Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS One ; 19(5): e0303789, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38768102

RESUMEN

Mucopolysaccharidosis type I (MPS I) is an inherited lysosomal disease caused by lowered activity of the enzyme alpha-L-iduronidase (IDUA). Current therapeutic options show limited efficacy and do not treat some important aspects of the disease. Therefore, it may be advantageous to identify strategies that could improve the efficacy of existing treatments. Pharmacological chaperones are small molecules that protect proteins from degradation, and their use in combination with enzyme replacement therapy (ERT) has been proposed as an alternative therapeutic strategy. Using the SEE-Tx® proprietary computational drug discovery platform, a new allosteric ligand binding cavity in IDUA was identified distal from the active site. Virtual high-throughput screening of approximately 5 million compounds using the SEE-Tx® docking platform identified a subset of small molecules that bound to the druggable cavity and functioned as novel allosteric chaperones of IDUA. Experimental validation by differential scanning fluorimetry showed an overall hit rate of 11.4%. Biophysical studies showed that one exemplary hit molecule GT-01803 bound to (Kd = 22 µM) and stabilized recombinant human IDUA (rhIDUA) in a dose-dependent manner. Co-administration of rhIDUA and GT-01803 increased IDUA activity in patient-derived fibroblasts. Preliminary in vivo studies have shown that GT-01803 improved the pharmacokinetic (PK) profile of rhIDUA, increasing plasma levels in a dose-dependent manner. Furthermore, GT-01803 also increased IDUA enzymatic activity in bone marrow tissue, which benefits least from standard ERT. Oral bioavailability of GT-01803 was found to be good (50%). Overall, the discovery and validation of a novel allosteric chaperone for rhIDUA presents a promising strategy to enhance the efficacy of existing treatments for MPS I. The compound's ability to increase rhIDUA activity in patient-derived fibroblasts and its good oral bioavailability underscore its potential as a potent adjunct to ERT, particularly for addressing aspects of the disease less responsive to standard treatment.


Asunto(s)
Iduronidasa , Mucopolisacaridosis I , Iduronidasa/metabolismo , Iduronidasa/genética , Mucopolisacaridosis I/tratamiento farmacológico , Humanos , Regulación Alostérica/efectos de los fármacos , Animales , Ratones , Terapia de Reemplazo Enzimático/métodos , Descubrimiento de Drogas , Fibroblastos/metabolismo , Fibroblastos/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Estabilidad de Enzimas , Simulación del Acoplamiento Molecular
2.
Cancer Cell ; 5(6): 565-74, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15193259

RESUMEN

R-etodolac, a nonsteroidal anti-inflammatory drug, inhibits the progression of CWRSA6 androgen-independent and LuCaP-35 androgen-dependent prostate cancer xenograft growth through downregulation of cyclin D1 expression via the PPARgamma pathway. PPARgamma protein degradation, observed post-R-etodolac treatment, resulted from phospho-MAP kinase (p44/42) induction by R-etodolac negatively regulating PPARgamma function. Negative regulation of PPARgamma was overcome by a combination regimen of R-etodolac with the HER-kinase axis inhibitor, rhuMab 2C4, which demonstrated an additive antitumor effect. We further show that the inhibition of HER-kinase activity by rhuMab 2C4 is sufficient to inhibit PPARgamma protein degradation. This study introduces a novel concept of an in vivo crosstalk between the HER-kinase axis and PPARgamma pathways, ultimately leading to negative regulation of PPARgamma activity and tumor growth inhibition.


Asunto(s)
Receptores ErbB/antagonistas & inhibidores , Etodolaco/farmacología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Transcripción/metabolismo , Animales , Antineoplásicos/farmacología , Compuestos Azo/farmacología , Western Blotting , Antígenos CD36/biosíntesis , Línea Celular , Línea Celular Tumoral , Colorantes/farmacología , Ciclina D1/biosíntesis , Ciclina D1/metabolismo , Regulación hacia Abajo , Activación Enzimática , Citometría de Flujo , Humanos , Metabolismo de los Lípidos , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Desnudos , Modelos Biológicos , Modelos Químicos , Monocitos/metabolismo , Células 3T3 NIH , Trasplante de Neoplasias , Neoplasias de la Próstata/patología , Isoformas de Proteínas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Activación Transcripcional , Transfección
3.
BJU Int ; 108(7): 1210-4, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21314886

RESUMEN

OBJECTIVE: • To study the pharmacokinetic and toxicity profile of intravesically administered TMX-101, with its active ingredient R-837, a synthetic Toll-like receptor (TLR)-7 agonist, in a pig model. MATERIALS AND METHODS: • TLR-7 expression was determined by immunohistochemistry in human and pig bladder tissue. • Four groups of six pigs received a 1-h intravesical instillation with R-837 of different formulations. • Pharmacokinetic analysis was performed on plasma. Toxicity evaluation included monitoring the well-being of the animals, peripheral blood cell counts, and interleukin-6 and creatinine measurements. Urine was collected for R-837 measurement and dipstick analysis. • In total, three pigs per group were sacrificed 24 h post-treatment, and the remaining animals were sacrificed after 1 week. Histopathological examination of the bladder wall was performed. RESULTS: • TLR-7 was homogeneously expressed in human and pig urothelium. • R-837 and vehicle were well tolerated without deterioration in animal well-being. • Systemic R-837 absorption was low. • Mean maximum plasma concentration of R-837 differed depending on the formulation. Post-treatment, plasma levels were negligible at 24 h. • Histopathological examination of the bladders did not show significant abnormalities, apart from the intended inflammatory reaction in the R-837 treated groups. CONCLUSION: • Intravesically administered R-837 in pigs, which showed a similar TLR-7 distribution in bladder tissue as humans, is well tolerated and causes no bladder wall toxicity, and formulations with poloxamer and hydroxypropyl-ß-cyclodextrin showed less systemic absorption.


Asunto(s)
Adyuvantes Inmunológicos/farmacocinética , Adyuvantes Inmunológicos/toxicidad , Aminoquinolinas/farmacocinética , Aminoquinolinas/toxicidad , Adyuvantes Inmunológicos/administración & dosificación , Administración Intravesical , Aminoquinolinas/administración & dosificación , Animales , Imiquimod , Porcinos
4.
Clin Adv Hematol Oncol ; 9(8 Suppl 19): 1-11, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22362008

RESUMEN

Bendamustine is a chemotherapeutic agent that displays a unique pattern of cytotoxicity compared with conventional alkylating agents. Bendamustine was originally synthesized in the former East German Democratic Republic in the 1960s. It was designed to have both alkylating and antimetabolite properties. The alkylating agent properties are similar to those seen with cyclophosphamide, chlorambucil, and melphalan, and the benzimidazole ring is similar to cladribine. Molecular analyses have revealed that bendamustine differs from other alkylating agents in its mechanism of action. Differences have been observed in regard to its effects on DNA repair and cell cycle progression. Moreover, bendamustine can induce cell death through both apoptotic and nonapoptotic pathways, thereby retaining activity even in cells without a functional apoptotic pathway. Bendamustine has demonstrated significant efficacy in patients with indolent lymphomas and chronic lymphocytic leukemia (CLL), including in patients with disease refractory to conventional alkylating agents and rituximab. The toxicity profile of bendamustine is also superior to that of conventional alkylating agents. Combination therapy with bendamustine and rituximab has demonstrated superior efficacy to a standard rituximab-containing chemotherapy regimen in patients with previously untreated indolent B-cell non-Hodgkin lymphoma, and it is currently being compared against the standard first-line regimen in CLL: fludarabine, cyclophosphamide, and rituximab. Ongoing and planned studies are evaluating new strategies in which bendamustine is being combined with existing agents and with novel therapies to optimize use in different clinical settings.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Neoplasias Hematológicas/tratamiento farmacológico , Compuestos de Mostaza Nitrogenada/farmacología , Compuestos de Mostaza Nitrogenada/farmacocinética , Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/farmacocinética , Clorhidrato de Bendamustina , Ensayos Clínicos como Asunto , Humanos , Compuestos de Mostaza Nitrogenada/química
5.
Int J Urol ; 17(5): 483-90, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20337728

RESUMEN

OBJECTIVE: To study the immune response caused by the intravesical administration of the immunomodulator R-837 in various formulations and to estimate its therapeutic potential for bladder cancer. METHODS: Female C57BL/6 mice were intravesically treated with different formulations of R-837, a Toll-like receptor 7 agonist used for treating genital warts and skin malignancy. The tested formulation mixtures contained different ratios of lactic acid, a thermosensitive poloxamer polymer (Lutrol F127) and 2-(hydroxypropyl)-beta-cyclodextrin (HPbetaCD). Induction of tumor necrosis factor alpha (TNFalpha) and keratinocyte-derived chemokine (KC) was analyzed by Luminex microbeads assay. The therapeutic potential of intravesical administration of R-837 was assessed in an orthotopic, syngeneic mouse model of bladder cancer using MB49 cells. RESULTS: Intravesical administration of R-837 in lactic acid alone induced systemic and bladder TNFalpha and KC in a dose-dependent manner. Formulations including poloxamer decreased systemic absorption of R-837 and significantly reduced systemic and local induction of KC. Addition of HPbetaCD in the poloxamer formulation particularly reversed levels of systemic and local levels of TNFalpha and KC. Histological examination showed that poloxamer-HPbetaCD formulation allowed infiltration of mononuclear cells into urothelium and lamina propria. In studies using orthotopic mouse bladder cancer, the tumor loads in R-837-treated mice were significantly lower than those in vehicle-treated or non-treated mice. CONCLUSION: The optimized poloxamer-HPbetaCD formulation of R-837 shows therapeutic potential for bladder cancer while avoiding adverse side-effects.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Aminoquinolinas/farmacología , Química Farmacéutica/métodos , Glicoproteínas de Membrana/agonistas , Receptor Toll-Like 7/agonistas , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , 2-Hidroxipropil-beta-Ciclodextrina , Administración Intravesical , Animales , Cistitis/prevención & control , Citocinas/metabolismo , Modelos Animales de Enfermedad , Excipientes/farmacología , Femenino , Imiquimod , Ácido Láctico/farmacología , Ratones , Ratones Endogámicos C57BL , Polietilenos/farmacología , Polipropilenos/farmacología , beta-Ciclodextrinas/farmacología
6.
Clin Cancer Res ; 14(1): 309-17, 2008 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-18172283

RESUMEN

PURPOSE: Bendamustine has shown clinical activity in patients with disease refractory to conventional alkylator chemotherapy. The purpose of this study was to characterize the mechanisms of action of bendamustine and to compare it with structurally related compounds. EXPERIMENTAL DESIGN: Bendamustine was profiled in the National Cancer Institute in vitro antitumor screen. Microarray-based gene expression profiling, real-time PCR, immunoblot, cell cycle, and functional DNA damage repair analyses were used to characterize response to bendamustine and compare it with chlorambucil and phosphoramide mustard. RESULTS: Bendamustine displays a distinct pattern of activity unrelated to other DNA-alkylating agents. Its mechanisms of action include activation of DNA-damage stress response and apoptosis, inhibition of mitotic checkpoints, and induction of mitotic catastrophe. In addition, unlike other alkylators, bendamustine activates a base excision DNA repair pathway rather than an alkyltransferase DNA repair mechanism. CONCLUSION: These results suggest that bendamustine possesses mechanistic features that differentiate it from other alkylating agents and may contribute to its distinct clinical efficacy profile.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Ciclo Celular/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Compuestos de Mostaza Nitrogenada/farmacología , Apoptosis/efectos de los fármacos , Clorhidrato de Bendamustina , Western Blotting , Línea Celular Tumoral , Clorambucilo/farmacología , Ciclofosfamida/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Citometría de Flujo , Humanos
8.
Cancer Chemother Pharmacol ; 60(4): 545-53, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17186240

RESUMEN

OBJECTIVE: SDX-101 is the non-cyclooxygenase 2-inhibiting R-enantiomer of the non-steroid anti-inflammatory drug etodolac, and has anti-tumour activity in chronic lymphocytic leukaemia (CLL). SDX-308 and SDX-309 are more potent, structurally related indole-pyran analogues of SDX-101. The current study was performed to investigate and quantify the cytotoxic potentiating effects resulting from a combination of either SDX-101, SDX-308 or SDX-309 with standard cytotoxic agents used in the CLL treatment today. METHODS: The lymphoma cell line U937-gtb was used, together with primary tumour cells isolated from seven CLL patients. Combinations between chlorambucil and each one of the agents etodolac, SDX-101, SDX-308 and SDX-309 were studied. In addition, SDX-309 was combined with fludarabine, doxorubicin or vincristine. Both simultaneous and sequential exposures were explored using the median-effect method. RESULTS: Most combinations were additive, which could be of clinical benefit since SDX-101 has been shown to be well tolerated. At the 70% effect level, synergy was observed between SDX-308 and chlorambucil in U937-gtb cells and in two-third of the CLL samples. Since chlorambucil is the most important drug in CLL therapy today and SDX-308 is presently targeted as the lead clinical candidate, this combination would be interesting for further studies. Vincristine and SDX-309 were synergistic in two-fourth of CLL samples. CONCLUSIONS: To conclude, the non-COX-inhibiting etodolac-derivatives SDX-101, SDX-308 and SDX-309 are potential candidates for combination treatment of CLL. Especially, SDX-308 in combination with chlorambucil warrants further evaluation.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Etodolaco/farmacología , Compuestos Heterocíclicos con 3 Anillos/farmacología , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Anciano , Anciano de 80 o más Años , Línea Celular Tumoral , Clorambucilo/administración & dosificación , Doxorrubicina/administración & dosificación , Sinergismo Farmacológico , Femenino , Humanos , Linfoma/tratamiento farmacológico , Masculino , Persona de Mediana Edad , Vincristina/administración & dosificación
9.
Clin Cancer Res ; 12(12): 3754-61, 2006 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-16778103

RESUMEN

PURPOSE: To determine the methylthioadenosine phosphorylase (MTAP) gene alterations in mantle cell lymphoma (MCL) and to investigate whether the targeted inactivation of the alternative de novo AMP synthesis pathway may be a useful therapeutic strategy in tumors with inactivation of this enzyme. EXPERIMENTAL DESIGN: MTAP gene deletion and protein expression were studied in 64 and 52 primary MCL, respectively, and the results were correlated with clinical behavior. Five MCL cell lines were analyzed for MTAP expression and for the in vitro sensitivity to L-alanosine, an inhibitor of adenylosuccinate synthetase, and hence de novo AMP synthesis. RESULTS: No protein expression was detected in 8 of 52 (15%) tumors and one cell line (Granta 519). Six of these MTAP negative tumors and Granta 519 cell line had a codeletion of MTAP and p16 genes; one case showed a deletion of MTAP, but not p16, and one tumor had no deletions in neither of these genes. Patients with MTAP deletions had a significant shorter overall survival (mean, 16.1 months) than patients with wild-type MTAP (mean, 63.6 months; P < 0.0001). L-Alanosine induced cytotoxicity and activation of the intrinsic mitochondrial-dependent apoptotic pathway in MCL cells. 9-beta-D-Erythrofuranosyladenine, an analogue of 5'-methylthioadenosine, selectively rescued MTAP-positive cells from L-alanosine toxicity. CONCLUSIONS: MTAP gene deletion and lack of protein expression are associated with poor prognosis in MCL and might identify patients who might benefit from treatment with de novo AMP synthesis pathway-targeted therapies.


Asunto(s)
Linfoma de Células del Manto/enzimología , Purina-Nucleósido Fosforilasa/deficiencia , Purina-Nucleósido Fosforilasa/genética , Secuencia de Bases , Línea Celular Tumoral , Cromosomas Humanos Par 11 , Cromosomas Humanos Par 14 , Cartilla de ADN , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Linfoma de Células del Manto/genética , Linfoma de Células del Manto/mortalidad , Linfoma de Células del Manto/patología , Estudios Retrospectivos , Análisis de Supervivencia , Factores de Tiempo , Translocación Genética
10.
Cancer Res ; 62(14): 4007-14, 2002 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-12124334

RESUMEN

We have shown previously that hypoestoxide (HE), a natural diterpenoid [a bicyclo (9, 3, 1) pentadecane], is a potent nonsteroidal anti-inflammatory drug. In this report, we demonstrate that HE also inhibits the growth of a variety of human and murine tumor cell lines in vitro at concentrations ranging from 0.3 to 10 microM and was inactive as a mutagen in the Ames test. HE exhibited highly potent (0.3-10 mg/kg dose ranges) activities against B16 melanoma growth in C57BL/6 mice and P388D1 leukemia in C57BL/6 x DBA/2 F(1) mice, respectively. At a low maximal effective dose of 5 mg/kg, HE induced significant in vivo antitumor activities that were better than or comparable with most of the standard chemotherapeutic antiangiogenic agents tested: cortisone acetate, vincristine, bleomycin, Adriamycin, 5-fluorouracil, cyclophosphamide, and etoposide. All of the agents, except vincristine, had much higher maximal effective doses than HE. HE arrested the growth of human Burkitt lymphoma CA46 cells and HeLa (cervical epitheloid carcinoma) cells in the G2-M phase of the cell cycle, which was caused by interference, either direct or indirect, with actin assembly. Thus, the cell cycle arrest occurred at cytokinesis, as demonstrated by an increase in the number of binucleate cells. Moreover, HE inhibited vascular endothelial growth factor-induced cell proliferation in vitro, with an IC(50) of 28.6 microM, and it significantly inhibited basic fibroblast growth factor-induced angiogenesis on the chick chorioallantoic membrane, with an IC50 of 10 microM. Furthermore, HE inhibited endothelial cell migration on vitronectin, collagen, and fibronectin. Besides its activity as a nonsteroidal anti-inflammatory drug, HE also has promise for the chemotherapy of cancer.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/farmacología , Diterpenos/farmacología , Actinas/metabolismo , Inhibidores de la Angiogénesis/toxicidad , Animales , Antineoplásicos/toxicidad , Adhesión Celular/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Embrión de Pollo , Diterpenos/toxicidad , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Inhibidores de Crecimiento/farmacología , Células HeLa , Humanos , Leucemia Experimental/tratamiento farmacológico , Leucemia Experimental/patología , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Pruebas de Mutagenicidad , Neovascularización Fisiológica/efectos de los fármacos , Conejos , Tubulina (Proteína)/metabolismo , Células Tumorales Cultivadas
11.
Mol Cancer Ther ; 4(12): 1860-6, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16373701

RESUMEN

The p16(INK4A)/CDKN2A gene on chromosome 9p21 is a site of frequent allelic loss in human cancers, and in a subset of cases, homozygous deletions at this locus encompass the telomeric methylthioadenosine phosphorylase (MTAP) gene. The MTAP gene product is the principal enzyme involved in purine synthesis via the salvage pathway, such that MTAP-negative cancers are solely dependent on de novo purine synthesis mechanisms. Inhibitors of the de novo pathway can then be used to selectively blockade purine synthesis in cancer cells while causing minimal collateral damage to normal cells. In this study, we determine that 10 of 28 (35%) biliary tract cancers show complete lack of Mtap protein expression. In vitro analysis using a selective inhibitor of the de novo purine synthesis pathway, L-alanosine, shows robust growth inhibition in MTAP-negative biliary cancer cell lines CAK-1 and GBD-1 accompanied by striking depletion of intracellular ATP and failure to rescue this depletion via addition of exogenous methylthioadenosine, the principal substrate of the MTAP gene product; in contrast, no significant effects were observed in MTAP-expressing HuCCT1 and SNU308 cell lines. Colony formation studies confirmed that L-alanosine reduced both number and size of CAK-1 colonies in soft agar assays. Knockdown of Mtap protein by RNA interference in L-alanosine-resistant HuCCT1 cells conferred sensitivity to this agent, confirming that intracellular Mtap protein levels determine response to L-alanosine. Inhibitors of de novo purine synthesis can be a potential mechanism-based strategy for treatment of biliary tract cancers, one third of which show complete loss of MTAP function.


Asunto(s)
Neoplasias del Sistema Biliar/genética , Eliminación de Gen , Homocigoto , Purina-Nucleósido Fosforilasa/genética , Secuencia de Bases , Neoplasias del Sistema Biliar/enzimología , Neoplasias del Sistema Biliar/patología , Línea Celular Tumoral , Cartilla de ADN , Humanos , Reacción en Cadena de la Polimerasa
12.
Oncogene ; 21(43): 6598-605, 2002 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-12242657

RESUMEN

The diverse receptor-ligand pairs of the Wnt and frizzled (Fz) families play important roles during embryonic development, and thus may be overexpressed in cancers that arise from immature cells. Hence, we investigated the expression and function of five Wnt (Wnt-1, 5a, 7a, 10b, 13) and two Fz (Fz-2, 5) genes in 10 head and neck squamous carcinoma cell lines (HNSCC). In comparison to normal bronchial or oral epithelial cells, all the HNSCC had markedly increased mRNA levels of Wnt-1, 7a, 10b, and 13, as well as Fz-2. Moreover, the levels of Wnt-1, 10b, and Fz-2 proteins were also markedly increased in HNSCC, relative to normal epithelial cells. Treatment of one HNSCC cell line (SNU 1076) with anti-Wnt-1 antibodies reduced the activity of the Wnt/Fz dependent transcription factor LEF/TCF, and diminished the expression of cyclin D1 and beta-catenin proteins. Blocking Wnt-1 signaling also inhibited proliferation and induced apoptosis in these cells. These results show that HNSCC cell lines often overexpress one or more Wnt and Fz genes, and suggest that the growth and survival of a subset of HNSCC may depend on the Wnt/Fz pathway. Hence, the Wnt and Fz receptors may be possible targets for immunotherapy therapy of this common cancer.


Asunto(s)
Carcinoma de Células Escamosas/terapia , Neoplasias de Cabeza y Cuello/terapia , Péptidos y Proteínas de Señalización Intercelular , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Receptores de Neurotransmisores/antagonistas & inhibidores , Proteínas de Pez Cebra , Anticuerpos/uso terapéutico , Carcinoma de Células Escamosas/metabolismo , Receptores Frizzled , Glicoproteínas/análisis , Glicoproteínas/antagonistas & inhibidores , Glicoproteínas/genética , Neoplasias de Cabeza y Cuello/metabolismo , Humanos , Inmunoterapia , Proteínas Proto-Oncogénicas/análisis , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/análisis , Receptores Acoplados a Proteínas G , Receptores de Neurotransmisores/análisis , Receptores de Neurotransmisores/genética , Células Tumorales Cultivadas , Proteínas Wnt , Proteína Wnt-5a , Proteína Wnt1
13.
Cancer Biol Ther ; 4(1): 83-6, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15662124

RESUMEN

Methylthioadenosine phosphorylase (MTAP) plays an important role in the salvage pathway for the synthesis of adenosine. Novel chemotherapeutic strategies exploiting the selective loss of MTAP function in cancers have been proposed. The MTAP gene, on chromosome 9p21, is frequently included within homozygous deletions of the p16INK4A/ CDKN2A gene. Biallelic deletions of the p16INK4A/CDKN2A gene are found in 40% of pancreatic cancers, suggesting that the MTAP gene may be frequently inactivated in pancreatic cancer and that selected patients with pancreatic cancer may benefit from therapies targeting this loss. We immunolabeled six xenografted pancreatic cancers with known MTAP and p16INK4A/CDKN2A gene status and found that immunolabeling mirrored gene status. Loss of expression of both MTAP and p16 was observed only in those pancreatic cancers with homozygous deletions that encompassed both the MTAP and p16INK4A/CDKN2A genes. We then immunolabeled a series of 320 microarrayed infiltrating pancreatic adenocarcinomas, 35 biliary adenocarcinomas, 54 ampullary cancers, and 35 noninvasive intraductal papillary mucinous neoplasms. Immunolabeling for MTAP was lost in 91 of the 300 (30%) evaluable pancreatic cancers, 9 of 54 (17%) ampullary cancers, 4 of 33 (12%) biliary cancers, and in 1 of 35 (3%) IPMNs. All neoplasms with loss of MTAP labeling also demonstrated loss of p16 labeling. These results suggest that MTAP expression is lost in approximately 30% of infiltrating pancreatic cancers and in a lower percentage of other periampullary neoplasms, that this loss is the result of homozygous deletions encompassing both the MTAP and p16INK4A/CDKN2A genes. Thus, pancreatic cancer is a promising cancer type in which to explore novel chemotherapeutic strategies to exploit the selective loss of MTAP function.


Asunto(s)
Adenocarcinoma/genética , Adenocarcinoma/patología , Neoplasias del Conducto Colédoco/genética , Neoplasias del Conducto Colédoco/patología , Eliminación de Gen , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Purina-Nucleósido Fosforilasa/genética , Adenocarcinoma/terapia , Adulto , Anciano , Anciano de 80 o más Años , Ampolla Hepatopancreática , Animales , Neoplasias del Conducto Colédoco/terapia , Femenino , Humanos , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Pancreáticas/terapia , Trasplante Heterólogo
14.
Am J Surg Pathol ; 29(11): 1497-504, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16224217

RESUMEN

The gene that encodes methylthioadenosine phosphorylase (MTAP), an enzyme involved in adenine and methionine salvage pathways, is located on chromosome 9p21 telomeric to the p16INK4A/CDKN2A tumor suppressor gene. Inactivation of the p16INK4A/CDKN2A gene occurs by three different mechanisms: hypermethylation of the gene promoter, intragenic mutation coupled with loss of the second allele, and homozygous deletion. Immunohistochemical labeling for the p16INK4A/CDKN2A gene product parallels gene status but does not elucidate the mechanism of gene inactivation. Since the MTAP gene is often co-deleted with p16INK4A/CDKN2A, concurrent immunolabeling for both proteins can identify cases with homozygous p16INK4A/CDKN2A gene deletion. MTAP loss itself has therapeutic implications since it may confer selective sensitivity to inhibitors of de novo purine biosynthesis, such as L-alanosine. Twelve tissue microarrays were constructed from 92 cases of Barrett-associated adenocarcinomas and precursor lesions and 112 cases of gastric adenocarcinoma and precursor lesions comprising 1161 individual cores. Multiple cores were arrayed from any given case, and when available, included the entire histologic spectrum of intestinal metaplasia-dysplasia-carcinoma. Tissue microarrays were labeled with monoclonal antibodies against MTAP protein (clone 6.9, Salmedix, Inc) and p16 (clone 16P07, Neomarkers). Complete loss of labeling was considered negative, while any labeling (p16: nuclear; MTAP: cytoplasmic and nuclear) was considered positive. Loss of MTAP labeling occurred exclusively in conjunction with loss of p16 labeling, confirming that the previous findings from this group that concurrent loss of MTAP and p16 labeling is a surrogate marker of 9p21 homozygous deletions. Complete loss of MTAP and p16 was seen in 4 of 25 (16%) patients with Barrett's esophagus, 4 of 18 (22%) with low-grade dysplasia, 5 of 39 (13%) with high-grade dysplasia, 17 of 78 (22%) with invasive adenocarcinoma, and 8 of 36 (22%) of metastases. There were 7 cases of esophageal adenocarcinoma with loss of both MTAP and p16 for which precursor lesions were available. In 6 on these 7 cases (85%), the precursor lesion(s) had loss of both MTAP and p16. Lack of MTAP and p16 expression was seen in 11 of 106 (10%) cases of gastric adenocarcinoma. All metaplastic (30 biopsies from 20 cases) and dysplastic (15 biopsies from 13 cases) gastric tissues had both intact MTAP and p16INK4A/CDKN2A gene products. No precursor lesions were available from the gastric cancers that had loss of both MTAP and p16. Two benign gastric hyperplastic polyps also had intact p16 and MTAP. Concurrent MTAP and p16 loss detected by immunohistochemistry can serve as a convenient surrogate for p16INK4A/CDKN2A gene homozygous deletion in archival tissues. Inactivation of p16INK4A/CDKN2A by homozygous deletion appears to be an early event in Barrett carcinogenesis, occurring in noninvasive precursor lesions, including nondysplastic Barrett mucosa, in subsets of cases. In the absence of MTAP, cells depend exclusively on the de novo synthesis pathway for production of adenosine. This loss of MTAP during 9p21 homozygous deletion might be exploited therapeutically using de novo purine synthesis antimetabolites to treat a subset of invasive gastroesophageal adenocarcinomas and esophageal precursor lesions.


Asunto(s)
Adenocarcinoma/genética , Neoplasias Esofágicas/genética , Genes p16/fisiología , Purina-Nucleósido Fosforilasa/genética , Neoplasias Gástricas/genética , Adenocarcinoma/patología , Esófago de Barrett/genética , Esófago de Barrett/patología , Neoplasias Esofágicas/patología , Eliminación de Gen , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Lesiones Precancerosas/genética , Lesiones Precancerosas/patología , Neoplasias Gástricas/patología
15.
Best Pract Res Clin Haematol ; 16(1): 83-9, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12670467

RESUMEN

Hairy-cell leukaemia cells have a low rate of growth but an even lower rate of apoptosis. Accordingly, this malignancy is an excellent model for studying the effects of drugs on the pathways of apoptosis independently of cell proliferation. The remarkable effectiveness of 2-chlorodeoxyadenosine in hairy-cell leukaemia affirms the feasibility of developing drugs that can destroy even non-proliferating malignant cells. The major nucleotide metabolite of 2-chlorodeoxyadenosine accumulates selectively in lymphocytes and co-activates two key apoptosis-regulating enzymes: poly(ADP-ribose) polymerase and Apaf-1/caspase-9. The ability of 2-chlorodeoxyadenosine to induce durable remissions in hairy-cell leukaemia may also be attributable to its effects on lymphocytes and monocytes in the microenvironment, although this latter effect remains to be proven experimentally.


Asunto(s)
Antineoplásicos/uso terapéutico , Cladribina/uso terapéutico , Evaluación de Medicamentos/métodos , Leucemia de Células Pilosas/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Factor Apoptótico 1 Activador de Proteasas , Caspasa 9 , Caspasas/efectos de los fármacos , Caspasas/metabolismo , Humanos , Leucemia de Células Pilosas/metabolismo , Leucemia de Células Pilosas/patología , Poli(ADP-Ribosa) Polimerasas/efectos de los fármacos , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas/efectos de los fármacos , Proteínas/metabolismo
16.
Artículo en Inglés | MEDLINE | ID: mdl-24110408

RESUMEN

We designed and tested a new rollable and implantable medical device to directly and continuously measure intraocular pressure. Since high intraocular pressure is a leading risk factor for glaucoma, such a system could solve the difficulties encountered in the management of this condition. In fact, glaucoma is one among those pathologies that could most benefit of an adaptive patient-specific medicine device. The presented prototype was realized with standard industrial microelectronic technologies (Flip-Chip on Kapton flexible PCB) and off-the-shelf IC components. Detailed system description and measurements, obtained during in-vitro and laboratory characterizations, are reported.


Asunto(s)
Ojo Artificial , Glaucoma/fisiopatología , Presión Intraocular/fisiología , Tonometría Ocular/instrumentación , Adaptación Ocular , Ritmo Circadiano/fisiología , Humanos , Polímeros/química , Prótesis e Implantes
17.
Semin Hematol ; 48 Suppl 1: S37-8, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21530770

RESUMEN

Bendamustine is a bifunctional mechlorethamine derivative that shares similarities to other alkylators; however, the presence of a benzimidazole ring may confer "nucleoside-like" properties and may allow the stabilization of the molecule leading to longer lasting DNA damage. Though bendamustine has demonstrated promising response rates in preclinical and clinical studies, particularly in follicular lymphoma, chronic lymphocytic leukemia, diffuse B-cell lymphoma, and mantle cell lymphoma, the unique and exact mechanism of action of this agent remains unclear. Several studies have been initiated to address this question, and it is hoped that emerging data will provide the basis for more effective utilization of this interesting drug. Several recent clinical trials have reported impressive results with bendamustine in lymphoid malignancies, and appropriate clinical use of this agent and the rationale behind its use are of growing importance. This review discussed emerging data and aimed to provide clinical updates and scientific rationales that are relevant to practicing clinicians who provide care to patients with lymphoid malignancies, and/or who are interested in understanding the evolving role of bendamustine in this setting.


Asunto(s)
Antineoplásicos/uso terapéutico , Linfoma/tratamiento farmacológico , Compuestos de Mostaza Nitrogenada/uso terapéutico , Animales , Antineoplásicos/farmacología , Clorhidrato de Bendamustina , Daño del ADN/efectos de los fármacos , Humanos , Compuestos de Mostaza Nitrogenada/farmacología
18.
Semin Hematol ; 48 Suppl 1: S4-11, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21530771

RESUMEN

Although the alkylating agent bendamustine was developed in Germany in the mid-twentieth century, it has only recently come to the forefront in the rest of the world as an effective chemotherapeutic agent for the treatment of several hematologic malignancies. Based on the activity demonstrated in single-arm and randomized trials, this nitrogen mustard is approved by the US Food and Drug Administration (FDA) for the treatment of chronic lymphocytic leukemia and rituximab-refractory indolent non-Hodgkin lymphoma. The unique structural and mechanistic features of bendamustine differentiate it from other alkylating agents, providing increased stability and potency in DNA cross linking and subsequent cytotoxicity. Due to its unusual development, few studies have closely examined the mechanisms of action for this nitrogen mustard and many unanswered questions remain. Additionally, phase I and pharmacokinetic studies are limited, although increased understanding of the clinical pharmacology of bendamustine led to development of dosing recommendations by international experts based on the available data. The clinical activity of bendamustine as a single agent and in combination with other chemotherapeutic and immunotherapeutic drugs, coupled with its potential lack of cross-resistance with many other chemotherapy agents, make bendamustine an attractive therapy for patients with newly diagnosed and refractory hematologic malignancies. This review will discuss the development of bendamustine, its structural and pharmacologic characteristics, and current data regarding the optimal dosing of this agent in specific clinical settings.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/uso terapéutico , Neoplasias Hematológicas/tratamiento farmacológico , Compuestos de Mostaza Nitrogenada/química , Compuestos de Mostaza Nitrogenada/uso terapéutico , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Clorhidrato de Bendamustina , Humanos , Compuestos de Mostaza Nitrogenada/farmacocinética , Compuestos de Mostaza Nitrogenada/farmacología
19.
Semin Hematol ; 48 Suppl 1: S12-23, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21530768

RESUMEN

Bendamustine has demonstrated substantial efficacy in the treatment of hematologic malignancies and continues to distinguish itself from other alkylating agents with regard to its activity in tumor cells. The mechanistic and clinical differences associated with bendamustine may be directly related to its unique structural features. Although the precise mechanisms of action are still poorly understood, bendamustine is associated with extensive and durable DNA damage. The increased potency of bendamustine may be due to secondary mechanisms such as inhibition of mitotic checkpoints, inefficient DNA repair, and initiation of p53-dependent DNA-damage stress response, all of which lead to mitotic catastrophe and apoptosis. It has also been hypothesized that the presence of a benzimidazole ring in addition to the nitrogen mustard group may influence the way bendamustine interacts with DNA and/or confer antimetabolite properties. Further elucidation of the mechanisms of action for bendamustine and the signaling pathways involved in the response to bendamustine-induced DNA damage is essential to maximize its therapeutic potential, identify biomarkers for response, and understand the potential for synergy with other agents involved in DNA damage and inhibition of DNA repair. This review will discuss the current understanding and hypotheses regarding bendamustine mechanisms of action and suggest future investigations that would shed light on the many unanswered questions.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Daño del ADN/efectos de los fármacos , Neoplasias Hematológicas/tratamiento farmacológico , Compuestos de Mostaza Nitrogenada/farmacología , Animales , Clorhidrato de Bendamustina , ADN/metabolismo , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA