Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 28
1.
J Med Chem ; 66(13): 8666-8686, 2023 07 13.
Article En | MEDLINE | ID: mdl-37403966

Hypoxia-inducible factors (HIFs) are heterodimeric transcription factors induced in diverse pathophysiological settings. Inhibition of HIF-2α has become a strategy for cancer treatment since the discovery that small molecules, upon binding into a small cavity of the HIF-2α PAS B domain, can alter its conformation and disturb the activity of the HIF dimer complex. Herein, the design, synthesis, and systematic SAR exploration of cycloalkyl[c]thiophenes as novel HIF-2α inhibitors are described, providing the first chemotype featuring an alkoxy-aryl scaffold. X-ray data confirmed the ability of these inhibitors to induce perturbation of key amino acids by appropriately presenting key pharmacophoric elements in the hydrophobic cavity. Selected compounds showed inhibition of VEGF-A secretion in cancer cells and prevention of Arg1 expression and activity in IL4-stimulated macrophages. Moreover, in vivo target gene modulation was demonstrated with compound 35r. Thus, the disclosed HIF-2α inhibitors represent valuable tools for investigating selective HIF-2α inhibition and its effect on tumor biology.


Basic Helix-Loop-Helix Transcription Factors , Thiophenes , Humans , Basic Helix-Loop-Helix Transcription Factors/metabolism , Thiophenes/pharmacology , Transcription Factors , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit
2.
JCI Insight ; 7(24)2022 12 22.
Article En | MEDLINE | ID: mdl-36413415

Metastatic clear cell renal cell carcinomas (ccRCCs) are resistant to DNA-damaging chemotherapies, limiting therapeutic options for patients whose tumors are resistant to tyrosine kinase inhibitors and/or immune checkpoint therapies. Here we show that mouse and human ccRCCs were frequently characterized by high levels of endogenous DNA damage and that cultured ccRCC cells exhibited intact cellular responses to chemotherapy-induced DNA damage. We identify that pharmacological inhibition of the DNA damage-sensing kinase ataxia telangiectasia and Rad3-related protein (ATR) with the orally administered, potent, and selective drug M4344 (gartisertib) induced antiproliferative effects in ccRCC cells. This effect was due to replication stress and accumulation of DNA damage in S phase. In some cells, DNA damage persisted into subsequent G2/M and G1 phases, leading to the frequent accumulation of micronuclei. Daily single-agent treatment with M4344 inhibited the growth of ccRCC xenograft tumors. M4344 synergized with chemotherapeutic drugs including cisplatin and carboplatin and the poly(ADP-ribose) polymerase inhibitor olaparib in mouse and human ccRCC cells. Weekly M4344 plus cisplatin treatment showed therapeutic synergy in ccRCC xenografts and was efficacious in an autochthonous mouse ccRCC model. These studies identify ATR inhibition as a potential novel therapeutic option for ccRCC.


Antineoplastic Agents , Carcinoma, Renal Cell , Humans , Animals , Mice , Carcinoma, Renal Cell/drug therapy , Cisplatin , Ataxia Telangiectasia Mutated Proteins , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use
3.
Oncotarget ; 11(48): 4490-4503, 2020 Dec 01.
Article En | MEDLINE | ID: mdl-33400734

Tumor cells typically enhance their metabolic capacity to sustain their higher rate of growth and proliferation. One way to elevate the nutrient intake into cancer cells is to increase the expression of genes encoding amino acid transporters, which may represent targetable vulnerabilities. Here, we study the regulation and function of the broad amino acid transporter SLC6A14 in combination with metabolic stress, providing insights into an uncharacterized aspect of the transporter activity. We analyze the pattern of transcriptional changes in a panel of breast cancer cell lines upon metabolic stress and found that SLC6A14 expression levels are increased in the absence of methionine. Methionine deprivation, which can be achieved via modulation of dietary methionine intake in tumor cells, in turn leads to a heightened activation of the AMP-activated kinase (AMPK) in SLC6A14-deficient cells. While SLC6A14 genetic deficiency does not have a major impact on cell proliferation, combined depletion of AMPK and SLC6A14 leads to an increase in apoptosis upon methionine starvation, suggesting that combined targeting of SLC6A14 and AMPK can be exploited as a therapeutic approach to starve tumor cells.

4.
Bioorg Med Chem Lett ; 29(4): 646-653, 2019 02 15.
Article En | MEDLINE | ID: mdl-30626557

In oncology, the "Warburg effect" describes the elevated production of energy by glycolysis in cancer cells. The ubiquitous and hypoxia-induced 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) plays a noteworthy role in the regulation of glycolysis by producing fructose-2,6-biphosphate (F-2,6-BP), a potent activator of the glycolysis rate-limiting phosphofructokinase PFK-1. Series of amides and sulfonamides derivatives based on a N-aryl 6-aminoquinoxaline scaffold were synthesized and tested for their inhibition of PFKFB3 in vitro in a biochemical assay as well as in HCT116 cells. The carboxamide series displayed satisfactory kinetic solubility and metabolic stability, and within this class, potent lead compounds with low nanomolar activity have been identified with a suitable profile for further in vivo evaluation.


Amides/chemistry , Phosphofructokinase-2/antagonists & inhibitors , Quinoxalines/chemistry , Quinoxalines/pharmacology , Sulfonamides/chemistry , HCT116 Cells , Humans , Kinetics , Solubility
5.
ChemMedChem ; 14(1): 169-181, 2019 01 08.
Article En | MEDLINE | ID: mdl-30378281

Energy and biomass production in cancer cells are largely supported by aerobic glycolysis in what is called the Warburg effect. The process is regulated by key enzymes, among which phosphofructokinase PFK-2 plays a significant role by producing fructose-2,6-biphosphate; the most potent activator of the glycolysis rate-limiting step performed by phosphofructokinase PFK-1. Herein, the synthesis, biological evaluation and structure-activity relationship of novel inhibitors of 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3), which is the ubiquitous and hypoxia-induced isoform of PFK-2, are reported. X-ray crystallography and docking were instrumental in the design and optimisation of a series of N-aryl 6-aminoquinoxalines. The most potent representative, N-(4-methanesulfonylpyridin-3-yl)-8-(3-methyl-1-benzothiophen-5-yl)quinoxalin-6-amine, displayed an IC50 of 14 nm for the target and an IC50 of 0.49 µm for fructose-2,6-biphosphate production in human colon carcinoma HCT116 cells. This work provides a new entry in the field of PFKFB3 inhibitors with potential for development in oncology.


Drug Discovery , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Phosphofructokinase-2/antagonists & inhibitors , Quinoxalines/chemistry , Quinoxalines/pharmacology , Cell Survival/drug effects , Crystallography, X-Ray , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , HCT116 Cells , Humans , Lactic Acid/antagonists & inhibitors , Lactic Acid/biosynthesis , Models, Molecular , Molecular Structure , Phosphofructokinase-2/metabolism , Quinoxalines/chemical synthesis , Structure-Activity Relationship
6.
Commun Biol ; 1: 210, 2018.
Article En | MEDLINE | ID: mdl-30511023

Cytotoxic activities of several Golgi-dispersing compounds including AMF-26/M-COPA, brefeldin A and golgicide A have previously been shown to induce autophagy or apoptosis. Here, we demonstrate that these Golgi disruptors also trigger ferroptosis, a non-apoptotic form of cell death characterized by iron-dependent oxidative degradation of lipids. Inhibitors of ferroptosis not only counteract cell death, but they also protect from Golgi dispersal and inhibition of protein secretion in response to several Golgi stress agents. Furthermore, the application of sublethal doses of ferroptosis-inducers such as erastin and sorafenib, low cystine growth conditions, or genetic knockdown of SLC7A11 and GPX4 all similarly protect cells from Golgi stress and lead to modulation of ACSL4, SLC7A5, SLC7A11 or GPX4 levels. Collectively, this study suggests a previously unrecognized function of the Golgi apparatus, which involves cellular redox control and prevents ferroptotic cell death.

7.
Mol Biol Cell ; 29(1): 42-52, 2018 01 01.
Article En | MEDLINE | ID: mdl-29118074

The secretory pathway is a major determinant of cellular homoeostasis. While research into secretory stress signaling has so far mostly focused on the endoplasmic reticulum (ER), emerging data suggest that the Golgi itself serves as an important signaling hub capable of initiating stress responses. To systematically identify novel Golgi stress mediators, we performed a transcriptomic analysis of cells exposed to three different pharmacological compounds known to elicit Golgi fragmentation: brefeldin A, golgicide A, and monensin. Subsequent gene-set enrichment analysis revealed a significant contribution of the ETS family transcription factors ELK1, GABPA/B, and ETS1 to the control of gene expression following compound treatment. Induction of Golgi stress leads to a late activation of the ETS upstream kinases MEK1/2 and ERK1/2, resulting in enhanced ETS factor activity and the transcription of ETS family target genes related to spliceosome function and cell death induction via alternate MCL1 splicing. Further genetic analyses using loss-of-function and gain-of-function experiments suggest that these transcription factors operate in parallel.


Alternative Splicing/genetics , Golgi Apparatus/metabolism , MAP Kinase Signaling System , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Proto-Oncogene Proteins c-ets/metabolism , Stress, Physiological , Transcription, Genetic , A549 Cells , Alternative Splicing/drug effects , Apoptosis/drug effects , Brefeldin A/pharmacology , Cytoprotection/drug effects , Gene Expression Profiling , Gene Knockdown Techniques , Golgi Apparatus/drug effects , HEK293 Cells , HeLa Cells , Humans , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/metabolism , Monensin/pharmacology , Pyridines/pharmacology , Quinolines/pharmacology , Small Molecule Libraries/pharmacology , Spliceosomes/drug effects , Spliceosomes/metabolism , Stress, Physiological/drug effects , Transcription, Genetic/drug effects , Transcriptome/drug effects , Transcriptome/genetics , Up-Regulation/drug effects
8.
Cell Death Dis ; 8(10): e3069, 2017 10 05.
Article En | MEDLINE | ID: mdl-28981087

Disruption of the Golgi apparatus can induce a distinct form of programmed cell death that has not been thoroughly characterized. We found that pharmacological application of Golgi stress leads to induction of death receptors (DRs) 4 and 5. DR4 appears to be primarily responsible for the initiation of cell death downstream of Golgi stress, whereas DR5 seems to be more important for cell death triggered by endoplasmic reticulum (ER) stress in specific cancer cell lines. DR induction downstream of either Golgi or ER stress mainly causes intracellular accumulation of DR4 presumably at the Golgi, rather than increased expression on the cell surface. Nevertheless, cells treated with secretory pathway stressors displayed an increased susceptibility to TRAIL (tumor necrosis factor related apoptosis inducing ligand), the endogenous ligand of DR4/5, probably due to intracellular sequestration of the caspase-8 regulator CFLAR (caspase-8 and FADD-like apoptosis regulator). These findings have implications for the treatment of cancer with DR agonists and our general understanding of DR signaling while highlighting the role of the Golgi apparatus as a cell death signaling platform.


Endoplasmic Reticulum Stress/drug effects , Neoplasms/drug therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Apoptosis/drug effects , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Caspase 8/genetics , Cell Line, Tumor , Endoplasmic Reticulum Stress/genetics , Golgi Apparatus/drug effects , Golgi Apparatus/genetics , Humans , Neoplasms/genetics , Neoplasms/pathology
9.
Mol Biol Cell ; 28(26): 3756-3772, 2017 Dec 15.
Article En | MEDLINE | ID: mdl-29074567

The Golgi apparatus is increasingly recognized as a major hub for cellular signaling and is involved in numerous pathologies, including neurodegenerative diseases and cancer. The study of Golgi stress-induced signaling pathways relies on the selectivity of the available tool compounds of which currently only a few are known. To discover novel Golgi-fragmenting agents, transcriptomic profiles of cells treated with brefeldin A, golgicide A, or monensin were generated and compared with a database of gene expression profiles from cells treated with other bioactive small molecules. In parallel, a phenotypic screen was performed for compounds that alter normal Golgi structure. Histone deacetylase (HDAC) inhibitors and DNA-damaging agents were identified as novel Golgi disruptors. Further analysis identified HDAC1/HDAC9 as well as BRD8 and DNA-PK as important regulators of Golgi breakdown mediated by HDAC inhibition. We provide evidence that combinatorial HDACi/(+)-JQ1 treatment spurs synergistic Golgi dispersal in several cancer cell lines, pinpointing a possible link between drug-induced toxicity and Golgi morphology alterations.


Azepines/pharmacology , Drug Evaluation, Preclinical/methods , Histone Deacetylase 1/metabolism , Histone Deacetylase Inhibitors/pharmacology , Triazoles/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Drug Synergism , Gene Expression Profiling/methods , Golgi Apparatus/drug effects , Histone Deacetylase 1/genetics , Histone Deacetylases/metabolism , Histones/metabolism , Humans
10.
Traffic ; 18(8): 530-544, 2017 08.
Article En | MEDLINE | ID: mdl-28485883

The Golgi apparatus is part of the secretory pathway and of central importance for modification, transport and sorting of proteins and lipids. ADP-ribosylation factors, whose activation can be blocked by brefeldin A (BFA), play a major role in functioning of the Golgi network and regulation of membrane traffic and are also involved in proliferation and migration of cancer cells. Due to high cytotoxicity and poor bioavailability, BFA has not passed the preclinical stage of drug development. Recently, AMF-26 and golgicide A have been described as novel inhibitors of the Golgi system with antitumor or bactericidal properties. We provide here further evidence that AMF-26 closely mirrors the mode of action of BFA but is less potent. Using several human cancer cell lines, we studied the effects of AMF-26, BFA and golgicide A on cell homeostasis including Golgi structure, endoplasmic reticulum (ER) stress markers, secretion and viability, and found overall a significant correlation between these parameters. Furthermore, modulation of ADP-ribosylation factor expression has a profound impact on Golgi organization and survival in response to Golgi stress inducers.


Cell Survival , Golgi Apparatus/metabolism , Stress, Physiological , ADP-Ribosylation Factors/drug effects , ADP-Ribosylation Factors/metabolism , Brefeldin A/pharmacology , Cell Line, Tumor , HEK293 Cells , Humans , Naphthols/pharmacology , Pyridines/pharmacology , Quinolines/pharmacology
11.
J Cell Sci ; 130(14): 2251-2265, 2017 Jul 15.
Article En | MEDLINE | ID: mdl-28536105

Tether complexes play important roles in endocytic and exocytic trafficking of lipids and proteins. In yeast, the multisubunit transport protein particle (TRAPP) tether regulates endoplasmic reticulum (ER)-to-Golgi and intra-Golgi transport and is also implicated in autophagy. In addition, the TRAPP complex acts as a guanine nucleotide exchange factor (GEF) for Ypt1, which is homologous to human Rab1a and Rab1b. Here, we show that human TRAPPC13 and other TRAPP subunits are critically involved in the survival response to several Golgi-disrupting agents. Loss of TRAPPC13 partially preserves the secretory pathway and viability in response to brefeldin A, in a manner that is dependent on ARF1 and the large GEF GBF1, and concomitant with reduced caspase activation and ER stress marker induction. TRAPPC13 depletion reduces Rab1a and Rab1b activity, impairs autophagy and leads to increased infectivity to the pathogenic bacterium Shigella flexneri in response to brefeldin A. Thus, our results lend support for the existence of a mammalian TRAPPIII complex containing TRAPPC13, which is important for autophagic flux under certain stress conditions.


Antigens, Neoplasm/metabolism , Golgi Apparatus/metabolism , Vesicular Transport Proteins/metabolism , A549 Cells , ADP-Ribosylation Factor 1/metabolism , Anti-Bacterial Agents/pharmacology , Antigens, Neoplasm/drug effects , Autophagy/physiology , Brefeldin A/pharmacology , Dysentery, Bacillary/drug therapy , Dysentery, Bacillary/metabolism , Gene Knockdown Techniques , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , HT29 Cells , HeLa Cells , Humans , Shigella flexneri/drug effects , Vesicular Transport Proteins/antagonists & inhibitors , Vesicular Transport Proteins/drug effects
12.
Cancer Discov ; 3(1): 82-95, 2013 Jan.
Article En | MEDLINE | ID: mdl-23242809

UNLABELLED: MYC deregulation is common in human cancer. IG-MYC translocations that are modeled in Eµ-Myc mice occur in almost all cases of Burkitt lymphoma as well as in other B-cell lymphoproliferative disorders. Deregulated expression of MYC results in increased mTOR complex 1 (mTORC1) signaling. As tumors with mTORC1 activation are sensitive to mTORC1 inhibition, we used everolimus, a potent and specific mTORC1 inhibitor, to test the requirement for mTORC1 in the initiation and maintenance of Eµ-Myc lymphoma. Everolimus selectively cleared premalignant B cells from the bone marrow and spleen, restored a normal pattern of B-cell differentiation, and strongly protected against lymphoma development. Established Eµ-Myc lymphoma also regressed after everolimus therapy. Therapeutic response correlated with a cellular senescence phenotype and induction of p53 activity. Therefore, mTORC1-dependent evasion of senescence is critical for cellular transformation and tumor maintenance by MYC in B lymphocytes. SIGNIFICANCE: This work provides novel insights into the requirements for MYC-induced oncogenesis by showing that mTORC1 activity is necessary to bypass senescence during transformation of B lymphocytes. Furthermore, tumor eradication through senescence elicited by targeted inhibition of mTORC1 identifies a previously uncharacterized mechanism responsible for significant anticancer activity of rapamycin analogues and serves as proof-of-concept that senescence can be harnessed for therapeutic benefit


Antineoplastic Agents/therapeutic use , Lymphoma/drug therapy , Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/metabolism , Sirolimus/analogs & derivatives , Animals , B-Lymphocytes/cytology , B-Lymphocytes/physiology , Cell Differentiation/drug effects , Cellular Senescence , Everolimus , Lymphoma/metabolism , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Transgenic , Multiprotein Complexes , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases
13.
Blood ; 114(2): 380-93, 2009 Jul 09.
Article En | MEDLINE | ID: mdl-19383971

LAQ824 and LBH589 (panobinostat) are histone deacetylase inhibitors (HDACi) developed as cancer therapeutics and we have used the Emu-myc lymphoma model to identify the molecular events required for their antitumor effects. Induction of tumor cell death was necessary for these agents to mediate therapeutic responses in vivo and both HDACi engaged the intrinsic apoptotic cascade that did not require p53. Death receptor pathway blockade had no effect on the therapeutic activities of LAQ824 and LBH589; however, overexpression of Bcl-2 or Bcl-X(L) protected lymphoma cells from HDACi-induced killing and suppressed their therapeutic activities. Deletion of Apaf-1 or Caspase-9 delayed HDACi-induced lymphoma killing in vitro and in vivo, associated with suppression of many biochemical indicators of apoptosis, but did not provide long-term resistance to these agents and failed to inhibit their therapeutic activities. Emu-myc lymphomas lacking a functional apoptosome displayed morphologic and biochemical features of autophagy after treatment with LAQ824 and LBH589, indicating that, in the absence of a complete intrinsic apoptosis pathway involving apoptosome formation, these HDACi can still mediate a therapeutic response. Our data indicate that damage to the mitochondria is the key event necessary for LAQ824 and LBH589 to mediate tumor cell death and a robust therapeutic response.


Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Lymphoma/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Histone Deacetylases/metabolism , Humans , Indoles , Lymphoma/pathology , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Neoplasm Transplantation , Panobinostat , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Death Domain/metabolism , Signal Transduction/drug effects , Survival Rate , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Suppressor Protein p53/metabolism
15.
Blood ; 113(9): 1982-91, 2009 Feb 26.
Article En | MEDLINE | ID: mdl-19060243

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


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biphenyl Compounds/pharmacology , Enzyme Inhibitors/administration & dosage , Histone Deacetylase Inhibitors , Lymphoma/drug therapy , Nitrophenols/pharmacology , Sulfonamides/pharmacology , Animals , Biphenyl Compounds/administration & dosage , Cell Survival/drug effects , Drug Delivery Systems , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Drug Synergism , Genes, bcl-2 , Genes, myc , Hydroxamic Acids/administration & dosage , Lymphoma/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nitrophenols/administration & dosage , Piperazines/administration & dosage , Piperazines/pharmacology , Substrate Specificity , Sulfonamides/administration & dosage , Vorinostat
16.
Proc Natl Acad Sci U S A ; 105(32): 11317-22, 2008 Aug 12.
Article En | MEDLINE | ID: mdl-18685088

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


Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Histone Deacetylase Inhibitors , Mammary Neoplasms, Experimental/drug therapy , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Animals , Antibodies, Monoclonal/pharmacology , CASP8 and FADD-Like Apoptosis Regulating Protein/biosynthesis , Cell Line, Tumor , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/pharmacology , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred BALB C , Mice, SCID , Neoplasm Proteins/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Vorinostat
17.
Mol Cancer Ther ; 7(5): 1066-79, 2008 May.
Article En | MEDLINE | ID: mdl-18483296

Histone deacetylase inhibitors (HDACi) are compounds that target the epigenome and cause tumor cell-selective apoptosis. A large number of these agents that have different chemical structures and can target multiple HDACs are being testing in clinical trials and vorinostat is now an approved drug for the treatment of cutaneous T-cell lymphoma. Although these agents are showing promise for the treatment of hematologic malignancies, it is possible that different drugs may have different mechanistic, biological, and therapeutic activities. When comparing an HDACi belonging to the hydroxamic acid class of compounds (vorinostat) with a cyclic tetrapeptide (romidepsin), we showed that these agents regulate the expression of a common set of cellular genes, but certain genes specifically responded to each agent. Using the Emu-myc mouse model of B-cell lymphoma, we showed previously that overexpression of the prosurvival proteins Bcl-2 and Bcl-XL inhibited the apoptotic and therapeutic activities of the vorinostat. Herein, we compared and contrasted the apoptotic-inducing activities of the hydroxamic acid oxamflatin with romidepsin. Like vorinostat, oxamflatin was unable to kill lymphomas overexpressing Bcl-2 and Bcl-XL, indicating that these proteins can generally protect cells against this class of HDACi. In contrast, romidepsin was able to induce apoptosis in lymphomas overexpressing Bcl-2 with delayed kinetics of cell death and could mediate therapeutic responses against these lymphomas. However, romidepsin was inactive when Bcl-XL was overexpressed. These data provide strong support that HDACi of different chemical classes may have subtle yet potentially important differences in their molecular and biological activities.


Antibiotics, Antineoplastic/pharmacology , Apoptosis , Depsipeptides/pharmacology , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Animals , Antibiotics, Antineoplastic/therapeutic use , Depsipeptides/therapeutic use , Dose-Response Relationship, Drug , Enzyme Inhibitors/therapeutic use , Hydroxamic Acids/pharmacology , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/enzymology , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-X Protein/metabolism
18.
Cancer Res ; 68(4): 961-4, 2008 Feb 15.
Article En | MEDLINE | ID: mdl-18281468

Hedgehog-mediated signaling has been shown to promote growth and dissemination of solid cancers, most prominently basal cell carcinomas and medulloblastoma. Recent findings indicate that hedgehog signals are also important for tumor growth in hematologic malignancies. Hedgehog ligands secreted by stromal cells could elicit Patched/Smoothened-mediated antiapoptotic signaling in mouse B-cell lymphomas. Inhibition of hedgehog signaling induced apoptosis in lymphoma cells and prolonged survival of lymphoma-bearing mice. Depletion of tumor cells proceeded in the absence of p53 via the mitochondrial apoptotic pathway. These and other recently published data on hedgehog inhibition in cancer cells and their implications will be discussed.


Hedgehog Proteins/metabolism , Lymphoma, B-Cell/metabolism , Animals , Humans , Lymphoma, B-Cell/pathology , Signal Transduction , Stromal Cells/metabolism , Stromal Cells/pathology
19.
Cell ; 129(2): 423-33, 2007 Apr 20.
Article En | MEDLINE | ID: mdl-17448999

Bid, a caspase-activated proapoptotic BH3-only protein, is essential for Fas-induced hepatocyte destruction. Recent studies published in Cell produced conflicting results, indicating that loss of Bid either protects or enhances apoptosis induced by DNA damage or replicative stress. To resolve this controversy, we generated novel Bid-deficient mice on an inbred C57BL/6 background and removed the drug-selection cassette from the targeted locus. Nine distinct cell types from these Bid-deficient mice underwent cell-cycle arrest and apoptosis in a manner indistinguishable from control WT cells in response to DNA damage or replicative stress. Moreover, we found that even cells from the original Bid-deficient mice responded normally to these stimuli, indicating that differences in genetic background or the presence of a strong promoter within the targeted locus are unlikely to explain the differences between our results and those reported previously. We conclude that Bid has no role in DNA damage- or replicative stress-induced apoptosis or cell-cycle arrest.


Apoptosis , BH3 Interacting Domain Death Agonist Protein/metabolism , Cell Cycle , DNA Damage , Animals , BH3 Interacting Domain Death Agonist Protein/genetics , Blood Cells/cytology , Cell Proliferation , Cytosol/chemistry , Female , Fibroblasts/cytology , Male , Mice , Mice, Inbred C57BL , Mitogens/metabolism
20.
Oncogene ; 24(4): 650-61, 2005 Jan 20.
Article En | MEDLINE | ID: mdl-15531915

PKCalpha and Ets1 are both associated with breast cancer progression. Our previous studies suggested that these proteins are likely to functionally interact with one another. Here, we show that attenuation of endogenous PKCalpha expression (siPalpha) by RNA interference leads to reduced Ets1 protein expression in a variety of cancer cells. Pulse-chase experiments and treatment with proteasome inhibitor MG-132 revealed that siPalpha interferes with both Ets1 protein synthesis and stability. The effect of siPalpha on Ets1 expression could be partially prevented by KN-93, suggesting that calcium/calmodulin-dependent kinase II (CaMKII), a modulator of Ets1 activity, may play a role in PKCalpha-dependent Ets1 regulation. In contrast, Ets1-regulating kinases ERK1/2 were not found to be involved in this process. To assess the importance of the PKCalpha/Ets1 interaction, we compared the biological responses of MDA-MB-231 cells to PKCalpha- and Ets1-specific siRNAs (siE1). While only siPalpha induced changes in cellular morphology and anchorage-independent growth, both siRNAs similarly affected cellular responses to the antitumor drug mithramycin A and to UV light. Microarray analyses further showed that the expression of a certain set of genes was equally affected by siPalpha and siE1. The data suggest that Ets1 serves as an effector for PKCalpha to fulfil certain functions in cancer cells.


Neoplasms/metabolism , Plicamycin/analogs & derivatives , Protein Kinase C/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line , Cell Shape , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/pathology , Oligonucleotide Array Sequence Analysis , Plicamycin/pharmacology , Protein Kinase C/genetics , Protein Kinase C-alpha , Proto-Oncogene Protein c-ets-1 , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ets , RNA Interference , Transcription Factors/biosynthesis , Transcription Factors/genetics , Ultraviolet Rays
...