Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Cell ; 146(6): 904-17, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21889194

ABSTRACT

MYC contributes to the pathogenesis of a majority of human cancers, yet strategies to modulate the function of the c-Myc oncoprotein do not exist. Toward this objective, we have targeted MYC transcription by interfering with chromatin-dependent signal transduction to RNA polymerase, specifically by inhibiting the acetyl-lysine recognition domains (bromodomains) of putative coactivator proteins implicated in transcriptional initiation and elongation. Using a selective small-molecule bromodomain inhibitor, JQ1, we identify BET bromodomain proteins as regulatory factors for c-Myc. BET inhibition by JQ1 downregulates MYC transcription, followed by genome-wide downregulation of Myc-dependent target genes. In experimental models of multiple myeloma, a Myc-dependent hematologic malignancy, JQ1 produces a potent antiproliferative effect associated with cell-cycle arrest and cellular senescence. Efficacy of JQ1 in three murine models of multiple myeloma establishes the therapeutic rationale for BET bromodomain inhibition in this disease and other malignancies characterized by pathologic activation of c-Myc.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Multiple Myeloma/drug therapy , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Azepines/chemistry , Azepines/pharmacology , Benzodiazepines/chemistry , Benzodiazepines/pharmacology , Cell Line, Tumor , Disease Models, Animal , Humans , Mice , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins c-myc/genetics , Transcriptional Activation/drug effects , Triazoles/chemistry , Triazoles/pharmacology
2.
Blood ; 119(15): e131-8, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-22289890

ABSTRACT

Conventional assays evaluating antitumor activity of immune effector cells have limitations that preclude their high-throughput application. We adapted the recently developed Compartment-Specific Bioluminescence Imaging (CS-BLI) technique to perform high-throughput quantification of innate antitumor activity and to show how pharmacologic agents (eg, lenalidomide, pomalidomide, bortezomib, and dexamethasone) and autologous BM stromal cells modulate that activity. CS-BLI-based screening allowed us to identify agents that enhance or inhibit innate antitumor cytotoxicity. Specifically, we identified compounds that stimulate immune effector cells against some tumor targets but suppressed their activity against other tumor cells. CS-BLI offers rapid, simplified, and specific evaluation of multiple conditions, including drug treatments and/or cocultures with stromal cells and highlights that immunomodulatory pharmacologic responses can be heterogeneous across different types of tumor cells. This study provides a framework to identify novel immunomodulatory agents and to prioritize compounds for clinical development on the basis of their effect on antitumor immunity.


Subject(s)
High-Throughput Screening Assays/methods , Immunity, Innate/physiology , Luminescent Measurements/methods , Neoplasms/diagnosis , Neoplasms/immunology , Animals , CD56 Antigen/metabolism , Cell Compartmentation/genetics , Cell Compartmentation/immunology , Cell Compartmentation/physiology , Cell Survival , Diagnostic Imaging/instrumentation , Diagnostic Imaging/methods , High-Throughput Screening Assays/instrumentation , Humans , Immunotherapy/methods , K562 Cells , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Luminescent Measurements/instrumentation , Neoplasms/pathology , Neoplasms/therapy , Substrate Specificity/immunology , Tumor Cells, Cultured
3.
Blood ; 117(17): 4409-19, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21321360

ABSTRACT

Recurrence of multiple myeloma (MM) after therapy suggests the presence of tumor-initiating subpopulations. In our study, we performed flow cytometry-based Hoechst 33342 staining to evaluate the existence of a MM population with stem-like features known as side population (SP) cells. SP cells exhibit substantial heterogeneity in MM cell lines and primary MM cells; express CD138 antigen in MM cell lines; display higher mRNA expression and functional activity of ABCG2 transporter; and have a higher proliferation index compared with non-SP cells. We observed evidence for clonogenic potential of SP cells, as well as the ability of SP cells to regenerate original population. Moreover, SP cells revealed higher tumorigenicity compared with non-SP cells. Importantly, lenalidomide decreased the percentage and clonogenicity of SP cells, and also induced phosphorylation changes in Akt, GSK-3α/ß, MEK1, c-Jun, p53, and p70S6K in SP cells. Adherence to bone marrow stromal cells (BMSCs) increased the percentage, viability, and proliferation potential of SP cells. Lenalidomide and thalidomide abrogated this stimulatory effect of BMSCs and significantly decreased the percentage of SP cells. Our studies demonstrate a novel mechanism of action for lenalidomide, namely targeting SP fraction, providing the framework for new therapeutic strategies targeting subpopulations of MM cells including presumptive stem cells.


Subject(s)
Antineoplastic Agents/pharmacology , Multiple Myeloma , Neoplastic Stem Cells/drug effects , Thalidomide/analogs & derivatives , ATP-Binding Cassette Transporters/genetics , Angiogenesis Inhibitors/pharmacology , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/physiology , Cell Division/drug effects , Cell Fractionation , Cell Line, Tumor , Cell Survival/drug effects , Colony-Forming Units Assay , Drug Resistance, Neoplasm , Humans , Lenalidomide , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Multiple Myeloma/physiopathology , Neoplasm Recurrence, Local/prevention & control , Neoplastic Stem Cells/pathology , Signal Transduction/drug effects , Stromal Cells/cytology , Stromal Cells/drug effects , Stromal Cells/physiology , Syndecan-1/metabolism , Thalidomide/pharmacology
4.
Nat Commun ; 14(1): 4812, 2023 08 09.
Article in English | MEDLINE | ID: mdl-37558654

ABSTRACT

Branched chain amino acid (BCAA) catabolic impairments have been implicated in several diseases. Branched chain ketoacid dehydrogenase (BCKDH) controls the rate limiting step in BCAA degradation, the activity of which is inhibited by BCKDH kinase (BDK)-mediated phosphorylation. Screening efforts to discover BDK inhibitors led to identification of thiophene PF-07208254, which improved cardiometabolic endpoints in mice. Structure-activity relationship studies led to identification of a thiazole series of BDK inhibitors; however, these inhibitors did not improve metabolism in mice upon chronic administration. While the thiophenes demonstrated sustained branched chain ketoacid (BCKA) lowering and reduced BDK protein levels, the thiazoles increased BCKAs and BDK protein levels. Thiazoles increased BDK proximity to BCKDH-E2, whereas thiophenes reduced BDK proximity to BCKDH-E2, which may promote BDK degradation. Thus, we describe two BDK inhibitor series that possess differing attributes regarding BDK degradation or stabilization and provide a mechanistic understanding of the desirable features of an effective BDK inhibitor.


Subject(s)
Amino Acids, Branched-Chain , Thiophenes , Mice , Animals , Amino Acids, Branched-Chain/metabolism , Phosphorylation , Thiophenes/pharmacology , Oxidoreductases/metabolism
5.
Br J Haematol ; 159(3): 340-51, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22970818

ABSTRACT

Jasmonates, plant stress hormones, have been demonstrated to be effective in killing various types of cancer cells. We therefore tested if methyljasmonate (MJ) has activity against multiple myeloma (MM) in vitro and in vivo. MM cell lines and primary MM tumour cells responded to MJ in vitro at concentrations that did not significantly affect normal haematopoietic cells, without stroma-mediated resistance. Brief MJ exposures of MM cells caused release of Hexokinase 2 (HK2) from mitochondria, rapid ATP depletion, perturbation of major intracellular signalling pathways, and ensuing mainly apoptotic cell death. Sensitivity to MJ correlated with lower cellular glucose consumption and lactate production, as well as lower intracellular protein levels of HK2, phosphorylated Voltage-dependent anion channel 2/3 (pVDAC2/3) and Aldo-keto reductase family 1 member C1 (AKR1C1), which represent potential biomarkers of responsiveness to MJ treatment, especially as AKR1C1 transcript levels also correlate with clinical outcome in bortezomib- or dexamethasone-treated MM patients. Interestingly, MJ synergized with bortezomib in vitro and prolonged survival of immunocompromised mice harbouring diffuse lesions of MM.1S cells compared to vehicle-treated mice (P = 0·0046). These studies indicate that jasmonates represent a new, promising strategy to treat MM.


Subject(s)
Acetates/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Cyclopentanes/pharmacology , Multiple Myeloma/metabolism , Oxylipins/pharmacology , Plant Growth Regulators/pharmacology , Acetates/administration & dosage , Adenosine Triphosphate/metabolism , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cyclopentanes/administration & dosage , Drug Synergism , Humans , Leukocytes, Mononuclear/drug effects , Mice , Mice, Inbred NOD , Mice, SCID , Mitochondria/drug effects , Mitochondria/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/mortality , Oxylipins/administration & dosage
6.
Blood ; 116(4): 593-602, 2010 Jul 29.
Article in English | MEDLINE | ID: mdl-20382846

ABSTRACT

Using a combination high-throughput screening technology, multiple classes of drugs and targeted agents were identified that synergize with dexamethasone (Dex) in multiple myeloma (MM) cells. Performing combination screening with these enhancers, we discovered an unexpected synergistic interaction between adenosine receptor agonists and phosphodiesterase (PDE) inhibitors that displays substantial activity in a panel of MM and diffuse large B-cell lymphoma (DLBCL) cell lines and tumor cells from MM patients. We have used selective adenosine receptor agonists, antagonists, and PDE inhibitors as well as small interfering RNAs targeting specific molecular isoforms of these proteins to dissect the molecular mechanism of this synergy. The adenosine A2A receptor and PDE2, 3, 4, and 7 are important for activity. Drug combinations induce cyclic AMP (cAMP) accumulation and up-regulate PDE4B. We also observe rigorous mathematical synergy in 3-way combinations containing A2A agonists, PDE inhibitors, and Dex at multiple concentrations and ratios. Taken together, these data suggest that A2A agonist/PDE inhibitor combinations may be attractive as an adjunctive to clinical glucocorticoid containing regiments for patients with MM or DLBCL and confer benefit in both glucocorticoid-sensitive and -resistant populations.


Subject(s)
Adenosine A2 Receptor Agonists , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Drug Screening Assays, Antitumor/methods , Hematologic Neoplasms/drug therapy , Phosphodiesterase Inhibitors/administration & dosage , B-Lymphocytes/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Dexamethasone/administration & dosage , Dexamethasone/pharmacology , Drug Delivery Systems , Drug Synergism , Glucocorticoids/administration & dosage , Glucocorticoids/pharmacology , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , High-Throughput Screening Assays/methods , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/pathology , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Phosphodiesterase Inhibitors/isolation & purification , Phosphodiesterase Inhibitors/pharmacology , Validation Studies as Topic
7.
J Nat Prod ; 75(4): 793-7, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22458669

ABSTRACT

CR1642D, an endophytic isolate of Penicillium sp. collected from a Costa Rican rainforest, was identified through a high-throughput approach to identify natural products with enhanced antitumor activity in the context of tumor-stromal interactions. Bioassay-guided separation led to the identification of five xanthones (1-5) from CR1642D. The structures of the xanthone dimer penexanthone A (1) and monomer penexanthone B (2) were elucidated on the basis of spectroscopic analyses, including 2D NMR experiments. All of the compounds were tested against a panel of tumor cell lines in the presence and absence of bone marrow stromal cells. Compound 3 was the most active, with IC(50) values of 1-17 µM, and its activity was enhanced 2-fold against tumor cell line RPMI8226 in the presence of stromal cells (IC(50) 1.2 µM, but 2.4 µM without stromal cells).


Subject(s)
Penicillium/chemistry , Xanthones/isolation & purification , Xanthones/pharmacology , Costa Rica , Humans , Molecular Sequence Data , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Stromal Cells/drug effects , Xanthones/chemistry
8.
Heliyon ; 8(10): e11091, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36303906

ABSTRACT

The AMP-activated protein kinase (AMPK) is a cellular sensor of energetics and when activated in skeletal muscle during contraction can impart changes in skeletal muscle metabolism. Therapeutics that selectively activate AMPK have been developed to lower glucose levels through increased glucose disposal rates as an approach to abrogate the hyperglycemic state of diabetes; however, the metabolic fate of glucose following AMPK activation remains unclear. We have used a combination of in vivo evaluation of glucose homeostasis and ex vivo skeletal muscle incubation to systematically evaluate metabolism following pharmacological activation of AMPK with PF-739, comparing this with AMPK activation through sustained intermittent electrical stimulation of contraction. These methods to activate AMPK result in increased glucose uptake but divergent metabolism of glucose: pharmacological activation results in increased glycogen accumulation while contraction-induced glucose uptake results in increased lactate formation and glucose oxidation. These results provide additional evidence to support a role for AMPK in control of skeletal muscle metabolism and additional insight into the potential for AMPK stimulation with small molecule direct activators.

9.
Br J Haematol ; 152(4): 420-32, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21223249

ABSTRACT

Cell cycle regulators, such as cyclin-dependent kinases (CDKs), are appealing targets for multiple myeloma (MM) therapy given the increased proliferative rates of tumour cells in advanced versus early stages of MM. We hypothesized that a multi-targeted CDK inhibitor with a different spectrum of activity compared to existing CDK inhibitors could trigger distinct molecular sequelae with therapeutic implications for MM. We therefore studied the small molecule heterocyclic compound NVP-LCQ195/AT9311 (LCQ195), which inhibits CDK1, CDK2 and CDK5, as well as CDK3 and CDK9. LCQ195 induced cell cycle arrest and eventual apoptotic cell death of MM cells, even at sub-µmol/l concentrations, spared non-malignant cells, and overcame the protection conferred to MM cells by stroma or cytokines of the bone marrow milieu. In MM cells, LCQ195 triggered decreased amplitude of transcriptional signatures associated with oncogenesis, drug resistance and stem cell renewal, including signatures of activation of key transcription factors for MM cells e.g. myc, HIF-1α, IRF4. Bortezomib-treated MM patients whose tumours had high baseline expression of genes suppressed by LCQ195 had significantly shorter progression-free and overall survival than those with low levels of these transcripts in their MM cells. These observations provide insight into the biological relevance of multi-targeted CDK inhibition in MM.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinases/antagonists & inhibitors , Multiple Myeloma/pathology , Apoptosis/drug effects , Boronic Acids/therapeutic use , Bortezomib , Cell Cycle/drug effects , Cell Survival/drug effects , Coculture Techniques , Cyclin-Dependent Kinases/metabolism , Cytokines/antagonists & inhibitors , Cytokines/pharmacology , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Drug Combinations , Drug Interactions , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/enzymology , Multiple Myeloma/genetics , Pyrazines/therapeutic use , Signal Transduction/drug effects , Stromal Cells/physiology , Survival Analysis , Transcription, Genetic , Treatment Outcome , Tumor Cells, Cultured
10.
Haematologica ; 96(8): 1170-9, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21712538

ABSTRACT

BACKGROUND: Isothiocyanates, a family of phytochemicals found in cruciferous vegetables, have cytotoxic effects against several types of tumor cells. Multiple myeloma is a fatal disease characterized by clonal proliferation of plasma cells in the bone marrow. The growing body of preclinical information on the anti-cancer activity of isothiocyanates led us to investigate their anti-myeloma properties. DESIGN AND METHODS: We evaluated the anti-myeloma activity of the isothiocyanates, sulforaphane and phenethyl isothiocyanate, on a panel of human myeloma cell lines as well as primary myeloma tumor cells. Cell viability, apoptosis, cell cycle alterations and cell proliferation were then analyzed in vitro and in a xenograft mouse model in vivo. The molecular sequelae of isothiocyanate treatment in multiple myeloma cells were evaluated by multiplex analyses using bead arrays and western blotting. RESULTS: We observed that sulforaphane and phenylethyl isothiocyanate have activity against myeloma cell lines and patients' myeloma cells both in vitro and in vivo using a myeloma xenograft mouse model. Isothiocyanates induced apoptotic death of myeloma cells; depletion of mitochondrial membrane potential; cleavage of PARP and caspases-3 and -9; as well as down-regulation of anti-apoptotic proteins including Mcl-1, X-IAP, c-IAP and survivin. Isothiocyanates induced G(2)/M cell cycle arrest accompanied by mitotic phosphorylation of histone H3. Multiplex analysis of phosphorylation of diverse components of signaling cascades revealed changes in MAPK activation; increased phosphorylation of c-jun and HSP27; as well as changes in the phosphorylation of Akt, and GSK3α/ß and p53. Isothiocyanates suppressed proliferation of myeloma cells alone and when co-cultured with HS-5 stromal cells. Sulforaphane and phenylethyl isothiocyanate enhanced the in vitro anti-myeloma activity of several conventional and novel therapies used in multiple myeloma. CONCLUSIONS: Our study shows that isothiocyanates have potent anti-myeloma activities and may enhance the activity of other anti-multiple myeloma agents. These results indicate that isothiocyanates may have therapeutic potential in multiple myeloma and provide the preclinical framework for future clinical studies of isothiocyanates in multiple myeloma.


Subject(s)
Antineoplastic Agents/pharmacology , Isothiocyanates/pharmacology , Multiple Myeloma/drug therapy , Signal Transduction/drug effects , Thiocyanates/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Isothiocyanates/therapeutic use , Isothiocyanates/toxicity , Mice , Mice, SCID , Multiple Myeloma/metabolism , Stromal Cells/drug effects , Sulfoxides , Thiocyanates/therapeutic use , Thiocyanates/toxicity , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
11.
Br J Haematol ; 147(5): 672-6, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19751238

ABSTRACT

This study characterized the preclinical anti-myeloma activity of VE465, a low molecular weight pan-Aurora kinase inhibitor. After 96-h drug exposure, several multiple myeloma (MM) cell lines were more sensitive to VE465 compared to non-malignant cells. The anti-MM activity of VE465 was maintained in the presence of interleukin-6 and, interestingly, enhanced by co-culture with stromal cells. However, primary MM cells were less responsive than cell lines. Combinations with dexamethasone (Dex), doxorubicin (Doxo) and bortezomib showed no antagonism. Our study highlights the potential role of the tumour microenvironment in modulating the activity of this drug class.


Subject(s)
Antineoplastic Agents/pharmacology , Multiple Myeloma/pathology , Piperazines/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Aurora Kinases , Cell Communication/drug effects , Cell Cycle/drug effects , Coculture Techniques , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor/methods , Hematopoietic Stem Cells/cytology , Humans , Tumor Cells, Cultured
13.
J Med Chem ; 61(16): 7273-7288, 2018 08 23.
Article in English | MEDLINE | ID: mdl-30036059

ABSTRACT

Studies on indole-3-carboxylic acid derivatives as direct activators of human adenosine monophosphate-activated protein kinase (AMPK) α1ß1γ1 isoform have culminated in the identification of PF-06409577 (1), PF-06885249 (2), and PF-06679142 (3) as potential clinical candidates. Compounds 1-3 are primarily cleared in animals and humans via glucuronidation. Herein, we describe the biosynthetic preparation, purification, and structural characterization of the glucuronide conjugates of 1-3. Spectral characterization of the purified glucuronides M1, M2, and M3 indicated that they were acyl glucuronide derivatives. In vitro pharmacological evaluation revealed that all three acyl glucuronides retained selective activation of ß1-containing AMPK isoforms. Inhibition of de novo lipogenesis with representative parent carboxylic acids and their respective acyl glucuronide conjugates in human hepatocytes demonstrated their propensity to activate cellular AMPK. Cocrystallization of the AMPK α1ß1γ1 isoform with 1-3 and M1-M3 provided molecular insights into the structural basis for AMPK activation by the glucuronide conjugates.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Indoles/chemistry , Indoles/metabolism , Lipogenesis/drug effects , AMP-Activated Protein Kinases/chemistry , Animals , Cells, Cultured , Crystallization/methods , Enzyme Activation/drug effects , Glucuronides/chemistry , Glucuronides/metabolism , Glucuronides/pharmacokinetics , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Indoles/pharmacology , Macaca fascicularis , Magnetic Resonance Spectroscopy , Male , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Rats, Wistar , Uridine Diphosphate Glucuronic Acid/pharmacology
14.
EBioMedicine ; 31: 122-132, 2018 May.
Article in English | MEDLINE | ID: mdl-29673898

ABSTRACT

Dysregulation of hepatic lipid and cholesterol metabolism is a significant contributor to cardiometabolic health, resulting in excessive liver lipid accumulation and ultimately non-alcoholic steatohepatitis (NASH). Therapeutic activators of the AMP-Activated Protein Kinase (AMPK) have been proposed as a treatment for metabolic diseases; we show that the AMPK ß1-biased activator PF-06409577 is capable of lowering hepatic and systemic lipid and cholesterol levels in both rodent and monkey preclinical models. PF-06409577 is able to inhibit de novo lipid and cholesterol synthesis pathways, and causes a reduction in hepatic lipids and mRNA expression of markers of hepatic fibrosis. These effects require AMPK activity in the hepatocytes. Treatment of hyperlipidemic rats or cynomolgus monkeys with PF-06409577 for 6weeks resulted in a reduction in circulating cholesterol. Together these data suggest that activation of AMPK ß1 complexes with PF-06409577 is capable of impacting multiple facets of liver disease and represents a promising strategy for the treatment of NAFLD and NASH in humans.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Enzyme Activators/pharmacology , Hepatocytes/enzymology , Indoles/pharmacology , Liver/enzymology , Non-alcoholic Fatty Liver Disease , Animals , Cell Line , Haplorhini , Hepatocytes/pathology , Humans , Liver/pathology , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/enzymology , Non-alcoholic Fatty Liver Disease/pathology , Rats
15.
J Clin Endocrinol Metab ; 92(12): 4845-52, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17848408

ABSTRACT

CONTEXT: The Bcl-2 family of proteins regulates apoptosis in various models and may represent a promising therapeutic target in human malignancies. OBJECTIVE/METHODS: We evaluated the sensitivity of thyroid carcinoma cell lines (two papillary, one follicular, two anaplastic, three medullary) in vitro to BH3I-1 and BH3I-2', two cell-permeable inhibitors of the Bcl-2 homology (BH)-3 domain-mediated interaction between proapoptotic and antiapoptotic Bcl-2 family members. The thyroid carcinoma cell line FRO was stably transfected with cDNA for Bcl-2 or constitutively active Akt and evaluated for sensitivity to BH3-domain inhibition. RESULTS: BH3-domain inhibition disrupted the mitochondrial membrane potential in thyroid carcinoma cells, induced caspase-dependent apoptosis, and potently sensitized them to sublethal concentrations of doxorubicin and the proteasome inhibitor bortezomib (Velcade). Overexpression of constitutively active Akt suppressed BH3I-1-induced cell death. Bcl-2-overexpressing FRO cells were more resistant to conventional chemotherapeutic agents (such as doxorubicin) but significantly more sensitive to BH3I-1 than control cells and were found to overexpress caspase-9, caspase-8, Bmf, Bok, and Bik transcripts and express less A1, BRaf, and FLIP transcripts. CONCLUSIONS: Bcl-2 expression protects thyroid carcinomas against chemotherapy-induced apoptosis. Nevertheless, overexpression of Bcl-2 may result in "oncogene addiction" of the cancer cell, which can be exploited by using BH3-domain inhibitors alone or in combination with other agents, including conventional chemotherapeutics (such as doxorubicin) or novel targeted therapies (such as the proteasome inhibitor bortezomib), for the treatment of aggressive thyroid cancer, including the medullary and anaplastic types.


Subject(s)
Carcinoma, Medullary/genetics , Carcinoma/genetics , Genes, bcl-2/genetics , Thyroid Neoplasms/genetics , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents/pharmacology , BH3 Interacting Domain Death Agonist Protein/genetics , Blotting, Western , Boronic Acids/pharmacology , Bortezomib , Cell Line, Tumor , Colorimetry , Doxorubicin/pharmacology , Humans , Membrane Potentials/physiology , Oncogene Protein v-akt/genetics , Protease Inhibitors/pharmacology , Pyrazines/pharmacology , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Tetrazolium Salts , Thiazoles , Transcription, Genetic
16.
Cell Metab ; 25(5): 1147-1159.e10, 2017 May 02.
Article in English | MEDLINE | ID: mdl-28467931

ABSTRACT

The AMP-activated protein kinase (AMPK) is a potential therapeutic target for metabolic diseases based on its reported actions in the liver and skeletal muscle. We evaluated two distinct direct activators of AMPK: a non-selective activator of all AMPK complexes, PF-739, and an activator selective for AMPK ß1-containing complexes, PF-249. In cells and animals, both compounds were effective at activating AMPK in hepatocytes, but only PF-739 was capable of activating AMPK in skeletal muscle. In diabetic mice, PF-739, but not PF-249, caused a rapid lowering of plasma glucose levels that was diminished in the absence of skeletal muscle, but not liver, AMPK heterotrimers and was the result of an increase in systemic glucose disposal with no impact on hepatic glucose production. Studies of PF-739 in cynomolgus monkeys confirmed translation of the glucose lowering and established activation of AMPK in skeletal muscle as a potential therapeutic approach to treat diabetic patients.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Aminopyridines/pharmacology , Enzyme Activators/pharmacology , Glucose/metabolism , Hypoglycemic Agents/pharmacology , Indoles/pharmacology , Aminopyridines/therapeutic use , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Enzyme Activation/drug effects , Enzyme Activators/therapeutic use , Female , Hypoglycemic Agents/therapeutic use , Indoles/therapeutic use , Liver/drug effects , Liver/metabolism , Macaca fascicularis , Male , Mice, Inbred C57BL , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism
17.
Cancer Chemother Pharmacol ; 71(5): 1357-68, 2013 May.
Article in English | MEDLINE | ID: mdl-23589314

ABSTRACT

PURPOSE: Bortezomib is an important agent in multiple myeloma treatment, but resistance in cell lines and patients has been described. The main mechanisms of resistance described in cancer fall into one of two categories, pharmacokinetic resistance (PK), e.g. over expression of drug efflux pumps and pharmacodynamic resistance, e.g. apoptosis resistance or altered survival pathways, where the agent reaches an appropriate concentration, but this fails to propagate an appropriate cell death response. Of the known pump mechanisms, P-glycoprotein (P-gp) is the best studied and considered to be the most important in contributing to general PK drug resistance. Resistance to bortezomib is multifactorial and there are conflicting indications that cellular overexpression of P-gp may contribute to resistance agent. Hence, better characterization of the interactions of this drug with classical resistance mechanisms should identify improved treatment applications. METHODS: Cell lines with different P-gp expression levels were used to determine the relationship between bortezomib and P-gp. Coculture system with stromal cells was used to determine the effect of the local microenvironment on the bortezomib-elacridar combination. To further assess P-gp function, intracellular accumulation of P-gp probe rhodamine-123 was utilised. RESULTS: In the present study, we show that bortezomib is a substrate for P-gp, but not for the other drug efflux transporters. Bortezomib activity is affected by P-gp expression and conversely, the expression of P-gp affect bortezomib's ability to act as a P-gp substrate. The local microenvironment did not alter the cellular response to bortezomib. We also demonstrate that bortezomib directly affects the expression and function of P-gp. CONCLUSIONS: Our findings strongly support a role for P-gp in bortezomib resistance and, therefore, suggest that combination of a P-gp inhibitor and bortezomib in P-gp positive myeloma would be a reasonable treatment combination to extend efficacy of this important drug.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Drug Resistance, Neoplasm , Multiple Myeloma/drug therapy , Pyrazines/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Biological Transport , Bortezomib , Cell Line , Cell Line, Tumor , Cellular Microenvironment , Coculture Techniques , Fluorescent Dyes/pharmacokinetics , Gene Expression Regulation/drug effects , Humans , Multiple Myeloma/pathology , Rhodamine 123/pharmacokinetics , Stromal Cells/metabolism
18.
Mol Cancer Ther ; 11(4): 942-51, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22246439

ABSTRACT

The NEDD8-activating enzyme is upstream of the 20S proteasome in the ubiquitin/proteasome pathway and catalyzes the first step in the neddylation pathway. NEDD8 modification of cullins is required for ubiquitination of cullin-ring ligases that regulate degradation of a distinct subset of proteins. The more targeted impact of NEDD8-activating enzyme on protein degradation prompted us to study MLN4924, an investigational NEDD8-activating enzyme inhibitor, in preclinical multiple myeloma models. In vitro treatment with MLN4924 led to dose-dependent decrease of viability (EC(50) = 25-150 nmol/L) in a panel of human multiple myeloma cell lines. MLN4924 was similarly active against a bortezomib-resistant ANBL-6 subline and its bortezomib-sensitive parental cells. MLN4924 had submicromolar activity (EC(50) values <500 nmol/L) against primary CD138(+) multiple myeloma patient cells and exhibited at least additive effect when combined with dexamethasone, doxorubicin, and bortezomib against MM.1S cells. The bortezomib-induced compensatory upregulation of transcripts for ubiquitin/proteasome was not observed with MLN4924 treatment, suggesting distinct functional roles of NEDD8-activating enzyme versus 20S proteasome. MLN4924 was well tolerated at doses up to 60 mg/kg 2× daily and significantly reduced tumor burden in both a subcutaneous and an orthotopic mouse model of multiple myeloma. These studies provide the framework for the clinical investigation of MLN4924 in multiple myeloma.


Subject(s)
Multiple Myeloma/drug therapy , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Ubiquitins/antagonists & inhibitors , Animals , Boronic Acids/pharmacology , Bortezomib , Cell Line, Tumor , Gene Expression Profiling , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Multiple Myeloma/enzymology , Multiple Myeloma/genetics , NEDD8 Protein , Pyrazines/pharmacology , Treatment Outcome , Ubiquitin-Activating Enzymes/metabolism , Ubiquitination , Ubiquitins/metabolism , Xenograft Model Antitumor Assays
19.
PLoS One ; 6(7): e20226, 2011.
Article in English | MEDLINE | ID: mdl-21750699

ABSTRACT

Polo-like kinases (PLKs) play an important role in cell cycle progression, checkpoint control and mitosis. The high mitotic index and chromosomal instability of advanced cancers suggest that PLK inhibitors may be an attractive therapeutic option for presently incurable advanced neoplasias with systemic involvement, such as multiple myeloma (MM). We studied the PLK 1, 2, 3 inhibitor BI 2536 and observed potent (IC50<40 nM) and rapid (commitment to cell death <24 hrs) in vitro activity against MM cells in isolation, as well as in vivo activity against a traditional subcutaneous xenograft mouse model. Tumor cells in MM patients, however, don't exist in isolation, but reside in and interact with the bone microenvironment. Therefore conventional in vitro and in vivo preclinical assays don't take into account how interactions between MM cells and the bone microenvironment can potentially confer drug resistance. To probe this question, we performed tumor cell compartment-specific bioluminescence imaging assays to compare the preclinical anti-MM activity of BI 2536 in vitro in the presence vs. absence of stromal cells or osteoclasts. We observed that the presence of these bone marrow non-malignant cells led to decreased anti-MM activity of BI 2536. We further validated these results in an orthotopic in vivo mouse model of diffuse MM bone lesions where tumor cells interact with non-malignant cells of the bone microenvironment. We again observed that BI 2536 had decreased activity in this in vivo model of tumor-bone microenvironment interactions highlighting that, despite BI 2536's promising activity in conventional assays, its lack of activity in microenvironmental models raises concerns for its clinical development for MM. More broadly, preclinical drug testing in the absence of relevant tumor microenvironment interactions may overestimate potential clinical activity, thus explaining at least in part the gap between preclinical vs. clinical efficacy in MM and other cancers.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Multiple Myeloma/drug therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Pteridines/pharmacology , Xenograft Model Antitumor Assays , Blotting, Western , Bone and Bones/drug effects , Bone and Bones/pathology , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Coculture Techniques , Dose-Response Relationship, Drug , Humans , Inhibitory Concentration 50 , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Osteoclasts/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Stromal Cells/drug effects , Time Factors , Tumor Burden/drug effects , Tumor Microenvironment/drug effects , Polo-Like Kinase 1
20.
Nat Med ; 16(4): 483-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20228816

ABSTRACT

Conventional anticancer drug screening is typically performed in the absence of accessory cells of the tumor microenvironment, which can profoundly alter antitumor drug activity. To address this limitation, we developed the tumor cell-specific in vitro bioluminescence imaging (CS-BLI) assay. Tumor cells (for example, myeloma, leukemia and solid tumors) stably expressing luciferase are cultured with nonmalignant accessory cells (for example, stromal cells) for selective quantification of tumor cell viability, in presence versus absence of stromal cells or drug treatment. CS-BLI is high-throughput scalable and identifies stroma-induced chemoresistance in diverse malignancies, including imatinib resistance in leukemic cells. A stroma-induced signature in tumor cells correlates with adverse clinical prognosis and includes signatures for activated Akt, Ras, NF-kappaB, HIF-1alpha, myc, hTERT and IRF4; for biological aggressiveness; and for self-renewal. Unlike conventional screening, CS-BLI can also identify agents with increased activity against tumor cells interacting with stroma. One such compound, reversine, shows more potent activity in an orthotopic model of diffuse myeloma bone lesions than in conventional subcutaneous xenografts. Use of CS-BLI, therefore, enables refined screening of candidate anticancer agents to enrich preclinical pipelines with potential therapeutics that overcome stroma-mediated drug resistance and can act in a synthetic lethal manner in the context of tumor-stroma interactions.


Subject(s)
Drug Screening Assays, Antitumor/methods , Luminescent Measurements/methods , Stromal Cells/metabolism , Tumor Cells, Cultured/drug effects , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Humans , Morpholines/pharmacology , Purines/pharmacology , Stromal Cells/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL