Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Oncotarget ; 9(47): 28547-28560, 2018 Jun 19.
Article in English | MEDLINE | ID: mdl-29983879

ABSTRACT

Despite the development of the novel Bruton tyrosine kinase inhibitor ibrutinib, mantle cell lymphoma (MCL) remains an incurable B-cell non-Hodgkin lymphoma. BMI-1 is required for the self-renewal and maintenance of MCL-initiating stem cells. Upregulation of BMI-1 has been reported in MCL patients, especially in those with refractory/relapsed disease. We studied the effects of a novel small-molecule selective inhibitor of BMI1 expression, PTC596, in MCL cells. Eight MCL cell lines and patient-derived samples were exposed to PTC596. PTC596 induced mitochondrial apoptosis, as evidenced by loss of mitochondrial membrane potential, caspase-3 cleavage, BAX activation, and phosphatidylserine externalization. There was a positive correlation between baseline BMI-1 protein levels and PTC596-induced apoptosis. p53 status did not affect sensitivity to PTC596. PTC596 effectively decreased BMI-1-expressing and tumor-initiating side population MCL cells (IC50: 138 nM) compared with ibrutinib, which modestly decreased side population cells. Interestingly, PTC596, reported to target cancer stem cells, decreased MCL-1 expression levels and antagonized ibrutinib-induced increase in MCL-1 expression, leading to synergistic apoptosis induction in MCL cells. There are currently no drugs that specifically target cancer stem cell fractions, and a reduction in BMI-1 protein by PTC596 may offer a novel therapeutic strategy for MCL.

2.
Mol Cancer Ther ; 13(7): 1848-59, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24739393

ABSTRACT

Aberrant activation of multiple signaling pathways is common in acute myelogenous leukemia (AML) cells, which can be linked to a poor prognosis for patients with this disease. Previous research with mTOR or MEK inhibitors revealed cytostatic, rather than cytotoxic, effects in in vitro and in vivo AML models. We evaluated the combination effect of the mTOR inhibitor AZD8055 and the MEK inhibitor selumetinib on human AML cell lines and primary AML samples. This combination demonstrated synergistic proapoptotic effects in AML cells with high basal activation of MEK and mTOR. We next incorporated the BH3 mimetic ABT-737 into this combination regimen to block Bcl-2, which further enhanced the apoptogenic effect of MEK/mTOR inhibition. The combination treatment also had a striking proapoptotic effect in CD33(+)/CD34(+) AML progenitor cells from primary AML samples with NRAS mutations. Mechanistically, upregulation of the proapoptotic protein Bim, accompanied by the downregulation of the antiapoptotic protein Mcl-1 (mainly via protein degradation), seemed to play critical roles in enhancing the combination drug effect. Furthermore, the modulation of survivin, Bax, Puma, and X-chromosome-linked inhibitor of apoptosis protein (XIAP) expression suggested a role for mitochondria-mediated apoptosis in the cytotoxicity of the drug combination. Consequently, the concomitant blockade of prosurvival MEK/mTOR signaling and the deactivation of Bcl-2 could provide a mechanism-based integrated therapeutic strategy for the eradication of AML cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzimidazoles/pharmacology , Leukemia, Myeloid, Acute/drug therapy , MAP Kinase Kinase Kinases/antagonists & inhibitors , Morpholines/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Apoptosis/drug effects , Benzimidazoles/administration & dosage , Cell Proliferation/drug effects , Drug Synergism , Gene Expression , Humans , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/pathology , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System/drug effects , Morpholines/administration & dosage , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Transfection , U937 Cells
3.
Blood ; 123(17): 2691-702, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24599548

ABSTRACT

Leukemia cells are protected from chemotherapy-induced apoptosis by their interactions with bone marrow mesenchymal stromal cells (BM-MSCs). Yet the underlying mechanisms associated with this protective effect remain unclear. Genome-wide gene expression profiling of BM-MSCs revealed that coculture with leukemia cells upregulated the transcription of genes associated with nuclear factor (NF)-κB signaling. Moreover, primary BM-MSCs from leukemia patients expressed NF-κB target genes at higher levels than their normal BM-MSC counterparts. The blockade of NF-κB activation via chemical agents or the overexpression of the mutant form of inhibitor κB-α (IκBα) in BM-MSCs markedly reduced the stromal-mediated drug resistance in leukemia cells in vitro and in vivo. In particular, our unique in vivo model of human leukemia BM microenvironment illustrated a direct link between NF-κB activation and stromal-associated chemoprotection. Mechanistic in vitro studies revealed that the interaction between vascular cell adhesion molecule 1 (VCAM-1) and very late antigen-4 (VLA-4) played an integral role in the activation of NF-κB in the stromal and tumor cell compartments. Together, these results suggest that reciprocal NF-κB activation in BM-MSCs and leukemia cells is essential for promoting chemoresistance in the transformed cells, and targeting NF-κB or VLA-4/VCAM-1 signaling could be a clinically relevant mechanism to overcome stroma-mediated chemoresistance in BM-resident leukemia cells.


Subject(s)
Drug Resistance, Neoplasm , Gene Expression Regulation, Leukemic , Integrin alpha4beta1/metabolism , NF-kappa B/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Animals , Bone and Bones/metabolism , Cell Adhesion , Cell Line, Transformed , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Endothelial Cells/cytology , Gene Expression Profiling , Humans , Mice , RNA, Messenger/metabolism , Signal Transduction , Stromal Cells/cytology
4.
Cancer Res ; 70(6): 2424-34, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20215498

ABSTRACT

Molecular aberrations of the Ras/Raf/mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) kinase (MEK)/ERK and/or Murine double minute (MDM2)/p53 signaling pathways have been reported in 80% and 50% of primary acute myeloid leukemia (AML) samples and confer poor outcome. In this study, antileukemic effects of combined MEK inhibition by AZD6244 and nongenotoxic p53 activation by MDM2 antagonist Nutlin-3a were investigated. Simultaneous blockade of MEK and MDM2 signaling by AZD6244 and Nutlin-3a triggered synergistic proapoptotic responses in AML cell lines [combination index (CI) = 0.06 +/- 0.03 and 0.43 +/- 0.03 in OCI/AML3 and MOLM13 cells, respectively] and in primary AML cells (CI = 0.52 +/- 0.01). Mechanistically, the combination upregulated levels of BH3-only proteins Puma and Bim, in part via transcriptional upregulation of the FOXO3a transcription factor. Suppression of Puma and Bim by short interfering RNA rescued OCI/AML3 cells from AZD/Nutlin-induced apoptosis. These results strongly indicate the therapeutic potential of combined MEK/MDM2 blockade in AML and implicate Puma and Bim as major regulators of AML cell survival.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Leukemia, Myeloid/drug therapy , Leukemia, Myeloid/enzymology , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Acute Disease , Apoptosis/drug effects , Apoptosis/physiology , Bcl-2-Like Protein 11 , Benzimidazoles/pharmacology , Drug Synergism , Extracellular Signal-Regulated MAP Kinases/metabolism , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , HL-60 Cells , Humans , Imidazoles/pharmacology , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , MAP Kinase Signaling System/drug effects , Myeloid Cell Leukemia Sequence 1 Protein , Phosphorylation , Piperazines/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , U937 Cells
5.
Blood ; 115(2): 306-14, 2010 Jan 14.
Article in English | MEDLINE | ID: mdl-19897582

ABSTRACT

Activation of p53 by murine double minute (MDM2) antagonist nutlin-3a or inhibition of X-linked inhibitor of apoptosis (XIAP) induces apoptosis in acute myeloid leukemia (AML) cells. We demonstrate that concomitant inhibition of MDM2 by nutlin-3a and of XIAP by small molecule antagonists synergistically induced apoptosis in p53 wild-type OCI-AML3 and Molm13 cells. Knockdown of p53 by shRNA blunted the synergy, and down-regulation of XIAP by antisense oligonucleotide (ASO) enhanced nutlin-3a-induced apoptosis, suggesting that the synergy was mediated by p53 activation and XIAP inhibition. This is supported by data showing that inhibition of both MDM2 and XIAP by their respective ASOs induced significantly more cell death than either ASO alone. Importantly, p53 activation and XIAP inhibition enhanced apoptosis in blasts from patients with primary AML, even when the cells were protected by stromal cells. Mechanistic studies demonstrated that XIAP inhibition potentiates p53-induced apoptosis by decreasing p53-induced p21 and that p53 activation enhances XIAP inhibition-induced cell death by promoting mitochondrial release of second mitochondria-derived activator of caspases (SMAC) and by inducing the expression of caspase-6. Because both XIAP and p53 are presently being targeted in ongoing clinical trials in leukemia, the combination strategy holds promise for expedited translation into the clinic.


Subject(s)
Apoptosis , Blast Crisis/metabolism , Leukemia, Myeloid, Acute/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism , Animals , Apoptosis Regulatory Proteins , Blast Crisis/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caspase 6/biosynthesis , Caspase 6/genetics , Cell Line, Tumor , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Leukemic/drug effects , Gene Expression Regulation, Leukemic/genetics , Gene Knockdown Techniques , Humans , Imidazoles/pharmacology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Leukemia, Myeloid, Acute/genetics , Mice , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Piperazines/pharmacology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Stromal Cells/metabolism , Tumor Suppressor Protein p53/genetics , X-Linked Inhibitor of Apoptosis Protein/genetics
6.
Mol Cancer Ther ; 8(9): 2509-16, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19723894

ABSTRACT

The advent of Bcr-Abl tyrosine kinase inhibitors (TKI) has revolutionized the treatment of chronic myelogenous leukemia (CML). However, resistance evolves due to BCR-ABL mutations and other mechanisms. Furthermore, patients with blast crisis CML are less responsive and quiescent CML stem cells are insensitive to these inhibitors. We found that triptolide, a diterpenoid, at nanomolar concentrations, promoted equally significant death of KBM5 cells, a cell line derived from a Bcr-Abl-bearing blast crisis CML patient and KBM5STI571 cells, an imatinib-resistant KBM5 subline bearing the T315I mutation. Similarly, Ba/F3 cells harboring mutated BCR-ABL were as sensitive as Ba/F3Bcr-Abl(p210wt) cells to triptolide. Importantly, triptolide induced apoptosis in primary samples from blast crisis CML patients, who showed resistance to Bcr-Abl TKIs in vivo, with less toxicity to normal cells. Triptolide decreased X-linked inhibitor of apoptosis protein, Mcl-1, and Bcr-Abl protein levels in K562, KBM5, and KBM5STI571 cells and in cells from blast crisis CML patients. It sensitized KBM5, but not KBM5STI571, cells to imatinib. More importantly, triptolide also induced death of quiescent CD34(+) CML progenitor cells, a major problem in the therapy of CML with TKIs. Collectively, these results suggest that triptolide potently induces blast crisis CML cell death independent of the cellular responses to Bcr-Abl TKIs, suggesting that triptolide could eradicate residual quiescent CML progenitor cells in TKI-treated patients and benefit TKI-resistant blast crisis CML patients.


Subject(s)
Antigens, CD34/immunology , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Blast Crisis/pathology , Diterpenes/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Phenanthrenes/pharmacology , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Benzamides , Blotting, Western , Cell Line, Tumor , Epoxy Compounds/pharmacology , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/genetics , Humans , Imatinib Mesylate , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Reverse Transcriptase Polymerase Chain Reaction
7.
Blood ; 111(7): 3742-50, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18187663

ABSTRACT

Acute myeloid leukemia (AML) cells are relatively resistant to tumor necrosis factor alpha-related apoptosis-inducing ligand (TRAIL). We previously reported that triptolide, a potent anticancer agent from a Chinese herb, decreases XIAP in leukemic cells. We evaluated the combination of triptolide and TRAIL and found synergistic promotion of apoptosis in AML cells. XIAP-overexpressing U937 cells (U937XIAP) were more resistant to TRAIL than U937neo cells, and inhibition of XIAP with the small-molecule inhibitor 1396-11 enhanced TRAIL-induced apoptosis, implying XIAP as a resistance factor in AML. Furthermore, triptolide increased DR5 levels in OCI-AML3, while the DR5 increase was blunted in p53-knockdown OCI-AML3 and p53-mutated U937 cells, confirming a role for p53 in the regulation of DR5. In support of this finding, disruption of MDM2-p53 binding with subsequent increase in p53 levels by nutlin3a increased DR5 levels and sensitized OCI-AML3 cells to TRAIL. The combination of 1396-11 plus nutlin3a plus TRAIL was more effective than either the 1396-11 and TRAIL or nutlin3a and TRAIL combinations in OCI-AML3 cells, further supporting the role of triptolide as a sensitizer to TRAIL-induced apoptosis in part by independent modulation of XIAP expression and p53 signaling. Thus, the combination of triptolide and TRAIL may provide a novel strategy for treating AML by overcoming critical mechanisms of apoptosis resistance.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Diterpenes/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Phenanthrenes/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Tumor Suppressor Protein p53/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism , Aniline Compounds/agonists , Aniline Compounds/pharmacology , Aniline Compounds/therapeutic use , Antineoplastic Agents, Alkylating/agonists , Antineoplastic Agents, Alkylating/therapeutic use , Diterpenes/agonists , Diterpenes/therapeutic use , Drug Synergism , Epoxy Compounds/agonists , Epoxy Compounds/pharmacology , Epoxy Compounds/therapeutic use , Female , Humans , Imidazoles/agonists , Imidazoles/pharmacology , Leukemia, Myeloid, Acute/metabolism , Male , Phenanthrenes/agonists , Phenanthrenes/therapeutic use , Piperazines/agonists , Piperazines/pharmacology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Signal Transduction/drug effects , TNF-Related Apoptosis-Inducing Ligand/agonists , Tumor Suppressor Protein p53/antagonists & inhibitors , U937 Cells , X-Linked Inhibitor of Apoptosis Protein/antagonists & inhibitors
8.
Leuk Lymphoma ; 48(4): 774-85, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17454637

ABSTRACT

Tetra-O-methyl nordihydroguaiaretic acid (M4N) was shown to induce G2 arrest and suppress human xenograft tumor growth by inhibiting Cdc2 and survivin. We examined the effect of M4N on leukemia and found that M4N inhibited growth and induced cell death in leukemic cell lines and blasts from AML patients. However, no significant changes in Cdc2 and survivin levels and G2 arrest were observed. Cell death and growth inhibition were dependent neither on XIAP, Bcl-2, and Bcl-X(L) levels nor on caspase-8. M4N did not promote cell differentiation in HL-60 cells. Interestingly, significant inhibition of AKT phosphorylation was observed in M4N treated OCI-AML3 cells. Collectively, our data showed that M4N inhibited cell growth and induced cell death in both leukemic cell lines and AML patient sample via a mechanism not mediated by Cdc2 and survivin inhibition and suggested that the extrinsic and the mitochondrial apoptotic pathways are not essential.


Subject(s)
Antioxidants/pharmacology , CDC2 Protein Kinase/metabolism , Leukemia/drug therapy , Masoprocol/pharmacology , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Apoptosis , Cell Cycle , Cell Line, Tumor , Cell Survival , Drug Screening Assays, Antitumor , HL-60 Cells , Humans , Inhibitor of Apoptosis Proteins , Jurkat Cells , Mitochondria/metabolism , Survivin , U937 Cells
9.
Cell Cycle ; 5(23): 2778-86, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17172851

ABSTRACT

Disruption of Mdm2-p53 interaction activates p53 signaling, disrupts the balance of antiapoptotic and proapoptotic Bcl-2 family proteins and induces apoptosis in acute myeloid leukemia (AML). Overexpression of Bcl-2 may inhibit this effect. Thus, functional inactivation of antiapoptotic Bcl-2 proteins may enhance apoptogenic effects of Mdm2 inhibition. We here investigate the potential therapeutic utility of combined targeting of Mdm2 by Nutlin-3a and Bcl-2 by ABT-737, recently developed inhibitors of protein-protein interactions. Nutlin-3a and ABT-737 induced Bax conformational change and mitochondrial apoptosis in AML cells in a strikingly synergistic fashion. Nutlin-3a induced p53-mediated apoptosis predominantly in S and G2/M cells, while cells in G1 were protected through induction of p21. In contrast, ABT-737 induced apoptosis predominantly in G1, the cell cycle phase with the lowest Bcl-2 protein levels and Bcl-2/Bax ratios. In addition, Bcl-2 phosphorylation on Ser70 was absent in G1 but detectable in G2/M, thus lower Bcl-2 levels and absence of Bcl-2 phosphorylation appeared to facilitate ABT-737-induced apoptosis of G1 cells. The complementary effects of Nutlin-3a and ABT-737 in different cell cycle phases could, in part, account for their synergistic activity. Our data suggest that combined targeting of Mdm2 and Bcl-2 proteins could offer considerable therapeutic promise in AML.


Subject(s)
Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Imidazoles/pharmacology , Leukemia, Myeloid, Acute/pathology , Mitochondria/metabolism , Nitrophenols/pharmacology , Piperazines/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Sulfonamides/pharmacology , Animals , Annexin A5/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm , G1 Phase/drug effects , G2 Phase/drug effects , Humans , Mice , Mitochondria/drug effects , Mitosis/drug effects , Mutant Proteins/metabolism , Phosphorylation/drug effects , Protein Conformation/drug effects , S Phase/drug effects , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism
10.
Cancer Cell ; 10(5): 375-88, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17097560

ABSTRACT

BCL-2 proteins are critical for cell survival and are overexpressed in many tumors. ABT-737 is a small-molecule BH3 mimetic that exhibits single-agent activity against lymphoma and small-cell lung cancer in preclinical studies. We here report that ABT-737 effectively kills acute myeloid leukemia blast, progenitor, and stem cells without affecting normal hematopoietic cells. ABT-737 induced the disruption of the BCL-2/BAX complex and BAK-dependent but BIM-independent activation of the intrinsic apoptotic pathway. In cells with phosphorylated BCL-2 or increased MCL-1, ABT-737 was inactive. Inhibition of BCL-2 phosphorylation and reduction of MCL-1 expression restored sensitivity to ABT-737. These data suggest that ABT-737 could be a highly effective antileukemia agent when the mechanisms of resistance identified here are considered.


Subject(s)
Apoptosis/physiology , Biphenyl Compounds , Drug Resistance, Neoplasm/physiology , Leukemia, Myeloid, Acute , Nitrophenols , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Sulfonamides , Animals , Biphenyl Compounds/metabolism , Biphenyl Compounds/therapeutic use , Cell Line , Dimerization , Hematopoietic Stem Cells/physiology , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Mice , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/metabolism , Nitrophenols/metabolism , Nitrophenols/therapeutic use , Piperazines/metabolism , Piperazines/therapeutic use , Protein Conformation , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/chemistry , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sulfonamides/metabolism , Sulfonamides/therapeutic use , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/chemistry , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
11.
Cell Cycle ; 5(19): 2223-9, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16969080

ABSTRACT

Patients with blast crisis (BC) CML frequently become resistant to Imatinib, a Bcr-Abl tyrosine kinase-targeting agent. Eg5, a microtubule-associated motor protein has been described to be highly expressed in BC CML by microarray analysis (Nowicki et al., Oncogene 2003; 22:3952-63). We investigated the regulation of Eg5 by Bcr-Abl tyrosine kinase and its potential as a therapeutic target in BC CML. Eg5 was highly expressed in all Philadelphia chromosome positive (Ph(+)) cell lines and BC CML patient samples. Inhibition of Bcr-Abl by Imatinib downregulated Eg5 expression in Imatinib-sensitive KBM5 and HL-60p185 cells, but not in Imatinib-resistant KBM5-STI571, harboring a T315I mutation, and Bcr-Abl-negative HL-60 cells. Blocking Eg5 expression with antisense oligonucleotide (Eg5-ASO) or inhibiting its activity with the small-molecule Eg5 inhibitor, S-trityl-L-cysteine induced G(2)/M cell cycle block and subsequent cell death in both Imatinib-sensitive and -resistant cells. Further, Eg5-ASO treatment of SCID mice harboring KBM5 cell xenografts significantly prolonged the median survival of the animals (p = 0.03). Our findings suggest that Eg5 is downstream of and regulated by Bcr-Abl tyrosine kinase in Philadelphia chromosome positive cells. Inhibition of Eg5 expression or its activity blocks cell cycle progression and induces cell death independent of the cellular response to Imatinib. Therefore, Eg5 could be a potential therapeutic target for the treatment of BC CML, in particular Imatinib-resistant BC CML.


Subject(s)
Drug Resistance, Neoplasm , Kinesins/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Piperazines/pharmacology , Pyrimidines/pharmacology , Animals , Apoptosis , Benzamides , Blast Crisis , Cell Cycle , Cell Line, Tumor , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/physiology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Imatinib Mesylate , Kinesins/antagonists & inhibitors , Kinesins/physiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Mice, SCID , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Transplantation, Heterologous
12.
Blood ; 108(2): 630-7, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16556893

ABSTRACT

Triptolide, a diterpenoid isolated from the Chinese herb Tripterygium wilfordii Hook.f, has shown antitumor activities in a broad range of solid tumors. Here, we examined its effects on leukemic cells and found that, at 100 nM or less, it potently induced apoptosis in various leukemic cell lines and primary acute myeloid leukemia (AML) blasts. We then attempted to identify its mechanisms of action. Triptolide induced caspase-dependent cell death accompanied by a significant decrease in XIAP levels. Forced XIAP overexpression attenuated triptolide-induced cell death. Triptolide also decreased Mcl-1 but not Bcl-2 and Bcl-X(L) levels. Bcl-2 overexpression suppressed triptolide-induced apoptosis. Further, triptolide induced loss of the mitochondrial membrane potential and cytochrome C release. Caspase-9 knock-out cells were resistant, while caspase-8-deficient cells were sensitive to triptolide, suggesting criticality of the mitochondrial but not the death receptor pathway for triptolide-induced apoptosis. Triptolide also enhanced cell death induced by other anticancer agents. Collectively, our results demonstrate that triptolide decreases XIAP and potently induces caspase-dependent apoptosis in leukemic cells mediated through the mitochondrial pathway at low nanomolar concentrations. The potent antileukemic activity of triptolide in vitro warrants further investigation of this compound for the treatment of leukemias and other malignancies.


Subject(s)
Apoptosis , Caspases/metabolism , Diterpenes/pharmacology , Leukemia/pathology , Mitochondria/metabolism , Phenanthrenes/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis Regulatory Proteins/analysis , Apoptosis Regulatory Proteins/drug effects , Caspase 8 , Caspase 9 , Cell Line, Tumor , Epoxy Compounds , Humans , Leukemia/drug therapy , X-Linked Inhibitor of Apoptosis Protein/analysis , X-Linked Inhibitor of Apoptosis Protein/drug effects , X-Linked Inhibitor of Apoptosis Protein/pharmacology
13.
Blood ; 107(4): 1546-54, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16223781

ABSTRACT

The multidrug resistance 1 (MDR1) gene product P-glycoprotein (P-gp) is frequently implicated in cross-resistance of tumors to chemotherapeutic drugs. In contrast, acute promyelocytic leukemia (APL) cells do not express MDR1 and are highly sensitive to anthracyclines. The combination of ATRA and the novel histone deacetylase inhibitor (HDACI) depsipeptide (FK228) induced P-gp expression and prevented growth inhibition and apoptosis in NB4 APL cells subsequently exposed to doxorubicin (DOX). ATRA/FK228 treatment after exposure to DOX, however, enhanced apoptosis. Both agents, ATRA or FK228, induced MDR1 mRNA. This effect was significantly enhanced by ATRA/FK228 administered in combination, due in part to increased H4 and H3-Lys9 acetylation of the MDR1 promoter and recruitment of the nuclear transcription factor Y alpha (NFYA) transcription activator to the CCAAT box. Cotreatment with specific P-gp inhibitor PSC833 reversed cytoprotective effects of ATRA/FK228. G1 cell-cycle arrest and p21 mRNA induction were also observed in response to ATRA/FK228, which may restrict DOX-induced apoptosis of cells in G2 phase. These results indicate that epigenetic mechanisms involving NF-YA transcription factor recruitment and histone acetylation are activated by ATRA and HDACI, induce MDR1 in APL cells, and point to the critical importance of mechanism-based sequential therapy in future clinical trials that combine HDAC inhibitors, ATRA, and anthracyclines.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Antibiotics, Antineoplastic/pharmacology , Depsipeptides/pharmacology , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/physiology , Leukemia, Promyelocytic, Acute/genetics , Cell Cycle/drug effects , Enzyme Inhibitors/pharmacology , Genes, MDR , Histone Deacetylase Inhibitors , Humans
14.
Blood ; 107(4): 1555-63, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16254145

ABSTRACT

KBM5 cells, derived from a patient with blast crisis Philadelphia chromosome-positive (Ph+) chronic myelogenous leukemia (CML), and imatinib-resistant KBM5 (KBM5-STI571) cells were found to express high levels of survivin. Inhibition of Bcr-Abl by imatinib significantly decreased survivin expression and cell viability in KBM5, but much less so in KBM5-STI571 cells. Inhibition of MEK, downstream of the Bcr-Abl signaling cascade decreased survivin expression and cell viability in both KBM5 and KBM5-STI571 cells. In addition, down-regulation of survivin by a survivin antisense oligonucleotide (Sur-AS-ODN) inhibited cell growth and induced maximal G2M block at 48 hours, whereas cell death was observed only at 72 hours in both KBM5 and KBM5-STI571 cells as shown by annexin V staining. Further, the combination of Sur-AS-ODN and imatinib induced more cell death in KBM5 cells than did either treatment alone. Down-regulating survivin also decreased colony-forming units (CFUs) in blast crisis CML patient samples. Our data therefore suggest that survivin is regulated by the Bcr-Abl/MAPK cascade in Ph+ CML. The facts that down-regulating survivin expression induced cell-growth arrest and subsequent cell death regardless of the cell response to imatinib and enhanced the sensitivity to imatinib suggest the potential therapeutic utility of this strategy in patients with CML, both imatinib sensitive and resistant.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Fusion Proteins, bcr-abl/metabolism , Gene Expression Regulation, Neoplastic , MAP Kinase Signaling System/physiology , Microtubule-Associated Proteins/genetics , Neoplasm Proteins/genetics , Piperazines/pharmacology , Pyrimidines/pharmacology , Benzamides , Cell Line, Tumor , Humans , Imatinib Mesylate , Inhibitor of Apoptosis Proteins , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , MAP Kinase Signaling System/drug effects , Survivin
15.
Blood ; 105(10): 4043-50, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15687241

ABSTRACT

We tested the effects of small-molecule XIAP antagonists based on a polyphenylurea pharmacophore on cultured acute myelogenous leukemia (AML) cell lines and primary patient samples. X-linked inhibitor of apoptosis protein (XIAP) antagonist N-[(5R)-6-[(anilinocarbonyl)amino]-5-((anilinocarbonyl){[(2R)-1-(4-cyclohexylbutyl)pyrrolidin-2-yl]methyl}amino)hexyl]-N-methyl-N'-phenylurea (1396-12), but not a structurally related control compound, induced apoptosis of primary leukemia samples with a lethal dose (LD50) of less than 10 microM in 16 of 27 (60%) samples. In contrast, XIAP antagonist 1396-12 was not lethal to the normal hematopoietic cells in short-term cytotoxicity assays. Response of primary AML specimens to XIAP inhibitor correlated with XIAP protein levels, with higher levels of XIAP associated with sensitivity. The XIAP antagonist 1396-12 induced activation of downstream caspases 3 and 7 prior to the activation of upstream caspase 8 and caspase 9. Apoptosis induction was also independent of B-cell lymphoma protein-2 (Bcl-2) or caspase 8, indicative of a downstream effect on apoptotic pathways. Thus, polyphenylurea-based XIAP antagonsists directly induce apoptosis of leukemia cells and AML patient samples at low micromolar concentrations through a mechanism of action distinct from conventional chemotherapeutic agents.


Subject(s)
Aniline Compounds/pharmacology , Apoptosis/drug effects , Caspases/metabolism , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Phenylurea Compounds/pharmacology , Proteins/antagonists & inhibitors , Cytarabine/pharmacology , Female , Humans , Male , Middle Aged , Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Cells, Cultured , X-Linked Inhibitor of Apoptosis Protein
16.
Mol Cancer Ther ; 3(10): 1249-62, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15486192

ABSTRACT

The peroxisome proliferator-activated receptor gamma (PPAR gamma) is a member of the nuclear receptor family that forms heterodimers with retinoid X receptor. These heterodimers bind to DNA and activate the transcription of target genes. Here, we report that the PPAR gamma receptor protein is expressed in primary myeloid and lymphoid leukemias and in lymphoma and myeloma cell lines. In this study, we compared the activity of several PPAR gamma ligands including BRL49653 (rosiglitazone), 15-deoxy-Delta 12,14-prostaglandin J(2), and the novel triterpenoid 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid on leukemia cells. Exposure to these PPAR gamma ligands induced apoptosis in myeloid (U937 and HL-60) and lymphoid (Su-DHL, Sup-M2, Ramos, Raji, Hodgkin's cell lines, and primary chronic lymphocytic leukemia) cells. A similar exposure to these PPAR gamma ligands induced the differentiation of myeloid leukemic cells. A combination of PPAR gamma ligands with a retinoid X receptor agonist (i.e., LG100268) or a retinoic acid receptor agonist (i.e., all trans-retinoic acid) enhanced differentiating and growth-inhibitory effects. 2-Cyano-3,12-dioxooleana-1,9-dien-28-oic acid induced differentiation and apoptosis with much greater potency than the other PPAR gamma ligands in established cell lines and primary chronic lymphocytic leukemia samples. Exposure to 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid induced mitochondrial depolarization and caspase activation, which was associated with apoptosis induction. In Bcl-2-overexpressing chronic lymphocytic leukemia cells, the small-molecule Bcl-2 inhibitor HA14-1 sensitized these cells to 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid-induced apoptosis. These results suggest that PPAR gamma ligation alone and in combination with retinoids holds promise as novel therapy for leukemias by activating the transcriptional activity of target genes that control apoptosis and differentiation in leukemias.


Subject(s)
Leukemia/pathology , Oleanolic Acid/analogs & derivatives , PPAR gamma/metabolism , Prostaglandin D2/analogs & derivatives , Retinoid X Receptors/metabolism , Agar/chemistry , Apoptosis , Blotting, Western , Cell Differentiation , Cell Line , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , Cell Survival , Dimerization , Fibrinolytic Agents/pharmacology , Flow Cytometry , HL-60 Cells , Humans , Imidazoles/pharmacology , Immunologic Factors/pharmacology , Jurkat Cells , Leukemia/drug therapy , Leukemia/metabolism , Ligands , Oleanolic Acid/pharmacology , Phagocytosis , Plasmids/metabolism , Prostaglandin D2/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/metabolism , Receptors, Retinoic Acid/metabolism , Retinoic Acid Receptor alpha , Rosiglitazone , Thiazolidinediones/pharmacology , Transcription, Genetic , Transcriptional Activation , Transfection , U937 Cells
17.
Cell Cycle ; 2(5): 488-93, 2003.
Article in English | MEDLINE | ID: mdl-12963850

ABSTRACT

Survivin, a member of inhibitor of apoptosis family of proteins, plays important roles in both cell proliferation and cell death. We previously observed that Survivin is overexpressed in leukemic cell lines and blasts from patients with acute myelogenous leukemia (AML). To understand the roles of Survivin in AML and search for new approaches to the treatment of AML, we inhibited Survivin expression in HL-60 cells with a Survivin anti-sense oligonucleotide (sur-AS-ODN) (ISIS 23722). This blocked significant numbers of HL-60 cells in G2/M phase, and halted cell proliferation at 24 hrs and progressing over time. There was only a slight increase in the number of apoptotic cells at 24 hrs compared with cells treated with nonsense oligonucleotide (NS-ODN). At 48 hrs, however, there were significant increases in sub-G1 phase and annexin V+ cells, suggesting that cell division defects caused cell death. This was supported by the finding that a reduction in the Survivin protein by sur-AS-ODN in cells under serum-free medium did not induce G2/M block and cell death compared to cells treated with NS-ODN. The formation of polyploid cells was observed 48 hrs after sur-AS-ODN treatment, as was the activation of caspase 3, which suggested that apoptotic cell death had occurred. The mitochondrial release of cytochrome C and Smac and the nuclear translocation of the apoptosis-inducing factor were also detected. Our results suggest that Survivin is essential for cell cycle progression in leukemic cells. Reduced Survivin expression causes a cell-cycle defect that leads to cell death through a mitochondrial pathway. This finding has potential utility for therapy of patients with AML.


Subject(s)
Apoptosis/physiology , Cell Death/physiology , Microtubule-Associated Proteins/metabolism , Cell Cycle/physiology , Cell Division/physiology , DNA, Antisense/metabolism , HL-60 Cells , Humans , Inhibitor of Apoptosis Proteins , Microtubule-Associated Proteins/drug effects , Mitochondria/metabolism , Mitochondrial Proteins , Neoplasm Proteins , Survivin
18.
Blood ; 102(12): 4179-86, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-12920030

ABSTRACT

Survivin and XIAP, members of the protein family known as the inhibitors of apoptosis, interfere with the activation of caspases, called the "cell death executioners." We examined Survivin (n = 116) and XIAP (n = 172) expression in primary acute myeloid leukemia (AML) blasts and assessed the impact of their expression on prognosis. They were detected in all samples analyzed. However, no correlation was observed with cytogenetics, remission attainment, or overall survival of patients with AML. To investigate the importance of caspases in chemotherapy-induced apoptosis in AML, we treated OCI-AML3 cells with Ara-C, doxorubicin, vincristine, and paclitaxel, which induced caspase cleavage and apoptosis. Blocking of caspase activation by pan-caspase inhibitor abolished poly(adenosine diphosphate [ADP]-ribose) polymerase cleavage and DNA fragmentation but did not prevent chemotherapy-induced cell death and did not inhibit, or only partially inhibited, mitochondrial release of cytochrome c, Smac, apoptosis-inducing factor (AIF), or loss of mitochondrial membrane potential. Caspase inhibition also did not protect AML blasts from chemotherapy-induced cell death in vitro. These results suggest that expression levels of Survivin or XIAP have no prognostic impact in AML patients. Although anticancer drugs induced caspase cleavage and apoptosis, cell killing was caspase independent. This may partially explain the lack of prognostic impact of XIAP and Survivin and may suggest caspase-independent mechanisms of cell death in AML.


Subject(s)
Caspase Inhibitors , Leukemia, Myeloid/pathology , Microtubule-Associated Proteins/analysis , Proteins/analysis , Acute Disease , Antineoplastic Agents/pharmacology , Bone Marrow Cells/pathology , Cell Death , Cell Survival , Cells, Cultured , Enzyme Inhibitors/pharmacology , Humans , Inhibitor of Apoptosis Proteins , Microtubule-Associated Proteins/physiology , Neoplasm Proteins , Prognosis , Proteins/physiology , Survivin , X-Linked Inhibitor of Apoptosis Protein
19.
Blood ; 99(9): 3461-4, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11964319

ABSTRACT

Recent studies suggest that the Bcl-2 and mitogen-activated protein kinase (MAPK) pathways together confer an aggressive, apoptosis-resistant phenotype on acute myelogenous leukemia (AML) cells. In this study, we analyzed the effects of simultaneous inhibition of these 2 pathways. In AML cell lines with constitutively activated MAPK, MAPK kinase (MEK) blockade by PD184352 strikingly potentiated the apoptosis induced by the small-molecule Bcl-2 inhibitor HA14-1 or by Bcl-2 antisense oligonucleotides. Isobologram analysis confirmed the synergistic nature of this interaction. Moreover, MEK blockade overcame Bcl-2 overexpression-mediated resistance to the proapoptotic effects of HA14-1. Most importantly, simultaneous exposure to PD184352 significantly (P =.01) potentiated HA14-1-mediated inhibition of clonogenic growth in all primary AML samples tested. These findings show that the Bcl-2 and MAPK pathways are relevant molecular targets in AML and that their concurrent inhibition could be developed into a new therapeutic strategy for this disease.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Leukemia, Myeloid, Acute/pathology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Benzamides/pharmacology , Benzopyrans/pharmacology , Drug Synergism , Enzyme Inhibitors/pharmacology , Humans , Nitriles/pharmacology , Oligonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , Tumor Cells, Cultured/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...