Skip to main content
Erschienen in: Tumor Biology 6/2012

01.12.2012 | Research Article

Synergistic induction of apoptosis and chemosensitization of human colorectal cancer cells by histone deacetylase inhibitor, scriptaid, and proteasome inhibitors: potential mechanisms of action

verfasst von: M. S. I. Abaza, A. M. Bahman, R. J. Al-Attiyah, A. M. Kollamparambil

Erschienen in: Tumor Biology | Ausgabe 6/2012

Einloggen, um Zugang zu erhalten

Abstract

Histone deacetylase inhibitors (HDACIs) exhibit modest results as single agents in preclinical and clinical studies against solid tumors; they often fall short and activate nuclear factor kappa-B (NFκB). Co-administration of HDACI with proteasome inhibitors (PIs), which interrupt NFκB pathways, may enhance HDACI-lethality. The goal of this study was to determine whether PIs could potentiate HDACI, scriptaid (SCP)-mediated lethality, to unravel the associated mechanisms and to assess the effects of the combined inhibition of HDAC and proteasome on chemotherapy response in human colorectal cancer cells. Cancer cells were exposed to agents alone or in combination; cell growth inhibition was determined by MTT and colony formation assays. HDAC-, proteasome-, NFκB-activities, and reactive oxygen species (ROS) were quantified. Induction of apoptosis and cell cycle alterations were monitored by flow cytometry. Expression of cell cycle/apoptosis and cytoprotective/stress-related genes was determined by real-time qRT-PCR and EIA, respectively. Potentiation of cancer cell sensitivity to chemotherapies by SCP/PIs was also evaluated. SCP and PIs: MG132, PI-1, or epoxomicin interact synergistically to potently inhibit cancer cell growth, alter cell cycle, induce apoptosis, reduce NFκB activity, and increase ROS generation. These events are associated with multiple perturbations in the expression of cell cycle, apoptosis, cytoprotective, and stress-related genes. Co-administration of SCP and PIs strikingly increases the chemosensitivity of cancer cells (122–2 × 105-fold) in a drug and SCP/PIs-dependent manner. This combination regimen markedly reduced the doses of chemotherapies with potent anticancer effects and less toxicity. A strategy combining HDAC/proteasome inhibition with chemotherapies warrants further investigation in colorectal cancer.
Anhänge
Nur mit Berechtigung zugänglich
Literatur
1.
Zurück zum Zitat Ellis L, Hammers H, Pili R. Targeting tumor angiogenesis with histone deacetylase inhibitors. Cancer Lett. 2009;280:145–53.PubMedCrossRef Ellis L, Hammers H, Pili R. Targeting tumor angiogenesis with histone deacetylase inhibitors. Cancer Lett. 2009;280:145–53.PubMedCrossRef
3.
Zurück zum Zitat Su GH, Sohn TA, Ryu B, Kern SE. A novel histone deacetylase inhibitor identified by high-throughput transcriptional screening of a computer library. Cancer Res. 2000;60:3137–42.PubMed Su GH, Sohn TA, Ryu B, Kern SE. A novel histone deacetylase inhibitor identified by high-throughput transcriptional screening of a computer library. Cancer Res. 2000;60:3137–42.PubMed
4.
Zurück zum Zitat Takai N, Ueda T, Nishida M, Nasu K, Narahara H. A novel histone deacetylase inhibitor, Scriptaid, induces growth inhibition, cell cycle arrest and apoptosis in human endometrial cancer and ovarian cancer cells. Int J Mol Med. 2006;17:323–9.PubMed Takai N, Ueda T, Nishida M, Nasu K, Narahara H. A novel histone deacetylase inhibitor, Scriptaid, induces growth inhibition, cell cycle arrest and apoptosis in human endometrial cancer and ovarian cancer cells. Int J Mol Med. 2006;17:323–9.PubMed
5.
Zurück zum Zitat Ellis L, Pili R. Histone deacetylase inhibitors: advancing therapeutic strategies in hematological and solid malignancies. Pharmaceuticals (Basel). 2010;3(8):2411–69. Ellis L, Pili R. Histone deacetylase inhibitors: advancing therapeutic strategies in hematological and solid malignancies. Pharmaceuticals (Basel). 2010;3(8):2411–69.
6.
Zurück zum Zitat Mayo MW, Denlinger CE, Broad RM, Yeung F, Reilly ET, Shi Y, Jones DR. Ineffectiveness of histone deacetylase inhibitors to induce apoptosis involves the transcriptional activation of NF-kappa B through the Akt pathway. J Biol Chem. 2003;278:18980–9.PubMedCrossRef Mayo MW, Denlinger CE, Broad RM, Yeung F, Reilly ET, Shi Y, Jones DR. Ineffectiveness of histone deacetylase inhibitors to induce apoptosis involves the transcriptional activation of NF-kappa B through the Akt pathway. J Biol Chem. 2003;278:18980–9.PubMedCrossRef
7.
Zurück zum Zitat Dai Y, Rahamani M, Dent P, Grant S. Blockade of histone deacetylase inhibitor-induced Rel A/p65 acetylation and NF-κB activation potentiate apoptosis in leukemia cells through a process mediated by oxidative damage, XIAP downregulation, and c-Jun N-terminal kinase 1 activation. Mol Cell Biol. 2005;25:5429–44.PubMedCrossRef Dai Y, Rahamani M, Dent P, Grant S. Blockade of histone deacetylase inhibitor-induced Rel A/p65 acetylation and NF-κB activation potentiate apoptosis in leukemia cells through a process mediated by oxidative damage, XIAP downregulation, and c-Jun N-terminal kinase 1 activation. Mol Cell Biol. 2005;25:5429–44.PubMedCrossRef
8.
Zurück zum Zitat Adams J, Palombella VJ, Elliot PJ. Proteasome inhibition: a new strategy in cancer treatment. Investig New Drugs. 2000;18:109–21.CrossRef Adams J, Palombella VJ, Elliot PJ. Proteasome inhibition: a new strategy in cancer treatment. Investig New Drugs. 2000;18:109–21.CrossRef
9.
Zurück zum Zitat Almond JB, Cohen GM. The proteasome: a novel target for cancer chemotherapy. Leukemia. 2002;16:433–43.PubMedCrossRef Almond JB, Cohen GM. The proteasome: a novel target for cancer chemotherapy. Leukemia. 2002;16:433–43.PubMedCrossRef
10.
Zurück zum Zitat An B, Goldfarb RH, Dou QP. Novel dipeptidyl proteasome inhibitors overcome Bcl-2 protective function and selectively accumulate the cyclin-dependent kinase inhibitor p27 and induce apoptosis in transformed, but not normal, human fibroblasts. Cell Death Diff. 1998;5:1062–75.CrossRef An B, Goldfarb RH, Dou QP. Novel dipeptidyl proteasome inhibitors overcome Bcl-2 protective function and selectively accumulate the cyclin-dependent kinase inhibitor p27 and induce apoptosis in transformed, but not normal, human fibroblasts. Cell Death Diff. 1998;5:1062–75.CrossRef
11.
Zurück zum Zitat Cusack Jr JC, Liu R, Houston M, et al. Enhanced chemosensitivity to CPT-11 with proteasome inhibitor PS-341: implication for systemic nuclear factor-kappaB inhibition. Cancer Res. 2001;61:3535–40.PubMed Cusack Jr JC, Liu R, Houston M, et al. Enhanced chemosensitivity to CPT-11 with proteasome inhibitor PS-341: implication for systemic nuclear factor-kappaB inhibition. Cancer Res. 2001;61:3535–40.PubMed
12.
Zurück zum Zitat Abaza MS, Al-Safar A, Al-Sawan S, Al-Attiyah R. c-myc antisense oligonucleotides sensitize human colorectal cancer cells to chemotherapeutic drugs. Tumor Biol. 2008;29:287–303.CrossRef Abaza MS, Al-Safar A, Al-Sawan S, Al-Attiyah R. c-myc antisense oligonucleotides sensitize human colorectal cancer cells to chemotherapeutic drugs. Tumor Biol. 2008;29:287–303.CrossRef
13.
Zurück zum Zitat Abaza MSI. Augmentation of the anticancer effects of proteasome inhibitors by combination with sodium butyrate in human colorectal cancer cells. Experimental and Therapeutic Medicine. 2010;1:675–93.CrossRef Abaza MSI. Augmentation of the anticancer effects of proteasome inhibitors by combination with sodium butyrate in human colorectal cancer cells. Experimental and Therapeutic Medicine. 2010;1:675–93.CrossRef
15.
Zurück zum Zitat Bolden JE, Perat MJ, Johnstone RW. Anticancer activities of histone deacetylase inhibitors. Nat Rev Drug Discov. 2006;5:769–84.PubMedCrossRef Bolden JE, Perat MJ, Johnstone RW. Anticancer activities of histone deacetylase inhibitors. Nat Rev Drug Discov. 2006;5:769–84.PubMedCrossRef
16.
Zurück zum Zitat Xu WS, Parmigiani RB, Marks PA. Histone deacetylase inhibitors: molecular mechanisms of action. Oncogene. 2007;26:5541–52.PubMedCrossRef Xu WS, Parmigiani RB, Marks PA. Histone deacetylase inhibitors: molecular mechanisms of action. Oncogene. 2007;26:5541–52.PubMedCrossRef
17.
Zurück zum Zitat Xu W, Ngo L, Perez G, Dokmanovic M, Marks PA. Intrinsic apoptotic and thioredoxin pathways in human prostate cancer cell response to histone deacetylase inhibitor. Proc Natl Acad Sci USA. 2006;103:15540–5.PubMedCrossRef Xu W, Ngo L, Perez G, Dokmanovic M, Marks PA. Intrinsic apoptotic and thioredoxin pathways in human prostate cancer cell response to histone deacetylase inhibitor. Proc Natl Acad Sci USA. 2006;103:15540–5.PubMedCrossRef
18.
Zurück zum Zitat Bots M, Johnstone RW. Rational combinations using HDAC inhibitors. Clin Cancer Res. 2009;15:3970–7.PubMedCrossRef Bots M, Johnstone RW. Rational combinations using HDAC inhibitors. Clin Cancer Res. 2009;15:3970–7.PubMedCrossRef
19.
Zurück zum Zitat Rasheed W, Bishton M, Johnstone RW, Prince HM. Histone deacetylase inhibitors in lymphoma and solid malignancies. Expert Rev Anticancer Ther. 2008;8:413–32.PubMedCrossRef Rasheed W, Bishton M, Johnstone RW, Prince HM. Histone deacetylase inhibitors in lymphoma and solid malignancies. Expert Rev Anticancer Ther. 2008;8:413–32.PubMedCrossRef
20.
Zurück zum Zitat Frew AJ, Johnstone RW, Bolden JE. Enhancing the apoptotic and therapeutic effects of HDAC inhibitors. Cancer Lett. 2008;125:125–33. Frew AJ, Johnstone RW, Bolden JE. Enhancing the apoptotic and therapeutic effects of HDAC inhibitors. Cancer Lett. 2008;125:125–33.
21.
Zurück zum Zitat Denlinger C, Keller M, Mayo M, Broad RM, Jones DR. Combined proteasome and histone deacetylase inhibition in non-small cell lung cancer. J Thorac Cardiovasc Surg. 2004;127:1078–86.PubMedCrossRef Denlinger C, Keller M, Mayo M, Broad RM, Jones DR. Combined proteasome and histone deacetylase inhibition in non-small cell lung cancer. J Thorac Cardiovasc Surg. 2004;127:1078–86.PubMedCrossRef
22.
Zurück zum Zitat Bhalla S, Balasubramanian S, David K, et al. PCI-24781 induces caspase and reactive oxygen species-dependent apoptosis through NFκB mechanisms and is synergistic with bortezomib in lymphoma cells. Clin Cancer Res. 2009;15:3354–65.PubMedCrossRef Bhalla S, Balasubramanian S, David K, et al. PCI-24781 induces caspase and reactive oxygen species-dependent apoptosis through NFκB mechanisms and is synergistic with bortezomib in lymphoma cells. Clin Cancer Res. 2009;15:3354–65.PubMedCrossRef
23.
Zurück zum Zitat Dasmahapatra G, Lembersky D, Son MP, Attkisson E, Dent P, Fisher RI, Friedberg JW, Grant S. Carfilzomib interacts synergistically with histone deacetylase inhibitors in mantle cell lymphoma cells in vitro and in vivo. Mol Cancer Ther. 2011;10:1686–97.PubMedCrossRef Dasmahapatra G, Lembersky D, Son MP, Attkisson E, Dent P, Fisher RI, Friedberg JW, Grant S. Carfilzomib interacts synergistically with histone deacetylase inhibitors in mantle cell lymphoma cells in vitro and in vivo. Mol Cancer Ther. 2011;10:1686–97.PubMedCrossRef
24.
Zurück zum Zitat Vigushin DM, Coomes RC. Histone deacetylase inhibitors in cancer treatment. Anticancer Drugs. 2002;13:1–13.PubMedCrossRef Vigushin DM, Coomes RC. Histone deacetylase inhibitors in cancer treatment. Anticancer Drugs. 2002;13:1–13.PubMedCrossRef
25.
Zurück zum Zitat Butler LM, Zhou X, Xu WS, Scher HI, Rifkind RA, Marks PA, et al. The histone deacetylase inhibitor SAHA arrests cancer cell growth, up-regulates thioredoxin-binding protein-2, and down-regulates thioredoxin. Proc Natl Acad Sci USA. 2002;99:11700–5.PubMedCrossRef Butler LM, Zhou X, Xu WS, Scher HI, Rifkind RA, Marks PA, et al. The histone deacetylase inhibitor SAHA arrests cancer cell growth, up-regulates thioredoxin-binding protein-2, and down-regulates thioredoxin. Proc Natl Acad Sci USA. 2002;99:11700–5.PubMedCrossRef
26.
Zurück zum Zitat Duan J, Friedman J, Nottingham L, Chen Z, Ara G, Van Waes C. Nuclear factor-κ p65 small interfering RNA or proteasome inhibitor bortezomib sensitizes head and neck squamous cell carcinomas to classic histone deacetylae inhibitors and novel histone deacetylase inhibitor PXD101. Mol Cancer Ther. 2007;6:37–50.PubMedCrossRef Duan J, Friedman J, Nottingham L, Chen Z, Ara G, Van Waes C. Nuclear factor-κ p65 small interfering RNA or proteasome inhibitor bortezomib sensitizes head and neck squamous cell carcinomas to classic histone deacetylae inhibitors and novel histone deacetylase inhibitor PXD101. Mol Cancer Ther. 2007;6:37–50.PubMedCrossRef
27.
28.
Zurück zum Zitat Denlinger CE, Mayo MW, Jones DR. Combination proteasome inhibition and gemcitabine chemo-sensitizes NSCLC to apoptosis by inhibition of NFκB. Proc Am Assoc Cancer Res. 2003;44:1058. Denlinger CE, Mayo MW, Jones DR. Combination proteasome inhibition and gemcitabine chemo-sensitizes NSCLC to apoptosis by inhibition of NFκB. Proc Am Assoc Cancer Res. 2003;44:1058.
29.
Zurück zum Zitat Zhang QL, Wang L, Zhang YW, Jiang XX, Yang F, Wu WL, Janin A, Chen Z, Shen ZX, Chen SJ, Zhao WL. The proteasome inhibitor bortezomib interacts synergistically with histone deacetylase inhibitor suberoylanilide hydroxamic acid to induce T-leukemia/lymphoma cells apoptosis. Leukemia. 2009;23:1507–14.PubMedCrossRef Zhang QL, Wang L, Zhang YW, Jiang XX, Yang F, Wu WL, Janin A, Chen Z, Shen ZX, Chen SJ, Zhao WL. The proteasome inhibitor bortezomib interacts synergistically with histone deacetylase inhibitor suberoylanilide hydroxamic acid to induce T-leukemia/lymphoma cells apoptosis. Leukemia. 2009;23:1507–14.PubMedCrossRef
30.
Zurück zum Zitat Heider U, von Metzler I, Kaiser M, et al. Synergistic interaction of the histone deacetylase inhibitor SAHA with the proteasome bortezomib in mantle cell lymphoma. Eur J Haematol. 2008;80:133–42.PubMedCrossRef Heider U, von Metzler I, Kaiser M, et al. Synergistic interaction of the histone deacetylase inhibitor SAHA with the proteasome bortezomib in mantle cell lymphoma. Eur J Haematol. 2008;80:133–42.PubMedCrossRef
31.
Zurück zum Zitat Ruefli AA, Ausserlechner MJ, Bernhard D, et al. The histone deacetylase inhibitor and chemotherapeutic agent suberoylanilide hydroxamic acid (SAHA) induces a cell-death pathway characterized by cleavage of Bid and production of reactive oxygen species. Proc Natl Acad Sci USA. 2001;98:10833–8.PubMedCrossRef Ruefli AA, Ausserlechner MJ, Bernhard D, et al. The histone deacetylase inhibitor and chemotherapeutic agent suberoylanilide hydroxamic acid (SAHA) induces a cell-death pathway characterized by cleavage of Bid and production of reactive oxygen species. Proc Natl Acad Sci USA. 2001;98:10833–8.PubMedCrossRef
32.
Zurück zum Zitat Heider U, Rademacher J, Lamottke B, Mieth M, Moebs M, von Metzler I, Assaf C, Sezer O. Synergistic interaction of the deacetylase inhibitor SAHA with the proteasome inhibitor bortezomib in cutaneous T cell lymphoma. Eur J Haematol. 2009;82:440–9.PubMedCrossRef Heider U, Rademacher J, Lamottke B, Mieth M, Moebs M, von Metzler I, Assaf C, Sezer O. Synergistic interaction of the deacetylase inhibitor SAHA with the proteasome inhibitor bortezomib in cutaneous T cell lymphoma. Eur J Haematol. 2009;82:440–9.PubMedCrossRef
33.
Zurück zum Zitat Flis S, Gnyszka A, Splawinski J. HDAC inhibitors, MS275 and SBHA, enhances cytotoxicity induced by oxaliplatin in the colorectal cancer cell lines. Biochem Biophys Res Comm. 2009;387:336–41.PubMedCrossRef Flis S, Gnyszka A, Splawinski J. HDAC inhibitors, MS275 and SBHA, enhances cytotoxicity induced by oxaliplatin in the colorectal cancer cell lines. Biochem Biophys Res Comm. 2009;387:336–41.PubMedCrossRef
34.
Zurück zum Zitat Sharma V, Koul N, Joseph C, Dixit D, Ghosh S, Sen E. HDAC inhibitor, scriptaid, induces glioma cell apoptosis through JNK activation and inhibits telomerase activity. J Cell Mol Med. 2010;14:2151–61.PubMedCrossRef Sharma V, Koul N, Joseph C, Dixit D, Ghosh S, Sen E. HDAC inhibitor, scriptaid, induces glioma cell apoptosis through JNK activation and inhibits telomerase activity. J Cell Mol Med. 2010;14:2151–61.PubMedCrossRef
35.
Zurück zum Zitat Orlowski RZ. The role of ubiquitin–proteasome pathway in apoptosis. Cell Death Diff. 1999;6:303–31.CrossRef Orlowski RZ. The role of ubiquitin–proteasome pathway in apoptosis. Cell Death Diff. 1999;6:303–31.CrossRef
36.
Zurück zum Zitat Pagabo M, Tam SW, Theodoras AM, Beer-Romero P, Del Sal G, Chau V, Yew PR. PR, et al. Role of the ubiquitin–proteasome pathway in regulating abundance of the cyclin-dependent kinase inhibitor p27. Science. 1995;269:682–5.CrossRef Pagabo M, Tam SW, Theodoras AM, Beer-Romero P, Del Sal G, Chau V, Yew PR. PR, et al. Role of the ubiquitin–proteasome pathway in regulating abundance of the cyclin-dependent kinase inhibitor p27. Science. 1995;269:682–5.CrossRef
37.
Zurück zum Zitat Chen YR, Wang X, Templeton D, Davis RJ, Tan TH. The role of c-jun N-terminal kinase (JNK) in apoptosis induced by ultraviolet C and γ radiation. Duration of JNK activation may determine cell death and proliferation. J Biol Chem. 1996;271:31929–36.PubMedCrossRef Chen YR, Wang X, Templeton D, Davis RJ, Tan TH. The role of c-jun N-terminal kinase (JNK) in apoptosis induced by ultraviolet C and γ radiation. Duration of JNK activation may determine cell death and proliferation. J Biol Chem. 1996;271:31929–36.PubMedCrossRef
38.
Zurück zum Zitat Nawrocki ST, Carew JS, Pino MS, et al. Aggresome disruption: a novel strategy to enhance bortezomib-induced apoptosis in pancreatic cancer cells. Cancer Res. 2006;66:3773–81.PubMedCrossRef Nawrocki ST, Carew JS, Pino MS, et al. Aggresome disruption: a novel strategy to enhance bortezomib-induced apoptosis in pancreatic cancer cells. Cancer Res. 2006;66:3773–81.PubMedCrossRef
39.
Zurück zum Zitat Xia Z, Dickens M, Raingeaud J, Davis RJ, Greenberg ME. Opposing effects of ERK and JNK-p38 MAP kinases on apoptosis. Science 1995;270:1326–31. Xia Z, Dickens M, Raingeaud J, Davis RJ, Greenberg ME. Opposing effects of ERK and JNK-p38 MAP kinases on apoptosis. Science 1995;270:1326–31.
40.
Zurück zum Zitat Toumier C, Hess P, Yang DD, et al. The Bax subfamily of Bcl2 related proteins is essential for apoptotic signal transduction by c-Jun NH2-terminal kinase. Mol Cell Biol. 2002;22:4929–42.CrossRef Toumier C, Hess P, Yang DD, et al. The Bax subfamily of Bcl2 related proteins is essential for apoptotic signal transduction by c-Jun NH2-terminal kinase. Mol Cell Biol. 2002;22:4929–42.CrossRef
41.
Zurück zum Zitat Tacchini L, Dansi P, Matteucci E, Bemelli-Zazzera A, Desiderio MA. Influence of proteasome and redox state on heat shock-induced activation of stress kinases, AP-1 and HSF. Biochim Biophy Acta. 2001;1538:76–89.CrossRef Tacchini L, Dansi P, Matteucci E, Bemelli-Zazzera A, Desiderio MA. Influence of proteasome and redox state on heat shock-induced activation of stress kinases, AP-1 and HSF. Biochim Biophy Acta. 2001;1538:76–89.CrossRef
42.
Zurück zum Zitat Shelton JG, Blalock WL, White ER, Steelman LS, McCubrey JA. Ability of the activated PI3/AKT oncoproteins to synergize with MEK1 and induce cell cycle progression and abrogate the cytokine-dependence of hematopoietic cells. Cell Cycle. 2004;3:503–12.PubMed Shelton JG, Blalock WL, White ER, Steelman LS, McCubrey JA. Ability of the activated PI3/AKT oncoproteins to synergize with MEK1 and induce cell cycle progression and abrogate the cytokine-dependence of hematopoietic cells. Cell Cycle. 2004;3:503–12.PubMed
43.
Zurück zum Zitat Baldini E, Gardin G, Giannessi P, Brema F, Camorriano A, Carnino F, Naso C, Pastorino G, Pronzato P, Rosso R, Rubagotti A, Torreta G, Conte PF. A randomized trial of chemotherapy with or without estrogenic recruitment in locally advanced breast cancer. North-West Oncology Group (GONO) study, Italy. Tumori. 1997;83:829–33.PubMed Baldini E, Gardin G, Giannessi P, Brema F, Camorriano A, Carnino F, Naso C, Pastorino G, Pronzato P, Rosso R, Rubagotti A, Torreta G, Conte PF. A randomized trial of chemotherapy with or without estrogenic recruitment in locally advanced breast cancer. North-West Oncology Group (GONO) study, Italy. Tumori. 1997;83:829–33.PubMed
44.
Zurück zum Zitat Wang CY, Cusack Jr JC, Liu R, Baldwin Jr AS. Control of inducible chemoresistance: enhanced anti-tumor therapy through increased apoptosis by inhibition of NFκB. Nat Med. 1999;5:412–7.PubMedCrossRef Wang CY, Cusack Jr JC, Liu R, Baldwin Jr AS. Control of inducible chemoresistance: enhanced anti-tumor therapy through increased apoptosis by inhibition of NFκB. Nat Med. 1999;5:412–7.PubMedCrossRef
45.
Zurück zum Zitat Vanden Berghe W, Francesconi E, De Bosscher K, Resche Rigon M, Haegeman G. Dissociated glucocorticoids with anti inflammatory potential repress interleukin-6 gene expression by a nuclear factor-κB-dependent mechanism. Mol Pharmacol. 1999;56:797–806. Vanden Berghe W, Francesconi E, De Bosscher K, Resche Rigon M, Haegeman G. Dissociated glucocorticoids with anti inflammatory potential repress interleukin-6 gene expression by a nuclear factor-κB-dependent mechanism. Mol Pharmacol. 1999;56:797–806.
46.
Zurück zum Zitat Mellits KH, Hay RT, Goodbourn S. Proteolytic degradation of MAD3 (IκBα) and enhanced processing of the NFκB precursor p105 steps in the activation of NFκB. Nucleic Acids Res. 1993;21:5059–66.PubMedCrossRef Mellits KH, Hay RT, Goodbourn S. Proteolytic degradation of MAD3 (IκBα) and enhanced processing of the NFκB precursor p105 steps in the activation of NFκB. Nucleic Acids Res. 1993;21:5059–66.PubMedCrossRef
47.
Zurück zum Zitat Huong PT, Moon DO, Kim KE, Jeong SJ, Lee KW, Lee KS, Jang JH, Erikson RL, Ahn JS, Kim BY. Proteasome inhibitor-I enhances tunicamycin-induced chemosensitization of prostate cancer cells through regulation of NFκB and CHOP expression. Cell Signal. 2011;23:857–65.PubMedCrossRef Huong PT, Moon DO, Kim KE, Jeong SJ, Lee KW, Lee KS, Jang JH, Erikson RL, Ahn JS, Kim BY. Proteasome inhibitor-I enhances tunicamycin-induced chemosensitization of prostate cancer cells through regulation of NFκB and CHOP expression. Cell Signal. 2011;23:857–65.PubMedCrossRef
48.
Zurück zum Zitat O'Connor OA, Wright J, Moskowitz C, et al. Phase II clinical experience with the novel proteasome inhibitor bortezomib in patients with indolent non-Hodgkin's lymphoma and mantle cell lymphoma. J Clin Oncol. 2005;23:676–84.PubMedCrossRef O'Connor OA, Wright J, Moskowitz C, et al. Phase II clinical experience with the novel proteasome inhibitor bortezomib in patients with indolent non-Hodgkin's lymphoma and mantle cell lymphoma. J Clin Oncol. 2005;23:676–84.PubMedCrossRef
Metadaten
Titel
Synergistic induction of apoptosis and chemosensitization of human colorectal cancer cells by histone deacetylase inhibitor, scriptaid, and proteasome inhibitors: potential mechanisms of action
verfasst von
M. S. I. Abaza
A. M. Bahman
R. J. Al-Attiyah
A. M. Kollamparambil
Publikationsdatum
01.12.2012
Verlag
Springer Netherlands
Erschienen in
Tumor Biology / Ausgabe 6/2012
Print ISSN: 1010-4283
Elektronische ISSN: 1423-0380
DOI
https://doi.org/10.1007/s13277-012-0456-6

Weitere Artikel der Ausgabe 6/2012

Tumor Biology 6/2012 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.