Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
2.
Nat Commun ; 14(1): 4812, 2023 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-37558654

RESUMEN

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.


Asunto(s)
Aminoácidos de Cadena Ramificada , Tiofenos , Ratones , Animales , Aminoácidos de Cadena Ramificada/metabolismo , Fosforilación , Tiofenos/farmacología , Oxidorreductasas/metabolismo
3.
Heliyon ; 8(10): e11091, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36303906

RESUMEN

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.

4.
J Med Chem ; 61(16): 7273-7288, 2018 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-30036059

RESUMEN

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.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Indoles/química , Indoles/metabolismo , Lipogénesis/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/química , Animales , Células Cultivadas , Cristalización/métodos , Activación Enzimática/efectos de los fármacos , Glucurónidos/química , Glucurónidos/metabolismo , Glucurónidos/farmacocinética , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Indoles/farmacología , Macaca fascicularis , Espectroscopía de Resonancia Magnética , Masculino , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Ratas Wistar , Uridina Difosfato Ácido Glucurónico/farmacología
5.
EBioMedicine ; 31: 122-132, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29673898

RESUMEN

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.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Activadores de Enzimas/farmacología , Hepatocitos/enzimología , Indoles/farmacología , Hígado/enzimología , Enfermedad del Hígado Graso no Alcohólico , Animales , Línea Celular , Haplorrinos , Hepatocitos/patología , Humanos , Hígado/patología , Ratones , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/enzimología , Enfermedad del Hígado Graso no Alcohólico/patología , Ratas
6.
Cell Metab ; 25(5): 1147-1159.e10, 2017 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-28467931

RESUMEN

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.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Aminopiridinas/farmacología , Activadores de Enzimas/farmacología , Glucosa/metabolismo , Hipoglucemiantes/farmacología , Indoles/farmacología , Aminopiridinas/uso terapéutico , Animales , Glucemia/metabolismo , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Activación Enzimática/efectos de los fármacos , Activadores de Enzimas/uso terapéutico , Femenino , Hipoglucemiantes/uso terapéutico , Indoles/uso terapéutico , Hígado/efectos de los fármacos , Hígado/metabolismo , Macaca fascicularis , Masculino , Ratones Endogámicos C57BL , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo
7.
Cancer Chemother Pharmacol ; 71(5): 1357-68, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23589314

RESUMEN

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.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Antineoplásicos/farmacología , Ácidos Borónicos/farmacología , Resistencia a Antineoplásicos , Mieloma Múltiple/tratamiento farmacológico , Pirazinas/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/efectos de los fármacos , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Transporte Biológico , Bortezomib , Línea Celular , Línea Celular Tumoral , Microambiente Celular , Técnicas de Cocultivo , Colorantes Fluorescentes/farmacocinética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Mieloma Múltiple/patología , Rodamina 123/farmacocinética , Células del Estroma/metabolismo
8.
Br J Haematol ; 159(3): 340-51, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22970818

RESUMEN

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.


Asunto(s)
Acetatos/farmacología , Antineoplásicos Fitogénicos/farmacología , Ciclopentanos/farmacología , Mieloma Múltiple/metabolismo , Oxilipinas/farmacología , Reguladores del Crecimiento de las Plantas/farmacología , Acetatos/administración & dosificación , Adenosina Trifosfato/metabolismo , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ciclopentanos/administración & dosificación , Sinergismo Farmacológico , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/mortalidad , Oxilipinas/administración & dosificación
9.
J Nat Prod ; 75(4): 793-7, 2012 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-22458669

RESUMEN

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).


Asunto(s)
Penicillium/química , Xantonas/aislamiento & purificación , Xantonas/farmacología , Costa Rica , Humanos , Datos de Secuencia Molecular , Estructura Molecular , Resonancia Magnética Nuclear Biomolecular , Células del Estroma/efectos de los fármacos , Xantonas/química
10.
Blood ; 119(15): e131-8, 2012 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-22289890

RESUMEN

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.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento/métodos , Inmunidad Innata/fisiología , Mediciones Luminiscentes/métodos , Neoplasias/diagnóstico , Neoplasias/inmunología , Animales , Antígeno CD56/metabolismo , Compartimento Celular/genética , Compartimento Celular/inmunología , Compartimento Celular/fisiología , Supervivencia Celular , Diagnóstico por Imagen/instrumentación , Diagnóstico por Imagen/métodos , Ensayos Analíticos de Alto Rendimiento/instrumentación , Humanos , Inmunoterapia/métodos , Células K562 , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/patología , Mediciones Luminiscentes/instrumentación , Neoplasias/patología , Neoplasias/terapia , Especificidad por Sustrato/inmunología , Células Tumorales Cultivadas
11.
Mol Cancer Ther ; 11(4): 942-51, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22246439

RESUMEN

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.


Asunto(s)
Mieloma Múltiple/tratamiento farmacológico , Enzimas Activadoras de Ubiquitina/antagonistas & inhibidores , Ubiquitinas/antagonistas & inhibidores , Animales , Ácidos Borónicos/farmacología , Bortezomib , Línea Celular Tumoral , Perfilación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mieloma Múltiple/enzimología , Mieloma Múltiple/genética , Proteína NEDD8 , Pirazinas/farmacología , Resultado del Tratamiento , Enzimas Activadoras de Ubiquitina/metabolismo , Ubiquitinación , Ubiquitinas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Cell ; 146(6): 904-17, 2011 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-21889194

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Descubrimiento de Drogas , Mieloma Múltiple/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Animales , Antineoplásicos/química , Azepinas/química , Azepinas/farmacología , Benzodiazepinas/química , Benzodiazepinas/farmacología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Ratones , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-myc/genética , Activación Transcripcional/efectos de los fármacos , Triazoles/química , Triazoles/farmacología
13.
PLoS One ; 6(7): e20226, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21750699

RESUMEN

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.


Asunto(s)
Proteínas de Ciclo Celular/antagonistas & inhibidores , Mieloma Múltiple/tratamiento farmacológico , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Pteridinas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Western Blotting , Huesos/efectos de los fármacos , Huesos/patología , Caspasa 3/metabolismo , Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Técnicas de Cocultivo , Relación Dosis-Respuesta a Droga , Humanos , Concentración 50 Inhibidora , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Osteoclastos/efectos de los fármacos , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Células del Estroma/efectos de los fármacos , Factores de Tiempo , Carga Tumoral/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Quinasa Tipo Polo 1
14.
Haematologica ; 96(8): 1170-9, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21712538

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Isotiocianatos/farmacología , Mieloma Múltiple/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Tiocianatos/farmacología , Animales , Antineoplásicos/uso terapéutico , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Humanos , Isotiocianatos/uso terapéutico , Isotiocianatos/toxicidad , Ratones , Ratones SCID , Mieloma Múltiple/metabolismo , Células del Estroma/efectos de los fármacos , Sulfóxidos , Tiocianatos/uso terapéutico , Tiocianatos/toxicidad , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Blood ; 117(17): 4409-19, 2011 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-21321360

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Mieloma Múltiple , Células Madre Neoplásicas/efectos de los fármacos , Talidomida/análogos & derivados , Transportadoras de Casetes de Unión a ATP/genética , Inhibidores de la Angiogénesis/farmacología , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/fisiología , División Celular/efectos de los fármacos , Fraccionamiento Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias , Resistencia a Antineoplásicos , Humanos , Lenalidomida , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/patología , Mieloma Múltiple/fisiopatología , Recurrencia Local de Neoplasia/prevención & control , Células Madre Neoplásicas/patología , Transducción de Señal/efectos de los fármacos , Células del Estroma/citología , Células del Estroma/efectos de los fármacos , Células del Estroma/fisiología , Sindecano-1/metabolismo , Talidomida/farmacología
16.
Br J Haematol ; 152(4): 420-32, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21223249

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Mieloma Múltiple/patología , Apoptosis/efectos de los fármacos , Ácidos Borónicos/uso terapéutico , Bortezomib , Ciclo Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Técnicas de Cocultivo , Quinasas Ciclina-Dependientes/metabolismo , Citocinas/antagonistas & inhibidores , Citocinas/farmacología , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Combinación de Medicamentos , Interacciones Farmacológicas , Ensayos de Selección de Medicamentos Antitumorales , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/enzimología , Mieloma Múltiple/genética , Pirazinas/uso terapéutico , Transducción de Señal/efectos de los fármacos , Células del Estroma/fisiología , Análisis de Supervivencia , Transcripción Genética , Resultado del Tratamiento , Células Tumorales Cultivadas
17.
Blood ; 116(4): 593-602, 2010 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-20382846

RESUMEN

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.


Asunto(s)
Agonistas del Receptor de Adenosina A2 , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Ensayos de Selección de Medicamentos Antitumorales/métodos , Neoplasias Hematológicas/tratamiento farmacológico , Inhibidores de Fosfodiesterasa/administración & dosificación , Linfocitos B/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Dexametasona/administración & dosificación , Dexametasona/farmacología , Sistemas de Liberación de Medicamentos , Sinergismo Farmacológico , Glucocorticoides/administración & dosificación , Glucocorticoides/farmacología , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/patología , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/patología , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/patología , Inhibidores de Fosfodiesterasa/aislamiento & purificación , Inhibidores de Fosfodiesterasa/farmacología , Estudios de Validación como Asunto
18.
Nat Med ; 16(4): 483-9, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20228816

RESUMEN

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.


Asunto(s)
Ensayos de Selección de Medicamentos Antitumorales/métodos , Mediciones Luminiscentes/métodos , Células del Estroma/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos , Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Humanos , Morfolinas/farmacología , Purinas/farmacología , Células del Estroma/efectos de los fármacos
19.
Br J Haematol ; 147(5): 672-6, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19751238

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Mieloma Múltiple/patología , Piperazinas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Aurora Quinasas , Comunicación Celular/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Técnicas de Cocultivo , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales/métodos , Células Madre Hematopoyéticas/citología , Humanos , Células Tumorales Cultivadas
20.
Cancer Res ; 69(14): 5835-42, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19584292

RESUMEN

The phosphatidylinositol 3-kinase (PI3K)-Akt-mammalian target of rapamycin (mTOR) pathway mediates proliferation, survival, and drug resistance in multiple myeloma (MM) cells. Here, we tested the anti-MM activity of NVP-BEZ235 (BEZ235), which inhibits PI3K/Akt/mTOR signaling at the levels of PI3K and mTOR. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide colorimetric survival assays showed that MM cell lines exhibited dose- and time-dependent decreased viability after exposure to BEZ235 (IC(50), 25-800 nmol/L for 48 hours). MM cells highly sensitive (IC(50), <25 nmol/L) to BEZ235 (e.g., MM.1S, MM.1R, Dox40, and KMS-12-PE) included both lines sensitive and resistant to conventional (dexamethasone, cytotoxic chemotherapeutics) agents. Pharmacologically relevant BEZ235 concentrations (25-400 nmol/L) induced rapid commitment to and induction of MM.1S and OPM-2 cell death. Furthermore, normal donor peripheral blood mononuclear cells were less sensitive (IC(50), >800 nmol/L) than the majority of MM cell lines tested, suggesting a favorable therapeutic index. In addition, BEZ235 was able to target MM cells in the presence of exogenous interleukin-6, insulin-like growth factor-1, stromal cells, or osteoclasts, which are known to protect against various anti-MM agents. Molecular profiling revealed that BEZ235 treatment decreased the amplitude of transcriptional signatures previously associated with myc, ribosome, and proteasome function, as well as high-risk MM and undifferentiated human embryonic stem cells. In vivo xenograft studies revealed significant reduction in tumor burden (P = 0.011) and survival (P = 0.028) in BEZ235-treated human MM tumor-bearing mice. Combinations of BEZ235 with conventional (e.g., dexamethasone and doxorubicin) or novel (e.g., bortezomib) anti-MM agents showed lack of antagonism. These results indicate that BEZ235 merits clinical testing, alone and in combination with other agents, in MM.


Asunto(s)
Imidazoles/farmacología , Mieloma Múltiple/tratamiento farmacológico , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Quinolinas/farmacología , Administración Oral , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Disponibilidad Biológica , Ciclo Celular/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Imidazoles/administración & dosificación , Imidazoles/farmacocinética , Immunoblotting , Factor I del Crecimiento Similar a la Insulina/farmacología , Interleucina-6/farmacología , Ratones , Ratones Desnudos , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacocinética , Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Quinolinas/administración & dosificación , Quinolinas/farmacocinética , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...