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










Publication year range
1.
Haematologica ; 102(9): 1537-1548, 2017 09.
Article in English | MEDLINE | ID: mdl-28659338

ABSTRACT

The bone marrow microenvironment is known to provide a survival advantage to residual acute myeloid leukemia cells, possibly contributing to disease recurrence. The mechanisms by which stroma in the microenvironment regulates leukemia survival remain largely unknown. Using reverse-phase protein array technology, we profiled 53 key protein molecules in 11 signaling pathways in 20 primary acute myeloid leukemia samples and two cell lines, aiming to understand stroma-mediated signaling modulation in response to the targeted agents temsirolimus (MTOR), ABT737 (BCL2/BCL-XL), and Nutlin-3a (MDM2), and to identify the effective combination therapy targeting acute myeloid leukemia in the context of the leukemia microenvironment. Stroma reprogrammed signaling networks and modified the sensitivity of acute myeloid leukemia samples to all three targeted inhibitors. Stroma activated AKT at Ser473 in the majority of samples treated with single-agent ABT737 or Nutlin-3a. This survival mechanism was partially abrogated by concomitant treatment with temsirolimus plus ABT737 or Nutlin-3a. Mapping the signaling networks revealed that combinations of two inhibitors increased the number of affected proteins in the targeted pathways and in multiple parallel signaling, translating into facilitated cell death. These results demonstrated that a mechanism-based selection of combined inhibitors can be used to guide clinical drug selection and tailor treatment regimens to eliminate microenvironment-mediated resistance in acute myeloid leukemia.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Resistance, Neoplasm/drug effects , Gene Expression Profiling , Gene Expression Regulation, Leukemic/drug effects , Leukemia, Myeloid, Acute/drug therapy , Signal Transduction/drug effects , Tumor Microenvironment/drug effects , Animals , Cell Line , Female , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Mice , Tumor Cells, Cultured
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.
Ann Hematol ; 91(12): 1861-70, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22893484

ABSTRACT

DNA methylation and BLC-2 are potential therapeutic targets in acute myeloid leukemia (AML). We investigated pharmacologic interaction between the DNA methyltransferase inhibitor 5-azacytidine (5-AZA) and the BCL-2 inhibitor ABT-737. Increased BCL-2 expression determined by reverse phase protein analysis was associated with poor survival in AML patients with unfavorable cytogenetics (n = 195). We found that 5-AZA, which itself has modest apoptotic activity, acts synergistically with ABT-737 to induce apoptosis. The 5-AZA/ABT-737 combination enhanced mitochondrial outer membrane permeabilization, as evidenced by effective conformational activation of BAX and ∆ψ(m) loss. Although absence of p53 limited apoptotic activities of 5-AZA and ABT-737 as single agents, the combination synergistically induced apoptosis independent of p53 expression. 5-AZA down-regulated MCL-1, known to mediate resistance to ABT-737, in a p53-independent manner. The 5-AZA/ABT-737 combination synergistically induced apoptosis in AML cells in seven of eight patients. 5-AZA significantly reduced MCL-1 levels in two of three samples examined. Our data provide a molecular rationale for this combination strategy in AML therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , DNA Modification Methylases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Mitochondria/drug effects , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis Regulatory Proteins/agonists , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/metabolism , Azacitidine/administration & dosage , Azacitidine/pharmacology , Azacitidine/therapeutic use , Biphenyl Compounds/administration & dosage , Biphenyl Compounds/pharmacology , Biphenyl Compounds/therapeutic use , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/therapeutic use , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred NOD , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nitrophenols/administration & dosage , Nitrophenols/pharmacology , Nitrophenols/therapeutic use , Piperazines/administration & dosage , Piperazines/pharmacology , Piperazines/therapeutic use , Sulfonamides/administration & dosage , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Tumor Cells, Cultured , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
4.
Blood ; 120(13): 2679-89, 2012 Sep 27.
Article in English | MEDLINE | ID: mdl-22826565

ABSTRACT

The interactions between the bone marrow (BM) microenvironment and acute myeloid leukemia (AML) is known to promote survival of AML cells. In this study, we used reverse phase-protein array (RPPA) technology to measure changes in multiple proteins induced by stroma in leukemic cells. We then investigated the potential of an mTOR kinase inhibitor, PP242, to disrupt leukemia/stroma interactions, and examined the effects of PP242 in vivo using a mouse model. Using RPPA, we confirmed that multiple survival signaling pathways, including the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR), were up-regulated in primary AML cells cocultured with stroma. PP242 effectively induced apoptosis in primary samples cultured with or without stroma. Mechanistically, PP242 attenuated the activities of mTORC1 and mTORC2, sequentially inhibited phosphorylated AKT, S6K, and 4EBP1, and concurrently suppressed chemokine receptor CXCR4 expression in primary leukemic cells and in stromal cells cultured alone or cocultured with leukemic cells. In the in vivo leukemia mouse model, PP242 inhibited mTOR signaling in leukemic cells and demonstrated a greater antileukemia effect than rapamycin. Our findings indicate that disrupting mTOR/AKT signaling with a selective mTOR kinase inhibitor can effectively target leukemic cells within the BM microenvironment.


Subject(s)
Apoptosis/drug effects , Bone Marrow/metabolism , Indoles/therapeutic use , Leukemia, Experimental/prevention & control , Leukemia, Myeloid, Acute/prevention & control , Mesenchymal Stem Cells/pathology , Multiprotein Complexes/antagonists & inhibitors , Purines/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antibiotics, Antineoplastic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols , Blotting, Western , Bone Marrow/pathology , Cell Proliferation , Coculture Techniques , Flow Cytometry , Humans , Leukemia, Experimental/mortality , Leukemia, Experimental/pathology , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mesenchymal Stem Cells/metabolism , Mice , Mice, SCID , Multiprotein Complexes/metabolism , Phosphorylation/drug effects , Protein Array Analysis , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/metabolism
5.
J Mol Med (Berl) ; 90(10): 1133-44, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22399013

ABSTRACT

In hematological malignancies, constitutive activation of the RAF/MEK/ERK pathway is frequently observed, conveys a poor prognosis, and constitutes a promising target for therapeutic intervention. Here, we investigated the molecular and functional effects of pharmacological MEK inhibition in cell line models of acute myeloid leukemia (AML) and freshly isolated primary AML samples. The small-molecule, ATP-non-competitive, MEK inhibitor PD0325901 markedly inhibited ERK phosphorylation and growth of several AML cell lines and approximately 70 % of primary AML samples. Growth inhibition was due to G(1)-phase arrest and induction of apoptosis. Transformation by constitutively active upstream pathway elements (HRAS, RAF-1, and MEK) rendered FDC-P1 cells exquisitely prone to PD0325901-induced apoptosis. Gene and protein expression profiling revealed a selective effect of PD0325901 on ERK phosphorylation and compensatory upregulation of the RAF/MEK and AKT/p70( S6K ) kinase modules, potentially mediating resistance to drug-induced growth inhibition. Consequently, in appropriate cellular contexts, both "vertical" (i.e., inhibition of RAF and MEK along the MAPK pathway) and "lateral" (i.e., simultaneous inhibition of the MEK/ERK and mTOR pathways) combination strategies may result in synergistic anti-leukemic effects. Overall, MEK inhibition exerts potent growth inhibitory and proapoptotic activity in preclinical models of AML, particularly in combination with other pathway inhibitors. Deeper understanding of the molecular mechanisms of action of MEK inhibitors will likely translate into more effective targeted strategies for the treatment of AML.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Benzenesulfonates/pharmacology , Diphenylamine/analogs & derivatives , Leukemia, Myeloid, Acute/drug therapy , MAP Kinase Kinase 1/antagonists & inhibitors , Pyridines/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Diphenylamine/pharmacology , Drug Resistance, Neoplasm , Drug Synergism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Inhibitory Concentration 50 , Intracellular Signaling Peptides and Proteins/metabolism , Niacinamide/analogs & derivatives , Oligonucleotide Array Sequence Analysis , Phenylurea Compounds , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Proto-Oncogene Proteins c-raf/metabolism , Signal Transduction , Sorafenib , Transcriptome/drug effects , Tumor Cells, Cultured/drug effects
6.
Cancer Res ; 70(12): 4949-60, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20501850

ABSTRACT

Peroxisome proliferator-activated receptor-gamma (PPARgamma) is a member of the nuclear receptor (NR) family of transcription factors with important regulatory roles in cellular growth, differentiation, and apoptosis. Using proteomic analysis, we showed expression of PPARgamma protein in a series of 260 newly diagnosed primary acute myelogenous leukemia (AML) samples. Forced expression of PPARgamma enhanced the sensitivity of myeloid leukemic cells to apoptosis induced by PPARgamma agonists 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid (CDDO) and 15-deoxy-(12,14)-15DPGJ(2), through preferential cleavage of caspase-8. No effects on cell cycle distribution or differentiation were noted, despite prominent induction of p21 in PPARgamma-transfected cells. In turn, antagonizing PPARgamma function by small interfering RNA or pharmacologic PPARgamma inhibitor significantly diminished apoptosis induction by CDDO. Overexpression of coactivator protein DRIP205 resulted in enhanced differentiation induction by CDDO in AML cells through PPARgamma activation. Studies with DRIP205 deletion constructs showed that the NR boxes of DRIP205 are not required for this coactivation. In a phase I clinical trial of CDDO (RTA-401) in leukemia, CDDO induced an increase in PPARgamma mRNA expression in six of nine patient samples; of those, induction of differentiation was documented in four patients and that of p21 in three patients, all expressing DRIP205 protein. In summary, these findings suggest that cellular levels of PPARgamma regulate induction of apoptosis via caspase-8 activation, whereas the coactivator DRIP205 is a determinant of induction of differentiation, in response to PPARgamma agonists in leukemic cells.


Subject(s)
Leukemia, Promyelocytic, Acute/metabolism , Leukemia, Promyelocytic, Acute/pathology , Mediator Complex Subunit 1/metabolism , Oleanolic Acid/analogs & derivatives , PPAR gamma/metabolism , Apoptosis/drug effects , Blotting, Western , Cell Cycle , Cell Differentiation/drug effects , Cell Line, Tumor , Chromatin Immunoprecipitation , Drug Resistance, Neoplasm , Flow Cytometry , Humans , Leukemia, Promyelocytic, Acute/genetics , Luciferases/metabolism , Mediator Complex Subunit 1/genetics , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Nitric Oxide/antagonists & inhibitors , Oleanolic Acid/pharmacology , PPAR gamma/antagonists & inhibitors , PPAR gamma/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction
7.
Blood ; 114(26): 5331-41, 2009 Dec 17.
Article in English | MEDLINE | ID: mdl-19850741

ABSTRACT

MicroRNAs (miRNAs) are associated with cytogenetics and molecular subtypes of acute myelogeneous leukemia (AML), but their impact on AML pathogenesis is poorly understood. We have previously shown that miR-29b expression is deregulated in primary AML blasts. In this work, we investigated the functional role of miR-29b in leukemogenesis. Restoration of miR-29b in AML cell lines and primary samples induces apoptosis and dramatically reduces tumorigenicity in a xenograft leukemia model. Transcriptome analysis after ectopic transfection of synthetic miR-29b into leukemia cells indicates that miR-29b target apoptosis, cell cycle, and proliferation pathways. A significant enrichment for apoptosis genes, including MCL-1, was found among the mRNAs inversely correlated with miR-29b expression in 45 primary AML samples. Together, the data support a tumor suppressor role for miR-29 and provide a rationale for the use of synthetic miR-29b oligonucleotides as a novel strategy to improve treatment response in AML.


Subject(s)
Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/genetics , MicroRNAs/genetics , Animals , Apoptosis/genetics , Blotting, Northern , Blotting, Western , Cell Proliferation , Female , Gene Expression , Humans , Mice , Mice, Nude , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Xenograft Model Antitumor Assays
8.
Blood ; 112(7): 2886-95, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18633130

ABSTRACT

Aberrant expression of Aurora kinases and inactivation of wild-type p53 by Mdm2 overexpression are frequent molecular events in acute myelogenous leukemia (AML), and preclinical data for inhibition of Aurora kinases or Mdm2 are promising. However, it remains largely unknown whether the viability of cells exposed to Aurora kinase inhibitors depends on the p53 status. We investigated the interaction of Aurora kinases and p53 pathways after their simultaneous blockades using a small-molecule pan-Aurora kinase inhibitor, MK-0457, and a selective small-molecule antagonist of Mdm2, Nutlin-3. We found that MK-0457, which itself activates p53 signaling, acts synergistically with Nutlin-3 to induce apoptosis in wild-type p53 AML cell lines OCI-AML-3 and MOLM-13 but not in p53-null HL-60 cells. MK-0457 and Nutlin-3 showed synergism in inducing p53, conformational change of Bax and Deltapsi(m) loss, suggesting an involvement of p53-mediated mitochondrial apoptosis. Nutlin-3 constrained endoreduplication after Aurora inhibition via activation of a p53-dependent postmitotic checkpoint and p21 induction in pseudo-G1 cells. Our findings provide the molecular rationale for concomitant targeting of Aurora kinases and Mdm2 in AML where TP53 mutations are rare and downstream p53 signaling is mostly intact.


Subject(s)
Apoptosis , Leukemia, Myeloid, Acute/enzymology , Mitochondria/pathology , Mitosis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Apoptosis/drug effects , Aurora Kinases , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA, Neoplasm/analysis , Drug Resistance, Neoplasm/drug effects , Drug Synergism , G1 Phase/drug effects , Gene Duplication/drug effects , Humans , Imidazoles/pharmacology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mitochondria/drug effects , Mitochondria/enzymology , Mitosis/drug effects , Piperazines/pharmacology , Protein Serine-Threonine Kinases/metabolism
9.
Cancer Res ; 68(9): 3413-20, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18451169

ABSTRACT

In this study, we investigated the mechanism of apoptosis induction of obatoclax (GX15-070), a novel Bcl-2 homology domain-3 (BH3) mimetic, in acute myeloid leukemia (AML) cell lines and primary AML samples. Obatoclax inhibited cell growth of HL-60, U937, OCI-AML3, and KG-1 cell lines. Apoptosis induction contributed to the observed antiproliferative effects at concentrations of this agent that mirror its affinity for antiapoptotic Bcl-2 proteins. We show that obatoclax can promote the release of cytochrome c from isolated leukemia cell mitochondria and that apoptosis induced by this agent is preceded by the release of Bak from Mcl-1, liberation of Bim from both Bcl-2 and Mcl-1, and the formation of an active Bak/Bax complex. Notably, apoptosis was diminished, but not fully prevented, in the absence of Bak/Bax or Bim, suggesting that obatoclax has additional targets that contribute to its cytotoxicity. At growth inhibitory doses that did not induce apoptosis or decrease viability, obatoclax induced an S-G(2) cell-cycle block. Obatoclax induced apoptosis in AML CD34+ progenitor cells with an average IC(50) of 3.59 +/- 1.23 micromol/L although clonogenicity was inhibited at concentrations of 75 to 100 nmol/L. Obatoclax synergized with the novel BH3 mimetic ABT-737 to induce apoptosis in OCI-AML3 cells and synergistically induced apoptosis in combination with AraC in leukemic cell lines and in primary AML samples. In conclusion, we show that obatoclax potently induces apoptosis and decreases leukemia cell proliferation and may be used in a novel therapeutic strategy for AML alone and in combination with other targeted agents and chemotherapeutics.


Subject(s)
Leukemia/drug therapy , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Pyrroles/pharmacology , Pyrroles/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , AraC Transcription Factor/pharmacology , Biomimetics , Biphenyl Compounds/pharmacology , Cells, Cultured , Drug Synergism , HL-60 Cells , Humans , Indoles , Mice , Mice, Knockout , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/metabolism , Nitrophenols/pharmacology , Piperazines/pharmacology , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/chemistry , Proto-Oncogene Proteins c-bcl-2/metabolism , Sulfonamides/pharmacology , Tumor Stem Cell Assay , U937 Cells
10.
Cancer Biol Ther ; 6(12): 1967-77, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18075297

ABSTRACT

Acute promyelocytic leukemia (APL) is associated with oncogenic PML-RARalpha that acts as a dominant negative transcriptional repressor of retinoic acid (RA) receptor target genes by recruiting histone deacetylase (HDAC). The peroxisome proliferator-activated receptor-gamma (PPARgamma) is a member of the nuclear receptor family that forms heterodimers with retinoid X receptor (RXR). In addition to RAR targets, PML-RARalpha silence a wide range of nuclear receptor target genes including PPARgamma targets. All-trans-retinoic acid (ATRA), a ligand for the RA receptor (RAR), restores normal retinoid signaling and induces terminal differentiation of APL cells; however, APL cells can develop resistance to ATRA. Using ATRA sensitive NB4 and ATRA-resistant derivative MR2 cell lines, we demonstrate that PPARgamma ligand 2-cyano-3, 12-dioxooleana-1, 9-dien-28-oic acid (CDDO) enhances pro-apoptotic and differentiating effects of ATRA in ATRA-sensitive NB4 cells and partially reverses ATRA resistance in MR2 cells. The CDDO/ATRA combination synergistically induces RARbeta2 expression both in ATRA-sensitive and -resistant APL cells. RARbeta2 MrNA induction by CDDO/ATRA was mediated in part by enhanced H3-Lys9 acetylation in the RARbeta2 promoter which in turn increased the affinity of RARbeta for betaRARE. PPARgamma specific inhibitor T007 and silencing of PPARgamma by siRNA diminished CDDO-induced maturation and RARbeta2 mRNA along with PPARgamma induction indicating that PPARgamma activation is at least partially responsible for the RARbeta2 transcription and maturation induction. In an in vivo mouse model of APL, CDDO derivative CDDO-methyl ester markedly enhanced ATRA-induced maturation and extended the survival of mice. In summary, these results provide rationale for the combined targeting of RAR and PPARgamma nuclear receptors in the therapy of APL.


Subject(s)
Antineoplastic Agents/pharmacology , Leukemia, Promyelocytic, Acute/drug therapy , Oleanolic Acid/analogs & derivatives , PPAR gamma/drug effects , Tretinoin/pharmacology , Acetylation/drug effects , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Histones/metabolism , Humans , Mice , Mice, Transgenic , Nicotinic Acids/pharmacology , Oleanolic Acid/pharmacology , Oleanolic Acid/therapeutic use , Promoter Regions, Genetic/drug effects , Protein Processing, Post-Translational/drug effects , RNA, Small Interfering/pharmacology , Receptors, Retinoic Acid/genetics , Response Elements/drug effects , Tetrahydronaphthalenes/pharmacology , Tretinoin/agonists
11.
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
12.
Mol Cancer Ther ; 5(2): 317-28, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16505105

ABSTRACT

HER2 overexpression is one of the most recognizable molecular alterations in breast tumors known to be associated with a poor prognosis. In the study described here, we explored the effect of HER2 overexpression on the sensitivity of breast cancer cells to the growth-inhibitory effects of 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid (CDDO), a synthetic triterpenoid, both in vitro and in vivo in a xenograft model of breast cancer. Both cell growth and colony formation in the soft agar assay, a hallmark of the transformation phenotype, were preferentially suppressed in HER2-overexpressing cell lines at low concentrations of CDDO, whereas growth-inhibitory effects at high concentrations did not correlate with the expression level of HER2. CDDO dose-dependently inhibited phosphorylation of HER2 in HER2-overexpressing cells and diminished HER2 kinase activity in vitro. CDDO induced the transactivation of the nuclear receptor peroxisome proliferator-activated receptor-gamma in both vector control and HER2-transfected MCF7 cells. Dose-response studies showed that the growth inhibition seen at lower concentrations of CDDO correlated with induction of the tumor suppressor gene caveolin-1, which is known to inhibit breast cancer cell growth. CDDO also reduced cyclin D1 mRNA and protein expression. In vivo studies with liposomally encapsulated CDDO showed complete abrogation of the growth of the highly tumorigenic MCF7/HER2 cells in a xenograft model of breast cancer. These findings provide the first in vitro and in vivo evidence that CDDO effectively inhibits HER2 tyrosine kinase activity and potently suppresses the growth of HER2-overexpressing breast cancer cells and suggest that CDDO has a therapeutic potential in advanced breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Caveolin 1/metabolism , Oleanolic Acid/analogs & derivatives , PPAR gamma/metabolism , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor, ErbB-2/antagonists & inhibitors , Animals , Breast Neoplasms/metabolism , Cell Proliferation/drug effects , Cyclin D1/genetics , Cyclin D1/metabolism , Female , Humans , Mice , Mice, Mutant Strains , Oleanolic Acid/pharmacology , Oleanolic Acid/therapeutic use , Phosphorylation/drug effects , RNA, Messenger/analysis , RNA, Messenger/metabolism , Transcriptional Activation , Xenograft Model Antitumor Assays
13.
Blood ; 106(9): 3150-9, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16014563

ABSTRACT

Although TP53 mutations are rare in acute myeloid leukemia (AML), inactivation of wild-type p53 protein frequently occurs through overexpression of its negative regulator MDM2 (murine double minute 2). Recently, small-molecule antagonists of MDM2, Nutlins, have been developed that inhibit the p53-MDM2 interaction and activate p53 signaling. Here, we study the effects of p53 activation by Nutlin-3 in AML cells. Treatment with MDM2 inhibitor triggered several molecular events consistent with induction of apoptosis: loss of mitochondrial membrane potential, caspase activation, phosphatidylserine externalization, and DNA fragmentation. There was a positive correlation in primary AML samples with wild-type p53 between baseline MDM2 protein levels and apoptosis induced by MDM2 inhibition. No induction of apoptosis was observed in AML samples harboring mutant p53. Colony formation of AML progenitors was inhibited in a dose-dependent fashion, whereas normal CD34+ progenitor cells were less affected. Mechanistic studies suggested that Nutlin-induced apoptosis was mediated by both transcriptional activation of proapoptotic Bcl-2 family proteins, and transcription-independent mitochondrial permeabilization resulting from mitochondrial p53 translocation. MDM2 inhibition synergistically enhanced cytotoxicity of cytosine arabinoside and doxorubicin in AML blasts but not in normal hematopoietic progenitor cells. p53 activation by targeting the p53-MDM2 interaction might offer a novel therapeutic strategy for AML that retain wild-type p53.


Subject(s)
Apoptosis/drug effects , Imidazoles/pharmacology , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Piperazines/pharmacology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Adult , Aged , Caspases/metabolism , Cell Nucleus/metabolism , Cytoplasm/metabolism , Doxorubicin/pharmacology , Enzyme Activation/drug effects , Female , Humans , Leukemia, Myeloid, Acute/therapy , Male , Middle Aged , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Cells, Cultured
14.
Cancer Res ; 64(21): 7927-35, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15520199

ABSTRACT

In acute myeloid leukemia (AML), resistance to chemotherapy is associated with defects in both the extrinsic and intrinsic pathways of apoptosis. Novel agents that activate endogenous apoptosis-inducing mechanisms directly may be potentially useful to overcome chemoresistance in AML. We examined the mechanisms of apoptosis induction by the novel synthetic triterpenoid 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid (CDDO) in AML cells. CDDO-induced apoptosis was associated with the loss of mitochondrial inner transmembrane potential, caspases activation, the translocation of apoptosis-inducing factor to the nucleus, and DNA fragmentation in AML cells. Apoptosis was equally evident in cells deficient in caspase-9 or caspase-8 after exposure to CDDO, suggesting caspase-independent cell death. The use of small interfering RNA to reduce the expression of apoptosis-inducing factor partially inhibited CDDO-induced apoptosis in AML cells. Cells overexpressing Bcl-2 were markedly resistant to CDDO-induced apoptosis. Moreover, CDDO promoted the release of cytochrome c from isolated mitochondria, suggesting that CDDO targets the mitochondria directly to trigger the intrinsic pathway of cell death in intact cells. Together, these results suggest that CDDO functions by activating the intrinsic pathway of apoptosis and initiates caspase-dependent and independent cell death. The direct modulation of mitochondrial-mediated, caspase-independent apoptosis by CDDO may be advantageous for overcoming chemoresistance in AML.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Caspases/physiology , Leukemia, Myeloid, Acute/drug therapy , Oleanolic Acid/analogs & derivatives , Oleanolic Acid/pharmacology , Apoptosis Inducing Factor , Cell Division/drug effects , Cell Line , Flavoproteins/antagonists & inhibitors , Flavoproteins/physiology , Humans , Leukemia, Myeloid, Acute/pathology , Membrane Potentials/drug effects , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/physiology , Mitochondria/drug effects , Mitochondria/physiology , PPAR gamma/physiology , Phosphatidylserines/metabolism , Proto-Oncogene Proteins c-bcl-2/physiology
15.
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
16.
Blood ; 103(5): 1815-22, 2004 Mar 01.
Article in English | MEDLINE | ID: mdl-14630813

ABSTRACT

Leptin is secreted by bone marrow (BM) adipocytes and stromal cells and was shown to stimulate myeloid proliferation. We here report that primary acute promyelocytic leukemia (APL) cells express high levels of the leptin-receptor (OB-R) long isoform. In cells with regulated promyelocytic leukemia-retinoic acid receptor (PML-RARalpha) expression, inducing PML-RARalpha was found to increase OB-R levels. We then investigated the effects of leptin produced by BM adipocytes on APL cells using a coculture system with mesenchymal stem cell (MSC)-derived adipocytes. In PML-RARalpha-expressing cells, all-trans retinoic acid (ATRA)- and doxorubicin-induced apoptosis were significantly reduced by coculture with adipocyte-differentiated MSCs. This antiapoptotic effect required direct cell-to-cell interactions, was associated with phosphorylation of signal transducer and activator of transcription-3 (STAT3) and mitogen-activated protein kinase (MAPK), and was reduced by blocking OB-R. This report provides a mechanistic basis for the BM adipocyte-leukemia cell interaction and suggests that OB-R receptor blockade may have therapeutic use in APL.


Subject(s)
Adipocytes/cytology , Leukemia, Promyelocytic, Acute/metabolism , Mesenchymal Stem Cells/metabolism , Neoplasm Proteins/biosynthesis , Oncogene Proteins, Fusion/biosynthesis , Receptors, Cell Surface/metabolism , Adipocytes/metabolism , Apoptosis , Blotting, Western , Cell Communication , Cell Differentiation , Cell Division , Coculture Techniques , DNA-Binding Proteins/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunohistochemistry , Leptin/metabolism , Phosphorylation , Protein Isoforms , Receptors, Leptin , Receptors, Retinoic Acid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor , Signal Transduction , Trans-Activators/metabolism , Tretinoin/metabolism , U937 Cells , Up-Regulation
17.
Cancer Res ; 63(18): 5926-39, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-14522919

ABSTRACT

Peroxisome proliferator-activated receptor gamma (PPARgamma) is a member of the nuclear hormonal receptor superfamily expressed in a large number of human cancers. Here, we demonstrate that PPARgamma is expressed and transcriptionally active in breast cancer cells independent of their p53, estrogen receptor, or human epidermal growth factor receptor 2 status. 2-Cyano-3,12-dioxooleana-1,9-dien-28-oic acid (CDDO), a novel synthetic triterpenoid, is a ligand for PPARgamma. We investigated the molecular mechanisms of CDDO on proliferation and apoptosis in breast cancer cells. In all breast cancer cell lines studied, CDDO transactivated PPARgamma, induced dose- and time-dependent cell growth inhibition, cell cycle arrest in G(1)-S and G(2)-M, and apoptosis. We then used differential cDNA array analysis to investigate the molecular changes induced by CDDO. After 16-h exposure of MCF-7 and MDA-MB-435 cells to CDDO, we found genes encoding the following proteins to be up-regulated in both cell lines: p21(Waf1/CIP1); GADD153; CAAT/enhancer binding protein transcription factor family members; and proteins involved in the ubiquitin-proteasome pathway. Among the down-regulated genes, we focused on the genes encoding cyclin D1, proliferating cell nuclear antigen, and the insulin receptor substrate 1. Using Western blot analysis and/or real-time PCR, we confirmed that CDDO regulated the expression of cyclin D1, p21(Waf1/CIP1), and Bcl-2. Cyclin D1 and p21(Waf1/CIP1) were additionally confirmed as important mediators of CDDO growth inhibition in genetically modified breast cancer cell lines. CDDO was able to significantly reduce the growth of MDA-MB-435 tumor cells in immunodeficient mice in vivo. The finding that CDDO can target genes critical for the regulation of cell cycle, apoptosis, and breast carcinogenesis suggests usage of CDDO as novel targeted therapy in breast cancer.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/drug therapy , Oleanolic Acid/analogs & derivatives , Oleanolic Acid/pharmacology , Receptors, Cytoplasmic and Nuclear/biosynthesis , Transcription Factors/biosynthesis , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Line, Tumor , Cyclin D1/biosynthesis , Cyclin D1/genetics , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Cyclins/genetics , Down-Regulation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunocompromised Host , Mice , Mice, Nude , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics
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.
Nucleic Acids Res ; 30(4): 1029-37, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-11842115

ABSTRACT

The Prp19p-associated complex is essential for the yeast pre-mRNA splicing reaction. The complex consists of at least eight protein components, but is not tightly associated with spliceosomal snRNAs. By a combination of genetic and biochemical methods we previously identified four components of this complex, Ntc25p, Ntc85p, Ntc30p and Ntc20p, all of them being novel splicing factors. We have now identified three other components of the complex, Ntc90p, Ntc77p and Ntc31p. These three proteins were also associated with the spliceosome during the splicing reaction in the same manner as Prp19p, concurrently with or immediately after dissociation of U4 snRNA. Two-hybrid analysis revealed that none of these proteins interacted with Prp19p or Ntc25p, but all interacted with Ntc85p. An interaction network between the identified components of the Prp19p-associated complex is demonstrated. Biochemical analysis revealed that Ntc90p, Ntc31p, Ntc30p and Ntc20p form a subcomplex, which, through interacting with Ntc85p and Ntc77p, can associate with Prp19p and Ntc25p to form the Prp19p-associated complex. Genetic analysis suggests that Ntc31p, Ntc30p and Ntc20p may play roles in modulating the function of Ntc90p.


Subject(s)
Fungal Proteins/metabolism , Fungal Proteins/physiology , Saccharomyces cerevisiae Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/physiology , Cell Division , Fungal Proteins/genetics , Macromolecular Substances , Models, Biological , Mutation , Precipitin Tests , RNA Splicing , RNA Splicing Factors , RNA, Fungal/metabolism , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Spliceosomes/metabolism , Two-Hybrid System Techniques
20.
Blood ; 99(1): 326-35, 2002 Jan 01.
Article in English | MEDLINE | ID: mdl-11756188

ABSTRACT

It has been shown that the novel synthetic triterpenoid CDDO inhibits proliferation and induces differentiation and apoptosis in myeloid leukemia cells. In the current study the effects of the C-28 methyl ester of CDDO, CDDO-Me, were analyzed on cell growth and apoptosis of leukemic cell lines and primary acute myelogenous leukemia (AML). CDDO-Me decreased the viability of leukemic cell lines, including multidrug resistant (MDR)-1-overexpressing, p53(null) HL-60-Dox and of primary AML cells, and it was 3- to 5-fold more active than CDDO. CDDO-Me induced a loss of mitochondrial membrane potential, induction of caspase-3 cleavage, increase in annexin V binding and DNA fragmentation, suggesting the induction of apoptosis. CDDO-Me induced pro-apoptotic Bax protein that preceded caspase activation. Furthermore, CDDO-Me inhibited the activation of ERK1/2, as determined by the inhibition of mitochondrial ERK1/2 phosphorylation, and it blocked Bcl-2 phosphorylation, rendering Bcl-2 less anti-apoptotic. CDDO-Me induced granulo-monocytic differentiation in HL-60 cells and monocytic differentiation in primary cells. Of significance, colony formation of AML progenitors was significantly inhibited in a dose-dependent fashion, whereas normal CD34(+) progenitor cells were less affected. Combinations with ATRA or the RXR-specific ligand LG100268 enhanced the effects of CDDO-Me on cell viability and terminal differentiation of myeloid leukemic cell lines. In conclusion, CDDO-Me is an MDR-1- and a p53-independent compound that exerts strong antiproliferative, apoptotic, and differentiating effects in myeloid leukemic cell lines and in primary AML samples when given in submicromolar concentrations. Differential effects of CDDO-Me on leukemic and normal progenitor cells suggest that CDDO-Me has potential as a novel compound in the treatment of hematologic malignancies.


Subject(s)
Apoptosis/drug effects , Cell Differentiation/drug effects , Leukemia, Myeloid, Acute/pathology , Oleanolic Acid/analogs & derivatives , Oleanolic Acid/pharmacology , Annexin A5/metabolism , Blast Crisis/pathology , Caspase 3 , Caspases/metabolism , Cell Survival/drug effects , Cytarabine/pharmacology , DNA Fragmentation/drug effects , Drug Interactions , Flow Cytometry , HL-60 Cells/drug effects , Humans , Membrane Potentials/drug effects , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Retinoids/pharmacology , Tretinoin/pharmacology , Tumor Cells, Cultured , bcl-2-Associated X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...