Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
N Engl J Med ; 369(19): 1783-96, 2013 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-24180494

RESUMEN

BACKGROUND: Ponatinib is a potent oral tyrosine kinase inhibitor of unmutated and mutated BCR-ABL, including BCR-ABL with the tyrosine kinase inhibitor-refractory threonine-to-isoleucine mutation at position 315 (T315I). We conducted a phase 2 trial of ponatinib in patients with chronic myeloid leukemia (CML) or Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph-positive ALL). METHODS: We enrolled 449 heavily pretreated patients who had CML or Ph-positive ALL with resistance to or unacceptable side effects from dasatinib or nilotinib or who had the BCR-ABL T315I mutation. Ponatinib was administered at an initial dose of 45 mg once daily. The median follow-up was 15 months. RESULTS: Among 267 patients with chronic-phase CML, 56% had a major cytogenetic response (51% of patients with resistance to or unacceptable side effects from dasatinib or nilotinib and 70% of patients with the T315I mutation), 46% had a complete cytogenetic response (40% and 66% in the two subgroups, respectively), and 34% had a major molecular response (27% and 56% in the two subgroups, respectively). Responses were observed regardless of the baseline BCR-ABL kinase domain mutation status and were durable; the estimated rate of a sustained major cytogenetic response of at least 12 months was 91%. No single BCR-ABL mutation conferring resistance to ponatinib was detected. Among 83 patients with accelerated-phase CML, 55% had a major hematologic response and 39% had a major cytogenetic response. Among 62 patients with blast-phase CML, 31% had a major hematologic response and 23% had a major cytogenetic response. Among 32 patients with Ph-positive ALL, 41% had a major hematologic response and 47% had a major cytogenetic response. Common adverse events were thrombocytopenia (in 37% of patients), rash (in 34%), dry skin (in 32%), and abdominal pain (in 22%). Serious arterial thrombotic events were observed in 9% of patients; these events were considered to be treatment-related in 3%. A total of 12% of patients discontinued treatment because of an adverse event. CONCLUSIONS: Ponatinib had significant antileukemic activity across categories of disease stage and mutation status. (Funded by Ariad Pharmaceuticals and others; PACE ClinicalTrials.gov number, NCT01207440 .).


Asunto(s)
Imidazoles/uso terapéutico , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Piridazinas/uso terapéutico , Trombosis/inducido químicamente , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Imidazoles/efectos adversos , Masculino , Persona de Mediana Edad , Inhibidores de Proteínas Quinasas/efectos adversos , Piridazinas/efectos adversos , Trombocitopenia/inducido químicamente , Adulto Joven
2.
Br J Cancer ; 108(5): 1021-6, 2013 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-23403817

RESUMEN

BACKGROUND: This open-label, multicentre, phase 2 trial evaluated the efficacy and tolerability of the mammalian target of rapamycin inhibitor ridaforolimus in women with advanced endometrial cancer. METHODS: Women with measurable recurrent or persistent endometrial cancer and documented disease progression were treated with ridaforolimus 12.5 mg intravenously once daily for 5 consecutive days every 2 weeks in a 4-week cycle. The primary end point was clinical benefit response, defined as an objective response or prolonged stable disease of 16 weeks or more. RESULTS: In all, 45 patients were treated with single-agent ridaforolimus. Clinical benefit was achieved by 13 patients (29%), including 5 (11%) with confirmed partial responses and 8 (18%) with prolonged stable disease. All patients with clinical benefit response received ridaforolimus for more than 4 months. In this heavily pretreated population, the 6-month progression-free survival was 18%. Ridaforolimus was generally well tolerated: adverse events were predictable and manageable, consistent with prior studies in other malignancies. Overall, the most common adverse events were diarrhoea (58%) and mouth sores (56%); most common grade 3 or higher adverse events were anaemia (27%) and hyperglycaemia (11%). CONCLUSION: Single-agent ridaforolimus has antitumor activity and acceptable tolerability in advanced endometrial cancer patients. Further clinical evaluation of ridaforolimus is warranted.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Neoplasias Endometriales/tratamiento farmacológico , Sirolimus/análogos & derivados , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos Alquilantes/administración & dosificación , Antineoplásicos Alquilantes/efectos adversos , Supervivencia sin Enfermedad , Esquema de Medicación , Neoplasias Endometriales/patología , Femenino , Humanos , Persona de Mediana Edad , Retratamiento , Sirolimus/administración & dosificación , Sirolimus/efectos adversos , Sirolimus/uso terapéutico
3.
Ann Oncol ; 24(4): 1104-11, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23211938

RESUMEN

BACKGROUND: Ridaforolimus is an inhibitor of mTOR with evidence of antitumor activity in an I.V. formulation. This multicenter, open-label, 3 + 3 design nonrandomized, dose-escalation, phase I/IIa trial was conducted to determine the safety, pharmacokinetic (PK) and pharmacodynamic parameters, maximum tolerated dose, and antitumor activity of oral ridaforolimus. PATIENTS AND METHODS: Patients with metastatic or unresectable solid tumors refractory to therapy were eligible. Seven different continuous and intermittent dosing regimens were examined. RESULTS: One hundred and forty-seven patients were enrolled in this study among which 85 were patients with sarcoma. Stomatitis was the most common DLT observed. The dosing regimen, 40 mg QD × 5 days/week, provided the best combination of cumulative dose, dose density, and cumulative exposure, and was the recommended dosing regimen for subsequent clinical development. PK was nonlinear, with less than proportional increases in day-1 blood AUC0-∞ and Cmax, particularly with doses >40 mg. The terminal half-life estimate of ridaforolimus (QD × 5 40 mg) was 42.0 h, and the mean half-life ∼30-60 h. The clinical benefit rate, (complete response, partial response, or stable disease for ≥4 months was 24.5% for all patients and 27.1% for patients with sarcoma. CONCLUSION: Oral ridaforolimus had an acceptable safety profile and exhibited antitumor activity in patients with sarcoma and other malignancies. ClinicalTrials.gov Identifier NCT00112372.


Asunto(s)
Neoplasias/tratamiento farmacológico , Sarcoma/tratamiento farmacológico , Sirolimus/análogos & derivados , Serina-Treonina Quinasas TOR/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Esquema de Medicación , Femenino , Humanos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias/patología , Sarcoma/patología , Sirolimus/administración & dosificación , Sirolimus/efectos adversos , Sirolimus/antagonistas & inhibidores , Sirolimus/farmacocinética , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Resultado del Tratamiento
4.
J Clin Pharm Ther ; 38(6): 440-4, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23888935

RESUMEN

WHAT IS KNOWN AND OBJECTIVE: Ponatinib is a potent oral tyrosine kinase inhibitor with activity against BCR-ABL, the primary driver of chronic myeloid leukaemia and Philadelphia chromosome-positive acute lymphoblastic leukaemia. This single-centre, single-dose, randomized, open-label, three-period crossover study evaluated the pharmacokinetics and bioavailability of a single oral dose of ponatinib (45-mg tablet) under fasting conditions and following consumption of high- and low-fat meals by healthy subjects. METHODS: Subjects were randomly assigned to one of the six possible treatment sequences, each evaluating three ponatinib 45-mg treatments: administered under fasting conditions; administered after a high-fat meal; or administered after a standardized low-fat meal. The high-fat meal derived approximately 50% of its total caloric content from fat, with approximately 150, 250 and 500-600 calories derived from protein, carbohydrates and fat, respectively (total of approximately 900-1000 calories). The standardized low-fat meal derived no more than 20% of total caloric content from fat, with approximately 56, 428 and 63 calories derived from protein, carbohydrates and fat, respectively (total of approximately 547 calories). During each of the three treatment periods, blood samples were collected predose and at 13 time points over the 96-h post-dose interval. Plasma concentrations of ponatinib were measured by liquid chromatography/tandem mass spectrometry. Mixed-model analyses of variance (anova) were performed on natural log-transformed PK parameters Cmax and AUC0-∞. RESULTS AND DISCUSSION: Geometric mean maximum plasma concentration (Cmax) values for the fasted, low-fat and high-fat regimens were 54·7, 51·6 and 51·5 ng/mL, respectively. Geometric mean area under the concentration-time curve from time zero to infinity (AUC0-∞) values for the fasted, low-fat and high-fat regimens were 1273, 1244 and 1392 h × ng/mL, respectively. All limits of the 90% CIs of the estimated geometric mean ratios for Cmax and all AUC comparisons fell within the 80%-125% margins. These results indicate that consumption of a high- or low-fat meal within 30 min prior to administration of ponatinib had no effect on the single-dose pharmacokinetics of ponatinib. WHAT IS NEW AND CONCLUSION: Food does not affect the single-dose pharmacokinetics of ponatinib. These data demonstrate that ponatinib may be administered with or without food.


Asunto(s)
Antineoplásicos/farmacocinética , Interacciones Alimento-Droga , Imidazoles/farmacocinética , Piridazinas/farmacocinética , Adulto , Análisis de Varianza , Área Bajo la Curva , Disponibilidad Biológica , Índice de Masa Corporal , Química Farmacéutica , Cromatografía Líquida de Alta Presión , Estudios Cruzados , Grasas de la Dieta/farmacología , Etnicidad , Ayuno/metabolismo , Femenino , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , Caracteres Sexuales , Espectrometría de Masas en Tándem , Adulto Joven
5.
Ann Oncol ; 21(6): 1315-1322, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19901013

RESUMEN

BACKGROUND: The additive cytotoxicity in vitro prompted a clinical study evaluating the non-prodrug rapamycin analogue ridaforolimus (AP23573; MK-8669; formerly deforolimus) administered i.v. combined with paclitaxel (PTX; Taxol). MATERIALS AND METHODS: Patients with taxane-sensitive solid tumors were eligible. The main dose escalation foresaw 50% ridaforolimus increments from 25 mg with a fixed PTX dose of 80 mg/m(2), both given weekly 3 weeks in a 4-week cycle. Collateral levels with a lower dose of either drug were planned upon achievement of the maximum tolerated dose in the main escalation. Pharmacodynamic studies in plasma, peripheral blood mononuclear cells (PBMCs) and skin biopsies and pharmacokinetic (PK) interaction studies at cycles 1 and 2 were carried out. RESULTS: Two recommended doses were determined: 37.5 mg ridaforolimus/60 mg/m(2) PTX and 12.5 mg/80 mg/m(2). Most frequent toxic effects were mouth sores (79%), anemia (79%), fatigue (59%), neutropenia (55%) and dermatitis (48%). Two partial responses were observed in pharyngeal squamous cell and pancreatic carcinoma. Eight patients achieved stable disease > or =4 months. No drug interaction emerged from PK studies. Decrease of eukaryotic initiation factor 4E-binding protein1 (4E-BP1) phosphorylation was shown in PBMCs. Similar inhibition of phosphorylation of 4E-BP1 and mitogen-activated protein kinase was present in reparative epidermis and vascular tissues, respectively. CONCLUSION: Potential antiangiogenic effects and encouraging antitumor activity justify further development of the combination.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias/tratamiento farmacológico , Paclitaxel/administración & dosificación , Sirolimus/análogos & derivados , Adulto , Anciano , Algoritmos , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Supervivencia sin Enfermedad , Esquema de Medicación , Interacciones Farmacológicas , Femenino , Humanos , Inyecciones Intravenosas , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Neoplasias/sangre , Neoplasias/metabolismo , Paclitaxel/efectos adversos , Paclitaxel/farmacocinética , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Sirolimus/administración & dosificación , Sirolimus/efectos adversos , Sirolimus/farmacocinética , Serina-Treonina Quinasas TOR , Resultado del Tratamiento
6.
Artículo en Inglés | MEDLINE | ID: mdl-2660190

RESUMEN

The body of this work illustrates the utility of the combined cytogenetic and molecular approach to lymphoid tumorigenesis. A number of tumor-specific translocations have proven amenable to dissection by molecular techniques. We have a firm grasp of the general principles that underlie lymphoid neoplasia; in particular, the activation of cellular oncogenes by translocation into genes of the immunoglobulin superfamily is a widespread phenomenon. However, numerous lymphopoietic malignancies are only poorly understood. These remain a challenge for the continued application of these methodologies.


Asunto(s)
Linfoma/genética , Translocación Genética , Secuencia de Bases , Linfoma de Burkitt/genética , Centrómero , ADN/genética , Humanos , Datos de Secuencia Molecular
7.
Cancer Res ; 60(7): 1800-4, 2000 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-10766161

RESUMEN

Both inactivation of the tumor suppressor gene, PTEN/MMAC1, and oncogenic activation of RAS have been described in human cutaneous melanoma. In mice, activation of a RAS-containing pathway is a necessary step in the pathogenesis of murine melanomas. Because PTEN negatively regulates on the downstream effects of phosphatidylinositol-3-kinase (PI3-K), we hypothesized that the loss of PTEN/MMAC1 and the activation of RAS may be largely equivalent because RAS is a known positive upstream regulator of PI3-K. We expanded our previous survey of PTEN/MMAC1 mutations and analyzed the RAS status of 53 cutaneous melanoma cell lines, 18 glioma cell lines, and 17 uncultured cutaneous melanoma metastasis. Overall, 51% of the cell lines had alterations in either PTEN/MMAC1 or RAS. We found 16 cell lines (30%) with alterations in PTEN/MMAC1 and 11 cell lines (21%) with activating NRAS mutations; only 1 cell line had concurrent alterations in both genes. Moreover, glioma cell lines with a high frequency of PTEN/MMAC1 inactivation had no identifiable RAS alterations. Ectopic expression of PTEN in several cutaneous melanoma cell lines suppressed colony formation irrespective of PTEN/MMAC1 status; furthermore, PTEN expression in cell lines carrying activated RAS also suppressed colony formation. The relative reciprocity of PTEN/MMAC1 abrogation and NRAS activation suggests that the two genetic changes, in a subset of cutaneous melanomas, are functionally overlapping.


Asunto(s)
Genes Supresores de Tumor , Genes ras , Melanoma/genética , Mutación , Monoéster Fosfórico Hidrolasas/genética , Neoplasias Cutáneas/genética , Proteínas Supresoras de Tumor , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Neoplasias Encefálicas/genética , Exones , Mutación del Sistema de Lectura , Eliminación de Gen , Glioma/genética , Humanos , Melanoma Experimental/genética , Ratones , Fosfohidrolasa PTEN , Mutación Puntual , Proteínas Proto-Oncogénicas p21(ras)/genética , Eliminación de Secuencia , Células Tumorales Cultivadas
8.
Cancer Res ; 59(1): 172-4, 1999 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-9892203

RESUMEN

A novel p53-related gene, p73, was recently isolated and cytogenetically mapped to chromosome region 1p36. Functionally, p73 expression induces p21waf and suppresses tumor cell growth. We mapped p73 using radiation hybrids and localized the gene to an interval that putatively harbors a melanoma tumor suppressor locus. We then analyzed p73 transcripts from 24 melanoma cell lines using reverse transcription-PCR/single strand conformation polymorphism and identified nine polymorphic sequence changes (three novel and six previously published polymorphisms); furthermore, we found evidence of biallelic transcription in our cell lines. However, we did not detect any deleterious mutations. These data suggest that the p73 gene is unlikely to be essential in melanoma tumorigenesis.


Asunto(s)
Proteínas de Unión al ADN/genética , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Melanoma/genética , Proteínas Nucleares/genética , Mapeo Cromosómico , Análisis Mutacional de ADN , Humanos , Transcripción Genética , Células Tumorales Cultivadas , Proteína Tumoral p73 , Proteínas Supresoras de Tumor
9.
Cancer Res ; 58(1): 109-13, 1998 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-9426066

RESUMEN

Mutations in genes that lie in the retinoblastoma pathway have been implicated in the pathogenesis of many tumor types. Two critical components that determine progression from G1 to S include p16/CDKN2A and CDK4. Alterations in p16/CDKN2A have been well documented in multiple cancers, including melanoma. However, changes in CDK4 are apparently more rare. Only two alterations, both at codon 24, have been identified in CDK4: an activating arginine-to-cysteine transition and a germ-line arginine-to-histidine substitution in one French kindred. In a survey of 20 neuroblastomas, 17 uncultured metastatic melanomas, 33 uncultured primary uveal melanomas, 8 colon cancer cell lines, and 20 primary colon cancer samples, we found no evidence of mutations in exon 2 of CDK4. From our cell lines derived from metastatic melanomas, we detected two alterations in the functionally critical exon 2 of CDK4: a lysine-to-glutamine transition at codon 22 and the arginine-to-histidine mutation at codon 24. These findings document several novel changes in the p16-binding region of CDK4.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Quinasas Ciclina-Dependientes/genética , Mutación Puntual , Proteínas Proto-Oncogénicas , Sitios de Unión/genética , Codón/genética , Neoplasias del Colon/genética , Quinasa 4 Dependiente de la Ciclina , Exones/genética , Mutación de Línea Germinal , Humanos , Melanoma/genética , Neuroblastoma/genética , Polimorfismo Conformacional Retorcido-Simple , Análisis de Secuencia de ADN , Células Tumorales Cultivadas
10.
Cancer Res ; 52(23): 6547-52, 1992 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-1330296

RESUMEN

To analyze the region upstream of c-myc, a number of novel probes were established. These were generated by chromosomal walking starting from the breakpoint of the chromosomal translocation of the B-cell line 380 and by cloning the breakpoint of the translocation of the Burkitt lymphoma cell line IARC/BL72. Using the newly isolated probes a detailed physical map of 500 kilobases of the region upstream of c-myc was established applying pulsed-field gel electrophoresis. The chromosomal breakpoint of IARC/BL72 cells was mapped to a site 55 kilobases 5' of c-myc. A region 20 kilobases in length and containing the breakpoints of 380, EW36, P3HR-1, and Daudi cells was identified 170-190 kilobases upstream of c-myc. In addition the HPV18 integration site in HeLa cells was located between 340 and 500 kilobases 5' of c-myc. The probes were used to define the c-myc amplification units in Colo320-HSR and HL60 cells as well as in four cases of small cell lung cancer. Evidence is provided that the amplicon of HL60 cells is discontinuously organized.


Asunto(s)
Linfoma de Burkitt/genética , Paseo de Cromosoma/métodos , Cromosomas Humanos Par 14 , Cromosomas Humanos Par 8 , Genes myc/genética , Mutagénesis Insercional/genética , Translocación Genética/genética , Secuencia de Bases , Carcinoma de Células Pequeñas/genética , Electroforesis en Gel de Campo Pulsado , Amplificación de Genes , Biblioteca Genómica , Humanos , Hibridación in Situ , Datos de Secuencia Molecular , Células Tumorales Cultivadas
11.
Cancer Res ; 55(13): 2713-8, 1995 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-7796391

RESUMEN

Sporadic and familial malignant melanoma susceptibility has been linked to defects in the chromosomal region 9p21. Recently, a putative 9p21 tumor suppressor gene, the cyclin dependent kinase inhibitor 2 (CDKN2) or p16 gene, has been shown to be deleted, mutated, or rearranged in a high percentage of sporadic melanoma cell lines, as well as mutated in the germline of a proportion of familial melanoma patients. CDKN2 encodes a M(r) 16,000 protein (p16) that plays a key role in cell cycle control by binding to the cyclin-dependent kinase 4 enzyme and inhibiting its ability to phosphorylate critical substrates necessary for transition past the G1 phase of the cell cycle. Thus, mutations or deletions of the CDKN2 gene could result in abnormal proliferation via defective cell cycle control. The correlation of 9p21 cytogenetic and molecular alterations with the clinical stages of melanoma progression suggests that dysfunction of a gene within this chromosomal region is critical to the evolution of melanoma. However, it remains unclear whether this gene is the CDKN2 gene. If so, then loss of p16 is potentially an initiating or early event in melanoma progression. To address the issues of what is the potential involvement of the CDKN2 gene in sporadic melanoma and precisely when during the clinically evident stages of melanoma progression defects in CDKN2 occur, we have evaluated by immunohistochemistry the expression of p16 protein in 103 melanocytic lesions representing all stages in the progression of melanoma. Our results suggest that loss of p16 protein expression is (a) not necessary for tumor initiation in malignant melanoma because all melanomas in situ and the majority of primary invasive melanomas retain expression of this protein; and (b) potentially more related to invasiveness or the ability to metastasize, because 52% of primary invasive tumors and 72% of metastatic lesions show partial or complete loss of expression of p16.


Asunto(s)
Proteínas Portadoras/metabolismo , Genes Supresores de Tumor , Melanocitos/metabolismo , Melanoma/metabolismo , Neoplasias Cutáneas/metabolismo , Western Blotting , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Humanos , Inmunohistoquímica , Melanocitos/patología , Metástasis de la Neoplasia
12.
Cancer Res ; 56(21): 5023-32, 1996 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-8895759

RESUMEN

Although homozygous deletions of the cyclin-dependent kinase inhibitor 2 gene p16INK4a on 9p21 have been reported frequently in metastatic melanoma cell lines, and intragenic mutations within the p16INK4a gene have been detected in familial melanoma kindreds, specific targeting of this gene in the development of sporadic melanoma in vivo remains controversial. Southern analyses were performed in this study to initially assess the frequency of hemi- or homozygous losses of p16INK4a, as well as its neighboring family member, p15INK4b, and other candidate regions within 9p21, in sporadic melanoma. Overall, 22 of 40 (55%) uncultured sporadic melanoma DNAs were determined to harbor deletions of 1-11 markers/genes located on 9p21. This included 10 tumors (25%; 10 of 40) with homozygous deletions limited to either the p16INK4a gene only (20%; 2 of 10), both the p16INK4a and p15INK4b genes (10%; 1 of 10), another novel 9p21 gene, FB19 (10%; 1 of 10), or all three of these genes plus surrounding markers (60%; 6 of 10). In subsequent single-strand conformation polymorphism and sequencing analyses, intragenic mutations in the p16INK4a gene were also revealed in two (10%; 2 of 21) melanoma DNAs that retained one copy of this locus. By comparison, the frequency of pl6INK4a and p15INK4b homozygous deletions, as well as p16INK4a mutations, in melanoma cell lines (analyzed in parallel) was 2-3-fold higher at 61 (23 of 38) and 24% (9 of 38), respectively. These findings indicate that (a) p16INK4a is inactivated in vivo in over one-fourth (27.5%; 11 of 40) of sporadic melanomas; (b) mutation/deletion of p16INK4a may confer a selective growth advantage in vitro; and (c) other 9p21 tumor suppressor genes could be targeted during the development of melanoma.


Asunto(s)
Proteínas Portadoras/genética , Proteínas de Ciclo Celular , Deleción Cromosómica , Cromosomas Humanos Par 9 , Eliminación de Gen , Genes Supresores de Tumor , Melanoma/genética , Proteínas Supresoras de Tumor , Inhibidor p15 de las Quinasas Dependientes de la Ciclina , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Marcadores Genéticos , Humanos , Melanoma/secundario , Mutación , Células Tumorales Cultivadas
13.
Oncogene ; 16(26): 3397-402, 1998 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-9692547

RESUMEN

A novel tumor suppressor gene, PTEN/MMAC1, has been recently shown to be mutated in gliomas, breast, prostate, kidney cancers and melanomas. Loss-of-heterozygosity studies in melanoma have suggested the presence of at least one chromosome 10q locus lost early in tumor progression. In this study, we screened 45 melanoma cell lines and 17 paired uncultured metastatic melanoma and peripheral blood specimens for PTEN/ MMAC1 alterations using PCR-SSCP and direct sequencing. We found nine melanoma cell lines with homozygous deletions (five with intragenic loss) and four cell lines with mutations (one nonsense and one frameshift; two intronic); from among our uncultured melanoma specimens, we found one tumor with a somatic 17 bp duplication in exon 7 leading to a premature stop codon and one tumor with a possible homozygous deletion. Furthermore, we have identified a novel intragenic polymorphism within intron 4 of PTEN/MMAC1. Taken together, these data suggest that PTEN/MMAC1 may be a chromosome 10q tumor suppressor important in melanoma tumor formation or progression.


Asunto(s)
Genes Supresores de Tumor , Melanoma/genética , Monoéster Fosfórico Hidrolasas , Proteínas Tirosina Fosfatasas/genética , Proteínas Supresoras de Tumor , Cromosomas Humanos Par 10 , Exones , Humanos , Pérdida de Heterocigocidad , Melanoma/etiología , Familia de Multigenes , Fosfohidrolasa PTEN , Polimorfismo Genético , Polimorfismo Conformacional Retorcido-Simple , Células Tumorales Cultivadas
14.
Oncogene ; 16(18): 2403-6, 1998 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-9620558

RESUMEN

PTEN/MMAC1/TEP1, encoding a dual-specificity phosphatase, is a tumor suppressor gene which has recently been cloned and mapped to chromosome 10q23.3. We have shown that germline mutations of PTEN are present in individuals with two hamartoma syndromes: Cowden Syndrome, associated with a predisposition to breast and thyroid cancers, and Bannayan-Zonana syndrome. Somatic mutations of PTEN have been reported in a variety of human cancer cell lines, suggesting a potential role for this gene in the pathogenesis of human malignancies. We report the identification of a highly conserved PTEN processed pseudogene, psiPTEN, which shares over 98% homology with the coding region of functional PTEN, and its localisation to chromosome 9p21. The high sequence homology of psiPTEN with the PTEN transcript may potentially lead to misinterpretation when performing mutation analyses based on cDNA templates. Caution should be exerted when using such screening approaches.


Asunto(s)
Cromosomas Humanos Par 9 , Monoéster Fosfórico Hidrolasas , Proteínas Tirosina Fosfatasas/genética , Seudogenes , Proteínas Supresoras de Tumor , Secuencia de Aminoácidos , Secuencia de Bases , Bandeo Cromosómico , Secuencia Conservada , Síndrome de Hamartoma Múltiple , Humanos , Datos de Secuencia Molecular , Fosfohidrolasa PTEN , Homología de Secuencia de Aminoácido , Homología de Secuencia de Ácido Nucleico , Especificidad de la Especie , Síndrome
15.
J Clin Oncol ; 16(2): 670-82, 1998 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9469357

RESUMEN

PURPOSE: A family history of melanoma is a significant risk factor for the disease, and recently several loci that determine susceptibility to the development of melanoma have been identified. The most important of these is p16/CDKN2A. We attempted to determine the degree to which the p16/CDKN2A gene has been implicated in the development of melanoma, and to identify other genetic factors that play a role as well. METHODS: We reviewed the literature published since the isolation of p16/CDKN2A and identified 13 studies that report the status of the gene in melanoma samples and 12 reports that examine p16/CDKN2A in melanoma kindreds. We also reviewed associated studies on CDK4 and RB1 involvement in melanoma, and examined the role of p16/CDKN2A in other inherited cancers. RESULTS: The evidence strongly implicates p16/CDKN2A in determining predisposition to malignant melanoma. Overall, approximately 20% of families that have been studied show mutations in the gene. However, because of clustering of sporadic cases in families, and potentially because of technical factors, this is likely an underestimate of the proportion of the genetic predisposition for melanoma that is due to p16/CDKN2A mutation. Rare families carry a mutated CDK4 gene that is also responsible for inherited melanoma. CONCLUSION: The gene p16/CDKN2A is an important determinant of melanoma risk. A commercial test is presently available to assess the status of this locus. However, because of uncertainties regarding the interpretation of the results of p16/CDKN2A genetic testing, we do not recommend routine clinical use of this test at this time.


Asunto(s)
Melanoma/genética , Neoplasias Cutáneas/genética , Cromosomas Humanos Par 1/genética , Cromosomas Humanos Par 9/genética , Quinasas Ciclina-Dependientes/genética , Genes p16/genética , Ligamiento Genético , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Humanos
16.
J Clin Oncol ; 19(3): 812-23, 2001 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-11157035

RESUMEN

PURPOSE: Although trials of adjuvant interferon alfa-2b (IFN alpha-2b) in high-risk melanoma patients suggest improvement in disease-free survival, it is unclear whether treatment offers improvement in overall survival. Widespread use of adjuvant IFN alpha-2b has been tempered by its significant toxicity. To quantify the trade-offs between IFN alpha-2b toxicity and survival, we assessed patient utilities for health states associated with IFN therapy. Utilities are measures of preference for a particular health state on a scale of 0 (death) to 1 (perfect health). PATIENTS AND METHODS: We assessed utilities for health states associated with adjuvant IFN among 107 low-risk melanoma patients using the standard gamble technique. Health states described four IFN alpha-2b toxicity scenarios and the following three posttreatment outcomes: disease-free health and melanoma recurrence (with or without IFN alpha-2b) leading to cancer death. We also asked patients the improvement in 5-year disease-free survival they would require to tolerate IFN. RESULTS: Utilities for melanoma recurrence with or without IFN alpha-2b were significantly lower than utilities for all IFN alpha-2b toxicities but were not significantly different from each other. At least half of the patients were willing to tolerate mild-moderate and severe IFN alpha-2b toxicity for 4% and 10% improvements, respectively, in 5-year disease-free survival. CONCLUSION: On average, patients rate quality of life with melanoma recurrence much lower than even severe IFN alpha-2b toxicity. These results suggest that recurrence-free survival is highly valued by patients. The utilities measured in our study can be applied directly to quality-of-life determinations in clinical trials of adjuvant IFN alpha-2b to measure the net benefit of therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Interferón-alfa/uso terapéutico , Melanoma/tratamiento farmacológico , Satisfacción del Paciente , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/efectos adversos , Actitud Frente a la Salud , Quimioterapia Adyuvante , Supervivencia sin Enfermedad , Femenino , Humanos , Interferón alfa-2 , Interferón-alfa/efectos adversos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/tratamiento farmacológico , Calidad de Vida , Proteínas Recombinantes , Factores de Riesgo , Encuestas y Cuestionarios , Tasa de Supervivencia
17.
Mol Endocrinol ; 9(10): 1356-66, 1995 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8544844

RESUMEN

WT1, a gene deleted in some Wilms' tumors, encodes a transcription factor with zinc fingers and shares homology with proteins in the early growth response gene family. Although defects in the WT1 gene are associated with nephroblastoma and genitourinary malformation, the specific function of WT1 in the gonads remains unclear. We investigated the expression of WT1 transcripts in rat ovary during follicle development by Northern blotting, RNase protection assay, and in situ hybridization. Abundant WT1 transcripts were found in the ovary, testis, uterus, and kidney, with lower levels in the heart and pancreas. Treatment with estrogen or gonadotropins did not affect the concentration of ovarian WT1 mRNA. In situ hybridization analysis indicated that ovarian WT1 mRNA is expressed exclusively in the surface epithelium and granulosa cells of primordial, primary, and secondary follicles, and its levels decrease during follicle growth. Although RNase protection assay suggested the presence of four alternatively spliced forms of WT1 mRNA, the ratio of these transcripts remains constant during ovarian growth. Developmental changes in the expression of two granulosa cell differentiation marker genes, inhibin-alpha and FSH receptor, were found to be inversely correlated with WT1 levels. Because potential WT1-binding sites were found in the promoter of inhibin-alpha gene, we further tested whether WT1 might regulate the expression of this gene. Cotransfection of a WT1 expression vector with a promoter reporter plasmid of inhibin-alpha resulted in the repression of promoter activities in CHO cells in a dose-dependent manner. These results suggest that WT1 is expressed in high levels in granulosa cells of primordial, primary, and secondary follicles but decreases with follicle development. This transcription factor might be a repressor of ovarian differentiation genes in the granulosa cells and play a role in arresting the differentiation of immature follicles.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Inhibinas/genética , Folículo Ovárico/metabolismo , Ovario/metabolismo , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular , Femenino , Regulación de la Expresión Génica , Hibridación in Situ , Inhibinas/metabolismo , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas WT1
18.
Arch Ophthalmol ; 101(5): 799-801, 1983 May.
Artículo en Inglés | MEDLINE | ID: mdl-6342583

RESUMEN

Ophthalmia nodosa is the nodular granulomatous inflammatory response of ocular tissue to caterpillar hairs. We experimentally simulated this condition by surgically implanting gypsy moth hairs in the rabbit cornea. We were not, however, able to produce ophthalmia nodosa solely by placing cilia in the rabbit cul-de-sac. The experimentally induced inflammation closely resembled human ophthalmia nodosa in both its clinical and histopathologic features.


Asunto(s)
Endoftalmitis/patología , Cuerpos Extraños en el Ojo/patología , Reacción a Cuerpo Extraño/patología , Lepidópteros , Mariposas Nocturnas , Animales , Córnea/patología , Femenino , Granuloma/patología , Cabello , Humanos , Masculino , Conejos
19.
Ann N Y Acad Sci ; 511: 196-206, 1987.
Artículo en Inglés | MEDLINE | ID: mdl-3326464

RESUMEN

Our understanding of the molecular biology of many tumors is still rudimentary. The genes involved in most solid tumors, and the mechanisms giving rise to their activation, are virtually unknown. In contrast, for hematopoietic malignancies, we have identified several genes important in oncogenesis, and we understand in at least a limited sense the mechanisms by which translocations impart these genes with the capacity to support malignant growth. It is becoming increasingly apparent that these mechanisms involve very subtle changes in the pattern of gene expression. Similarly, the studies on the mechanisms of chromosome translocation described here underscore the proposition that malignancy may arise by slight perturbations of normal function. The enzymes that physiologically recombine Ig and TCR genes do so with high fidelity under normal circumstances. But occasionally misrecognition of target sequences may lead to chromosome translocation and neoplasia. A deeper understanding of this process will be facilitated by a more certain grasp of normal B- and T-lymphocyte differentiation and proliferation.


Asunto(s)
Leucemia/genética , Linfoma/genética , Translocación Genética , Linfocitos B , Secuencia de Bases , Linfoma de Burkitt/genética , Diferenciación Celular , Cromosomas Humanos/ultraestructura , Humanos , Leucemia Linfoide/genética , Linfoma Folicular/genética , Datos de Secuencia Molecular , Linfocitos T
20.
Arch Surg ; 134(4): 381-7, 1999 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10199310

RESUMEN

HYPOTHESIS: Patients with melanoma and histologically negative sentinel lymph nodes identified by lymphatic mapping have a very good prognosis. DESIGN: Cohort study with follow-up information obtained from medical records and telephone interviews. SETTING AND PATIENTS: Of all patients with cutaneous melanoma who underwent intraoperative sentinel lymph node mapping between November 15, 1993, and April 18, 1997, at the Massachusetts General Hospital, Boston, 89 were found to have no evidence of melanoma in their sentinel nodes. Forty-six lesions (51%) were on an extremity and 44 (49%) were of axial location. The median tumor thickness was 1.8 mm (range, 0.36-12.0 mm) and 11 tumors (12%) were ulcerated. INTERVENTIONS: Patients underwent intraoperative sentinel lymph node mapping with lymphazurin and radiolabeled sulfur colloid. Sentinel lymph nodes were analyzed by standard hematoxylin-eosin staining. Only 2 patients received adjuvant therapy following wide excision of the primary lesion. MAIN OUTCOME MEASURES: Site of initial recurrence and time to initial recurrence. RESULTS: The median follow-up for all patients was 23 months (range, 2-54 months). Eleven patients (12%) developed melanoma recurrences, and 78 (88%) patients remain disease free. Regional lymph nodes were the initial site of recurrence in 7 (8%) of 89 patients, and 7 (7%) of 106 mapped basins. Four patients had recurrence without involvement of regional lymph nodes: 2 with distant metastases and 2 with in transit metastases. The median time to recurrence was 12 months (range, 2-35 months). Sentinel lymph nodes were reanalyzed using serial sections and immunoperoxidase stains in 7 patients with recurrence and metastatic melanoma was identified in 3 (43%). CONCLUSIONS: The risk for melanoma recurrence is relatively low in patients with histologically negative sentinel nodes identified by lymphatic mapping. Longer follow-up will improve our understanding of the prognostic value of this procedure.


Asunto(s)
Ganglios Linfáticos/patología , Melanoma/patología , Neoplasias Cutáneas/patología , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Cohortes , Femenino , Estudios de Seguimiento , Humanos , Inmunohistoquímica , Cuidados Intraoperatorios , Metástasis Linfática , Masculino , Melanoma/mortalidad , Melanoma/secundario , Persona de Mediana Edad , Recurrencia Local de Neoplasia/epidemiología , Neoplasias Cutáneas/mortalidad , Tasa de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA