Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-p2v8j Total loading time: 0 Render date: 2024-06-09T01:38:45.653Z Has data issue: false hasContentIssue false

29 - NF-κB and cancer

from Part 2.1 - Molecular pathways underlying carcinogenesis: signal transduction

Published online by Cambridge University Press:  05 February 2015

Willscott E. Naugler
Affiliation:
Oregon Health and Sciences University, Department of Medicine, Division of GI and Hepatology, Portland, OR, USA
Michael Karin
Affiliation:
Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology and Pathology, Moores Cancer Center, UCSD School of Medicine, La Jolla, CA, USA
Edward P. Gelmann
Affiliation:
Columbia University, New York
Charles L. Sawyers
Affiliation:
Memorial Sloan-Kettering Cancer Center, New York
Frank J. Rauscher, III
Affiliation:
The Wistar Institute Cancer Centre, Philadelphia
Get access

Summary

Introduction

Nuclear factor-κB (NF-κB) transcription factors and their signaling pathways have come to the forefront of the cancer field as mechanistic links connecting chronic inflammation and oncogenesis (1). These master transcription factors integrate multiple stimuli and co-ordinate innate and adaptive immune responses involved in acute and chronic inflammation (2). Epidemiological studies which pointed out that chronic inflammation and persistent infections greatly increase the risk of cancers of stomach, colon, and liver first suggested a link between inflammation, the innate immune response, and cancer (3). NF-κB was suggested as the molecular culprit that bridges these pathophysiological states and responses (1). However, establishing the association between NF-κB signaling and oncogenesis has been a challenging task because NF-κB and its activating machinery are rarely mutated in cancer cells in the same way as classical oncogenes (like Ras) or tumor-suppressor genes (like p53). Nonetheless, much evidence has been gathered, both through correlative studies and through direct experimentation, that NF-κB signaling does indeed contribute to cancer development and progression, mainly in inflammation-associated cancers, but also in cancers where underlying chronic inflammation plays little or no role (for example, breast and prostate cancers). The list of human cancers that were found to exhibit constitutive NF-κB activation is long (see Table 29.1). Experimental evidence providing causality for NF-κB signaling in oncogenesis has accumulated over the past several years (4), and will be detailed in this chapter.

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 336 - 352
Publisher: Cambridge University Press
Print publication year: 2013

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Karin, M, Cao, Y, Greten, FR, Li, ZW. NF-kappaB in cancer: from innocent bystander to major culprit. Nature Reviews Cancer 2002;2:301–10.CrossRef
Bonizzi, G, Karin, M. The two NF-kappaB activation pathways and their role in innate and adaptive immunity. Trends in Immunology 2004;25:280–8.CrossRef
Balkwill, F, Mantovani, A. Inflammation and cancer: back to Virchow? Lancet 2001;357:539–45.
Karin, M. Nuclear factor-kappaB in cancer development and progression. Nature 2006;441:431–6.CrossRef
Naugler, WE, Karin, M. NF-kappaB and cancer-identifying targets and mechanisms. Current Opinion in Genetic Development 2008; 18:19–26.CrossRef
Hanahan, D, Weinberg, RA. The hallmarks of cancer. Cell 2000;100:57–70.CrossRef
Sen, R, Baltimore, D. Inducibility of kappa immunoglobulin enhancer-binding protein Nf-kappa B by a posttranslational mechanism. Cell 1986;47:921–8.CrossRef
Brownell, E, O’Brien, SJ, Nash, WG, Rice, N. Genetic characterization of human c-rel sequences. Molecular and Cellular Biology 1985;5:2826–31.CrossRef
Gilmore, TD. The Re1/NF-kappa B/I kappa B signal transduction pathway and cancer. Cancer Treatment and Research 2003;115:241–65.CrossRef
Gilmore, TD. NF-kappa B, KBF1, dorsal, and related matters. Cell 1990;62:841–3.CrossRef
Hoffmann, A, Baltimore, D. Circuitry of nuclear factor kappaB signaling. Immunology Reviews 2006;210:171–86.CrossRef
Hoffmann, A, Natoli, G, Ghosh, G. Transcriptional regulation via the NF-kappaB signaling module. Oncogene 2006;25:6706–16.CrossRef
Chen, ZJ, Parent, L, Maniatis, T. Site-specific phosphorylation of IkappaBalpha by a novel ubiquitination-dependent protein kinase activity. Cell 1996;84:853–62.CrossRef
Karin, M, Ben-Neriah, Y. Phosphorylation meets ubiquitination: the control of NF-[kappa]B activity. Annual Review of Immunology 2000;18:621–63.CrossRef
DiDonato, JA, Hayakawa, M, Rothwarf, DM, Zandi, E, Karin, M. A cytokine-responsive IkappaB kinase that activates the transcription factor NF-kappaB. Nature 1997;388:548–54.CrossRef
Zandi, E, Rothwarf, DM, Delhase, M, Hayakawa, M, Karin, M. The IkappaB kinase complex (IKK) contains two kinase subunits, IKKalpha and IKKbeta, necessary for IkappaB phosphorylation and NF-kappaB activation. Cell 1997;91:243–52.CrossRef
Rothwarf, DM, Zandi, E, Natoli, G, Karin, M. IKK-gamma is an essential regulatory subunit of the IkappaB kinase complex. Nature 1998;395:297–300.CrossRef
Hacker, H, Karin, M. Regulation and function of IKK and IKK-related kinases. Science STKE 2006;2006:re13.
Senftleben, U, Cao, Y, Xiao, G, et al. Activation by IKKalpha of a second, evolutionary conserved, NF-kappa B signaling pathway. Science 2001;293:1495–9.CrossRef
Karin, M, Lawrence, T, Nizet, V. Innate immunity gone awry: linking microbial infections to chronic inflammation and cancer. Cell 2006;124:823–35.CrossRef
Sen, R, Baltimore, D. Multiple nuclear factors interact with the immunoglobulin enhancer sequences. Cell 1986;46:705–16.CrossRef
Gilmore, TD. Introduction to NF-kappaB: players, pathways, perspectives. Oncogene 2006;25:6680–4.CrossRef
Perkins, ND. Post-translational modifications regulating the activity and function of the nuclear factor kappa B pathway. Oncogene 2006;25:6717–30.CrossRef
Greten, FR, Arkan, MC, Bollrath, J, et al. NF-kappaB is a negative regulator of IL-1beta secretion as revealed by genetic and pharmacological inhibition of IKKbeta. Cell 2007;130:918–31.CrossRef
Dejardin, E, Droin, NM, Delhase, M, et al. The lymphotoxin-beta receptor induces different patterns of gene expression via two NF-kappaB pathways. Immunity 2002;17:525–35.CrossRef
Claudio, E, Brown, K, Park, S, Wang, H, Siebenlist, U. BAFF-induced NEMO-independent processing of NF-kappa B2 in maturing B cells. Nature Immunology 2002;3:958–65.CrossRef
Coope, HJ, Atkinson, PG, Huhse, B, et al. CD40 regulates the processing of NF-kappaB2 p100 to p52. EMBO Journal 2002;21:5375–85.CrossRef
Gerondakis, S, Grumont, R, Gugasyan, R, et al. Unravelling the complexities of the NF-kappaB signalling pathway using mouse knockout and transgenic models. Oncogene 2006;25:6781–99.CrossRef
Luedde, T, Beraza, N, Trautwein, C. Evaluation of the role of nuclear factor-kappaB signaling in liver injury using genetic animal models. Journal of Gastroenterology and Hepatology 2006;21 Suppl 3:S43–6.CrossRefGoogle ScholarPubMed
Maeda, S, Kamata, H, Luo, JL, Leffert, H, Karin, M. IKKbeta couples hepatocyte death to cytokine-driven compensatory proliferation that promotes chemical hepatocarcinogenesis. Cell 2005;121:977–90.CrossRef
Sha, WC, Liou, HC, Tuomanen, EI, Baltimore, D. Targeted disruption of the p50 subunit of NF-kappa B leads to multifocal defects in immune responses. Cell 1995;80:321–30.CrossRef
Caamano, JH, Rizzo, CA, Durham, SK, et al. Nuclear factor (NF)-kappa B2 (p100/p52) is required for normal splenic microarchitecture and B cell-mediated immune responses. Journal of Experimental Medicine 1998;187:185–96.CrossRefGoogle ScholarPubMed
Burkly, L, Hession, C, Ogata, L, et al. Expression of relB is required for the development of thymic medulla and dendritic cells. Nature 1995;373:531–6.CrossRef
Grumont, RJ, Rourke, IJ, O’Reilly, LA, et al. B lymphocytes differentially use the Rel and nuclear factor kappaB1 (NF-kappaB1) transcription factors to regulate cell cycle progression and apoptosis in quiescent and mitogen-activated cells. Journal of Experimental Medicine 1998;187:663–74.CrossRefGoogle ScholarPubMed
Beg, AA, Sha, WC, Bronson, RT, Ghosh, S, Baltimore, D. Embryonic lethality and liver degeneration in mice lacking the RelA component of NF-kappa B. Nature 1995;376:167–70.CrossRef
Doi, TS, Marino, MW, Takahashi, T, et al. Absence of tumor necrosis factor rescues RelA-deficient mice from embryonic lethality. Proceedings of the National Academy of Sciences USA 1999;96:2994–9.CrossRef
Alcamo, E, Mizgerd, JP, Horwitz, BH, et al. Targeted mutation of TNF receptor I rescues the RelA-deficient mouse and reveals a critical role for NF-kappa B in leukocyte recruitment. Journal of Immunology 2001;167:1592–600.CrossRefGoogle ScholarPubMed
Hu, Y, Baud, V, Delhase, M, et al. Abnormal morphogenesis but intact IKK activation in mice lacking the IKKalpha subunit of IkappaB kinase. Science 1999;284:316–20.CrossRef
Hu, Y, Baud, V, Oga, T, et al. IKKalpha controls formation of the epidermis independently of NF-kappaB. Nature 2001;410:710–14.CrossRef
Sil, AK, Maeda, S, Sano, Y, Roop, DR, Karin, M. IkappaB kinase-alpha acts in the epidermis to control skeletal and craniofacial morphogenesis. Nature 2004;428:660–4.CrossRef
Li, Q, Van Antwerp, D, Mercurio, F, Lee, KF, Verma, IM. Severe liver degeneration in mice lacking the IkappaB kinase 2 gene. Science 1999;284:321–5.CrossRef
Rudolph, D, Yeh, WC, Wakeham, A, et al. Severe liver degeneration and lack of NF-kappaB activation in NEMO/IKKgamma-deficient mice. Genes and Development 2000;14:854–62.
Makris, C, Godfrey, VL, Krahn-Senftleben, G, et al. Female mice heterozygous for IKK gamma/NEMO deficiencies develop a dermatopathy similar to the human X-linked disorder incontinentia pigmenti. Molecular Cell 2000;5:969–79.CrossRef
Nelson, DL. NEMO, NFkappaB signaling and incontinentia pigmenti. Current Opinion in Genetic Development 2006;16:282–8.CrossRef
Beg, AA, Sha, WC, Bronson, RT, Baltimore, D. Constitutive NF-kappa B activation, enhanced granulopoiesis, and neonatal lethality in I kappa B alpha-deficient mice. Genes and Development 1995;9:2736–46.CrossRef
Parkin, DM, Bray, F, Ferlay, J, Pisani, P. Estimating the world cancer burden: Globocan 2000. International Journal of Cancer 2001;94:153–6.CrossRefGoogle ScholarPubMed
Coussens, LM, Werb, Z. Inflammation and cancer. Nature 2002;420:860–7.CrossRef
Itzkowitz, SH, Harpaz, N. Diagnosis and management of dysplasia in patients with inflammatory bowel diseases. Gastroenterology 2004;126:1634–48.CrossRef
Ekbom, A, Helmick, C, Zack, M, Adami, HO. Ulcerative colitis and colorectal cancer: a population-based study. New England Journal of Medicine 1990;323:1228–33.CrossRefGoogle ScholarPubMed
Jemal, A, Siegel, R, Ward, E, et al. Cancer statistics, 2008. CA, A Cancer Journal for Clinicians 2008;58:71–96.CrossRef
Okayasu, I, Ohkusa, T, Kajiura, K, Kanno, J, Sakamoto, S. Promotion of colorectal neoplasia in experimental murine ulcerative colitis. Gut 1996;39:87–92.CrossRef
Chen, J, Huang, XF. The signal pathways in azoxymethane-induced colon cancer and preventive implications. Cancer Biology and Therapy 2009;8:1313–17.CrossRef
Tanaka, T, Kohno, H, Suzuki, R, et al. A novel inflammation-related mouse colon carcinogenesis model induced by azoxymethane and dextran sodium sulfate. Cancer Science 2003;94:965–73.CrossRef
Greten, FR, Eckmann, L, Greten, TF, et al. IKKbeta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell 2004;118:285–96.CrossRef
Van Antwerp, DJ, Martin, SJ, Kafri, T, Green, DR, Verma, IM. Suppression of TNF-alpha-induced apoptosis by NF-kappaB. Science 1996;274:787–9.CrossRef
Liu, ZG, Hsu, H, Goeddel, DV, Karin, M. Dissection of TNF receptor 1 effector functions: JNK activation is not linked to apoptosis while NF-kappaB activation prevents cell death. Cell 1996;87:565–76.CrossRef
Beg, AA, Baltimore, D. An essential role for NF-kappaB in preventing TNF-alpha-induced cell death. Science 1996;274:782–4.CrossRef
Naugler, WE, Karin, M. The wolf in sheep's clothing: the role of interleukin-6 in immunity, inflammation and cancer. Trends in Molecular Medicine 2008;14:109–19.CrossRef
Becker, C, Fantini, MC, Schramm, C, et al. TGF-beta suppresses tumor progression in colon cancer by inhibition of IL-6 trans-signaling. Immunity 2004;21:491–501.CrossRef
Grivennikov, S, Karin, E, Terzic, J, et al. IL-6 and Stat3 are required for survival of intestinal epithelial cells and development of colitis-associated cancer. Cancer Cell 2009;15:103–13.CrossRef
Bollrath, J, Phesse, TJ, von Burstin, VA, et al. gp130-mediated Stat3 activation in enterocytes regulates cell survival and cell-cycle progression during colitis-associated tumorigenesis. Cancer Cell 2009;15:91–102.CrossRef
Pickert, G, Neufert, C, Leppkes, M, et al. STAT3 links IL-22 signaling in intestinal epithelial cells to mucosal wound healing. Journal of Experimental Medicine 2009;206:1465–72.CrossRefGoogle ScholarPubMed
Rubinfeld, B, Albert, I, Porfiri, E, et al. Binding of GSK3beta to the APC-beta-catenin complex and regulation of complex assembly. Science 1996;272:1023–6.CrossRef
Munemitsu, S, Albert, I, Souza, B, Rubinfeld, B, Polakis, P. Regulation of intracellular beta-catenin levels by the adenomatous polyposis coli (APC) tumor-suppressor protein. Proceedings of the National Academy of Sciences USA 1995;92:3046–50.CrossRef
Rakoff-Nahoum, S, Medzhitov, R. Regulation of spontaneous intestinal tumorigenesis through the adaptor protein MyD88. Science 2007;317:124–7.CrossRef
Eberhart, CE, Coffey, RJ, Radhika, A, et al. Up-regulation of cyclooxygenase 2 gene expression in human colorectal adenomas and adenocarcinomas. Gastroenterology 1994;107:1183–8.CrossRef
Tazawa, R, Xu, XM, Wu, KK, Wang, LH. Characterization of the genomic structure, chromosomal location and promoter of human prostaglandin H synthase-2 gene. Biochemical and Biophysical Research Communications 1994;203:190–9.CrossRef
Plummer, SM, Holloway, KA, Manson, MM, et al. Inhibition of cyclo-oxygenase 2 expression in colon cells by the chemopreventive agent curcumin involves inhibition of NF-kappaB activation via the NIK/IKK signalling complex. Oncogene 1999;18:6013–20.CrossRef
Williams, JL, Nath, N, Chen, J, et al. Growth inhibition of human colon cancer cells by nitric oxide (NO)-donating aspirin is associated with cyclooxygenase-2 induction and beta-catenin/T-cell factor signaling, nuclear factor-kappaB, and NO synthase 2 inhibition: implications for chemoprevention. Cancer Research 2003;63:7613–18.
Sheehan, KM, Sabah, M, Cummins, RJ, et al. Cyclooxygenase-2 expression in stromal tumors of the gastrointestinal tract. Human Pathology 2003;34:1242–6.CrossRef
Liu, ES, Shin, VY, Ye, YN, et al. Cyclooxygenase-2 in cancer cells and macrophages induces colon cancer cell growth by cigarette smoke extract. European Journal of Pharmacology 2005;518:47–55.CrossRefGoogle ScholarPubMed
Adegboyega, PA, Ololade, O, Saada, J, et al. Subepithelial myofibroblasts express cyclooxygenase-2 in colorectal tubular adenomas. Clinical Cancer Research 2004;10:5870–9.CrossRef
Sonoshita, M, Takaku, K, Oshima, M, Sugihara, K, Taketo, MM. Cyclooxygenase-2 expression in fibroblasts and endothelial cells of intestinal polyps. Cancer Research 2002;62:6846–9.
Brown, JR, DuBois, RN. COX-2: a molecular target for colorectal cancer prevention. Journal of Clinical Oncology 2005;23:2840–55.CrossRefGoogle ScholarPubMed
Charalambous, MP, Lightfoot, T, Speirs, V, Horgan, K, Gooderham, NJ. Expression of COX-2, NF-kappaB-p65, NF-kappaB-p50 and IKKalpha in malignant and adjacent normal human colorectal tissue. British Journal of Cancer 2009;101:106–15.CrossRefGoogle ScholarPubMed
Hagemann, T, Lawrence, T, McNeish, I, et al. “Re-educating” tumor-associated macrophages by targeting NF-kappaB. Journal of Experimental Medicine 2008;205:1261–8.CrossRefGoogle ScholarPubMed
Fattovich, G, Stroffolini, T, Zagni, I, Donato, F. Hepatocellular carcinoma in cirrhosis: incidence and risk factors. Gastroenterology 2004;127:S35–50.
Naugler, WE, Sakurai, T, Kim, S, et al. Gender disparity in liver cancer due to sex differences in MyD88-dependent IL-6 production. Science 2007;317:121–4.CrossRef
Sakurai, T, Maeda, S, Chang, L, Karin, M. Loss of hepatic NF-(kappa)B activity enhances chemical hepatocarcinogenesis through sustained c-Jun N-terminal kinase 1 activation. Proceedings of the National Academy of Sciences USA 2006;103:10 544–51.
Pikarsky, E, Porat, RM, Stein, I, et al. NF-kappaB functions as a tumour promoter in inflammation-associated cancer. Nature 2004;431:461–6.CrossRef
Lee, JS, Chu, IS, Mikaelyan, A, et al. Application of comparative functional genomics to identify best-fit mouse models to study human cancer. Nature Genetics 2004;36:1306–11.CrossRef
Sakurai, T, He, G, Matsuzawa, A, et al. Hepatocyte necrosis induced by oxidative stress and IL-1 alpha release mediate carcinogen-induced compensatory proliferation and liver tumorigenesis. Cancer Cell 2008;14:156–65.CrossRef
Bosch, FX, Ribes, J, Diaz, M, Cleries, R. Primary liver cancer: worldwide incidence and trends. Gastroenterology 2004;127:S5–16.
Khoruts, A, Stahkne, L, McClain, C, Logan, G, Allen, J. Circulating tumor necrosis factor, interleukin-1 and interleukin-6 in chronic alcoholic patients. Hepatology 1991;13:267–76.CrossRef
Abiru, S, Migita, K, Maeda, Y, et al. Serum cytokine and soluble cytokine receptor levels in patients with non-alcoholic steatohepatitis. Liver International 2006;26:39–45.CrossRef
Kakumu, S, Shinagawa, T, Ishikawa, T, et al. Serum interleukin 6 levels in patients with chronic hepatitis B. American Journal of Gastroenterology 1991;86:1804–8.Google ScholarPubMed
Malaguarnera, M, Di Fazio, I, Laurino, A, et al. Serum interleukin 6 concentrations in chronic hepatitis C patients before and after interferon-alpha treatment. International Journal of Clinical Pharmacology and Therapeutics 1997;35:385–8.Google ScholarPubMed
Giannitrapani, L, Cervello, M, Soresi, M, et al. Circulating IL-6 and sIL-6R in patients with hepatocellular carcinoma. Annals of the New York Academy of Science 2002;963:46–52.CrossRef
Malaguarnera, M, Di Fazio, I, Laurino, A, et al. [Role of interleukin 6 in hepatocellular carcinoma]. Bulletin du Cancer 1996;83:379–84.
Soresi, M, Giannitrapani, L, D’Antona, F, et al. Interleukin-6 and its soluble receptor in patients with liver cirrhosis and hepatocellular carcinoma. World Journal of Gastroenterology 2006;12:2563–8.CrossRefGoogle ScholarPubMed
Wong, VW, Yu, J, Cheng, AS, et al. High serum interleukin-6 level predicts future hepatocellular carcinoma development in patients with chronic hepatitis B. International Journal of Cancer 2009;124:2766–70.CrossRefGoogle ScholarPubMed
Nakagawa, H, Maeda, S, Yoshida, H, et al. Serum IL-6 levels and the risk for hepatocarcinogenesis in chronic hepatitis C patients: an analysis based on gender differences. International Journal of Cancer 2009;125:2264–9.CrossRefGoogle ScholarPubMed
Calvisi, DF, Ladu, S, Gorden, A, et al. Ubiquitous activation of Ras and Jak/Stat pathways in human HCC. Gastroenterology 2006;130:1117–28.CrossRef
Hui, L, Bakiri, L, Mairhorfer, A, et al. p38alpha suppresses normal and cancer cell proliferation by antagonizing the JNK-c-Jun pathway. Nature Genetics 2007;39:741–9.CrossRef
Kamata, H, Honda, S, Maeda, S, et al. Reactive oxygen species promote TNFalpha-induced death and sustained JNK activation by inhibiting MAP kinase phosphatases. Cell 2005;120:649–61.CrossRef
Luedde, T, Beraza, N, Kotsikoris, V, et al. Deletion of NEMO/IKKgamma in liver parenchymal cells causes steatohepatitis and hepatocellular carcinoma. Cancer Cell 2007;11:119–32.CrossRef
Mauad, TH, van Nieuwkerk, CM, Dingemans, KP, et al. Mice with homozygous disruption of the mdr2 P-glycoprotein gene. A novel animal model for studies of nonsuppurative inflammatory cholangitis and hepatocarcinogenesis. American Journal of Pathology 1994;145:1237–45.Google ScholarPubMed
Haybaeck, J, Zeller, N, Wolf, MJ, et al. A lymphotoxin-driven pathway to hepatocellular carcinoma. Cancer Cell 2009;16:295–308.CrossRef
Singh, S, Shi, Q, Bailey, ST, et al. Nuclear factor-kappaB activation: a molecular therapeutic target for estrogen receptor-negative and epidermal growth factor receptor family receptor-positive human breast cancer. Molecular Cancer Therapeutics 2007;6:1973–82.CrossRef
Cao, Y, Luo, JL, Karin, M. IkappaB kinase alpha kinase activity is required for self-renewal of ERBB2/Her2-transformed mammary tumor-initiating cells. Proceedings of the National Academy of Sciences USA 2007;104:15 852–7.
Cabannes, E, Khan, G, Aillet, F, Jarrett, RF, Hay, RT. Mutations in the IkBa gene in Hodgkin's disease suggest a tumour suppressor role for IkappaBalpha. Oncogene 1999;18:3063–70.CrossRef
Annunziata, CM, Davis, RE, Demchenko, Y, et al. Frequent engagement of the classical and alternative NF-kappaB pathways by diverse genetic abnormalities in multiple myeloma. Cancer Cell 2007;12:115–30.CrossRef
Keats, JJ, Fonseca, R, Chesi, M, et al. Promiscuous mutations activate the noncanonical NF-kappaB pathway in multiple myeloma. Cancer Cell 2007;12:131–44.CrossRef
Vallabhapurapu, S, Matsuzawa, A, Zhang, W, et al. Nonredundant and complementary functions of TRAF2 and TRAF3 in a ubiquitination cascade that activates NIK-dependent alternative NF-kappaB signaling. Nature Immunology 2008;9:1364–70.CrossRef
Lam, LT, Davis, RE, Ngo, VN, et al. Compensatory IKKalpha activation of classical NF-kappaB signaling during IKKbeta inhibition identified by an RNA interference sensitization screen. Proceedings of the National Academy of Sciences USA 2008;105:20 798–803.
Jourdan, M, Moreaux, J, Vos, JD, et al. Targeting NF-kappaB pathway with an IKK2 inhibitor induces inhibition of multiple myeloma cell growth. British Journal of Haematology 2007;138:160–8.CrossRefGoogle ScholarPubMed
Rahman, KW, Ali, S, Aboukameel, A, et al. Inactivation of NF-(kappa)B by 3,3’-diindolylmethane contributes to increased apoptosis induced by chemotherapeutic agent in breast cancer cells. Molecular Cancer Therapeutics 2007;6:2757–65.CrossRef
Tew, GW, Lorimer, EL, Berg, TJ, et al. SmgGDS regulates cell proliferation, migration, and NF-kappa B transcriptional activity in non-small cell lung carcinoma. Journal of Biological Chemistry 2007;283:963–76.CrossRefGoogle ScholarPubMed
Fernandez-Majada, V, Aguilera, C, Villanueva, A, et al. Nuclear IKK activity leads to dysregulated notch-dependent gene expression in colorectal cancer. Proceedings of the National Academy of Sciences USA 2007;104:276–81.CrossRef
Yang, J, Pan, WH, Clawson, GA, Richmond, A. Systemic targeting inhibitor of kappaB kinase inhibits melanoma tumor growth. Cancer Research 2007;67:3127–34.CrossRef
Zou, P, Kawada, J, Pesnicak, L, Cohen, JI. Bortezomib induces apoptosis of Epstein-Barr virus (EBV)-transformed B cells and prolongs survival of mice inoculated with EBV-transformed B cells. Journal of Virology 2007;81:10 029–36.CrossRefGoogle ScholarPubMed
Westerheide, SD, Mayo, MW, Anest, V, Hanson, JL, Baldwin, AS. The putative oncoprotein Bcl-3 induces cyclin D1 to stimulate G(1) transition. Molecular and Cellular Biology 2001;21:8428–36.CrossRef
Cao, Y, Bonizzi, G, Seagroves, TN, et al. IKKalpha provides an essential link between RANK signaling and cyclin D1 expression during mammary gland development. Cell 2001;107:763–75.CrossRef
Jung, YJ, Isaacs, JS, Lee, S, Trepel, J, Neckers, L. IL-1beta-mediated up-regulation of HIF-1alpha via an NFkappaB/COX-2 pathway identifies HIF-1 as a critical link between inflammation and oncogenesis. FASEB Journal 2003;17:2115–17.CrossRef
Pahl, HL. Activators and target genes of Rel/NF-kappaB transcription factors. Oncogene 1999;18:6853–66.CrossRef
Luo, JL, Maeda, S, Hsu, LC, Yagita, H, Karin, M. Inhibition of NF-kappaB in cancer cells converts inflammation- induced tumor growth mediated by TNFalpha to TRAIL-mediated tumor regression. Cancer Cell 2004;6:297–305.CrossRef
Chen, H, Li, M, Campbell, RA, et al. Interference with nuclear factor kappa B and c-Jun NH2-terminal kinase signaling by TRAF6C small interfering RNA inhibits myeloma cell proliferation and enhances apoptosis. Oncogene 2006;25:6520–7.CrossRef
Chauhan, D, Catley, L, Li, G, et al. A novel orally active proteasome inhibitor induces apoptosis in multiple myeloma cells with mechanisms distinct from Bortezomib. Cancer Cell 2005;8:407–19.CrossRef
Mi, J, Zhang, X, Liu, Y, et al. NF-kappaB inhibition by an adenovirus expressed aptamer sensitizes TNFalpha-induced apoptosis. Biochemical and Biophysical Research Communications 2007;359:475–80.CrossRef
Tapia, MA, Gonzalez-Navarrete, I, Dalmases, A, et al. Inhibition of the canonical IKK/NF kappa B pathway sensitizes human cancer cells to doxorubicin. Cell Cycle 2007;6:2284–92.CrossRef
Carvalho, G, Fabre, C, Braun, T, et al. Inhibition of NEMO, the regulatory subunit of the IKK complex, induces apoptosis in high-risk myelodysplastic syndrome and acute myeloid leukemia. Oncogene 2007;26:2299–307.CrossRef
Bernal-Mizrachi, L, Lovly, CM, Ratner, L. The role of NF-(kappa)B-1 and NF-(kappa)B-2-mediated resistance to apoptosis in lymphomas. Proceedings of the National Academy of Sciences USA 2006;103:9220–5.CrossRef
Zhang, B, Wang, Z, Li, T, Tsitsikov, EN, Ding, HF. NF-kappaB2 mutation targets TRAF1 to induce lymphomagenesis. Blood 2007;110:743–51.CrossRef
Janssens, S, Tinel, A, Lippens, S, Tschopp, J. PIDD mediates NF-kappaB activation in response to DNA damage. Cell 2005;123:1079–92.CrossRef
Basseres, DS, Baldwin, AS. Nuclear factor-kappaB and inhibitor of kappaB kinase pathways in oncogenic initiation and progression. Oncogene 2006;25:6817–30.CrossRef
Huber, MA, Azoitei, N, Baumann, B, et al. NF-kappaB is essential for epithelial-mesenchymal transition and metastasis in a model of breast cancer progression. Journal of Clinical Investigation 2004;114:569–81.CrossRefGoogle Scholar
Wang, X, Belguise, K, Kersual, N, et al. Oestrogen signalling inhibits invasive phenotype by repressing RelB and its target BCL2. Nature Cell Biology 2007;9:470–8.CrossRef
Zhang, Q, Helfand, BT, Jang, TL, et al. Nuclear factor-kappaB-mediated transforming growth factor-beta-induced expression of vimentin is an independent predictor of biochemical recurrence after radical prostatectomy. Clinical Cancer Research 2009;15:3557–67.CrossRef
Yang, J, Mani, SA, Donaher, JL, et al. Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell 2004;117:927–39.CrossRef
Horikawa, T, Yang, J, Kondo, S, et al. Twist and epithelial-mesenchymal transition are induced by the EBV oncoprotein latent membrane protein 1 and are associated with metastatic nasopharyngeal carcinoma. Cancer Research 2007;67:1970–8.CrossRef
Luo, JL, Tan, W, Ricono, JM, et al. Nuclear cytokine-activated IKKalpha controls prostate cancer metastasis by repressing Maspin. Nature 2007;446:690–4.CrossRef
Maeda, G, Chiba, T, Kawashiri, S, Satoh, T, Imai, K. Epigenetic inactivation of IkappaB Kinase-alpha in oral carcinomas and tumor progression. Clinical Cancer Research 2007;13:5041–7.CrossRef
Van Waes, C, Yu, M, Nottingham, L, Karin, M. Inhibitor-kappaB kinase in tumor promotion and suppression during progression of squamous cell carcinoma. Clinical Cancer Research 2007;13:4956–9.CrossRef
Schmidt, D, Textor, B, Pein, OT, et al. Critical role for NF-kappaB-induced JunB in VEGF regulation and tumor angiogenesis. EMBO Journal 2007;26:710–19.CrossRef
Cummins, EP, Berra, E, Comerford, KM, et al. Prolyl hydroxylase-1 negatively regulates IkappaB kinase-beta, giving insight into hypoxia-induced NFkappaB activity. Proceedings of the National Academy of Sciences USA 2006;103:18 154–9.
Rius, J, Guma, M, Schachtrup, C, et al. NF-kappaB links innate immunity to the hypoxic response through transcriptional regulation of HIF-1alpha. Nature 2008;453:807–11.CrossRef
Gilmore, TD, Herscovitch, M. Inhibitors of NF-kappaB signaling: 785 and counting. Oncogene 2006;25:6887–99.CrossRef
Wagner, AD, Arnold, D, Grothey, AA, Haerting, J, Unverzagt, S. Anti-angiogenic therapies for metastatic colorectal cancer. Cochrane Database System Reviews 2009:CD005392.
Raab, MS, Podar, K, Breitkreutz, I, Richardson, PG, Anderson, KC. Multiple myeloma. Lancet 2009;374:324–39.CrossRef
Cardoso, F, Durbecq, V, Laes, JF, et al. Bortezomib (PS-341, Velcade) increases the efficacy of trastuzumab (Herceptin) in HER-2-positive breast cancer cells in a synergistic manner. Molecular Cancer Therapeutics 2006;5:3042–51.CrossRef

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

  • NF-κB and cancer
    • By Willscott E. Naugler, Oregon Health and Sciences University, Department of Medicine, Division of GI and Hepatology, Portland, OR, USA, Michael Karin, Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology and Pathology, Moores Cancer Center, UCSD School of Medicine, La Jolla, CA, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.030
Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

  • NF-κB and cancer
    • By Willscott E. Naugler, Oregon Health and Sciences University, Department of Medicine, Division of GI and Hepatology, Portland, OR, USA, Michael Karin, Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology and Pathology, Moores Cancer Center, UCSD School of Medicine, La Jolla, CA, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.030
Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

  • NF-κB and cancer
    • By Willscott E. Naugler, Oregon Health and Sciences University, Department of Medicine, Division of GI and Hepatology, Portland, OR, USA, Michael Karin, Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology and Pathology, Moores Cancer Center, UCSD School of Medicine, La Jolla, CA, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.030
Available formats
×