Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Rep ; 5(4): 1047-59, 2013 Nov 27.
Article in English | MEDLINE | ID: mdl-24268771

ABSTRACT

To design rational therapies for JAK2-driven hematological malignancies, we functionally dissected the key survival pathways downstream of hyperactive JAK2. In tumors driven by mutant JAK2, Stat1, Stat3, Stat5, and the Pi3k and Mek/Erk pathways were constitutively active, and gene expression profiling of TEL-JAK2 T-ALL cells revealed the upregulation of prosurvival Bcl-2 family genes. Combining the Bcl-2/Bcl-xL inhibitor ABT-737 with JAK2 inhibitors mediated prolonged disease regressions and cures in mice bearing primary human and mouse JAK2 mutant tumors. Moreover, combined targeting of JAK2 and Bcl-2/Bcl-xL was able to circumvent and overcome acquired resistance to single-agent JAK2 inhibitor treatment. Thus, inhibiting the oncogenic JAK2 signaling network at two nodal points, at the initiating stage (JAK2) and the effector stage (Bcl-2/Bcl-xL), is highly effective and provides a clearly superior therapeutic benefit than targeting just one node. Therefore, we have defined a potentially curative treatment for hematological malignancies expressing constitutively active JAK2.


Subject(s)
Drug Resistance, Neoplasm , Janus Kinase 2/antagonists & inhibitors , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , bcl-X Protein/antagonists & inhibitors , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Biphenyl Compounds/pharmacology , Cell Line, Tumor , Cell Survival , Gene Expression Profiling , Humans , Janus Kinase 2/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Nitriles , Nitrophenols/pharmacology , Piperazines/pharmacology , Proto-Oncogene Proteins/genetics , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects , Sulfonamides/pharmacology , Transplantation, Heterologous , bcl-X Protein/genetics
2.
Mol Cancer Ther ; 12(12): 2709-21, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24092806

ABSTRACT

Histone deacetylase inhibitors (HDACi) are anticancer agents that induce hyperacetylation of histones, resulting in chromatin remodeling and transcriptional changes. In addition, nonhistone proteins, such as the chaperone protein Hsp90, are functionally regulated through hyperacetylation mediated by HDACis. Histone acetylation is thought to be primarily regulated by HDACs 1, 2, and 3, whereas the acetylation of Hsp90 has been proposed to be specifically regulated through HDAC6. We compared the molecular and biologic effects induced by an HDACi with broad HDAC specificity (vorinostat) with agents that predominantly inhibited selected class I HDACs (MRLB-223 and romidepsin). MRLB-223, a potent inhibitor of HDACs 1 and 2, killed tumor cells using the same apoptotic pathways as the HDAC 1, 2, 3, 6, and 8 inhibitor vorinostat. However, vorinostat induced histone hyperacetylation and killed tumor cells more rapidly than MRLB-223 and had greater therapeutic efficacy in vivo. FDCP-1 cells dependent on the Hsp90 client protein Bcr-Abl for survival, were killed by all HDACis tested, concomitant with caspase-dependent degradation of Bcr-Abl. These studies provide evidence that inhibition of HDAC6 and degradation of Bcr-Abl following hyperacetylation of Hsp90 is likely not a major mechanism of action of HDACis as had been previously posited.


Subject(s)
Antineoplastic Agents/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Acetylation/drug effects , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Enzyme Activation/drug effects , Fusion Proteins, bcr-abl/metabolism , HSP90 Heat-Shock Proteins/metabolism , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 2/antagonists & inhibitors , Histone Deacetylase 6 , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/pharmacology , Lymphoma/drug therapy , Lymphoma/metabolism , Lymphoma/mortality , Lymphoma/pathology , Mice , Vorinostat , Xenograft Model Antitumor Assays
3.
Cancer Res ; 71(10): 3603-15, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21398407

ABSTRACT

The concept of personalized anticancer therapy is based on the use of targeted therapeutics through in-depth knowledge of the molecular mechanisms of action of these agents when used alone and in combination. We have identified the apoptotic proteins and pathways necessary for synergistic tumor cell apoptosis and in vivo antitumor responses seen when the HDAC inhibitor vorinostat is combined with the BH3-mimetic ABT-737 in lymphomas overexpressing Bcl-2. Vorinostat "primes" tumors overexpressing Bcl-2 for rapid ABT-737-mediated apoptosis by inducing expression of the BH3-only gene bmf. Moreover, these synergistic effects of vorinostat/ABT-737 were blunted in cells with an inactive p53 pathway or in cells lacking expression of the p53 target gene, noxa. These studies show the important and complex functional interaction between specific proapoptotic BH3-only proteins and the BH3-mimetic compound ABT-737 and provide the most comprehensive functional link between tumor genotype and the apoptotic and therapeutic effects of HDACi combined with ABT-737.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Animals , Apoptosis , Biphenyl Compounds/pharmacology , Cell Line, Tumor , Female , Genes, p53 , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Nitrophenols/pharmacology , Peptide Fragments/metabolism , Piperazines/pharmacology , Proto-Oncogene Proteins/metabolism , Sulfonamides/pharmacology , Tumor Suppressor Protein p53/genetics , Vorinostat
4.
Proc Natl Acad Sci U S A ; 108(10): 4141-6, 2011 Mar 08.
Article in English | MEDLINE | ID: mdl-21368108

ABSTRACT

Histone deacetylase inhibitors (HDACi) have been successfully used as monotherapies for the treatment of hematological malignancies; however, the single agent effects of HDACi against solid tumors are less robust. Using preclinical models of lymphoma, we have recently demonstrated that HDACi induce tumor cell-specific apoptosis and that this is essential for the therapeutic effects of these agents. Herein, we demonstrate that HDACi can be combined with immune-activating antibodies designed to promote the function of antigen-presenting cells (APCs) and enhance proliferation and survival of cytotoxic T cells (CTL) to stimulate a host antitumor immune response resulting in eradication of established solid tumors. This unique combination therapy was dependent on tumor cell apoptosis mediated by HDACi that stimulated the uptake of dead tumor cells by APCs. Tumor eradication was mediated by CD8(+) CTL that used perforin as the key immune effector molecule. This combination therapy was well tolerated and induced long-term immunological antitumor memory capable of mediating spontaneous tumor eradication upon rechallenge. These studies indicate that the ability of HDACi to mediate subtherapeutic levels of tumor cell apoptosis can be exploited by combining with antibodies that augment host antitumor immune responses to mediate robust and prolonged eradication of solid tumors.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Neoplasms, Experimental/therapy , Animals , Antigen-Presenting Cells/immunology , Combined Modality Therapy , Mice , Neoplasms, Experimental/drug therapy , T-Lymphocytes, Cytotoxic/immunology , Vorinostat
5.
Blood ; 113(9): 1982-91, 2009 Feb 26.
Article in English | MEDLINE | ID: mdl-19060243

ABSTRACT

The apoptotic and therapeutic activities of the histone deacetylase inhibitor (HDACi) vorinostat are blocked by overexpression of Bcl-2 or Bcl-X(L). Herein, we used the small molecule inhibitor ABT-737 to restore sensitivity of Emu-myc lymphomas overexpressing Bcl-2 or Bcl-X(L) to vorinostat and valproic acid (VPA). Combining low-dose ABT-737 with vorinostat or VPA resulted in synergistic apoptosis of these cells. ABT-737 was ineffective against Emu-myc/Mcl-1 and Emu-myc/A1 cells either as a single agent or in combination with HDACi. However, in contrast to the reported binding specificity data, Emu-myc/Bcl-w lymphomas were insensitive to ABT-737 used alone or in combination with HDACi, indicating that the regulatory activity of ABT-737 is restricted to Bcl-2 and Bcl-X(L). Emu-myc lymphomas that expressed Bcl-2 throughout the tumorigenesis process were especially sensitive to ABT-737, while those forced to overexpress Mcl-1 were not. This supports the notion that tumor cells "addicted" to ABT-737 target proteins (ie, Bcl-2 or Bcl-X(L)) are likely to be the most sensitive target cell population. Our studies provide important preclinical data on the binding specificity of ABT-737 and its usefulness against primary hematologic malignancies when used as a single agent and in combination with HDACi.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biphenyl Compounds/pharmacology , Enzyme Inhibitors/administration & dosage , Histone Deacetylase Inhibitors , Lymphoma/drug therapy , Nitrophenols/pharmacology , Sulfonamides/pharmacology , Animals , Biphenyl Compounds/administration & dosage , Cell Survival/drug effects , Drug Delivery Systems , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Drug Synergism , Genes, bcl-2 , Genes, myc , Hydroxamic Acids/administration & dosage , Lymphoma/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nitrophenols/administration & dosage , Piperazines/administration & dosage , Piperazines/pharmacology , Substrate Specificity , Sulfonamides/administration & dosage , Vorinostat
6.
Proc Natl Acad Sci U S A ; 105(32): 11317-22, 2008 Aug 12.
Article in English | MEDLINE | ID: mdl-18685088

ABSTRACT

Histone deacetylase inhibitors (HDACi) and agents such as recombinant tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and agonistic anti-TRAIL receptor (TRAIL-R) antibodies are anticancer agents that have shown promise in preclinical settings and in early phase clinical trials as monotherapies. Although HDACi and activators of the TRAIL pathway have different molecular targets and mechanisms of action, they share the ability to induce tumor cell-selective apoptosis. The ability of HDACi to induce expression of TRAIL-R death receptors 4 and 5 (DR4/DR5), and induce tumor cell death via the intrinsic apoptotic pathway provides a molecular rationale to combine these agents with activators of the TRAIL pathway that activate the alternative (death receptor) apoptotic pathway. Herein, we demonstrate that the HDACi vorinostat synergizes with the mouse DR5-specific monoclonal antibody MD5-1 to induce rapid and robust tumor cell apoptosis in vitro and in vivo. Importantly, using a preclinical mouse breast cancer model, we show that the combination of vorinostat and MD5-1 is safe and induces regression of established tumors, whereas single agent treatment had little or no effect. Functional analyses revealed that rather than mediating enhanced tumor cell apoptosis via the simultaneous activation of the intrinsic and extrinsic apoptotic pathways, vorinostat augmented MD5-1-induced apoptosis concomitant with down-regulation of the intracellular apoptosis inhibitor cellular-FLIP (c-FLIP). These data demonstrate that combination therapies involving HDACi and activators of the TRAIL pathway can be efficacious for the treatment of cancer in experimental mouse models.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Histone Deacetylase Inhibitors , Mammary Neoplasms, Experimental/drug therapy , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Animals , Antibodies, Monoclonal/pharmacology , CASP8 and FADD-Like Apoptosis Regulating Protein/biosynthesis , Cell Line, Tumor , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/pharmacology , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred BALB C , Mice, SCID , Neoplasm Proteins/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Vorinostat
SELECTION OF CITATIONS
SEARCH DETAIL
...