23 results on '"Raje, N"'
Search Results
2. Ricolinostat (ACY-1215) induced inhibition of aggresome formation accelerates carfilzomib-induced multiple myeloma cell death.
- Author
-
Mishima Y, Santo L, Eda H, Cirstea D, Nemani N, Yee AJ, O'Donnell E, Selig MK, Quayle SN, Arastu-Kapur S, Kirk C, Boise LH, Jones SS, and Raje N
- Subjects
- Animals, Antineoplastic Agents pharmacology, Autophagy drug effects, Cell Line, Tumor, Cell Survival drug effects, Disease Models, Animal, Drug Synergism, Endoplasmic Reticulum Stress drug effects, Female, Heterografts, Histone Deacetylase Inhibitors pharmacology, Humans, Mice, Multiple Myeloma genetics, Multiple Myeloma pathology, Phagosomes metabolism, Proteasome Inhibitors pharmacology, Apoptosis drug effects, Hydroxamic Acids pharmacology, Multiple Myeloma metabolism, Oligopeptides pharmacology, Pyrimidines pharmacology
- Abstract
Proteasome inhibition induces the accumulation of aggregated misfolded/ubiquitinated proteins in the aggresome; conversely, histone deacetylase 6 (HDAC6) inhibition blocks aggresome formation. Although this rationale has been the basis of proteasome inhibitor (PI) and HDAC6 inhibitor combination studies, the role of disruption of aggresome formation by HDAC6 inhibition has not yet been studied in multiple myeloma (MM). The present study aimed to evaluate the impact of carfilzomib (CFZ) in combination with a selective HDAC6 inhibitor (ricolinostat) in MM cells with respect to the aggresome-proteolysis pathway. We observed that combination treatment of CFZ with ricolinostat triggered synergistic anti-MM effects, even in bortezomib-resistant cells. Immunofluorescent staining showed that CFZ increased the accumulation of ubiquitinated proteins and protein aggregates in the cytoplasm, as well as the engulfment of aggregated ubiquitinated proteins by autophagosomes, which was blocked by ricolinostat. Electron microscopy imaging showed increased autophagy triggered by CFZ, which was inhibited by the addition of ACY-1215. Finally, an in vivo mouse xenograft study confirmed a decrease in tumour volume, associated with apoptosis, following treatment with CFZ in combination with ricolinostat. Our results suggest that ricolinostat inhibits aggresome formation, caused by CFZ-induced inhibition of the proteasome pathway, resulting in enhanced apoptosis in MM cells., (© 2015 John Wiley & Sons Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
3. Induction of differential apoptotic pathways in multiple myeloma cells by class-selective histone deacetylase inhibitors.
- Author
-
Hideshima T, Mazitschek R, Santo L, Mimura N, Gorgun G, Richardson PG, Raje N, and Anderson KC
- Subjects
- Antineoplastic Agents toxicity, Cell Line, Tumor, Endoplasmic Reticulum Stress drug effects, Histone Deacetylase Inhibitors toxicity, Humans, Oligopeptides pharmacology, Antineoplastic Agents pharmacology, Apoptosis drug effects, Histone Deacetylase Inhibitors pharmacology, Multiple Myeloma metabolism, Signal Transduction drug effects
- Published
- 2014
- Full Text
- View/download PDF
4. A novel vascular disrupting agent plinabulin triggers JNK-mediated apoptosis and inhibits angiogenesis in multiple myeloma cells.
- Author
-
Singh AV, Bandi M, Raje N, Richardson P, Palladino MA, Chauhan D, and Anderson KC
- Subjects
- Animals, Blotting, Western, Cell Proliferation drug effects, Diketopiperazines, Fluorescent Antibody Technique, Humans, Male, Mice, Mice, SCID, Mitogen-Activated Protein Kinase 8 antagonists & inhibitors, Mitogen-Activated Protein Kinase 8 genetics, RNA, Small Interfering genetics, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Angiogenesis Inhibitors pharmacology, Apoptosis drug effects, Imidazoles pharmacology, Mitogen-Activated Protein Kinase 8 metabolism, Multiple Myeloma blood supply, Multiple Myeloma drug therapy, Neovascularization, Pathologic prevention & control, Piperazines pharmacology
- Abstract
Previous studies have established a role of vascular-disrupting agents as anti- cancer agents. Plinabulin is a novel vascular-disrupting agent that exhibits potent interruption of tumor blood flow because of the disruption of tumor vascular endothelial cells, resulting in tumor necrosis. In addition, plinabulin exerts a direct action on tumor cells, resulting in apoptosis. In the present study, we examined the anti-multiple myeloma (MM) activity of plinabulin. We show that low concentrations of plinabulin exhibit a potent antiangiogenic action on vascular endothelial cells. Importantly, plinabulin also induces apoptotic cell death in MM cell lines and tumor cells from patients with MM, associated with mitotic growth arrest. Plinabulin-induced apoptosis is mediated through activation of caspase-3, caspase-8, caspase-9, and poly(ADP-ribose) polymerase cleavage. Moreover, plinabulin triggered phosphorylation of stress response protein JNK, as a primary target, whereas blockade of JNK with a biochemical inhibitor or small interfering RNA strategy abrogated plinabulin-induced mitotic block or MM cell death. Finally, in vivo studies show that plinabulin was well tolerated and significantly inhibited tumor growth and prolonged survival in a human MM.1S plasmacytoma murine xenograft model. Our study therefore provides the rationale for clinical evaluation of plinabulin to improve patient outcome in MM.
- Published
- 2011
- Full Text
- View/download PDF
5. A novel Aurora-A kinase inhibitor MLN8237 induces cytotoxicity and cell-cycle arrest in multiple myeloma.
- Author
-
Görgün G, Calabrese E, Hideshima T, Ecsedy J, Perrone G, Mani M, Ikeda H, Bianchi G, Hu Y, Cirstea D, Santo L, Tai YT, Nahar S, Zheng M, Bandi M, Carrasco RD, Raje N, Munshi N, Richardson P, and Anderson KC
- Subjects
- Animals, Antineoplastic Combined Chemotherapy Protocols pharmacology, Aurora Kinase A, Aurora Kinases, Azepines therapeutic use, Boronic Acids pharmacology, Bortezomib, Cell Cycle, Cell Line, Tumor, Cellular Senescence drug effects, Clinical Trials, Phase I as Topic, Clinical Trials, Phase II as Topic, Dexamethasone pharmacology, Doxorubicin pharmacology, Gene Expression Regulation, Enzymologic drug effects, Gene Expression Regulation, Neoplastic drug effects, Humans, Mice, Mice, SCID, Multiple Myeloma enzymology, Neoplasm Transplantation, Protein Kinase Inhibitors therapeutic use, Protein Serine-Threonine Kinases biosynthesis, Pyrazines pharmacology, Pyrimidines therapeutic use, Spindle Apparatus metabolism, Time Factors, Transplantation, Heterologous, Tumor Suppressor Proteins biosynthesis, Xenograft Model Antitumor Assays, Apoptosis drug effects, Azepines pharmacology, Multiple Myeloma drug therapy, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases antagonists & inhibitors, Pyrimidines pharmacology
- Abstract
Aurora-A is a mitotic kinase that regulates mitotic spindle formation and segregation. In multiple myeloma (MM), high Aurora-A gene expression has been correlated with centrosome amplification and proliferation; thus, inhibition of Aurora-A in MM may prove to be therapeutically beneficial. Here we assess the in vitro and in vivo anti-MM activity of MLN8237, a small-molecule Aurora-A kinase inhibitor. Treatment of cultured MM cells with MLN8237 results in mitotic spindle abnormalities, mitotic accumulation, as well as inhibition of cell proliferation through apoptosis and senescence. In addition, MLN8237 up-regulates p53 and tumor suppressor genes p21 and p27. Combining MLN8237 with dexamethasone, doxorubicin, or bortezomib induces synergistic/additive anti-MM activity in vitro. In vivo anti-MM activity of MLN8237 was confirmed using a xenograft-murine model of human-MM. Tumor burden was significantly reduced (P = .007) and overall survival was significantly increased (P < .005) in animals treated with 30 mg/kg MLN8237 for 21 days. Induction of apoptosis and cell death by MLN8237 were confirmed in tumor cells excised from treated animals by TdT-mediated dUTP nick end labeling assay. MLN8237 is currently in phase 1 and phase 2 clinical trials in patients with advanced malignancies, and our preclinical results suggest that MLN8237 may be a promising novel targeted therapy in MM.
- Published
- 2010
- Full Text
- View/download PDF
6. AT7519, A novel small molecule multi-cyclin-dependent kinase inhibitor, induces apoptosis in multiple myeloma via GSK-3beta activation and RNA polymerase II inhibition.
- Author
-
Santo L, Vallet S, Hideshima T, Cirstea D, Ikeda H, Pozzi S, Patel K, Okawa Y, Gorgun G, Perrone G, Calabrese E, Yule M, Squires M, Ladetto M, Boccadoro M, Richardson PG, Munshi NC, Anderson KC, and Raje N
- Subjects
- Animals, Cell Cycle drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Dose-Response Relationship, Drug, Enzyme Activation drug effects, Glycogen Synthase Kinase 3 beta, Humans, Male, Mice, Mice, SCID, Multiple Myeloma pathology, Apoptosis drug effects, Cyclin-Dependent Kinases antagonists & inhibitors, Glycogen Synthase Kinase 3 metabolism, Multiple Myeloma drug therapy, Piperidines pharmacology, Pyrazoles pharmacology, RNA Polymerase II antagonists & inhibitors
- Abstract
Dysregulated cell cycling is a universal hallmark of cancer and is often mediated by abnormal activation of cyclin-dependent kinases (CDKs) and their cyclin partners. Overexpression of individual complexes are reported in multiple myeloma (MM), making them attractive therapeutic targets. In this study, we investigate the preclinical activity of a novel small-molecule multi-CDK inhibitor, AT7519, in MM. We show the anti-MM activity of AT7519 displaying potent cytotoxicity and apoptosis; associated with in vivo tumor growth inhibition and prolonged survival. At the molecular level, AT7519 inhibited RNA polymerase II (RNA pol II) phosphorylation, a CDK9, 7 substrate, associated with decreased RNA synthesis confirmed by [(3)H] Uridine incorporation. In addition, AT7519 inhibited glycogen synthase kinase 3beta (GSK-3beta) phosphorylation; conversely pretreatment with a selective GSK-3 inhibitor and shRNA GSK-3beta knockdown restored MM survival, suggesting the involvement of GSK-3beta in AT7519-induced apoptosis. GSK-3beta activation was independent of RNA pol II dephosphorylation confirmed by alpha-amanitin, a specific RNA pol II inihibitor, showing potent inhibition of RNA pol II phosphorylation without corresponding effects on GSK-3beta phosphorylation. These results offer new insights into the crucial, yet controversial role of GSK-3beta in MM and show significant anti-MM activity of AT7519, providing the rationale for its clinical evaluation in MM.
- Published
- 2010
- Full Text
- View/download PDF
7. 5-Azacytidine, a DNA methyltransferase inhibitor, induces ATR-mediated DNA double-strand break responses, apoptosis, and synergistic cytotoxicity with doxorubicin and bortezomib against multiple myeloma cells.
- Author
-
Kiziltepe T, Hideshima T, Catley L, Raje N, Yasui H, Shiraishi N, Okawa Y, Ikeda H, Vallet S, Pozzi S, Ishitsuka K, Ocio EM, Chauhan D, and Anderson KC
- Subjects
- Bortezomib, Cell Division drug effects, Cell Line, Tumor, Cell Survival drug effects, Drug Synergism, Humans, Antineoplastic Agents pharmacology, Apoptosis drug effects, Azacitidine pharmacology, Boronic Acids pharmacology, DNA Damage, DNA, Neoplasm drug effects, Doxorubicin pharmacology, Enzyme Inhibitors pharmacology, Multiple Myeloma pathology, Pyrazines pharmacology
- Abstract
In this study, we investigated the cytotoxicity of 5-azacytidine, a DNA methyltransferase inhibitor, against multiple myeloma (MM) cells, and characterized DNA damage-related mechanisms of cell death. 5-Azacytidine showed significant cytotoxicity against both conventional therapy-sensitive and therapy-resistant MM cell lines, as well as multidrug-resistant patient-derived MM cells, with IC(50) of approximately 0.8-3 micromol/L. Conversely, 5-azacytidine was not cytotoxic to peripheral blood mononuclear cells or patient-derived bone marrow stromal cells (BMSC) at these doses. Importantly, 5-azacytidine overcame the survival and growth advantages conferred by exogenous interleukin-6 (IL-6), insulin-like growth factor-I (IGF-I), or by adherence of MM cells to BMSCs. 5-Azacytidine treatment induced DNA double-strand break (DSB) responses, as evidenced by H2AX, Chk2, and p53 phosphorylations, and apoptosis of MM cells. 5-Azacytidine-induced apoptosis was both caspase dependent and independent, with caspase 8 and caspase 9 cleavage; Mcl-1 cleavage; Bax, Puma, and Noxa up-regulation; as well as release of AIF and EndoG from the mitochondria. Finally, we show that 5-azacytidine-induced DNA DSB responses were mediated predominantly by ATR, and that doxorubicin, as well as bortezomib, synergistically enhanced 5-azacytidine-induced MM cell death. Taken together, these data provide the preclinical rationale for the clinical evaluation of 5-azacytidine, alone and in combination with doxorubicin and bortezomib, to improve patient outcome in MM.
- Published
- 2007
- Full Text
- View/download PDF
8. Up-regulation of c-Jun inhibits proliferation and induces apoptosis via caspase-triggered c-Abl cleavage in human multiple myeloma.
- Author
-
Podar K, Raab MS, Tonon G, Sattler M, Barilà D, Zhang J, Tai YT, Yasui H, Raje N, DePinho RA, Hideshima T, Chauhan D, and Anderson KC
- Subjects
- Adamantane pharmacology, Apoptosis drug effects, Benzamides, Boronic Acids pharmacology, Bortezomib, Caspase Inhibitors, Cell Growth Processes drug effects, Cell Growth Processes physiology, Dexamethasone pharmacology, Humans, Imatinib Mesylate, Leukemia, Erythroblastic, Acute drug therapy, Leukemia, Erythroblastic, Acute metabolism, Leukemia, Erythroblastic, Acute pathology, Melphalan pharmacology, Multiple Myeloma genetics, Multiple Myeloma metabolism, Piperazines pharmacology, Proto-Oncogene Proteins c-abl genetics, Proto-Oncogene Proteins c-jun genetics, Proto-Oncogene Proteins c-jun metabolism, Pyrazines pharmacology, Pyrimidines pharmacology, Transfection, Up-Regulation drug effects, Adamantane analogs & derivatives, Apoptosis physiology, Caspases metabolism, Hydroquinones pharmacology, Multiple Myeloma drug therapy, Multiple Myeloma pathology, Proto-Oncogene Proteins c-abl metabolism, Proto-Oncogene Proteins c-jun biosynthesis
- Abstract
Here we show the antimyeloma cytotoxicity of adaphostin and carried out expression profiling of adaphostin-treated multiple myeloma (MM) cells to identify its molecular targets. Surprisingly, c-Jun was the most up-regulated gene even at the earliest point of analysis (2 h). We also observed adaphostin-induced c-Abl cleavage in immunoblot analysis. Proteasome inhibitor bortezomib, but not melphalan or dexamethasone, induced similar effects, indicating unique agent-dependent mechanisms. Using caspase inhibitors, as well as caspase-resistant mutants of c-Abl (TM-c-Abl and D565A-Abl), we then showed that c-Abl cleavage in MM cells requires caspase activity. Importantly, both overexpression of the c-Abl fragment or c-Jun and knockdown of c-Abl and c-Jun expression by small interfering RNA confirmed that adaphostin-induced c-Jun up-regulation triggers downstream caspase-mediated c-Abl cleavage, inhibition of MM cell growth, and induction of apoptosis. Finally, our data suggest that this mechanism may not only be restricted to MM but may also be important in a broad range of malignancies including erythroleukemia and solid tumors.
- Published
- 2007
- Full Text
- View/download PDF
9. Didox, a ribonucleotide reductase inhibitor, induces apoptosis and inhibits DNA repair in multiple myeloma cells.
- Author
-
Raje N, Kumar S, Hideshima T, Ishitsuka K, Yasui H, Chhetri S, Vallet S, Vonescu E, Shiraishi N, Kiziltepe T, Elford HL, Munshi NC, and Anderson KC
- Subjects
- Antineoplastic Agents, Alkylating pharmacology, Caspases physiology, Cell Cycle drug effects, Cell Survival drug effects, DNA Repair genetics, Dose-Response Relationship, Drug, Down-Regulation drug effects, Drug Evaluation, Preclinical, Drug Synergism, Enzyme Inhibitors pharmacology, Gene Expression Regulation, Neoplastic drug effects, Humans, Melphalan pharmacology, Multiple Myeloma genetics, Multiple Myeloma metabolism, Neoplasm Proteins metabolism, Proto-Oncogene Proteins c-bcl-2 metabolism, Ribonucleotide Reductases antagonists & inhibitors, Tumor Cells, Cultured, Antineoplastic Agents pharmacology, Apoptosis drug effects, DNA Repair drug effects, Hydroxamic Acids pharmacology, Multiple Myeloma pathology
- Abstract
Ribonucleotide reductase (RR) is the enzyme that catalyses the rate-limiting step in DNA synthesis, the production of deoxynucleotides. RR activity is markedly elevated in tumour tissue and is crucial for cell division. It is therefore an excellent target for cancer chemotherapy. This study examined the anti-myeloma activity of Didox (3,4-Dihydroxybenzohydroxamic acid), a novel RR inhibitor (RRI). Our data showed that Didox induced caspase-dependent multiple myeloma (MM) cell apoptosis. Didox, unlike other RRIs that mainly target the pyrimidine metabolism pathway, targets both purine and pyrimidine metabolism pathways in MM, as demonstrated by transcriptional profiling using the Affymetrix U133A 2.0 gene chip. Specifically, a >or=2-fold downregulation of genes in these anabolic pathways was shown as early as 12 h after exposure to Didox. Furthermore, apoptosis was accompanied by downregulation of bcl family proteins including bcl-2, bcl(xl), and XIAP. Importantly, RR M1 component transcript was also downregulated, associated with decreased protein expression. Genes involved in DNA repair mechanisms, specifically RAD 51 homologue, were also downregulated. As Didox acts on MM cells by inhibiting DNA synthesis and repair, combination studies with melphalan, an agent commonly used in MM, were performed. A strong in vitro synergism was shown, with combination indices of <0.7 as determined by the Chou-Talalay method. These studies therefore provide the preclinical rationale for evaluation of Didox, alone and in combination with DNA-damaging agents, to improve patient outcome in MM.
- Published
- 2006
- Full Text
- View/download PDF
10. MLN120B, a novel IkappaB kinase beta inhibitor, blocks multiple myeloma cell growth in vitro and in vivo.
- Author
-
Hideshima T, Neri P, Tassone P, Yasui H, Ishitsuka K, Raje N, Chauhan D, Podar K, Mitsiades C, Dang L, Munshi N, Richardson P, Schenkein D, and Anderson KC
- Subjects
- Animals, Antineoplastic Agents pharmacology, Cell Adhesion drug effects, Enzyme Inhibitors, Humans, Insulin-Like Growth Factor I pharmacology, Interleukin-6 pharmacology, Mice, Mice, SCID, Multiple Myeloma pathology, NF-kappa B metabolism, Phosphorylation drug effects, Stromal Cells drug effects, Stromal Cells metabolism, Tumor Cells, Cultured, Tumor Necrosis Factor-alpha pharmacology, Vascular Endothelial Growth Factor A metabolism, Apoptosis drug effects, Bone Marrow pathology, Cell Proliferation drug effects, I-kappa B Kinase antagonists & inhibitors, Multiple Myeloma drug therapy, Protein Kinase Inhibitors pharmacology
- Abstract
Purpose: The purpose of this study is to delineate the biological significance of IkappaB kinase (IKK) beta inhibition in multiple myeloma cells in the context of bone marrow stromal cells (BMSC) using a novel IKKbeta inhibitor MLN120B., Experimental Design: Growth-inhibitory effect of MLN120B in multiple myeloma cells in the presence of cytokines [interleukin-6 (IL-6) and insulin-like growth factor-I (IGF-1)], conventional agents (dexamethasone, melphalan, and doxorubicin), or BMSC was assessed in vitro. In vivo anti-multiple myeloma activity of MLN120B was evaluated in severe combined immunodeficient (SCID)-hu model., Results: MLN120B inhibits both baseline and tumor necrosis factor-alpha-induced nuclear factor-kappaB activation, associated with down-regulation of IkappaBalpha and p65 nuclear factor-kappaB phosphorylation. MLN120B triggers 25% to 90% growth inhibition in a dose-dependent fashion in multiple myeloma cell lines and significantly augments tumor necrosis factor-alpha-induced cytotoxicity in MM.1S cells. MLN120B augments growth inhibition triggered by doxorubicin and melphalan in both RPMI 8226 and IL-6-dependent INA6 cell lines. Neither IL-6 nor IGF-1 overcomes the growth-inhibitory effect of MLN120B. MLN120B inhibits constitutive IL-6 secretion by BMSCs by 70% to 80% without affecting viability. Importantly, MLN120B almost completely blocks stimulation of MM.1S, U266, and INA6 cell growth, as well as IL-6 secretion from BMSCs, induced by multiple myeloma cell adherence to BMSCs. MLN120B overcomes the protective effect of BMSCs against conventional (dexamethasone) therapy., Conclusions: Our data show that the novel IKKbeta inhibitor MLN120B induces growth inhibition of multiple myeloma cells in SCID-hu mouse model. These studies provide the framework for clinical evaluation of MLN120B, alone and in combined therapies, trials of these novel agents to improve patient outcome in multiple myeloma.
- Published
- 2006
- Full Text
- View/download PDF
11. Honokiol overcomes conventional drug resistance in human multiple myeloma by induction of caspase-dependent and -independent apoptosis.
- Author
-
Ishitsuka K, Hideshima T, Hamasaki M, Raje N, Kumar S, Hideshima H, Shiraishi N, Yasui H, Roccaro AM, Richardson P, Podar K, Le Gouill S, Chauhan D, Tamura K, Arbiser J, and Anderson KC
- Subjects
- Antineoplastic Agents, Phytogenic pharmacology, Antineoplastic Agents, Phytogenic therapeutic use, Biphenyl Compounds chemistry, Biphenyl Compounds therapeutic use, Bone Marrow Cells drug effects, Cell Cycle drug effects, Cell Line, Tumor, Cell Proliferation drug effects, DNA biosynthesis, DNA drug effects, Dose-Response Relationship, Drug, Endothelial Cells drug effects, Humans, Insulin-Like Growth Factor I pharmacology, Interleukin-6 pharmacology, Lignans chemistry, Lignans therapeutic use, Multiple Myeloma drug therapy, Neovascularization, Pathologic chemically induced, Stromal Cells drug effects, Structure-Activity Relationship, Apoptosis drug effects, Biphenyl Compounds pharmacology, Caspases pharmacology, Drug Resistance, Multiple, Lignans pharmacology, Multiple Myeloma pathology
- Abstract
Honokiol (HNK) is an active component purified from magnolia, a plant used in traditional Chinese and Japanese medicine. Here we show that HNK significantly induces cytotoxicity in human multiple myeloma (MM) cell lines and tumor cells from patients with relapsed refractory MM. Neither coculture with bone marrow stromal cells nor cytokines (interleukin-6 and insulin-like growth factor-1) protect against HNK-induced cytotoxicity. Although activation of caspases 3, 7, 8, and 9 is triggered by HNK, the pan-caspase inhibitor z-VAD-fmk does not abrogate HNK-induced apoptosis. Importantly, release of an executioner of caspase-independent apoptosis, apoptosis-inducing factor (AIF), from mitochondria is induced by HNK treatment. HNK induces apoptosis in the SU-DHL4 cell line, which has low levels of caspase 3 and 8 associated with resistance to both conventional and novel drugs. These results suggest that HNK induces apoptosis via both caspase-dependent and -independent pathways. Furthermore, HNK enhances MM cell cytotoxicity and apoptosis induced by bortezomib. In addition to its direct cytotoxicity to MM cells, HNK also represses tube formation by endothelial cells, suggesting that HNK inhibits neovascurization in the bone marrow microenvironment. Taken together, our results provide the preclinical rationale for clinical protocols of HNK to improve patient outcome in MM.
- Published
- 2005
- Full Text
- View/download PDF
12. Novel inosine monophosphate dehydrogenase inhibitor VX-944 induces apoptosis in multiple myeloma cells primarily via caspase-independent AIF/Endo G pathway.
- Author
-
Ishitsuka K, Hideshima T, Hamasaki M, Raje N, Kumar S, Podar K, Le Gouill S, Shiraishi N, Yasui H, Roccaro AM, Tai YZ, Chauhan D, Fram R, Tamura K, Jain J, and Anderson KC
- Subjects
- Apoptosis physiology, Apoptosis Inducing Factor, Blotting, Western, Bone Marrow Cells, Caspases drug effects, Caspases metabolism, Cell Line, Tumor, Cell Proliferation drug effects, Coculture Techniques, Endodeoxyribonucleases drug effects, Enzyme Inhibitors pharmacology, Flavoproteins drug effects, Humans, Insulin-Like Growth Factor I metabolism, Interleukin-6 metabolism, Membrane Proteins drug effects, Antineoplastic Agents pharmacology, Apoptosis drug effects, Endodeoxyribonucleases metabolism, Flavoproteins metabolism, IMP Dehydrogenase antagonists & inhibitors, Membrane Proteins metabolism, Multiple Myeloma pathology, Organic Chemicals pharmacology
- Abstract
Inosine monophosphate dehydrogenase (IMPDH) is a rate-limiting enzyme required for the de novo synthesis of guanine nucleotides from IMP. VX-944 (Vertex Pharmaceuticals, Cambridge, MA, USA) is a small-molecule, selective, noncompetitive inhibitor directed against human IMPDH. In this report, we show that VX-944 inhibits in vitro growth of human multiple myeloma (MM) cell lines via induction of apoptosis. Interleukin-6, insulin-like growth factor-1, or co-culture with bone marrow stromal cells (BMSCs) do not protect against VX-944-induced MM cell growth inhibition. VX-944 induced apoptosis in MM cell lines with only modest activation of caspases 3, 8, and 9. Furthermore, the pan-caspase inhibitor z-VAD-fmk did not inhibit VX-944-induced apoptosis and cell death. During VX-944-induced apoptosis, expressions of Bax and Bak were enhanced, and both apoptosis-inducing factor (AIF) and endonuclease G (Endo G) were released from the mitochondria to cytosol, suggesting that VX-944 triggers apoptosis in MM cells primarily via a caspase-independent, Bax/AIF/Endo G pathway. Importantly, VX-944 augments the cytotoxicity of doxorubicin and melphalan even in the presence of BMSCs. Taken together, our data demonstrate a primarily non-caspase-dependent apoptotic pathway triggered by VX-944, thereby providing a rationale to enhance MM cell cytotoxicity by combining this agent with conventional agents which trigger caspase activation.
- Published
- 2005
- Full Text
- View/download PDF
13. FTY720 induces apoptosis in multiple myeloma cells and overcomes drug resistance.
- Author
-
Yasui H, Hideshima T, Raje N, Roccaro AM, Shiraishi N, Kumar S, Hamasaki M, Ishitsuka K, Tai YT, Podar K, Catley L, Mitsiades CS, Richardson PG, Albert R, Brinkmann V, Chauhan D, and Anderson KC
- Subjects
- Apoptosis physiology, Bone Marrow Cells cytology, Caspases metabolism, Cell Growth Processes physiology, Coculture Techniques, Drug Resistance, Neoplasm, Fingolimod Hydrochloride, Immunosuppressive Agents pharmacology, Insulin-Like Growth Factor I pharmacology, Interleukin-6 pharmacology, Membrane Potentials drug effects, Mitochondria drug effects, Mitochondria physiology, Multiple Myeloma pathology, Proto-Oncogene Proteins c-bcl-2 metabolism, Signal Transduction drug effects, Sphingosine analogs & derivatives, Stromal Cells cytology, bcl-2-Associated X Protein, Antineoplastic Agents pharmacology, Apoptosis drug effects, Multiple Myeloma drug therapy, Propylene Glycols pharmacology
- Abstract
The novel immunomodulator FTY720 down-modulates sphingosine-1-phosphate receptor 1 on lymphocytes at low nanomolar concentrations, thereby inhibiting sphingosine-1-phosphate receptor 1-dependent egress of lymphocytes from lymph nodes into efferent lymphatics and blood. At high micromolar concentration, FTY720 has been shown to induce growth inhibition and/or apoptosis in human cancer cells in vitro. In this study, we investigated the biological effects of FTY720 on multiple myeloma cells. We found that FTY720 induces potent cytotoxicity against drug-sensitive and drug-resistant multiple myeloma cell lines as well as freshly isolated tumor cells from multiple myeloma patients who do not respond to conventional agents. FTY720 triggers activation of caspase-8, -9, and -3, followed by poly(ADP-ribose) polymerase cleavage. Interestingly, FTY720 induces alterations in mitochondrial membrane potential (DeltaPsim) and Bax cleavage, followed by translocation of cytochrome c and Smac/Diablo from mitochondria to the cytosol. In combination treatment studies, both dexamethasone and anti-Fas antibodies augment anti-multiple myeloma activity induced by FTY720. Neither interleukin-6 nor insulin-like growth factor-I, which both induce multiple myeloma cell growth and abrogate dexamethasone-induced apoptosis, protect against FTY720-induced growth inhibition. Importantly, growth of multiple myeloma cells adherent to bone marrow stromal cells is also significantly inhibited by FTY720. Finally, it down-regulates interleukin-6-induced phosphorylation of Akt, signal transducers and activators of transcription 3, and p42/44 mitogen-activated protein kinase; insulin-like growth factor-I-triggered Akt phosphorylation; and tumor necrosis factor alpha-induced IkappaBalpha and nuclear factor-kappaB p65 phosphorylation. These results suggest that FTY720 overcomes drug resistance in multiple myeloma cells and provide the rationale for its clinical evaluation to improve patient outcome in multiple myeloma.
- Published
- 2005
- Full Text
- View/download PDF
14. VEGF induces Mcl-1 up-regulation and protects multiple myeloma cells against apoptosis.
- Author
-
Le Gouill S, Podar K, Amiot M, Hideshima T, Chauhan D, Ishitsuka K, Kumar S, Raje N, Richardson PG, Harousseau JL, and Anderson KC
- Subjects
- Animals, Bone Marrow pathology, Cell Line, Cell Proliferation, Culture Media, Serum-Free pharmacology, Humans, Interleukin-6 pharmacology, Mice, Myeloid Cell Leukemia Sequence 1 Protein, Neoplasm Proteins genetics, Proto-Oncogene Proteins c-bcl-2 genetics, RNA, Small Interfering pharmacology, Serum Albumin, Bovine pharmacology, Tumor Cells, Cultured, Up-Regulation drug effects, Apoptosis drug effects, Multiple Myeloma pathology, Neoplasm Proteins biosynthesis, Neoplasm Proteins physiology, Proto-Oncogene Proteins c-bcl-2 biosynthesis, Proto-Oncogene Proteins c-bcl-2 physiology, Vascular Endothelial Growth Factor A pharmacology
- Abstract
Interleukin-6 (IL-6) triggers multiple myeloma (MM) cell proliferation and protects against apoptosis by up-regulating myeloid cell leukemia 1 (Mcl-1). Vascular endothelial growth factor (VEGF) induces modest proliferation of MM cells and induces IL-6 secretion in a paracrine loop involving MM cells and bone marrow stromal cells. Using murine embryonic fibroblast cell lines as a model (Mcl-1(wt/wt) and Mcl-1(Delta/null) MEFs), we here demonstrate that deletion of Mcl-1 reduces fetal bovine serum (FBS)-, VEGF-, and IL-6-induced proliferation. We also show that VEGF up-regulates Mcl-1 expression in a time- and dose-dependent manner in 3 human MM cell lines and MM patient cells. Importantly, we demonstrate that the pan-VEGF inhibitor, GW654652, inhibits VEGF-induced up-regulation of Mcl-1 and, as with Mcl-1 siRNA, is associated with decreased proliferation and induction of apoptosis. Finally, we show that VEGF protects MM patient cells against FBS starvation-induced apoptosis. Our studies therefore demonstrate that VEGF-induced MM cell proliferation and survival are mediated via Mcl-1, providing the preclinical framework for novel therapeutics targeting Mcl-1 and/or VEGF to improve patient outcome in MM.
- Published
- 2004
- Full Text
- View/download PDF
15. Thalidomide and its analogs overcome drug resistance of human multiple myeloma cells to conventional therapy.
- Author
-
Hideshima T, Chauhan D, Shima Y, Raje N, Davies FE, Tai YT, Treon SP, Lin B, Schlossman RL, Richardson P, Muller G, Stirling DI, and Anderson KC
- Subjects
- Cell Cycle drug effects, DNA Replication drug effects, Dexamethasone toxicity, Doxorubicin toxicity, Female, G1 Phase drug effects, Humans, Male, Melphalan toxicity, Thalidomide analogs & derivatives, Tumor Cells, Cultured, Antineoplastic Agents toxicity, Apoptosis drug effects, Drug Resistance, Multiple, Immunosuppressive Agents toxicity, Multiple Myeloma pathology, Thalidomide therapeutic use, Thalidomide toxicity
- Abstract
Although thalidomide (Thal) was initially used to treat multiple myeloma (MM) because of its known antiangiogenic effects, the mechanism of its anti-MM activity is unclear. These studies demonstrate clinical activity of Thal against MM that is refractory to conventional therapy and delineate mechanisms of anti-tumor activity of Thal and its potent analogs (immunomodulatory drugs [IMiDs]). Importantly, these agents act directly, by inducing apoptosis or G1 growth arrest, in MM cell lines and in patient MM cells that are resistant to melphalan, doxorubicin, and dexamethasone (Dex). Moreover, Thal and the IMiDs enhance the anti-MM activity of Dex and, conversely, are inhibited by interleukin 6. As for Dex, apoptotic signaling triggered by Thal and the IMiDs is associated with activation of related adhesion focal tyrosine kinase. These studies establish the framework for the development and testing of Thal and the IMiDs in a new treatment paradigm to target both the tumor cell and the microenvironment, overcome classical drug resistance, and achieve improved outcome in this presently incurable disease.
- Published
- 2000
16. SHP2 mediates the protective effect of interleukin-6 against dexamethasone-induced apoptosis in multiple myeloma cells.
- Author
-
Chauhan D, Pandey P, Hideshima T, Treon S, Raje N, Davies FE, Shima Y, Tai YT, Rosen S, Avraham S, Kharbanda S, and Anderson KC
- Subjects
- Apoptosis drug effects, Focal Adhesion Kinase 2, Humans, Intracellular Signaling Peptides and Proteins, Kinetics, Multiple Myeloma, Phosphorylation, Protein Tyrosine Phosphatase, Non-Receptor Type 11, Protein Tyrosine Phosphatase, Non-Receptor Type 6, Protein-Tyrosine Kinases metabolism, Recombinant Proteins metabolism, SH2 Domain-Containing Protein Tyrosine Phosphatases, Transfection, Tumor Cells, Cultured, src Homology Domains, Apoptosis physiology, Dexamethasone pharmacology, Interleukin-6 pharmacology, Protein Tyrosine Phosphatases metabolism
- Abstract
Our previous studies have shown that activation of a related adhesion focal tyrosine kinase (RAFTK) (also known as Pyk2) is required for dexamethasone (Dex)-induced apoptosis in multiple myeloma (MM) cells and that human interleukin-6 (IL-6), a known growth and survival factor for MM cells, blocks both RAFTK activation and apoptosis induced by Dex. However, the mechanism whereby IL-6 inhibits Dex-induced apoptosis is undefined. In this study, we demonstrate that protein-tyrosine phosphatase SHP2 mediates this protective effect. We show that IL-6 triggers selective activation of SHP2 and its association with RAFTK in Dex-treated MM cells. SHP2 interacts with RAFTK through a region other than its Src homology 2 domains. We demonstrate that RAFTK is a direct substrate of SHP2 both in vitro and in vivo, and that Tyr(906) in the C-terminal domain of RAFTK mediates its interaction with SHP2. Moreover, overexpression of dominant negative SHP2 blocked the protective effect of IL-6 against Dex-induced apoptosis. These findings demonstrate that SHP2 mediates the anti-apoptotic effect of IL-6 and suggest SHP2 as a novel therapeutic target in MM.
- Published
- 2000
- Full Text
- View/download PDF
17. RAFTK/PYK2-dependent and -independent apoptosis in multiple myeloma cells.
- Author
-
Chauhan D, Hideshima T, Pandey P, Treon S, Teoh G, Raje N, Rosen S, Krett N, Husson H, Avraham S, Kharbanda S, and Anderson KC
- Subjects
- Dexamethasone pharmacology, Enzyme Activation, Focal Adhesion Kinase 2, Humans, Multiple Myeloma metabolism, Phosphorylation, Radiation, Ionizing, Tumor Cells, Cultured, Apoptosis physiology, Multiple Myeloma pathology, Protein-Tyrosine Kinases physiology
- Abstract
Related Adhesion Focal Tyrosine Kinase (RAFTK; also known as Pyk2), is a member of the Focal Adhesion Kinase (FAK) subfamily and is activated by TNF alpha, UV light and increases in intracellular calcium levels. However, the function of RAFTK remains largely unknown. Our previous studies demonstrated that treatment with dexamethasone (Dex), ionizing radiation (IR), and anti-Fas mAb induces apoptosis in multiple myeloma (MM) cells. In the present study, we examined the potential role of RAFTK during induction of apoptosis in human MM cells triggered by these three stimuli. Dex-induced apoptosis, in contrast to apoptosis triggered by anti-Fas mAb or IR, is associated with activation of RAFTK. Transient overexpression of RAFTK wild type (RAFTK WT) induces apoptosis, whereas transient overexpression of Kinase inactive RAFTK (RAFTK K-M) blocks Dex-induced apoptosis. In contrast, transient overexpression of RAFTK K-M has no effect on apoptosis triggered by IR or Fas. In Dex-resistant cells, Dex does not trigger either RAFTK activation or apoptosis. Finally, interleukin-6 (IL-6), a known survival factor for MM cells, inhibits both activation of RAFTK and apoptosis of MM.1S cells triggered by Dex. Our studies therefore demonstrate Dex-induced RAFTK-dependent, and IR or Fas induced RAFTK-independent apoptotic signaling cascades in MM cells.
- Published
- 1999
- Full Text
- View/download PDF
18. Small-molecule multi-targeted kinase inhibitor RGB-286638 triggers P53-dependent and -independent anti-multiple myeloma activity through inhibition of transcriptional CDKs
- Author
-
Cirstea, D, Hideshima, T, Santo, L, Eda, H, Mishima, Y, Nemani, N, Hu, Y, Mimura, N, Cottini, F, Gorgun, G, Ohguchi, H, Suzuki, R, Loferer, H, Munshi, N C, Anderson, K C, and Raje, N
- Published
- 2013
- Full Text
- View/download PDF
19. A novel Bcl-2/Bcl-XL/Bcl-w inhibitor ABT-737 as therapy in multiple myeloma
- Author
-
Chauhan, D, Velankar, M, Brahmandam, M, Hideshima, T, Podar, K, Richardson, P, Schlossman, R, Ghobrial, I, Raje, N, Munshi, N, and Anderson, K C
- Published
- 2007
- Full Text
- View/download PDF
20. Novel etodolac analog SDX-308 (CEP-18082) induces cytotoxicity in multiple myeloma cells associated with inhibition of β-catenin/TCF pathway
- Author
-
Yasui, H, Hideshima, T, Ikeda, H, Ocio, E M, Kiziltepe, T, Vallet, S, Okawa, Y, Neri, P, Sukhdeo, K, Podar, K, Chauhan, D, Richardson, P G, Raje, N, Carrasco, D R, and Anderson, K C
- Published
- 2007
- Full Text
- View/download PDF
21. Antimyeloma activity of two novel N-substituted and tetraflourinated thalidomide analogs
- Author
-
Kumar, S, Raje, N, Hideshima, T, Ishitsuka, K, Roccaro, A, Shiraishi, N, Hamasaki, M, Yasui, H, Munshi, N C, Richardson, P, Figg, W D, and Anderson, K C
- Published
- 2005
- Full Text
- View/download PDF
22. Preclinical activity of P276-00, a novel small-molecule cyclin-dependent kinase inhibitor in the therapy of multiple myeloma.
- Author
-
Raje, N., Hideshima, T., Mukherjee, S., Raab, M., Vallet, S., Chhetri, S., Cirstea, D., Pozzi, S., Mitsiades, C., Rooney, M., Kiziltepe, T., Podar, K., Okawa, Y., Ikeda, H., Carrasco, R., Richardson, P. G., Chauhan, D., Munshi, N. C., Sharma, S., and Parikh, H.
- Subjects
- *
CYCLIN-dependent kinases , *MULTIPLE myeloma , *PLASMA cells , *CELL cycle regulation , *DRUG synergism , *APOPTOSIS , *PHYSICAL therapy - Abstract
Cyclin D dysregulation and overexpression is noted in the majority of multiple myeloma (MM) patients, suggesting its critical role in MM pathogenesis. Here, we sought to identify the effects of targeting cyclin D in MM. We first confirmed cyclin D mRNA overexpression in 42 of 64 (65%) patient plasma cells. Silencing cyclin D1 resulted in >50% apoptotic cell death suggesting its validity as a potential therapeutic target. We next evaluated P276-00, a clinical-grade small-molecule cyclin-dependent kinase inhibitor as a way to target the cyclins. P276-00 resulted in dose-dependent cytotoxicity in MM cells. Cell-cycle analysis confirmed either growth arrest or caspase-dependent apoptosis; this was preceded by inhibition of Rb-1 phosphorylation with associated downregulation of a range of cyclins suggesting a regulatory role of P276-00 in cell-cycle progression through broad activity. Proliferative stimuli such as interleukin-6, insulin-like growth factor-1 and bone-marrow stromal cell adherence induced cyclins; P276-00 overcame these growth, survival and drug resistance signals. Because the cyclins are substrates of proteasome degradation, combination studies with bortezomib resulted in synergism. Finally, in vivo efficacy of P276-00 was confirmed in an MM xenograft model. These studies form the basis of an ongoing phase I study in the treatment of relapsed/refractory MM.Leukemia (2009) 23, 961–970; doi:10.1038/leu.2008.378; published online 8 January 2009 [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
23. A novel Bcl-2/Bcl-XL/Bcl-w inhibitor ABT-737 as therapy in multiple myeloma.
- Author
-
Chauhan, D., Velankar, M., Brahmandam, M., Hideshima, T., Podar, K., Richardson, P., Schlossman, R., Ghobrial, I., Raje, N., Munshi, N., and Anderson, K. C.
- Subjects
MULTIPLE myeloma treatment ,CHEMICAL inhibitors ,DRUG therapy ,PROTEINS ,APOPTOSIS ,B cell lymphoma - Abstract
Bcl-2 or Bcl-X
L confers resistance to chemotherapy in multiple myeloma (MM). Here we characterized the effects of ABT-737, a potent small-molecule inhibitor of antiapoptotic proteins Bcl-2, Bcl-XL and Bcl-w with markedly higher affinity than previously reported compounds, on human MM cells. ABT-737 induces apoptosis in MM cells, including those resistant to conventional therapy. Examination of purified patient MM cells demonstrated similar results, without significant toxicity against normal peripheral blood mononuclear cells and MM bone marrow stromal cells. Importantly, ABT-737 decreases the viability of bortezomib-, dexamethasone-(Dex) and thalidomide-refractory patient MM cells. Additionally, ABT-737 abrogates MM cell growth triggered by interleukin-6 or insulin-like growth factor-1. Mechanistic studies show that ABT-737-induced apoptosis is associated with activation of caspase-8, caspase-9 and caspase-3, followed by poly(ADP-ribose) polymerase cleavage. Combining ABT-737 with proteasome inhibitor bortezomib, melphalan or dexamethasone induces additive anti-MM activity. Taken together, our study provides the rationale for clinical protocols evaluating ABT-737, alone and together with botezomib, mephalan or dexamethasone, to enhance MM cell killing, overcome drug resistance conferred by Bcl-2 and improve patient outcome in MM.Oncogene (2007) 26, 2374–2380. doi:10.1038/sj.onc.1210028; published online 2 October 2006 [ABSTRACT FROM AUTHOR]- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.