Skip to main content
Top
Published in: Investigational New Drugs 1/2010

Open Access 01-12-2010 | SPECIAL ISSUE ARTICLE

The DAC system and associations with multiple myeloma

Authors: Enrique M. Ocio, Jesús F. San Miguel

Published in: Investigational New Drugs | Special Issue 1/2010

Login to get access

Summary

Despite the clear progress achieved in recent years in the treatment of MM, most patients eventually relapse and therefore novel therapeutic options are still necessary for these patients. In this regard, several drugs that target specific mechanisms of the tumor cells are currently being explored in the preclinical and clinical setting. This manuscripts offers a review of the rationale and current status of the antimyeloma activity of one of the most relevant examples of these targeted drugs: deacetylase inhibitors (DACi). Several studies have demonstrated the prooncogenic activity of deacetylases (DACs) through the targeting not only of histones but also of non histone proteins relevant to tumor progression, such as p53, E2F family members, Bcl-6, Hsp90, HIF-1α or Nur77. This fact together with the DACs overexpression present in several tumors, has prompted the development of some DACi with potential antitumor effect. This situation is also evident in the case of MM as two mechanisms of DACi, the inhibition of the epigenetic inactivation of p53 and the blockade of the unfolded protein response, through the inhibition of the aggressome formation (by targeting DAC6) and the inactivation of the chaperone system (by acetylating HSP-90), provides the rationale for the exploration of the potential antimyeloma activity of these compounds. Several DACi with different chemical structure and different selectivity for targeting the DAC families have been tested in MM. Their preclinical activity in monotherapy has been quite exciting and has been described to be mediated by various mechanisms: the induction of apoptosis and cell cycle arrest mainly by the upregulation of p21; the interferece with the interaction between plasma cells and the microenvironment, by reducing the expression and signalling of several cytokines or by inhibiting angiogenesis. Finally they also have a role in protecting murine models from myeloma bone disease. Neverteless, the clinical activity in monotherapy of these drugs in relapsed/refractory MM patients has been very modest. This has prompted the development of combinations such as the one with bortezomib or lenalidomide and dexamethasone, which have already been taken into the clinics with positive preliminary results.
Literature
2.
go back to reference Petrelli NJ, Winer EP, Brahmer J et al (2009) Clinical Cancer Advances 2009: major research advances in cancer treatment, prevention, and screening–a report from the American Society of Clinical Oncology. J Clin Oncol 27:6052–6069CrossRefPubMed Petrelli NJ, Winer EP, Brahmer J et al (2009) Clinical Cancer Advances 2009: major research advances in cancer treatment, prevention, and screening–a report from the American Society of Clinical Oncology. J Clin Oncol 27:6052–6069CrossRefPubMed
4.
go back to reference Kumar SK, Rajkumar SV, Dispenzieri A et al (2008) Improved survival in multiple myeloma and the impact of novel therapies. Blood 111:2516–2520CrossRefPubMed Kumar SK, Rajkumar SV, Dispenzieri A et al (2008) Improved survival in multiple myeloma and the impact of novel therapies. Blood 111:2516–2520CrossRefPubMed
5.
go back to reference Richardson PG, Sonneveld P, Schuster MW et al (2005) Bortezomib or high-dose dexamethasone for relapsed multiple myeloma. N Engl J Med 352:2487–2498CrossRefPubMed Richardson PG, Sonneveld P, Schuster MW et al (2005) Bortezomib or high-dose dexamethasone for relapsed multiple myeloma. N Engl J Med 352:2487–2498CrossRefPubMed
6.
go back to reference Singhal S, Mehta J, Desikan R et al (1999) Antitumor activity of thalidomide in refractory multiple myeloma. N Engl J Med 341:1565–1571CrossRefPubMed Singhal S, Mehta J, Desikan R et al (1999) Antitumor activity of thalidomide in refractory multiple myeloma. N Engl J Med 341:1565–1571CrossRefPubMed
7.
go back to reference Dimopoulos M, Spencer A, Attal M et al (2007) Lenalidomide plus dexamethasone for relapsed or refractory multiple myeloma. N Engl J Med 357:2123–2132CrossRefPubMed Dimopoulos M, Spencer A, Attal M et al (2007) Lenalidomide plus dexamethasone for relapsed or refractory multiple myeloma. N Engl J Med 357:2123–2132CrossRefPubMed
8.
go back to reference Weber DM, Chen C, Niesvizky R et al (2007) Lenalidomide plus dexamethasone for relapsed multiple myeloma in North America. N Engl J Med 357:2133–2142CrossRefPubMed Weber DM, Chen C, Niesvizky R et al (2007) Lenalidomide plus dexamethasone for relapsed multiple myeloma in North America. N Engl J Med 357:2133–2142CrossRefPubMed
9.
go back to reference Ocio EM, Mateos MV, Maiso P, Pandiella A, San-Miguel JF (2008) New drugs in multiple myeloma: mechanisms of action and phase I/II clinical findings. Lancet Oncol 9:1157–1165CrossRefPubMed Ocio EM, Mateos MV, Maiso P, Pandiella A, San-Miguel JF (2008) New drugs in multiple myeloma: mechanisms of action and phase I/II clinical findings. Lancet Oncol 9:1157–1165CrossRefPubMed
10.
go back to reference Mitsiades CS, Hideshima T, Chauhan D et al (2009) Emerging treatments for multiple myeloma: beyond immunomodulatory drugs and bortezomib. Semin Hematol 46:166–175CrossRefPubMed Mitsiades CS, Hideshima T, Chauhan D et al (2009) Emerging treatments for multiple myeloma: beyond immunomodulatory drugs and bortezomib. Semin Hematol 46:166–175CrossRefPubMed
11.
go back to reference Gregoretti IV, Lee YM, Goodson HV (2004) Molecular evolution of the histone deacetylase family: functional implications of phylogenetic analysis. J Mol Biol 338:17–31CrossRefPubMed Gregoretti IV, Lee YM, Goodson HV (2004) Molecular evolution of the histone deacetylase family: functional implications of phylogenetic analysis. J Mol Biol 338:17–31CrossRefPubMed
12.
go back to reference Dokmanovic M, Clarke C, Marks PA (2007) Histone deacetylase inhibitors: overview and perspectives. Mol Cancer Res 5:981–989CrossRefPubMed Dokmanovic M, Clarke C, Marks PA (2007) Histone deacetylase inhibitors: overview and perspectives. Mol Cancer Res 5:981–989CrossRefPubMed
13.
go back to reference de Ruijter AJ, van Gennip AH, Caron HN, Kemp S, van Kuilenburg AB (2003) Histone deacetylases (HDACs): characterization of the classical HDAC family. Biochem J 370:737–749CrossRefPubMed de Ruijter AJ, van Gennip AH, Caron HN, Kemp S, van Kuilenburg AB (2003) Histone deacetylases (HDACs): characterization of the classical HDAC family. Biochem J 370:737–749CrossRefPubMed
14.
go back to reference Witt O, Deubzer HE, Milde T, Oehme I (2009) HDAC family: What are the cancer relevant targets? Cancer Lett 277:8–21CrossRefPubMed Witt O, Deubzer HE, Milde T, Oehme I (2009) HDAC family: What are the cancer relevant targets? Cancer Lett 277:8–21CrossRefPubMed
15.
go back to reference Bolden JE, Peart MJ, Johnstone RW (2006) Anticancer activities of histone deacetylase inhibitors. Nat Rev Drug Discov 5:769–784CrossRefPubMed Bolden JE, Peart MJ, Johnstone RW (2006) Anticancer activities of histone deacetylase inhibitors. Nat Rev Drug Discov 5:769–784CrossRefPubMed
16.
go back to reference Minucci S, Pelicci PG (2006) Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer. Nat Rev Cancer 6:38–51CrossRefPubMed Minucci S, Pelicci PG (2006) Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer. Nat Rev Cancer 6:38–51CrossRefPubMed
17.
go back to reference Xu WS, Parmigiani RB, Marks PA (2007) Histone deacetylase inhibitors: molecular mechanisms of action. Oncogene 26:5541–5552CrossRefPubMed Xu WS, Parmigiani RB, Marks PA (2007) Histone deacetylase inhibitors: molecular mechanisms of action. Oncogene 26:5541–5552CrossRefPubMed
18.
go back to reference Hubbert C, Guardiola A, Shao R et al (2002) HDAC6 is a microtubule-associated deacetylase. Nature 417:455–458CrossRefPubMed Hubbert C, Guardiola A, Shao R et al (2002) HDAC6 is a microtubule-associated deacetylase. Nature 417:455–458CrossRefPubMed
19.
go back to reference Tran AD, Marmo TP, Salam AA et al (2007) HDAC6 deacetylation of tubulin modulates dynamics of cellular adhesions. J Cell Sci 120:1469–1479CrossRefPubMed Tran AD, Marmo TP, Salam AA et al (2007) HDAC6 deacetylation of tubulin modulates dynamics of cellular adhesions. J Cell Sci 120:1469–1479CrossRefPubMed
20.
go back to reference Bali P, Pranpat M, Bradner J et al (2005) Inhibition of histone deacetylase 6 acetylates and disrupts the chaperone function of heat shock protein 90: a novel basis for antileukemia activity of histone deacetylase inhibitors. J Biol Chem 280:26729–26734CrossRefPubMed Bali P, Pranpat M, Bradner J et al (2005) Inhibition of histone deacetylase 6 acetylates and disrupts the chaperone function of heat shock protein 90: a novel basis for antileukemia activity of histone deacetylase inhibitors. J Biol Chem 280:26729–26734CrossRefPubMed
22.
go back to reference Weichert W, Roske A, Gekeler V et al (2008) Association of patterns of class I histone deacetylase expression with patient prognosis in gastric cancer: a retrospective analysis. Lancet Oncol 9:139–148CrossRefPubMed Weichert W, Roske A, Gekeler V et al (2008) Association of patterns of class I histone deacetylase expression with patient prognosis in gastric cancer: a retrospective analysis. Lancet Oncol 9:139–148CrossRefPubMed
23.
go back to reference Weichert W, Roske A, Niesporek S et al (2008) Class I histone deacetylase expression has independent prognostic impact in human colorectal cancer: specific role of class I histone deacetylases in vitro and in vivo. Clin Cancer Res 14:1669–1677CrossRefPubMed Weichert W, Roske A, Niesporek S et al (2008) Class I histone deacetylase expression has independent prognostic impact in human colorectal cancer: specific role of class I histone deacetylases in vitro and in vivo. Clin Cancer Res 14:1669–1677CrossRefPubMed
24.
go back to reference Weichert W, Roske A, Gekeler V et al (2008) Histone deacetylases 1, 2 and 3 are highly expressed in prostate cancer and HDAC2 expression is associated with shorter PSA relapse time after radical prostatectomy. Br J Cancer 98:604–610CrossRefPubMed Weichert W, Roske A, Gekeler V et al (2008) Histone deacetylases 1, 2 and 3 are highly expressed in prostate cancer and HDAC2 expression is associated with shorter PSA relapse time after radical prostatectomy. Br J Cancer 98:604–610CrossRefPubMed
25.
go back to reference Marks P, Rifkind RA, Richon VM, Breslow R, Miller T, Kelly WK (2001) Histone deacetylases and cancer: causes and therapies. Nat Rev Cancer 1:194–202CrossRefPubMed Marks P, Rifkind RA, Richon VM, Breslow R, Miller T, Kelly WK (2001) Histone deacetylases and cancer: causes and therapies. Nat Rev Cancer 1:194–202CrossRefPubMed
26.
go back to reference Ozdag H, Teschendorff AE, Ahmed AA et al (2006) Differential expression of selected histone modifier genes in human solid cancers. BMC Genomics 7:90CrossRefPubMed Ozdag H, Teschendorff AE, Ahmed AA et al (2006) Differential expression of selected histone modifier genes in human solid cancers. BMC Genomics 7:90CrossRefPubMed
27.
go back to reference Sakuma T, Uzawa K, Onda T et al (2006) Aberrant expression of histone deacetylase 6 in oral squamous cell carcinoma. Int J Oncol 29:117–124PubMed Sakuma T, Uzawa K, Onda T et al (2006) Aberrant expression of histone deacetylase 6 in oral squamous cell carcinoma. Int J Oncol 29:117–124PubMed
28.
go back to reference Saji S, Kawakami M, Hayashi S et al (2005) Significance of HDAC6 regulation via estrogen signaling for cell motility and prognosis in estrogen receptor-positive breast cancer. Oncogene 24:4531–4539CrossRefPubMed Saji S, Kawakami M, Hayashi S et al (2005) Significance of HDAC6 regulation via estrogen signaling for cell motility and prognosis in estrogen receptor-positive breast cancer. Oncogene 24:4531–4539CrossRefPubMed
29.
go back to reference Zhang Z, Yamashita H, Toyama T et al (2004) HDAC6 expression is correlated with better survival in breast cancer. Clin Cancer Res 10:6962–6968CrossRefPubMed Zhang Z, Yamashita H, Toyama T et al (2004) HDAC6 expression is correlated with better survival in breast cancer. Clin Cancer Res 10:6962–6968CrossRefPubMed
30.
go back to reference Linggi BE, Brandt SJ, Sun ZW, Hiebert SW (2005) Translating the histone code into leukemia. J Cell Biochem 96:938–950CrossRefPubMed Linggi BE, Brandt SJ, Sun ZW, Hiebert SW (2005) Translating the histone code into leukemia. J Cell Biochem 96:938–950CrossRefPubMed
31.
go back to reference Choi Y, Elagib KE, Goldfarb AN (2005) AML-1-ETO-Mediated erythroid inhibition: new paradigms for differentiation blockade by a leukemic fusion protein. Crit Rev Eukaryot Gene Expr 15:207–216PubMed Choi Y, Elagib KE, Goldfarb AN (2005) AML-1-ETO-Mediated erythroid inhibition: new paradigms for differentiation blockade by a leukemic fusion protein. Crit Rev Eukaryot Gene Expr 15:207–216PubMed
32.
go back to reference Amann JM, Nip J, Strom DK et al (2001) ETO, a target of t(8;21) in acute leukemia, makes distinct contacts with multiple histone deacetylases and binds mSin3A through its oligomerization domain. Mol Cell Biol 21:6470–6483CrossRefPubMed Amann JM, Nip J, Strom DK et al (2001) ETO, a target of t(8;21) in acute leukemia, makes distinct contacts with multiple histone deacetylases and binds mSin3A through its oligomerization domain. Mol Cell Biol 21:6470–6483CrossRefPubMed
33.
go back to reference Atsumi A, Tomita A, Kiyoi H, Naoe T (2006) Histone deacetylase 3 (HDAC3) is recruited to target promoters by PML-RARalpha as a component of the N-CoR co-repressor complex to repress transcription in vivo. Biochem Biophys Res Commun 345:1471–1480CrossRefPubMed Atsumi A, Tomita A, Kiyoi H, Naoe T (2006) Histone deacetylase 3 (HDAC3) is recruited to target promoters by PML-RARalpha as a component of the N-CoR co-repressor complex to repress transcription in vivo. Biochem Biophys Res Commun 345:1471–1480CrossRefPubMed
34.
go back to reference Chauchereau A, Mathieu M, de Saintignon SJ et al (2004) HDAC4 mediates transcriptional repression by the acute promyelocytic leukaemia-associated protein PLZF. Oncogene 23:8777–8784CrossRefPubMed Chauchereau A, Mathieu M, de Saintignon SJ et al (2004) HDAC4 mediates transcriptional repression by the acute promyelocytic leukaemia-associated protein PLZF. Oncogene 23:8777–8784CrossRefPubMed
35.
go back to reference Xiong W, Wu X, Starnes S et al (2008) An analysis of the clinical and biologic significance of TP53 loss and the identification of potential novel transcriptional targets of TP53 in multiple myeloma. Blood 112:4235–4246CrossRefPubMed Xiong W, Wu X, Starnes S et al (2008) An analysis of the clinical and biologic significance of TP53 loss and the identification of potential novel transcriptional targets of TP53 in multiple myeloma. Blood 112:4235–4246CrossRefPubMed
36.
go back to reference Gutierrez NC, Castellanos MV, Martin ML et al (2007) Prognostic and biological implications of genetic abnormalities in multiple myeloma undergoing autologous stem cell transplantation: t(4;14) is the most relevant adverse prognostic factor, whereas RB deletion as a unique abnormality is not associated with adverse prognosis. Leukemia 21:143–150CrossRefPubMed Gutierrez NC, Castellanos MV, Martin ML et al (2007) Prognostic and biological implications of genetic abnormalities in multiple myeloma undergoing autologous stem cell transplantation: t(4;14) is the most relevant adverse prognostic factor, whereas RB deletion as a unique abnormality is not associated with adverse prognosis. Leukemia 21:143–150CrossRefPubMed
37.
go back to reference Chang H, Qi C, Yi QL, Reece D, Stewart AK (2005) p53 gene deletion detected by fluorescence in situ hybridization is an adverse prognostic factor for patients with multiple myeloma following autologous stem cell transplantation. Blood 105:358–360CrossRefPubMed Chang H, Qi C, Yi QL, Reece D, Stewart AK (2005) p53 gene deletion detected by fluorescence in situ hybridization is an adverse prognostic factor for patients with multiple myeloma following autologous stem cell transplantation. Blood 105:358–360CrossRefPubMed
38.
go back to reference Chng WJ, Price-Troska T, Gonzalez-Paz N et al (2007) Clinical significance of TP53 mutation in myeloma. Leukemia 21:582–584CrossRefPubMed Chng WJ, Price-Troska T, Gonzalez-Paz N et al (2007) Clinical significance of TP53 mutation in myeloma. Leukemia 21:582–584CrossRefPubMed
39.
go back to reference Ocio EM, Maiso P, Chen X et al (2009) Zalypsis: a novel marine-derived compound with potent antimyeloma activity that reveals high sensitivity of malignant plasma cells to DNA double-strand breaks. Blood 113:3781–3791CrossRefPubMed Ocio EM, Maiso P, Chen X et al (2009) Zalypsis: a novel marine-derived compound with potent antimyeloma activity that reveals high sensitivity of malignant plasma cells to DNA double-strand breaks. Blood 113:3781–3791CrossRefPubMed
40.
go back to reference Hurt EM, Thomas SB, Peng B, Farrar WL (2006) Reversal of p53 epigenetic silencing in multiple myeloma permits apoptosis by a p53 activator. Cancer Biol Ther 5:1154–1160CrossRefPubMed Hurt EM, Thomas SB, Peng B, Farrar WL (2006) Reversal of p53 epigenetic silencing in multiple myeloma permits apoptosis by a p53 activator. Cancer Biol Ther 5:1154–1160CrossRefPubMed
41.
go back to reference Davenport EL, Moore HE, Dunlop AS et al (2007) Heat shock protein inhibition is associated with activation of the unfolded protein response pathway in myeloma plasma cells. Blood 110:2641–2649CrossRefPubMed Davenport EL, Moore HE, Dunlop AS et al (2007) Heat shock protein inhibition is associated with activation of the unfolded protein response pathway in myeloma plasma cells. Blood 110:2641–2649CrossRefPubMed
42.
go back to reference Davenport EL, Morgan GJ, Davies FE (2008) Untangling the unfolded protein response. Cell Cycle 7:865–869CrossRefPubMed Davenport EL, Morgan GJ, Davies FE (2008) Untangling the unfolded protein response. Cell Cycle 7:865–869CrossRefPubMed
43.
go back to reference Bennett EJ, Bence NF, Jayakumar R, Kopito RR (2005) Global impairment of the ubiquitin-proteasome system by nuclear or cytoplasmic protein aggregates precedes inclusion body formation. Mol Cell 17:351–365CrossRefPubMed Bennett EJ, Bence NF, Jayakumar R, Kopito RR (2005) Global impairment of the ubiquitin-proteasome system by nuclear or cytoplasmic protein aggregates precedes inclusion body formation. Mol Cell 17:351–365CrossRefPubMed
44.
go back to reference Kopito RR (2000) Aggresomes, inclusion bodies and protein aggregation. Trends Cell Biol 10:524–530CrossRefPubMed Kopito RR (2000) Aggresomes, inclusion bodies and protein aggregation. Trends Cell Biol 10:524–530CrossRefPubMed
45.
go back to reference Kawaguchi Y, Kovacs JJ, McLaurin A, Vance JM, Ito A, Yao TP (2003) The deacetylase HDAC6 regulates aggresome formation and cell viability in response to misfolded protein stress. Cell 115:727–738CrossRefPubMed Kawaguchi Y, Kovacs JJ, McLaurin A, Vance JM, Ito A, Yao TP (2003) The deacetylase HDAC6 regulates aggresome formation and cell viability in response to misfolded protein stress. Cell 115:727–738CrossRefPubMed
46.
47.
go back to reference Rasheed WK, Johnstone RW, Prince HM (2007) Histone deacetylase inhibitors in cancer therapy. Expert Opin Investig Drugs 16:659–678CrossRefPubMed Rasheed WK, Johnstone RW, Prince HM (2007) Histone deacetylase inhibitors in cancer therapy. Expert Opin Investig Drugs 16:659–678CrossRefPubMed
48.
go back to reference Haggarty SJ, Koeller KM, Wong JC, Grozinger CM, Schreiber SL (2003) Domain-selective small-molecule inhibitor of histone deacetylase 6 (HDAC6)-mediated tubulin deacetylation. Proc Natl Acad Sci USA 100:4389–4394CrossRefPubMed Haggarty SJ, Koeller KM, Wong JC, Grozinger CM, Schreiber SL (2003) Domain-selective small-molecule inhibitor of histone deacetylase 6 (HDAC6)-mediated tubulin deacetylation. Proc Natl Acad Sci USA 100:4389–4394CrossRefPubMed
49.
go back to reference Mitsiades N, Mitsiades CS, Richardson PG et al (2003) Molecular sequelae of histone deacetylase inhibition in human malignant B cells. Blood 101:4055–4062CrossRefPubMed Mitsiades N, Mitsiades CS, Richardson PG et al (2003) Molecular sequelae of histone deacetylase inhibition in human malignant B cells. Blood 101:4055–4062CrossRefPubMed
50.
go back to reference Mitsiades CS, Mitsiades NS, McMullan CJ et al (2004) Transcriptional signature of histone deacetylase inhibition in multiple myeloma: biological and clinical implications. Proc Natl Acad Sci USA 101:540–545CrossRefPubMed Mitsiades CS, Mitsiades NS, McMullan CJ et al (2004) Transcriptional signature of histone deacetylase inhibition in multiple myeloma: biological and clinical implications. Proc Natl Acad Sci USA 101:540–545CrossRefPubMed
51.
go back to reference Lavelle D, Chen YH, Hankewych M, DeSimone J (2001) Histone deacetylase inhibitors increase p21(WAF1) and induce apoptosis of human myeloma cell lines independent of decreased IL-6 receptor expression. Am J Hematol 68:170–178CrossRefPubMed Lavelle D, Chen YH, Hankewych M, DeSimone J (2001) Histone deacetylase inhibitors increase p21(WAF1) and induce apoptosis of human myeloma cell lines independent of decreased IL-6 receptor expression. Am J Hematol 68:170–178CrossRefPubMed
52.
go back to reference Catley L, Weisberg E, Tai YT et al (2003) NVP-LAQ824 is a potent novel histone deacetylase inhibitor with significant activity against multiple myeloma. Blood 102:2615–2622CrossRefPubMed Catley L, Weisberg E, Tai YT et al (2003) NVP-LAQ824 is a potent novel histone deacetylase inhibitor with significant activity against multiple myeloma. Blood 102:2615–2622CrossRefPubMed
53.
go back to reference Khan SB, Maududi T, Barton K, Ayers J, Alkan S (2004) Analysis of histone deacetylase inhibitor, depsipeptide (FR901228), effect on multiple myeloma. Br J Haematol 125:156–161CrossRefPubMed Khan SB, Maududi T, Barton K, Ayers J, Alkan S (2004) Analysis of histone deacetylase inhibitor, depsipeptide (FR901228), effect on multiple myeloma. Br J Haematol 125:156–161CrossRefPubMed
54.
go back to reference Kaiser M, Zavrski I, Sterz J et al (2006) The effects of the histone deacetylase inhibitor valproic acid on cell cycle, growth suppression and apoptosis in multiple myeloma. Haematologica 91:248–251PubMed Kaiser M, Zavrski I, Sterz J et al (2006) The effects of the histone deacetylase inhibitor valproic acid on cell cycle, growth suppression and apoptosis in multiple myeloma. Haematologica 91:248–251PubMed
55.
go back to reference Schwartz C, Palissot V, Aouali N et al (2007) Valproic acid induces non-apoptotic cell death mechanisms in multiple myeloma cell lines. Int J Oncol 30:573–582PubMed Schwartz C, Palissot V, Aouali N et al (2007) Valproic acid induces non-apoptotic cell death mechanisms in multiple myeloma cell lines. Int J Oncol 30:573–582PubMed
56.
go back to reference Kitazoe K, Abe M, Hiasa M et al (2009) Valproic acid exerts anti-tumor as well as anti-angiogenic effects on myeloma. Int J Hematol 89:45–57CrossRefPubMed Kitazoe K, Abe M, Hiasa M et al (2009) Valproic acid exerts anti-tumor as well as anti-angiogenic effects on myeloma. Int J Hematol 89:45–57CrossRefPubMed
57.
go back to reference Neri P, Tagliaferri P, Di Martino MT et al (2008) In vivo anti-myeloma activity and modulation of gene expression profile induced by valproic acid, a histone deacetylase inhibitor. Br J Haematol 143:520–531PubMed Neri P, Tagliaferri P, Di Martino MT et al (2008) In vivo anti-myeloma activity and modulation of gene expression profile induced by valproic acid, a histone deacetylase inhibitor. Br J Haematol 143:520–531PubMed
58.
go back to reference Catley L, Weisberg E, Kiziltepe T et al (2006) Aggresome induction by proteasome inhibitor bortezomib and alpha-tubulin hyperacetylation by tubulin deacetylase (TDAC) inhibitor LBH589 are synergistic in myeloma cells. Blood 108:3441–3449CrossRefPubMed Catley L, Weisberg E, Kiziltepe T et al (2006) Aggresome induction by proteasome inhibitor bortezomib and alpha-tubulin hyperacetylation by tubulin deacetylase (TDAC) inhibitor LBH589 are synergistic in myeloma cells. Blood 108:3441–3449CrossRefPubMed
59.
go back to reference Maiso P, Carvajal-Vergara X, Ocio EM et al (2006) The histone deacetylase inhibitor LBH589 is a potent antimyeloma agent that overcomes drug resistance. Cancer Res 66:5781–5789CrossRefPubMed Maiso P, Carvajal-Vergara X, Ocio EM et al (2006) The histone deacetylase inhibitor LBH589 is a potent antimyeloma agent that overcomes drug resistance. Cancer Res 66:5781–5789CrossRefPubMed
60.
go back to reference Ocio EM, Vilanova D, Atadja P et al (2009) In vitro and in vivo rationale for the triple combination of panobinostat (LBH589) and dexamethasone with either bortezomib or lenalidomide in multiple myeloma. Haematologica Ocio EM, Vilanova D, Atadja P et al (2009) In vitro and in vivo rationale for the triple combination of panobinostat (LBH589) and dexamethasone with either bortezomib or lenalidomide in multiple myeloma. Haematologica
61.
go back to reference Feng R, Oton A, Mapara MY, Anderson G, Belani C, Lentzsch S (2007) The histone deacetylase inhibitor, PXD101, potentiates bortezomib-induced anti-multiple myeloma effect by induction of oxidative stress and DNA damage. Br J Haematol 139:385–397CrossRefPubMed Feng R, Oton A, Mapara MY, Anderson G, Belani C, Lentzsch S (2007) The histone deacetylase inhibitor, PXD101, potentiates bortezomib-induced anti-multiple myeloma effect by induction of oxidative stress and DNA damage. Br J Haematol 139:385–397CrossRefPubMed
62.
go back to reference Arts J, Angibaud P, Marien A et al (2007) R306465 is a novel potent inhibitor of class I histone deacetylases with broad-spectrum antitumoral activity against solid and haematological malignancies. Br J Cancer 97:1344–1353CrossRefPubMed Arts J, Angibaud P, Marien A et al (2007) R306465 is a novel potent inhibitor of class I histone deacetylases with broad-spectrum antitumoral activity against solid and haematological malignancies. Br J Cancer 97:1344–1353CrossRefPubMed
63.
go back to reference Feng R, Ma H, Hassig CA et al (2008) KD5170, a novel mercaptoketone-based histone deacetylase inhibitor, exerts antimyeloma effects by DNA damage and mitochondrial signaling. Mol Cancer Ther 7:1494–1505CrossRefPubMed Feng R, Ma H, Hassig CA et al (2008) KD5170, a novel mercaptoketone-based histone deacetylase inhibitor, exerts antimyeloma effects by DNA damage and mitochondrial signaling. Mol Cancer Ther 7:1494–1505CrossRefPubMed
64.
go back to reference Deleu S, Lemaire M, Arts J et al (2009) The effects of JNJ-26481585, a novel hydroxamate-based histone deacetylase inhibitor, on the development of multiple myeloma in the 5T2MM and 5T33MM murine models. Leukemia 23:1894–1903CrossRefPubMed Deleu S, Lemaire M, Arts J et al (2009) The effects of JNJ-26481585, a novel hydroxamate-based histone deacetylase inhibitor, on the development of multiple myeloma in the 5T2MM and 5T33MM murine models. Leukemia 23:1894–1903CrossRefPubMed
65.
go back to reference Todoerti K, Barbui V, Pedrini O et al Pleiotropic anti-myeloma activity of ITF2357: inhibition of interleukin-6 receptor signaling and repression of miR-19a and miR-19b. Haematologica 95:260–269 Todoerti K, Barbui V, Pedrini O et al Pleiotropic anti-myeloma activity of ITF2357: inhibition of interleukin-6 receptor signaling and repression of miR-19a and miR-19b. Haematologica 95:260–269
66.
go back to reference Mandl-Weber S, Meinel F, Jankowsky R, Oduncu F, Schmidmaier R, Baumann P The novel inhibitor of histone deacetylase resminostat (RAS2410) inhibits proliferation and induces apoptosis in multiple myeloma (MM) cells. Br J Haematol Mandl-Weber S, Meinel F, Jankowsky R, Oduncu F, Schmidmaier R, Baumann P The novel inhibitor of histone deacetylase resminostat (RAS2410) inhibits proliferation and induces apoptosis in multiple myeloma (MM) cells. Br J Haematol
67.
go back to reference Fandy TE, Shankar S, Ross DD, Sausville E, Srivastava RK (2005) Interactive effects of HDAC inhibitors and TRAIL on apoptosis are associated with changes in mitochondrial functions and expressions of cell cycle regulatory genes in multiple myeloma. Neoplasia 7:646–657CrossRefPubMed Fandy TE, Shankar S, Ross DD, Sausville E, Srivastava RK (2005) Interactive effects of HDAC inhibitors and TRAIL on apoptosis are associated with changes in mitochondrial functions and expressions of cell cycle regulatory genes in multiple myeloma. Neoplasia 7:646–657CrossRefPubMed
68.
go back to reference Akiyama M, Hideshima T, Hayashi T et al (2002) Cytokines modulate telomerase activity in a human multiple myeloma cell line. Cancer Res 62:3876–3882PubMed Akiyama M, Hideshima T, Hayashi T et al (2002) Cytokines modulate telomerase activity in a human multiple myeloma cell line. Cancer Res 62:3876–3882PubMed
69.
go back to reference Chen S, Dai Y, Pei XY, Grant S (2009) Bim upregulation by histone deacetylase inhibitors mediates interactions with the Bcl-2 antagonist ABT-737: evidence for distinct roles for Bcl-2, Bcl-xL, and Mcl-1. Mol Cell Biol 29:6149–6169CrossRefPubMed Chen S, Dai Y, Pei XY, Grant S (2009) Bim upregulation by histone deacetylase inhibitors mediates interactions with the Bcl-2 antagonist ABT-737: evidence for distinct roles for Bcl-2, Bcl-xL, and Mcl-1. Mol Cell Biol 29:6149–6169CrossRefPubMed
70.
go back to reference Damiano JS, Cress AE, Hazlehurst LA, Shtil AA, Dalton WS (1999) Cell adhesion mediated drug resistance (CAM-DR): role of integrins and resistance to apoptosis in human myeloma cell lines. Blood 93:1658–1667PubMed Damiano JS, Cress AE, Hazlehurst LA, Shtil AA, Dalton WS (1999) Cell adhesion mediated drug resistance (CAM-DR): role of integrins and resistance to apoptosis in human myeloma cell lines. Blood 93:1658–1667PubMed
71.
go back to reference Hideshima T, Mitsiades C, Tonon G, Richardson PG, Anderson KC (2007) Understanding multiple myeloma pathogenesis in the bone marrow to identify new therapeutic targets. Nat Rev Cancer 7:585–598CrossRefPubMed Hideshima T, Mitsiades C, Tonon G, Richardson PG, Anderson KC (2007) Understanding multiple myeloma pathogenesis in the bone marrow to identify new therapeutic targets. Nat Rev Cancer 7:585–598CrossRefPubMed
72.
go back to reference Wolf JL, Siegel D, Matous J et al (2008) A Phase II Study of Oral Panobinostat (LBH589) in Adult Patients with Advanced Refractory Multiple Myeloma. ASH Annu Meet Abstr 112:2774 Wolf JL, Siegel D, Matous J et al (2008) A Phase II Study of Oral Panobinostat (LBH589) in Adult Patients with Advanced Refractory Multiple Myeloma. ASH Annu Meet Abstr 112:2774
73.
go back to reference Richardson P, Mitsiades C, Colson K et al (2008) Phase I trial of oral vorinostat (suberoylanilide hydroxamic acid, SAHA) in patients with advanced multiple myeloma. Leuk Lymphoma 49:502–507CrossRefPubMed Richardson P, Mitsiades C, Colson K et al (2008) Phase I trial of oral vorinostat (suberoylanilide hydroxamic acid, SAHA) in patients with advanced multiple myeloma. Leuk Lymphoma 49:502–507CrossRefPubMed
74.
go back to reference Galli M, Salmoiraghi S, Golay J et al A phase II multiple dose clinical trial of histone deacetylase inhibitor ITF2357 in patients with relapsed or progressive multiple myeloma. Ann Hematol 89:185–190 Galli M, Salmoiraghi S, Golay J et al A phase II multiple dose clinical trial of histone deacetylase inhibitor ITF2357 in patients with relapsed or progressive multiple myeloma. Ann Hematol 89:185–190
75.
go back to reference Niesvizky R, Ely S, DiLiberto M et al (2005) Multicenter Phase II Trial of the Histone Deacetylase Inhibitor Depsipeptide (FK228) for the Treatment of Relapsed or Refractory Multiple Myeloma (MM). ASH Annu Meet Abstr 106:2574- Niesvizky R, Ely S, DiLiberto M et al (2005) Multicenter Phase II Trial of the Histone Deacetylase Inhibitor Depsipeptide (FK228) for the Treatment of Relapsed or Refractory Multiple Myeloma (MM). ASH Annu Meet Abstr 106:2574-
76.
go back to reference Campbell RA, Sanchez E, Steinberg J et al Vorinostat enhances the antimyeloma effects of melphalan and bortezomib. Eur J Haematol 84:201–211 Campbell RA, Sanchez E, Steinberg J et al Vorinostat enhances the antimyeloma effects of melphalan and bortezomib. Eur J Haematol 84:201–211
77.
go back to reference Deleu S, Lemaire M, Arts J et al (2009) Bortezomib alone or in combination with the histone deacetylase inhibitor JNJ-26481585: effect on myeloma bone disease in the 5T2MM murine model of myeloma. Cancer Res 69:5307–5311CrossRefPubMed Deleu S, Lemaire M, Arts J et al (2009) Bortezomib alone or in combination with the histone deacetylase inhibitor JNJ-26481585: effect on myeloma bone disease in the 5T2MM murine model of myeloma. Cancer Res 69:5307–5311CrossRefPubMed
78.
go back to reference Hideshima T, Bradner JE, Wong J et al (2005) Small-molecule inhibition of proteasome and aggresome function induces synergistic antitumor activity in multiple myeloma. Proc Natl Acad Sci USA 102:8567–8572CrossRefPubMed Hideshima T, Bradner JE, Wong J et al (2005) Small-molecule inhibition of proteasome and aggresome function induces synergistic antitumor activity in multiple myeloma. Proc Natl Acad Sci USA 102:8567–8572CrossRefPubMed
79.
go back to reference Pei XY, Dai Y, Grant S (2004) Synergistic induction of oxidative injury and apoptosis in human multiple myeloma cells by the proteasome inhibitor bortezomib and histone deacetylase inhibitors. Clin Cancer Res 10:3839–3852CrossRefPubMed Pei XY, Dai Y, Grant S (2004) Synergistic induction of oxidative injury and apoptosis in human multiple myeloma cells by the proteasome inhibitor bortezomib and histone deacetylase inhibitors. Clin Cancer Res 10:3839–3852CrossRefPubMed
80.
go back to reference Badros A, Burger AM, Philip S et al (2009) Phase I study of vorinostat in combination with bortezomib for relapsed and refractory multiple myeloma. Clin Cancer Res 15:5250–5257CrossRefPubMed Badros A, Burger AM, Philip S et al (2009) Phase I study of vorinostat in combination with bortezomib for relapsed and refractory multiple myeloma. Clin Cancer Res 15:5250–5257CrossRefPubMed
81.
go back to reference Weber D, Badros AZ, Jagannath S et al (2008) Vorinostat plus bortezomib for the treatment of relapsed/refractory multiple myeloma: early clinical experience. ASH Annu Meet Abstr 112:871 Weber D, Badros AZ, Jagannath S et al (2008) Vorinostat plus bortezomib for the treatment of relapsed/refractory multiple myeloma: early clinical experience. ASH Annu Meet Abstr 112:871
82.
go back to reference Weber DM, Jagannath S, Sobecks R et al (2008) Combination of vorinostat plus bortezomib for the treatment of patients with multiple myeloma who have previously received bortezomib. ASH Annu Meet Abstr 112:3711 Weber DM, Jagannath S, Sobecks R et al (2008) Combination of vorinostat plus bortezomib for the treatment of patients with multiple myeloma who have previously received bortezomib. ASH Annu Meet Abstr 112:3711
83.
go back to reference San-Miguel JF, Sezer O, Siegel D et al (2009) A phase IB, multi-center, open-label dose-escalation study of oral panobinostat (LBH589) and I.V. Bortezomib in patients with relapsed multiple myeloma. ASH Annu Meet Abstr 114:3852- San-Miguel JF, Sezer O, Siegel D et al (2009) A phase IB, multi-center, open-label dose-escalation study of oral panobinostat (LBH589) and I.V. Bortezomib in patients with relapsed multiple myeloma. ASH Annu Meet Abstr 114:3852-
84.
go back to reference Harrison SJ, Quach H, Yuen K et al (2008) High response rates with the combination of bortezomib, dexamethasone and the pan-histone deacetylase inhibitor romidepsin in patients with relapsed or refractory multiple myeloma in a phase I/II clinical trial. ASH Annu Meet Abstr 112:3698 Harrison SJ, Quach H, Yuen K et al (2008) High response rates with the combination of bortezomib, dexamethasone and the pan-histone deacetylase inhibitor romidepsin in patients with relapsed or refractory multiple myeloma in a phase I/II clinical trial. ASH Annu Meet Abstr 112:3698
85.
go back to reference Voorhees PM, Gasparetto C, Richards KL et al (2009) Vorinostat in combination with pegylated liposomal doxorubicin and bortezomib for patients with relapsed/refractory multiple myeloma: results of a phase I study. ASH Annu Meet Abstr 114:306- Voorhees PM, Gasparetto C, Richards KL et al (2009) Vorinostat in combination with pegylated liposomal doxorubicin and bortezomib for patients with relapsed/refractory multiple myeloma: results of a phase I study. ASH Annu Meet Abstr 114:306-
86.
go back to reference Spencer A, Lonial S, Taylor K et al (2009) Panobinostat and lenalidomide combination phase I trial in myeloma. Clin Lymphoma Myeloma. Abstract 329 Spencer A, Lonial S, Taylor K et al (2009) Panobinostat and lenalidomide combination phase I trial in myeloma. Clin Lymphoma Myeloma. Abstract 329
87.
go back to reference Siegel D, Weber DM, Mitsiades CS et al (2009) Combined vorinostat, lenalidomide and dexamethasone therapy in patients with relapsed or refractory multiple myeloma: a phase I study. ASH Annu Meet Abstr 114:305- Siegel D, Weber DM, Mitsiades CS et al (2009) Combined vorinostat, lenalidomide and dexamethasone therapy in patients with relapsed or refractory multiple myeloma: a phase I study. ASH Annu Meet Abstr 114:305-
88.
go back to reference Berenson JR, Yellin O, Boccia RV, Nassir Y, Rothstein S, Swift RA (2009) A phase I study of oral melphalan combined with LBH589 for patients with relapsed or refractory multiple myeloma (MM). ASH Annu Meet Abstr 114:1855- Berenson JR, Yellin O, Boccia RV, Nassir Y, Rothstein S, Swift RA (2009) A phase I study of oral melphalan combined with LBH589 for patients with relapsed or refractory multiple myeloma (MM). ASH Annu Meet Abstr 114:1855-
Metadata
Title
The DAC system and associations with multiple myeloma
Authors
Enrique M. Ocio
Jesús F. San Miguel
Publication date
01-12-2010
Publisher
Springer US
Published in
Investigational New Drugs / Issue Special Issue 1/2010
Print ISSN: 0167-6997
Electronic ISSN: 1573-0646
DOI
https://doi.org/10.1007/s10637-010-9589-x

Other articles of this Special Issue 1/2010

Investigational New Drugs 1/2010 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