Estrov, M. J. Keating, Z. Jin, P. Huang, OSW-1: a natural compound with potent anticancer activity and a novel mechanism of action. J. Natl. Cancer Inst. 97, 1781–1785 (2005)CrossRefPubMed Y. Zhou, C. Garcia-Prieto, D. A. Carney, R. H. Xu, H. Pelicano, Y. Kang, W. Yu, C. Lou, S. Kondo, J. Liu, D. M. Harris, Z. Estrov, M. J. Keating, Z. Jin, P. Huang, OSW-1: a natural compound with potent anticancer activity and a novel mechanism of action. J. Natl. Cancer Inst. 97, 1781–1785 (2005)CrossRefPubMed
40.
go back to reference V. Yadav, S. Sultana, J. Yadav, N. Saini, Gatifloxacin induces S and G2-phase cell cycle arrest in pancreatic cancer cells via p21/p27/p53. PLoS One 7, e47796 (2012)CrossRefPubMedPubMedCentral V. Yadav, S. Sultana, J. Yadav, N. Saini, Gatifloxacin induces S and G2-phase cell cycle arrest in pancreatic cancer cells via p21/p27/p53. PLoS One 7, e47796 (2012)CrossRefPubMedPubMedCentral
41.
go back to reference D. Lavelle, J. DeSimone, M. Hankewych, T. Kousnetzova, Y. H. Chen, Decitabine induces cell cycle arrest at the G1 phase via p21(WAF1) and the G2/M phase via the p38 MAP kinase pathway. Leuk. Res. 27, 999–1007 (2003)CrossRefPubMed D. Lavelle, J. DeSimone, M. Hankewych, T. Kousnetzova, Y. H. Chen, Decitabine induces cell cycle arrest at the G1 phase via p21(WAF1) and the G2/M phase via the p38 MAP kinase pathway. Leuk. Res. 27, 999–1007 (2003)CrossRefPubMed
42.
go back to reference T. Fujimoto, M. Onda, H. Nagai, T. Nagahata, K. Ogawa, M. Emi, Upregulation and overexpression of human X-box binding protein 1 (hXBP-1) gene in primary breast cancers. Breast Cancer 10, 301–306 (2003)CrossRefPubMed T. Fujimoto, M. Onda, H. Nagai, T. Nagahata, K. Ogawa, M. Emi, Upregulation and overexpression of human X-box binding protein 1 (hXBP-1) gene in primary breast cancers. Breast Cancer 10, 301–306 (2003)CrossRefPubMed
43.
go back to reference C. Jamora, G. Dennert, A. S. Lee, Inhibition of tumor progression by suppression of stress protein GRP78/BiP induction in fibrosarcoma B/C10ME. Proc. Natl. Acad. Sci. U. S. A. 93, 7690–7694 (1996)CrossRefPubMedPubMedCentral C. Jamora, G. Dennert, A. S. Lee, Inhibition of tumor progression by suppression of stress protein GRP78/BiP induction in fibrosarcoma B/C10ME. Proc. Natl. Acad. Sci. U. S. A. 93, 7690–7694 (1996)CrossRefPubMedPubMedCentral
44.
go back to reference M. Shuda, N. Kondoh, N. Imazeki, K. Tanaka, T. Okada, K. Mori, A. Hada, M. Arai, T. Wakatsuki, O. Matsubara, N. Yamamoto, M. Yamamoto, Activation of the ATF6, XBP1 and grp78 genes in human hepatocellular carcinoma: a possible involvement of the ER stress pathway in hepatocarcinogenesis. J. Hepatol. 38, 605–614 (2003)CrossRefPubMed M. Shuda, N. Kondoh, N. Imazeki, K. Tanaka, T. Okada, K. Mori, A. Hada, M. Arai, T. Wakatsuki, O. Matsubara, N. Yamamoto, M. Yamamoto, Activation of the ATF6, XBP1 and grp78 genes in human hepatocellular carcinoma: a possible involvement of the ER stress pathway in hepatocarcinogenesis. J. Hepatol. 38, 605–614 (2003)CrossRefPubMed
46.
go back to reference C. Garcia-Prieto, K. B. R. Ahmed, Z. Chen, Y. Zhou, N. Hammoudi, Y. Kang, C. Lou, Y. Mei, Z. Jin, P. Huang, Effective killing of leukemia cells by the natural product OSW-1 through disruption of cellular calcium homeostasis. J. Biol. Chem. 288, 3240–3250 (2013)CrossRefPubMed C. Garcia-Prieto, K. B. R. Ahmed, Z. Chen, Y. Zhou, N. Hammoudi, Y. Kang, C. Lou, Y. Mei, Z. Jin, P. Huang, Effective killing of leukemia cells by the natural product OSW-1 through disruption of cellular calcium homeostasis. J. Biol. Chem. 288, 3240–3250 (2013)CrossRefPubMed
47.
go back to reference J. Jin, X. Jin, C. Qian, Y. Ruan, H. Jiang, Signaling network of OSW-1-induced apoptosis and necroptosis in hepatocellular carcinoma. Mol. Med. Rep. 7, 1646–1650 (2013) J. Jin, X. Jin, C. Qian, Y. Ruan, H. Jiang, Signaling network of OSW-1-induced apoptosis and necroptosis in hepatocellular carcinoma. Mol. Med. Rep. 7, 1646–1650 (2013)
48.
go back to reference Y. Inami, S. Waguri, A. Sakamoto, T. Kouno, K. Nakada, O. Hino, S. Watanabe, J. Ando, M. Iwadate, M. Yamamoto, M. S. Lee, K. Tanaka, M. Komatsu, Persistent activation of Nrf2 through p62 in hepatocellular carcinoma cells. J. Cell Biol. 193, 275–284 (2011)CrossRefPubMedPubMedCentral Y. Inami, S. Waguri, A. Sakamoto, T. Kouno, K. Nakada, O. Hino, S. Watanabe, J. Ando, M. Iwadate, M. Yamamoto, M. S. Lee, K. Tanaka, M. Komatsu, Persistent activation of Nrf2 through p62 in hepatocellular carcinoma cells. J. Cell Biol. 193, 275–284 (2011)CrossRefPubMedPubMedCentral
49.
go back to reference J. M. Luk, C. T. Lam, A. F. Siu, B. Y. Lam, I. O. Ng, M. Y. Hu, C. M. Che, S. T. Fan, Proteomic profiling of hepatocellular carcinoma in Chinese cohort reveals heat-shock proteins (Hsp27, Hsp70, GRP78) up-regulation and their associated prognostic values. Proteomics 6, 1049–1057 (2006)CrossRefPubMed J. M. Luk, C. T. Lam, A. F. Siu, B. Y. Lam, I. O. Ng, M. Y. Hu, C. M. Che, S. T. Fan, Proteomic profiling of hepatocellular carcinoma in Chinese cohort reveals heat-shock proteins (Hsp27, Hsp70, GRP78) up-regulation and their associated prognostic values. Proteomics 6, 1049–1057 (2006)CrossRefPubMed
50.
go back to reference S. Derks, B. Diosdado, Personalized cancer medicine: next steps in the genomic era. Cell. Oncol. 38, 1–2 (2015)CrossRef S. Derks, B. Diosdado, Personalized cancer medicine: next steps in the genomic era. Cell. Oncol. 38, 1–2 (2015)CrossRef
Metadata
Title
NFκB activation demarcates a subset of hepatocellular carcinoma patients for targeted therapy
Authors
Vignesh Ramesh
Karthikeyan Selvarasu
Jaishree Pandian
Soundarajan Myilsamy
Chidambaranathan Shanmugasundaram
Kumaresan Ganesan
Publication date
01-12-2016
Publisher
Springer Netherlands
Published in
Cellular Oncology / Issue 6/2016
Print ISSN: 2211-3428
Electronic ISSN: 2211-3436
DOI
https://doi.org/10.1007/s13402-016-0294-4

Other articles of this Issue 6/2016

Cellular Oncology 6/2016 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