Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Haematologica ; 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38572562

ABSTRACT

Azacitidine/venetoclax is an active regimen in patients with newly diagnosed AML. However, primary or secondary resistance to azacitidine/venetoclax is an area of unmet need and overexpression of MCL-1 is suggested to be a potential resistance mechanism. Pevonedistat inhibits MCL-1 through activation of NOXA, and pevonedistat/azacitidine has previously shown activity in AML. To assess the tolerability and efficacy of adding pevonedistat to azacitidine/venetoclax in relapsed/refractory AML, we conducted a phase I multicenter openlabel study in 16 adults with relapsed/refractory AML. Patients were treated with azacitidine, venetoclax along with pevonedistat intravenously on days 1, 3 and 5 of each 28-day cycle at 10, 15 or 20 mg/m2 in successive cohorts in the dose escalation phase. The impact of treatment on protein neddylation as well as expression of pro-apoptotic BCL2 family members was assessed. The recommended phase II dose of pevonedistat was 20 mg/m2. Grade 3 or higher adverse events included neutropenia (31%), thrombocytopenia (13%), febrile neutropenia (19%), anemia (19%), hypertension (19%) and sepsis (19%). The overall response rate was 46.7% for the whole cohort including complete remission (CR) in 5 of 7 (71.4%) patients who were naïve to the hypomethylating agent/venetoclax. No measurable residual disease (MRD) was detected in 80.0% of the patients who achieved CR. The median time to best response was 50 (range: 23 - 77) days. Four patients were bridged to allogeneic stem cell transplantation. The combination of azacitidine, venetoclax and pevonedistat is safe and shows encouraging preliminary activity in patients with relapsed/refractory AML. (NCT04172844).

2.
Genes Dev ; 29(20): 2140-52, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26494789

ABSTRACT

Mitochondrial outer membrane permeabilization (MOMP), a key step in the intrinsic apoptotic pathway, is incompletely understood. Current models emphasize the role of BH3-only BCL2 family members in BAX and BAK activation. Here we demonstrate concentration-dependent BAK autoactivation under cell-free conditions and provide evidence that this autoactivation plays a key role in regulating the intrinsic apoptotic pathway in intact cells. In particular, we show that up to 80% of BAK (but not BAX) in lymphohematopoietic cell lines is oligomerized and bound to anti-apoptotic BCL2 family members in the absence of exogenous death stimuli. The extent of this constitutive BAK oligomerization is diminished by BAK knockdown and unaffected by BIM or PUMA down-regulation. Further analysis indicates that sensitivity of cells to BH3 mimetics reflects the identity of the anti-apoptotic proteins to which BAK is constitutively bound, with extensive BCLXL•BAK complexes predicting navitoclax sensitivity, and extensive MCL1•BAK complexes predicting A1210477 sensitivity. Moreover, high BAK expression correlates with sensitivity of clinical acute myelogenous leukemia to chemotherapy, whereas low BAK levels correlate with resistance and relapse. Collectively, these results inform current understanding of MOMP and provide new insight into the ability of BH3 mimetics to induce apoptosis without directly activating BAX or BAK.


Subject(s)
Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Transcriptional Activation/genetics , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism , Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Jurkat Cells , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/physiopathology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sulfonamides/pharmacology , bcl-X Protein/metabolism
4.
Blood ; 127(22): 2711-22, 2016 06 02.
Article in English | MEDLINE | ID: mdl-26917778

ABSTRACT

The mammalian target of rapamycin (mTOR), a kinase that regulates proliferation and apoptosis, has been extensively evaluated as a therapeutic target in multiple malignancies. Rapamycin analogs, which partially inhibit mTOR complex 1 (mTORC1), exhibit immunosuppressive and limited antitumor activity, but sometimes activate survival pathways through feedback mechanisms involving mTORC2. Thus, attention has turned to agents targeting both mTOR complexes by binding the mTOR active site. Here we show that disruption of either mTOR-containing complex is toxic to acute lymphocytic leukemia (ALL) cells and identify 2 previously unrecognized pathways leading to this cell death. Inhibition of mTORC1-mediated 4EBP1 phosphorylation leads to decreased expression of c-MYC and subsequent upregulation of the proapoptotic BCL2 family member PUMA, whereas inhibition of mTORC2 results in nuclear factor-κB-mediated expression of the Early Growth Response 1 (EGR1) gene, which encodes a transcription factor that binds and transactivates the proapoptotic BCL2L11 locus encoding BIM. Importantly, 1 or both pathways contribute to death of malignant lymphoid cells after treatment with dual mTORC1/mTORC2 inhibitors. Collectively, these observations not only provide new insight into the survival roles of mTOR in lymphoid malignancies, but also identify alterations that potentially modulate the action of mTOR dual inhibitors in ALL.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11/metabolism , Early Growth Response Protein 1/metabolism , Enzyme Inhibitors/pharmacology , NF-kappa B/metabolism , Phosphoproteins/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Humans , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , TOR Serine-Threonine Kinases/antagonists & inhibitors
5.
Nucleic Acids Res ; 44(6): 2816-26, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-26917015

ABSTRACT

A number of established and investigational anticancer drugs slow the religation step of DNA topoisomerase I (topo I). These agents induce cytotoxicity by stabilizing topo I-DNA covalent complexes, which in turn interact with advancing replication forks or transcription complexes to generate lethal lesions. Despite the importance of topo I-DNA covalent complexes, it has been difficult to detect these lesions within intact cells and tumors. Here, we report development of a monoclonal antibody that specifically recognizes covalent topo I-DNA complexes, but not free topo I or DNA, by immunoblotting, immunofluorescence or flow cytometry. Utilizing this antibody, we demonstrate readily detectable topo I-DNA covalent complexes after treatment with camptothecins, indenoisoquinolines and cisplatin but not nucleoside analogues. Topotecan-induced topo I-DNA complexes peak at 15-30 min after drug addition and then decrease, whereas indotecan-induced complexes persist for at least 4 h. Interestingly, simultaneous staining for covalent topo I-DNA complexes, phospho-H2AX and Rad51 suggests that topotecan-induced DNA double-strand breaks occur at sites distinct from stabilized topo I-DNA covalent complexes. These studies not only provide new insight into the action of topo I-directed agents, but also illustrate a strategy that can be applied to study additional topoisomerases and their inhibitors in vitro and in vivo.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Antineoplastic Agents, Phytogenic/pharmacology , DNA Topoisomerases, Type I/genetics , DNA/genetics , Gene Expression Regulation, Neoplastic , Topoisomerase I Inhibitors/pharmacology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Apoptosis/drug effects , Benzodioxoles/pharmacology , Cell Line, Tumor , Cisplatin/pharmacology , DNA/metabolism , DNA Breaks, Double-Stranded , DNA Topoisomerases, Type I/metabolism , HCT116 Cells , Histones/genetics , Histones/metabolism , Humans , Isoquinolines/pharmacology , K562 Cells , Mice , Molecular Sequence Data , Protein Binding/drug effects , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Sequence Alignment , Structure-Activity Relationship , Topotecan/pharmacology
6.
Blood ; 125(4): 658-67, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-25452615

ABSTRACT

Follicular lymphoma (FL), an indolent neoplasm caused by a t(14;18) chromosomal translocation that juxtaposes the BCL2 gene and immunoglobulin locus, has a variable clinical course and frequently undergoes transformation to an aggressive lymphoma. Although BCL2 mutations have been previously described, their relationship to FL progression remains unclear. In this study, we evaluated the frequency and nature of BCL2 mutations in 2 independent cohorts of grade 1 and 2 FLs, along with the correlation between BCL2 mutations, transformation risk, and survival. The prevalence of BCL2 coding sequence mutations was 12% in FL at diagnosis and 53% at transformation (P < .0001). The presence of these BCL2 mutations at diagnosis correlated with an increased risk of transformation (hazard ratio 3.6; 95% CI, 2.0-6.2; P < .0001) and increased risk of death due to lymphoma (median survival of 9.5 years with BCL2 mutations vs 20.4 years without; P = .012). In a multivariate analysis, BCL2 mutations and high FL international prognostic index were independent risk factors for transformation and death due to lymphoma. Some mutant Bcl-2 proteins exhibited enhanced antiapoptotic capacity in vitro. Accordingly, BCL2 mutations can affect antiapoptotic Bcl-2 function, are associated with increased activation-induced cytidine deaminase expression, and correlate with increased risk of transformation and death due to lymphoma.


Subject(s)
Cell Transformation, Neoplastic/genetics , Lymphoma, Follicular/genetics , Lymphoma, Follicular/mortality , Mutation , Proto-Oncogene Proteins c-bcl-2/genetics , Adult , Age Factors , Aged , Aged, 80 and over , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Chromosomes, Human, Pair 14/genetics , Chromosomes, Human, Pair 14/metabolism , Chromosomes, Human, Pair 18/genetics , Chromosomes, Human, Pair 18/metabolism , Cohort Studies , Cytidine Deaminase/biosynthesis , Cytidine Deaminase/genetics , Disease-Free Survival , Female , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoglobulins/genetics , Immunoglobulins/metabolism , Lymphoma, Follicular/metabolism , Male , Middle Aged , Prevalence , Proto-Oncogene Proteins c-bcl-2/metabolism , Risk Factors , Survival Rate
7.
J Biol Chem ; 289(30): 20543-58, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-24895135

ABSTRACT

Recombinant human tumor necrosis factor-α-related apoptosis inducing ligand (TRAIL), agonistic monoclonal antibodies to TRAIL receptors, and small molecule TRAIL receptor agonists are in various stages of preclinical and early phase clinical testing as potential anticancer drugs. Accordingly, there is substantial interest in understanding factors that affect sensitivity to these agents. In the present study we observed that the poly(ADP-ribose) polymerase (PARP) inhibitors olaparib and veliparib sensitize the myeloid leukemia cell lines ML-1 and K562, the ovarian cancer line PEO1, non-small cell lung cancer line A549, and a majority of clinical AML isolates, but not normal marrow, to TRAIL. Further analysis demonstrated that PARP inhibitor treatment results in activation of the FAS and TNFRSF10B (death receptor 5 (DR5)) promoters, increased Fas and DR5 mRNA, and elevated cell surface expression of these receptors in sensitized cells. Chromatin immunoprecipitation demonstrated enhanced binding of the transcription factor Sp1 to the TNFRSF10B promoter in the presence of PARP inhibitor. Knockdown of PARP1 or PARP2 (but not PARP3 and PARP4) not only increased expression of Fas and DR5 at the mRNA and protein level, but also recapitulated the sensitizing effects of the PARP inhibition. Conversely, Sp1 knockdown diminished the PARP inhibitor effects. In view of the fact that TRAIL is part of the armamentarium of natural killer cells, these observations identify a new facet of PARP inhibitor action while simultaneously providing the mechanistic underpinnings of a novel therapeutic combination that warrants further investigation.


Subject(s)
Apoptosis/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors , Receptors, TNF-Related Apoptosis-Inducing Ligand/biosynthesis , fas Receptor/biosynthesis , Humans , K562 Cells , Neoplasms/genetics , Neoplasms/metabolism , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Response Elements , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , fas Receptor/genetics
8.
Mol Pharmacol ; 85(5): 723-34, 2014 May.
Article in English | MEDLINE | ID: mdl-24569089

ABSTRACT

Signaling through the phosphatidylinositol-3 kinase (PI3K)/Akt pathway, which is aberrantly activated in >50% of carcinomas, inhibits apoptosis and contributes to drug resistance. Accordingly, several Akt inhibitors are currently undergoing preclinical or early clinical testing. To examine the effect of Akt inhibition on the activity of multiple widely used classes of antineoplastic agents, human cancer cell lines were treated with the Akt inhibitor A-443654 [(2S)-1-(1H-indol-3-yl)-3-[5-(3-methyl-2H-indazol-5-yl)pyridin-3-yl]oxypropan-2-amine; ATP-competitive] or MK-2206 (8-[4-(1-aminocyclobutyl)phenyl]-9-phenyl-2H-[1,2,4]triazolo[3,4-f][1,6]naphthyridin-3-one;dihydrochloride; allosteric inhibitor) or with small interfering RNA (siRNA) targeting phosphoinositide-dependent kinase 1 (PDK1) along with cisplatin, melphalan, camptothecin, or etoposide and assayed for colony formation. Surprisingly different results were observed when Akt inhibitors were combined with different drugs. Synergistic effects were observed in multiple cell lines independent of PI3K pathway status when A-443654 or MK-2206 was combined with the DNA cross-linking agents cisplatin or melphalan. In contrast, effects of the Akt inhibitors in combination with camptothecin or etoposide were more complicated. In HCT116 and DLD1 cells, which harbor activating PI3KCA mutations, A-443654 over a broad concentration range enhanced the effects of camptothecin or etoposide. In contrast, in cell lines lacking activating PI3KCA mutations, partial inhibition of Akt signaling synergized with camptothecin or etoposide, but higher A-443654 or MK-2206 concentrations (>80% inhibition of Akt signaling) or PDK1 siRNA antagonized the topoisomerase poisons by diminishing DNA synthesis, a process that contributes to effective DNA damage and killing by these agents. These results indicate that the effects of combining inhibitors of the PI3K/Akt pathway with certain classes of chemotherapeutic agents might be more complicated than previously recognized.


Subject(s)
Antineoplastic Agents/pharmacology , Heterocyclic Compounds, 3-Ring/pharmacology , Indazoles/pharmacology , Indoles/pharmacology , Poisons/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Topoisomerase II Inhibitors/pharmacology , Antineoplastic Agents/metabolism , Cell Line, Tumor , Drug Synergism , HCT116 Cells , Heterocyclic Compounds, 3-Ring/metabolism , Humans , Indazoles/metabolism , Indoles/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors , Poisons/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Topoisomerase I Inhibitors/metabolism , Topoisomerase I Inhibitors/pharmacology , Topoisomerase II Inhibitors/metabolism
9.
J Biol Chem ; 288(32): 22899-914, 2013 Aug 09.
Article in English | MEDLINE | ID: mdl-23798675

ABSTRACT

The CXCR4 chemokine receptor promotes survival of many different cell types. Here, we describe a previously unsuspected role for CXCR4 as a potent inducer of apoptosis in acute myeloid leukemia (AML) cell lines and a subset of clinical AML samples. We show that SDF-1, the sole ligand for CXCR4, induces the expected migration and ERK activation in the KG1a AML cell line transiently overexpressing CXCR4, but ERK activation did not lead to survival. Instead, SDF-1 treatment led via a CXCR4-dependent mechanism to apoptosis, as evidenced by increased annexin V staining, condensation of chromatin, and cleavage of both procaspase-3 and PARP. This SDF-1-induced death pathway was partially inhibited by hypoxia, which is often found in the bone marrow of AML patients. SDF-1-induced apoptosis was inhibited by dominant negative procaspase-9 but not by inhibition of caspase-8 activation, implicating the intrinsic apoptotic pathway. Further analysis showed that this pathway was activated by multiple mechanisms, including up-regulation of Bak at the level of mRNA and protein, stabilization of the Bak activator Noxa, and down-regulation of antiapoptotic Bcl-XL. Furthermore, adjusting expression levels of Bak, Bcl-XL, or Noxa individually altered the level of apoptosis in AML cells, suggesting that the combined modulation of these family members by SDF-1 coordinates their interplay to produce apoptosis. Thus, rather than mediating survival, SDF-1 may be a means to induce apoptosis of CXCR4-expressing AML cells directly in the SDF-1-rich bone marrow microenvironment if the survival cues of the bone marrow are disrupted.


Subject(s)
Apoptosis , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/metabolism , MAP Kinase Signaling System , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Receptors, CXCR4/metabolism , bcl-2 Homologous Antagonist-Killer Protein/biosynthesis , bcl-X Protein/biosynthesis , Annexin A5/genetics , Annexin A5/metabolism , Cell Survival/genetics , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Down-Regulation/genetics , Female , HEK293 Cells , Humans , Jurkat Cells , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , Protein Stability , Proto-Oncogene Proteins c-bcl-2/genetics , Receptors, CXCR4/genetics , U937 Cells , Up-Regulation/genetics , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-X Protein/genetics
10.
Blood ; 119(2): 476-87, 2012 Jan 12.
Article in English | MEDLINE | ID: mdl-22080480

ABSTRACT

The mammalian target of rapamycin (mTOR) plays crucial roles in proliferative and antiapoptotic signaling in lymphoid malignancies. Rapamycin analogs, which are allosteric mTOR complex 1 (mTORC1) inhibitors, are active in mantle cell lymphoma and other lymphoid neoplasms, but responses are usually partial and short-lived. In the present study we compared the effects of rapamycin with the dual mTORC1/mTORC2 inhibitor OSI-027 in cell lines and clinical samples representing divers lymphoid malignancies. In contrast to rapamycin, OSI-027 markedly diminished proliferation and induced apoptosis in a variety of lymphoid cell lines and clinical samples, including specimens of B-cell acute lymphocytic leukemia (ALL), mantle cell lymphoma, marginal zone lymphoma and Sezary syndrome. Additional analysis demonstrated that OSI-027-induced apoptosis depended on transcriptional activation of the PUMA and BIM genes. Overexpression of Bcl-2, which neutralizes Puma and Bim, or loss of procaspase 9 diminished OSI-027-induced apoptosis in vitro. Moreover, OSI-027 inhibited phosphorylation of mTORC1 and mTORC2 substrates, up-regulated Puma, and induced regressions in Jeko xenografts. Collectively, these results not only identify a pathway that is critical for the cytotoxicity of dual mTORC1/mTORC2 inhibitors, but also suggest that simultaneously targeting mTORC1 and mTORC2 might be an effective anti-lymphoma strategy in vivo.


Subject(s)
Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis/drug effects , Imidazoles/pharmacology , Lymphoma/pathology , Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Triazines/pharmacology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Blotting, Western , Cell Proliferation/drug effects , Humans , Immunoprecipitation , Immunosuppressive Agents/pharmacology , Lymphoma/drug therapy , Lymphoma/metabolism , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Phosphorylation/drug effects , Proteins/genetics , Proteins/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Cells, Cultured
11.
Haematologica ; 99(1): 60-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23996484

ABSTRACT

Although farnesyltransferase inhibitors have shown promising activity in relapsed lymphoma and sporadic activity in acute myelogenous leukemia, their mechanism of cytotoxicity is incompletely understood, making development of predictive biomarkers difficult. In the present study, we examined the action of tipifarnib in human acute myelogenous leukemia cell lines and clinical samples. In contrast to the Ras/MEK/ERK pathway-mediated Bim upregulation that is responsible for tipifarnib-induced killing of malignant lymphoid cells, inhibition of Rheb-induced mTOR signaling followed by dose-dependent upregulation of Bax and Puma occurred in acute myelogenous leukemia cell lines undergoing tipifarnib-induced apoptosis. Similar Bax and Puma upregulation occurred in serial bone marrow samples harvested from a subset of acute myelogenous leukemia patients during tipifarnib treatment. Expression of FTI-resistant Rheb M184L, like knockdown of Bax or Puma, diminished tipifarnib-induced killing. Further analysis demonstrated that increased Bax and Puma levels reflect protein stabilization rather than increased gene expression. In U937 cells selected for tipifarnib resistance, neither inhibition of signaling downstream of Rheb nor Bax and Puma stabilization occurred. Collectively, these results not only identify a pathway downstream from Rheb that contributes to tipifarnib cytotoxicity in human acute myelogenous leukemia cells, but also demonstrate that FTI-induced killing of lymphoid versus myeloid cells reflects distinct biochemical mechanisms downstream of different farnesylated substrates. (ClinicalTrials.gov identifier NCT00602771).


Subject(s)
Antineoplastic Agents/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Leukemia, Myeloid, Acute/metabolism , Monomeric GTP-Binding Proteins/metabolism , Neuropeptides/metabolism , Quinolones/pharmacology , bcl-2-Associated X Protein/metabolism , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , Farnesyltranstransferase/metabolism , Humans , Prenylation/drug effects , Protein Stability/drug effects , Proto-Oncogene Proteins/metabolism , Ras Homolog Enriched in Brain Protein , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , U937 Cells
12.
Cell Death Differ ; 31(4): 405-416, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38538744

ABSTRACT

BH3 mimetics, including the BCL2/BCLXL/BCLw inhibitor navitoclax and MCL1 inhibitors S64315 and tapotoclax, have undergone clinical testing for a variety of neoplasms. Because of toxicities, including thrombocytopenia after BCLXL inhibition as well as hematopoietic, hepatic and possible cardiac toxicities after MCL1 inhibition, there is substantial interest in finding agents that can safely sensitize neoplastic cells to these BH3 mimetics. Building on the observation that BH3 mimetic monotherapy induces AMP kinase (AMPK) activation in multiple acute leukemia cell lines, we report that the AMPK inhibitors (AMPKis) dorsomorphin and BAY-3827 sensitize these cells to navitoclax or MCL1 inhibitors. Cell fractionation and phosphoproteomic analyses suggest that sensitization by dorsomorphin involves dephosphorylation of the proapoptotic BCL2 family member BAD at Ser75 and Ser99, leading BAD to translocate to mitochondria and inhibit BCLXL. Consistent with these results, BAD knockout or mutation to BAD S75E/S99E abolishes the sensitizing effects of dorsomorphin. Conversely, dorsomorphin synergizes with navitoclax or the MCL1 inhibitor S63845 to induce cell death in primary acute leukemia samples ex vivo and increases the antitumor effects of navitoclax or S63845 in several xenograft models in vivo with little or no increase in toxicity in normal tissues. These results suggest that AMPK inhibition can sensitize acute leukemia to multiple BH3 mimetics, potentially allowing administration of lower doses while inducing similar antineoplastic effects.


Subject(s)
AMP-Activated Protein Kinases , Aniline Compounds , Myeloid Cell Leukemia Sequence 1 Protein , Pyrimidines , Sulfonamides , bcl-X Protein , Humans , Animals , Aniline Compounds/pharmacology , Sulfonamides/pharmacology , AMP-Activated Protein Kinases/metabolism , Mice , bcl-X Protein/metabolism , bcl-X Protein/antagonists & inhibitors , Cell Line, Tumor , Pyrimidines/pharmacology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Pyrazoles/pharmacology , bcl-Associated Death Protein/metabolism , Apoptosis/drug effects , Cell Death/drug effects , Leukemia/drug therapy , Leukemia/pathology , Leukemia/metabolism , Phosphorylation/drug effects , Peptide Fragments/pharmacology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Drug Synergism
13.
J Biol Chem ; 287(6): 4198-210, 2012 Feb 03.
Article in English | MEDLINE | ID: mdl-22158865

ABSTRACT

Poly(ADP-ribose) polymerase-1 (PARP1) plays critical roles in the regulation of DNA repair. Accordingly, small molecule inhibitors of PARP are being developed as agents that could modulate the activity of genotoxic chemotherapy, such as topoisomerase I poisons. In this study we evaluated the ability of the PARP inhibitor veliparib to enhance the cytotoxicity of the topoisomerase I poisons topotecan and camptothecin (CPT). Veliparib increased the cell cycle and cytotoxic effects of topotecan in multiple cell line models. Importantly, this sensitization occurred at veliparib concentrations far below those required to substantially inhibit poly(ADP-ribose) polymer synthesis and at least an order of magnitude lower than those involved in selective killing of homologous recombination-deficient cells. Further studies demonstrated that veliparib enhanced the effects of CPT in wild-type mouse embryonic fibroblasts (MEFs) but not Parp1(-/-) MEFs, confirming that PARP1 is the critical target for this sensitization. Importantly, parental and Parp1(-/-) MEFs had indistinguishable CPT sensitivities, ruling out models in which PARP1 catalytic activity plays a role in protecting cells from topoisomerase I poisons. To the contrary, cells were sensitized to CPT in a veliparib-independent manner upon transfection with PARP1 E988K, which lacks catalytic activity, or the isolated PARP1 DNA binding domain. These results are consistent with a model in which small molecule inhibitors convert PARP1 into a protein that potentiates the effects of topoisomerase I poisons by binding to damaged DNA and preventing its normal repair.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , DNA Topoisomerases, Type I , Neoplasms/drug therapy , Neoplasms/enzymology , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Benzimidazoles/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , Embryo, Mammalian/enzymology , Embryo, Mammalian/pathology , Fibroblasts/enzymology , Fibroblasts/pathology , Humans , Mice , Mice, Knockout , Neoplasms/genetics , Neoplasms/pathology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Topoisomerase I Inhibitors/pharmacology , Topotecan/pharmacology
14.
Blood ; 118(18): 4872-81, 2011 Nov 03.
Article in English | MEDLINE | ID: mdl-21673341

ABSTRACT

The mechanism of cytotoxicity of farnesyltransferase inhibitors is incompletely understood and seems to vary depending on the cell type. To identify potential determinants of sensitivity or resistance for study in the accompanying clinical trial (Witzig et al, page 4882), we examined the mechanism of cytotoxicity of tipifarnib in human lymphoid cell lines. Based on initial experiments showing that Jurkat variants lacking Fas-associated death domain or procaspase-8 undergo tipifarnib-induced apoptosis, whereas cells lacking caspase-9 or overexpressing Bcl-2 do not, we examined changes in Bcl-2 family members. Tipifarnib caused dose-dependent up-regulation of Bim in lymphoid cell lines (Jurkat, Molt3, H9, DoHH2, and RL) that undergo tipifarnib-induced apoptosis but not in lines (SKW6.4 and Hs445) that resist tipifarnib-induced apoptosis. Further analysis demonstrated that increased Bim levels reflect inhibition of signaling from c-Raf to MEK1/2 and ERK1/2. Additional experiments showed that down-regulation of the Ras guanine nucleotide exchange factor RasGRP1 diminished tipifarnib sensitivity, suggesting that H-Ras or N-Ras is a critical farnesylation target upstream of c-Raf in lymphoid cells. These results not only trace a pathway through c-Raf to Bim that contributes to tipifarnib cytotoxicity in human lymphoid cells but also identify potential determinants of sensitivity to this agent.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Cytotoxins/pharmacology , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Lymphocytes/drug effects , MAP Kinase Signaling System/drug effects , Membrane Proteins/genetics , Proto-Oncogene Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/drug effects , Drug Evaluation, Preclinical , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Jurkat Cells , Lymphocytes/metabolism , Lymphocytes/physiology , MAP Kinase Signaling System/physiology , Membrane Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins/metabolism , Up-Regulation/drug effects
15.
Blood ; 118(18): 4882-9, 2011 Nov 03.
Article in English | MEDLINE | ID: mdl-21725056

ABSTRACT

A phase 2 study of the oral farnesyltransferase inhibitor tipifarnib was conducted in 93 adult patients with relapsed or refractory lymphoma. Patients received tipifarnib 300 mg twice daily on days 1-21 of each 28-day cycle. The median number of prior therapies was 5 (range, 1-17). For the aggressive B-cell, indolent B-cell, and T-cell and Hodgkin lymphoma (HL/T) groups, the response rates were 17% (7/42), 7% (1/15), and 31% (11/36), respectively. Of the 19 responders, 7 were diffuse large B-cell non-Hodgkin lymphoma (NHL), 7 T-cell NHL, 1 follicular grade 2, and 4 HL. The median response duration for the 19 responders was 7.2 months (mean, 15.8 months; range, 1.8-62), and 5 patients in the HL/T group are still receiving treatment at 29-64+ months. The grade 3/4 toxicities observed were fatigue and reversible myelosuppression. Correlative studies suggest that Bim and Bcl-2 should be examined as potential predictors of response in future studies. These results indicate that tipifarnib has activity in lymphoma, particularly in heavily pretreated HL/T types, with little activity in follicular NHL. In view of its excellent toxicity profile and novel mechanism of action, further studies in combination with other agents appear warranted. This trial is registered at www.clinicaltrials.gov as #NCT00082888.


Subject(s)
Lymphoma/drug therapy , Quinolones/therapeutic use , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Resistance, Neoplasm/drug effects , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/therapeutic use , Farnesyltranstransferase/antagonists & inhibitors , Female , Humans , Lymphoma/pathology , Male , Middle Aged , Quinolones/administration & dosage , Quinolones/adverse effects , Recurrence , Treatment Outcome , Young Adult
16.
Virol J ; 10: 229, 2013 Jul 10.
Article in English | MEDLINE | ID: mdl-23842430

ABSTRACT

Vaccinia virus protein A33 (A33VACV) plays an important role in protection against orthopoxviruses, and hence is included in experimental multi-subunit smallpox vaccines. In this study we show that single-dose vaccination with recombinant Sindbis virus expressing A33VACV, is sufficient to protect mice against lethal challenge with vaccinia virus WR (VACV-WR) and ectromelia virus (ECTV) but not against cowpox virus (CPXV), a closely related orthopoxvirus. Moreover, a subunit vaccine based on the cowpox virus A33 ortholog (A33CPXV) failed to protect against cowpox and only partially protected mice against VACV-WR challenge. We mapped regions of sequence variation between A33VACV and A33CPXVand analyzed the role of such variations in protection. We identified a single protective region located between residues 104-120 that harbors a putative H-2Kd T cell epitope as well as a B cell epitope - a target for the neutralizing antibody MAb-1G10 that blocks spreading of extracellular virions. Both epitopes in A33CPXV are mutated and predicted to be non-functional. Whereas vaccination with A33VACV did not induce in-vivo CTL activity to the predicted epitope, inhibition of virus spread in-vitro, and protection from lethal VACV challenge pointed to the B cell epitope highlighting the critical role of residue L118 and of adjacent compensatory residues in protection. This epitope's critical role in protection, as well as its modifications within the orthopoxvirus genus should be taken in context with the failure of A33 to protect against CPXV as demonstrated here. These findings should be considered when developing new subunit vaccines and monoclonal antibody based therapeutics against orthopoxviruses, especially variola virus, the etiologic agent of smallpox.


Subject(s)
Cowpox virus/immunology , Ectromelia virus/immunology , Ectromelia, Infectious/prevention & control , Membrane Glycoproteins/immunology , Vaccinia virus/immunology , Vaccinia/prevention & control , Viral Envelope Proteins/immunology , Viral Vaccines/immunology , Adaptive Immunity , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Disease Models, Animal , Drug Carriers , Epitopes, B-Lymphocyte/genetics , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , Genetic Variation , Genetic Vectors , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Sindbis Virus/genetics , Viral Envelope Proteins/genetics , Viral Vaccines/administration & dosage
17.
JMIR Infodemiology ; 3: e43646, 2023 Jun 01.
Article in English | MEDLINE | ID: mdl-37261891

ABSTRACT

BACKGROUND: To respond to the need to establish infodemic management functions at the national public health institute in Germany (Robert Koch Institute, RKI), we explored and assessed available data sources, developed a social listening and integrated analysis framework, and defined when infodemic management functions should be activated during emergencies. OBJECTIVE: We aimed to establish a framework for social listening and integrated analysis for public health in the German context using international examples and technical guidance documents for infodemic management. METHODS: This study completed the following objectives: identified (potentially) available data sources for social listening and integrated analysis; assessed these data sources for their suitability and usefulness for integrated analysis in addition to an assessment of their risk using the RKI's standardized data protection requirements; developed a framework and workflow to combine social listening and integrated analysis to report back actionable infodemic insights for public health communications by the RKI and stakeholders; and defined criteria for activating integrated analysis structures in the context of a specific health event or health emergency. RESULTS: We included and classified 38% (16/42) of the identified and assessed data sources for social listening and integrated analysis at the RKI into 3 categories: social media and web-based listening data, RKI-specific data, and infodemic insights. Most data sources can be analyzed weekly to detect current trends and narratives and to inform a timely response by reporting insights that include a risk assessment and scalar judgments of different narratives and themes. CONCLUSIONS: This study identified, assessed, and prioritized a wide range of data sources for social listening and integrated analysis to report actionable infodemic insights, ensuring a valuable first step in establishing and operationalizing infodemic management at the RKI. This case study also serves as a roadmap for others. Ultimately, once operational, these activities will inform better and targeted public health communication at the RKI and beyond.

18.
Cancer Med ; 12(23): 21229-21239, 2023 12.
Article in English | MEDLINE | ID: mdl-37960985

ABSTRACT

BACKGROUND: Despite recent approval of several new agents, relapsed acute lymphoblastic leukemia (ALL) remains challenging to treat. Sapanisertib (MLN0128/TAK-228) is an oral TORC1/2 inhibitor that exhibited preclinical activity against ALL. METHODS: We conducted a single-arm multi-center Phase II study of sapanisertib monotherapy (3 mg orally daily of the milled formulation for 21 days every 28 days) in patients with ALL through the Experimental Therapeutics Clinical Trials Network (NCI-9775). RESULTS: Sixteen patients, 15 of whom were previously treated (median 3 prior lines of therapy), were enrolled. Major grade 3-4 non-hematologic toxicities included mucositis (3 patients) and hyperglycemia (2 patients) as well as hepatic failure, seizures, confusion, pneumonitis, and anorexia (1 patient each). Grade >2 hematological toxicity included leukopenia (3), lymphopenia (2), thrombocytopenia, and neutropenia (1). The best response was stable disease in 2 patients (12.5%), while only 3 patients (19%) were able to proceed to Cycle 2. Pharmacokinetic analysis demonstrated drug exposures similar to those observed in solid tumor patients. Immunoblotting in serially collected samples indicated limited impact of treatment on phosphorylation of mTOR pathway substrates such as 4EBP1, S6, and AKT. CONCLUSION: In summary, single-agent sapanisertib had a good safety profile but limited target inhibition or efficacy in ALL as a single agent. This trial was registered at ClinicalTrials.gov as NCT02484430.


Subject(s)
Benzoxazoles , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy
19.
Blood Cancer J ; 13(1): 81, 2023 05 17.
Article in English | MEDLINE | ID: mdl-37193683

ABSTRACT

How to identify follicular lymphoma (FL) patients with low disease burden but high risk for early progression is unclear. Building on a prior study demonstrating the early transformation of FLs with high variant allele frequency (VAF) BCL2 mutations at activation-induced cytidine deaminase (AICDA) sites, we examined 11 AICDA mutational targets, including BCL2, BCL6, PAX5, PIM1, RHOH, SOCS, and MYC, in 199 newly diagnosed grade 1 and 2 FLs. BCL2 mutations with VAF ≥20% occurred in 52% of cases. Among 97 FL patients who did not initially receive rituximab-containing therapy, nonsynonymous BCL2 mutations at VAF ≥20% were associated with increased transformation risk (HR 3.01, 95% CI 1.04-8.78, p = 0.043) and a trend toward shorter event-free survival (EFS, median 20 months with mutations versus 54 months without, p = 0.052). Other sequenced genes were less frequently mutated and did not increase the prognostic value of the panel. Across the entire population, nonsynonymous BCL2 mutations at VAF ≥20% were associated with decreased EFS (HR 1.55, 95% CI 1.02-2.35, p = 0.043 after correction for FLIPI and treatment) and decreased overall survival after median 14-year follow-up (HR 1.82, 95% CI 1.05-3.17, p = 0.034). Thus, high VAF nonsynonymous BCL2 mutations remain prognostic even in the chemoimmunotherapy era.


Subject(s)
Lymphoma, Follicular , Humans , Lymphoma, Follicular/drug therapy , Lymphoma, Follicular/genetics , Mutation , Prognosis , Progression-Free Survival , Proto-Oncogene Proteins c-bcl-2/genetics
20.
J Biol Chem ; 286(41): 35823-35833, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21865165

ABSTRACT

Previous studies have suggested that there are two signaling pathways leading from ligation of the Fas receptor to induction of apoptosis. Type I signaling involves Fas ligand-induced recruitment of large amounts of FADD (FAS-associated death domain protein) and procaspase 8, leading to direct activation of caspase 3, whereas type II signaling involves Bid-mediated mitochondrial perturbation to amplify a more modest death receptor-initiated signal. The biochemical basis for this dichotomy has previously been unclear. Here we show that type I cells have a longer half-life for Fas message and express higher amounts of cell surface Fas, explaining the increased recruitment of FADD and subsequent signaling. Moreover, we demonstrate that cells with type II Fas signaling (Jurkat or HCT-15) can signal through a type I pathway upon forced receptor overexpression and that shRNA-mediated Fas down-regulation converts cells with type I signaling (A498) to type II signaling. Importantly, the same cells can exhibit type I signaling for Fas and type II signaling for TRAIL (TNF-α-related apoptosis-inducing ligand), indicating that the choice of signaling pathway is related to the specific receptor, not some other cellular feature. Additional experiments revealed that up-regulation of cell surface death receptor 5 levels by treatment with 7-ethyl-10-hydroxy-camptothecin converted TRAIL signaling in HCT116 cells from type II to type I. Collectively, these results suggest that the type I/type II dichotomy reflects differences in cell surface death receptor expression.


Subject(s)
Fas-Associated Death Domain Protein/biosynthesis , Gene Expression Regulation/physiology , Signal Transduction/physiology , TNF-Related Apoptosis-Inducing Ligand/biosynthesis , fas Receptor/metabolism , Animals , Fas-Associated Death Domain Protein/genetics , Humans , Jurkat Cells , Mice , TNF-Related Apoptosis-Inducing Ligand/genetics , fas Receptor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL