Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
N Engl J Med ; 369(19): 1783-96, 2013 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-24180494

RESUMO

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 .).


Assuntos
Imidazóis/uso terapêutico , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Piridazinas/uso terapêutico , Trombose/induzido quimicamente , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Imidazóis/efeitos adversos , Masculino , Pessoa de Meia-Idade , Inibidores de Proteínas Quinases/efeitos adversos , Piridazinas/efeitos adversos , Trombocitopenia/induzido quimicamente , Adulto Jovem
2.
Oncogene ; 28(23): 2289-98, 2009 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-19398955

RESUMO

BRAF, a cellular oncogene and effector of RAS-mediated signaling, is activated by mutation in approximately 60% of melanomas. Most of these mutations consist of a V600E substitution resulting in constitutive kinase activation. Mutant BRAF thus represents an important therapeutic target in melanoma. In an effort to produce a pre-clinical model of mutant BRAF function in melanoma, we have generated a mouse expressing BRAF V600E targeted to melanocytes. We show that in these transgenic mice, widespread benign melanocytic hyperplasia with histological features of nevi occurs, with biochemical evidence of senescence. Melanocytic hyperplasia progresses to overt melanoma with an incidence dependent on BRAF expression levels. Melanomas show CDKN2A loss, and genetic disruption of the CDKN2A locus greatly enhances melanoma formation, consistent with collaboration between BRAF activation and CDKN2A loss suggested from studies of human melanoma. The development of melanoma also involves activation of the Mapk and Akt signaling pathways and loss of senescence, findings that faithfully recapitulate those seen in human melanomas. This murine model of mutant BRAF-induced melanoma formation thus provides an important tool for identifying further genetic alterations that cooperates with BRAF and that may be useful in enhancing susceptibility to BRAF-targeted therapeutics in melanoma.


Assuntos
Melanócitos/patologia , Melanoma/patologia , Nevo/patologia , Proteínas Proto-Oncogênicas B-raf/genética , Neoplasias Cutâneas/patologia , Substituição de Aminoácidos , Animais , Animais Recém-Nascidos , Southern Blotting , Linhagem Celular Tumoral , Células Cultivadas , Senescência Celular , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Perfilação da Expressão Gênica , Humanos , Melanócitos/metabolismo , Melanoma/genética , Melanoma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mutação , Nevo/genética , Nevo/metabolismo , Proteínas Proto-Oncogênicas B-raf/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células de Schwann/metabolismo , Células de Schwann/patologia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
3.
J Clin Oncol ; 19(3): 812-23, 2001 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11157035

RESUMO

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.


Assuntos
Antineoplásicos/uso terapêutico , Interferon-alfa/uso terapêutico , Melanoma/tratamento farmacológico , Satisfação do Paciente , Adulto , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos/efeitos adversos , Atitude Frente a Saúde , Quimioterapia Adjuvante , Intervalo Livre de Doença , Feminino , Humanos , Interferon alfa-2 , Interferon-alfa/efeitos adversos , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/tratamento farmacológico , Qualidade de Vida , Proteínas Recombinantes , Fatores de Risco , Inquéritos e Questionários , Taxa de Sobrevida
4.
Oncologist ; 6(1): 34-55, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11161227

RESUMO

UNLABELLED: For the past 40 years, various forms of interferon (IFN) have been evaluated as therapy in a number of malignant and non-malignant diseases. With the advent of gene cloning, large quantities of pure IFN became available for clinical study. This paper reviews the biology, pharmacology, and clinical applications of IFN formulations most commonly used in oncology. It then reviews the most common side effects seen in patients treated with IFN, and makes recommendations for the management of IFN-induced toxicity. The major oncological indications for IFN include melanoma, renal cell carcinoma, AIDS-related Kaposi's sarcoma, follicular lymphoma, hairy cell leukemia, and chronic myelogenous leukemia. Unfortunately, IFN therapy is associated with significant toxicity, which can be divided into constitutional, neuropsychiatric, hematologic, and hepatic effects. These toxicities have a major impact on the patient's quality of life, and on the physician's ability to optimally treat the patient. Careful attention to all aspects of patient care can result in improved tolerability of this difficult but promising therapy. CONCLUSION: a better understanding of IFN biology, indications, side effect profiles, and toxicity management will aid in optimizing its use in the treatment of patients with cancer.


Assuntos
Carcinoma de Células Renais/tratamento farmacológico , Interferons , Neoplasias Renais/tratamento farmacológico , Leucemia de Células Pilosas/tratamento farmacológico , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Linfoma Folicular/tratamento farmacológico , Melanoma/tratamento farmacológico , Sarcoma de Kaposi/tratamento farmacológico , Neoplasias Cutâneas/tratamento farmacológico , Transplante de Medula Óssea , Quimioterapia Adjuvante , Terapia Combinada , Humanos , Interferons/efeitos adversos , Interferons/farmacocinética , Interferons/farmacologia , Oncologia/tendências , Qualidade de Vida
5.
Arch Dermatol ; 136(9): 1118-22, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10987867

RESUMO

BACKGROUND: In patients with cutaneous melanoma, early age at disease onset is characteristic in familial cases and in individuals with multiple primary melanomas. Both subsets of patients with melanoma are at risk for harboring germline CDKN2A or CDK4 mutations. OBJECTIVE: We set out to prospectively determine the prevalence of CDKN2A and CDK4 mutations in a group of young patients with melanoma. DESIGN: We prospectively screened 913 patients over a 6-month period and identified 519 patients with invasive melanomas. We invited 172 patients with melanoma who were younger than 40 years to participate in the study, and 49 patients consented and donated peripheral blood samples. Forty-nine percent (n = 24) of our patients developed cutaneous melanoma before the age of 30 years. SETTING: A melanoma clinic in the Boston, Mass, area. MAIN OUTCOME MEASURE: We used a combination of single-strand conformation analysis and direct sequencing of samples of peripheral blood leukocyte DNA to search for mutations in exons 1alpha, 1beta, 2, and 3 of CDKN2A and in exon 2 of CDK4. RESULTS: The mean and median ages at diagnosis in our group were 30 and 32 years, respectively. Among a group of 49 patients, we detected 1 (2%; 95% confidence interval, 0.07%-10.8%) Met 53 Ile CDKN2A mutation, which was found in a patient with a strong family history of melanoma. This alteration has been previously shown to impair p16 function. One patient had an Ala 148 Thr change in CDKN2A, which has also been shown to be a polymorphism. We also detected a sequence polymorphism (in the 3' untranslated region [3'UTR] of CDKN2A) in 27% of our patients. A similar incidence of this 3'UTR polymorphism was observed in a control population. We found no CDK4 mutations. CONCLUSIONS: Germline CDKN2A and CDK4 mutations are not common in patients who develop melanoma at an early age. This finding contrasts with other cancer-predisposition syndromes, in which there is an increased incidence of germline mutations among young patients. Selection of patients with melanoma for genetic testing based solely on age at onset may not be warranted at the current time.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/genética , Quinases Ciclina-Dependentes/genética , Mutação em Linhagem Germinativa/genética , Melanoma/genética , Proteínas Proto-Oncogênicas , Neoplasias Cutâneas/genética , Adolescente , Adulto , Quinase 4 Dependente de Ciclina , Feminino , Predisposição Genética para Doença/genética , Testes Genéticos , Humanos , Masculino , Melanoma/diagnóstico , Melanoma/patologia , Estudos Prospectivos , Sensibilidade e Especificidade , Neoplasias Cutâneas/diagnóstico , Neoplasias Cutâneas/patologia
6.
Cell Immunol ; 204(1): 29-37, 2000 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-11006015

RESUMO

HLA-A2.1/K(b) transgenic mice (A2.1/K(b) mice) were used to investigate the processing of human gp100 melanoma antigen by murine antigen presenting cells (APC). Bone marrow-derived dendritic cells (DC) from A2.1/K(b) mice were transduced with adenovirus encoding human gp100 (Ad2/hugp100v2). The Ad2/hugp100v2-transduced DC express human gp100, as documented by immunoperoxidase staining. Flow cytometric analysis demonstrates that Ad vector transduction does not downregulate expression of several markers, including MHC class I. We show that Ad2/hugp100v2-transduced DC are recognized by peptide-specific, A2.1-restricted CTL, suggesting correct processing and presentation of the hugp100 antigen by murine DC. To assess dominance among the various A2.1-restricted epitopes encoded by hugp100, A2.1/K(b) transgenic mice were immunized with Ad2/hugp100v2-transduced DC. Resulting effector cytotoxic T lymphocytes (CTL) were assayed for peptide specificity using a panel of six synthetic peptides known to encode A2.1-restricted epitopes of human gp100 (denoted G154, G177, G209, G280, G457, G476). CTL obtained from Ad2/hugp100v2-transduced DC immunized A2.1/K(b) mouse lysed target cells presenting five of the six epitopes, supporting the observation that murine cells correctly process the hugp100 antigen. The immunogenicity of individual gp100 epitopes correlates with their binding affinity to A2.1. CTL generated from A2.1/K(b) mice immunized with Ad2/hugp100v2-transduced DC also specifically recognize A2.1(+)/gp100(+) human melanoma cells. These data suggest that murine APC process and present the same set of HLA-restricted peptides, similar to human APC. HLA transgenic mice serve as a useful model system to study class I-restricted epitopes of human tumor-associated antigens.


Assuntos
Apresentação de Antígeno , Antígenos de Neoplasias/imunologia , Células Dendríticas/imunologia , Antígeno HLA-A2/imunologia , Glicoproteínas de Membrana/imunologia , Proteínas de Neoplasias/imunologia , Adenoviridae/genética , Animais , Citotoxicidade Imunológica , Epitopos , Genes MHC Classe I , Antígenos H-2/genética , Antígenos H-2/imunologia , Antígeno HLA-A2/genética , Humanos , Melanoma/imunologia , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Proteínas de Neoplasias/genética , Oligopeptídeos/imunologia , Proteínas Recombinantes de Fusão/imunologia , Especificidade da Espécie , Linfócitos T Citotóxicos , Transformação Genética , Antígeno gp100 de Melanoma
7.
Cancer ; 89(2): 356-62, 2000 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-10918166

RESUMO

BACKGROUND: The use of a high dose regimen of interferon-alpha-2b (IFN) has recently been demonstrated to benefit patients with resected high risk melanoma. The incidence of melanoma is rising rapidly, and the use of this regimen is becoming increasingly common. IFN has been associated with numerous psychiatric side effects. METHODS: The authors describe four melanoma patients treated with adjuvant IFN who developed a manic-depressive syndrome or mood instability with therapy, and they review the literature on mania and the mixed affective syndromes associated with IFN. RESULTS: The authors suggest that IFN may induce a mixed affective instability, and that patients risk developing hypomania or mania as IFN doses fluctuate or as IFN-induced depression is treated with antidepressants alone. Mania is particularly associated with dose reductions or pauses in IFN treatment. The risk of mood fluctuation continues after treatment with IFN stops, and patients should be monitored for 6 months following completion of therapy. Gabapentin appeared effective as monotherapy for acute mania, as an antianxiety agent, as a hypnotic, and as a mood stabilizer in these individual cases. CONCLUSIONS: Mania and mood instability can occur in patients being treated with IFN therapy for melanoma. In this study, gabapentin was an effective mood-stabilizing agent for these patients.


Assuntos
Acetatos/uso terapêutico , Aminas , Ansiolíticos/uso terapêutico , Antimaníacos/uso terapêutico , Antineoplásicos/efeitos adversos , Transtorno Bipolar/induzido quimicamente , Transtorno Bipolar/tratamento farmacológico , Ácidos Cicloexanocarboxílicos , Interferon-alfa/efeitos adversos , Melanoma/tratamento farmacológico , Ácido gama-Aminobutírico , Adulto , Antineoplásicos/uso terapêutico , Transtorno Bipolar/prevenção & controle , Quimioterapia Adjuvante , Depressão/induzido quimicamente , Depressão/tratamento farmacológico , Feminino , Gabapentina , Humanos , Interferon alfa-2 , Interferon-alfa/uso terapêutico , Masculino , Pessoa de Meia-Idade , Transtornos do Humor/induzido quimicamente , Transtornos do Humor/tratamento farmacológico , Estudos Prospectivos , Proteínas Recombinantes
8.
Cancer J ; 6(3): 139-45, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10882328

RESUMO

We performed an analysis of toxicity and survival in stage III melanoma patients receiving adjuvant interferon alfa-2b (IFN). This was a retrospective single-arm analysis of 40 patients with stage III melanoma who received (IFN) administered at maximum tolerated doses of 20 mU/m2/day intravenously (i.v.) for 1 month and 10 mU/m2 three times per week subcutaneously (s.c.) for 48 weeks. Toxicity in our series is comparable to that experienced in the Eastern Cooperative Oncology Group (ECOG) 1684 trial, except for higher rates of dose-limiting myelosuppression and hepatotoxicity. All 40 patients experienced constitutional symptoms, but only 14/40 (35%) experienced grade 3 to 4 symptoms. Of the 40 patients, 36 (90%) experienced neurologic symptoms, but only seven (17.5%) experienced grade 3 to 4 neurotoxicity. Two patients stopped treatment because of severe psychiatric symptoms; one patient attempted suicide, and a psychosis developed in another. Thirty-nine (97.5%) patients experienced myelosuppression; 31 (77.5%) developing grade 3 to 4 myelosuppression. Hepatotoxicity was evident in 39 (97.5%) patients, and 26 (65%) experienced grade 3 to 4 hepatotoxicity. Three patients (7.5%) experienced mild renal toxicity. At a median follow-up of 27 months from initiation of therapy, there have been 19 relapses (47.5% disease-free survival [DFS]) and 10 deaths (75% OS) resulting from progression of disease. The DFS compares with the treatment arm in ECOG 1684 at 27 months, but overall survival is higher in our series of patients at the same time point. In a single program setting, IFN can be administered with similar side effects and outcome profiles seen in multi-institutional studies. Modifications in the induction regimen resulted in notably higher hematologic and hepatic toxicities but did not preclude administering further therapy and did not result in increased attrition rate among patients: only nine patients (22.5%) had their treatment stopped as a result of IFN-related toxicity. In comparison, 26% of patients had to have their treatment discontinued because of toxicity in ECOG 1684.


Assuntos
Antineoplásicos/uso terapêutico , Quimioterapia Adjuvante , Interferon-alfa/uso terapêutico , Melanoma/tratamento farmacológico , Adulto , Antineoplásicos/efeitos adversos , Antineoplásicos/toxicidade , Ensaios Clínicos como Assunto , Intervalo Livre de Doença , Relação Dose-Resposta a Droga , Feminino , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/mortalidade , Humanos , Interferon alfa-2 , Interferon-alfa/efeitos adversos , Interferon-alfa/toxicidade , Metástase Linfática , Masculino , Melanoma/mortalidade , Pessoa de Meia-Idade , Proteínas Recombinantes , Estudos Retrospectivos , Fatores de Risco , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/mortalidade , Fatores de Tempo
9.
Cancer Res ; 60(7): 1800-4, 2000 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-10766161

RESUMO

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.


Assuntos
Genes Supressores de Tumor , Genes ras , Melanoma/genética , Mutação , Monoéster Fosfórico Hidrolases/genética , Neoplasias Cutâneas/genética , Proteínas Supressoras de Tumor , Processamento Alternativo , Sequência de Aminoácidos , Animais , Sequência de Bases , Neoplasias Encefálicas/genética , Éxons , Mutação da Fase de Leitura , Deleção de Genes , Glioma/genética , Humanos , Melanoma Experimental/genética , Camundongos , PTEN Fosfo-Hidrolase , Mutação Puntual , Proteínas Proto-Oncogênicas p21(ras)/genética , Deleção de Sequência , Células Tumorais Cultivadas
10.
J Immunol ; 164(6): 3402-12, 2000 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-10706736

RESUMO

Replication-deficient recombinant adenovirus (Ad) encoding human gp100 or MART-1 melanoma Ag was used to transduce human dendritic cells (DC) ex vivo as a model system for cancer vaccine therapy. A second generation E1/E4 region deleted Ad which harbors the CMV immediate-early promoter/enhancer and a unique E4-ORF6/pIX chimeric gene was employed as the backbone vector. We demonstrate that human monocyte-derived DC are permissive to Ad infection at multiplicity of infection between 100 and 500 and occurs independent of the coxsackie Ad receptor. Fluorescent-labeled Ad was used to assess the kinetics and distribution of viral vector within DC. Ad-transduced DC show peak transgene expression at 24-48 h and expression remains detectable for at least 7 days. DC transduced with replication-deficient Ad do not exhibit any unusual phenotypic characteristics or cytopathic effects. DC transduced with Ad2/gp100v2 can elicit tumor-specific CTL in vitro from patients bearing gp100+ metastatic melanoma. Using a panel of gp100-derived synthetic peptides, we show that Ad2/gp100v2-transduced DC elicit Ag-specific CTL that recognize only the G209 and G280 epitopes, both of which display relatively short half-lives ( approximately 7-8 h) on the surface of HLA-A*0201+ cells. Thus, patients with metastatic melanoma are not tolerant to gp100 Ag based on the detection of CD8+ T cells specific for multiple HLA-A*0201-restricted, gp100-derived epitopes.


Assuntos
Adenovírus Humanos/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Epitopos de Linfócito T/imunologia , Antígeno HLA-A2/imunologia , Melanoma/imunologia , Glicoproteínas de Membrana/imunologia , Proteínas de Neoplasias/imunologia , Adenovírus Humanos/genética , Antígenos de Neoplasias , Células Cultivadas , Células Dendríticas/metabolismo , Enterovirus Humano B/genética , Epitopos de Linfócito T/genética , Corantes Fluorescentes/metabolismo , Vetores Genéticos/imunologia , Antígeno HLA-A2/genética , Meia-Vida , Humanos , Epitopos Imunodominantes/genética , Epitopos Imunodominantes/imunologia , Imunofenotipagem , Antígeno MART-1 , Melanoma/genética , Glicoproteínas de Membrana/genética , Proteínas de Neoplasias/genética , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Receptores Virais/genética , Linfócitos T Citotóxicos/imunologia , Vírion/genética , Vírion/metabolismo , Replicação Viral/genética , Antígeno gp100 de Melanoma
11.
Arch Surg ; 134(4): 381-7, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10199310

RESUMO

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.


Assuntos
Linfonodos/patologia , Melanoma/patologia , Neoplasias Cutâneas/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Coortes , Feminino , Seguimentos , Humanos , Imuno-Histoquímica , Cuidados Intraoperatórios , Metástase Linfática , Masculino , Melanoma/mortalidade , Melanoma/secundário , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/epidemiologia , Neoplasias Cutâneas/mortalidade , Taxa de Sobrevida
12.
Biochem Biophys Res Commun ; 255(1): 59-63, 1999 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-10082655

RESUMO

The tumor suppressor gene MEN1 and several oncogenes including CCND1/cyclin D1/PRAD1 map to chromosome 11q13. However, molecular and cytogenetic analysis suggests the presence of a second tumor suppressor locus at this chromosome region. We have identified a novel gene from chromosome 11q13, which encodes a protein of 126 amino acids sharing an overall 57% identity with the p12(DOC-1) protein encoded by the DOC-1 gene, the human homolog of hamster putative tumor suppressor doc-1 (deleted in oral cancer-1). We therefore designated the novel gene as DOC-1R for DOC-1-related. The cytogenetic location was confirmed by chromosome fluorescent in situ hybridization. Northern blot analysis indicated that it was expressed in all the tissues examined. DOC-1R protein showed heterogeneous subcellular localization. RT-PCR-SSCP analysis failed to detect deleterious mutations of the DOC-1R transcript in four premalignant oral keratinocyte lines and 20 different cancer cell lines from tumor types which frequently harbor LOH at chromosome 11q13.


Assuntos
Cromossomos Humanos Par 11 , Genes Supressores de Tumor , Proteínas Oncogênicas/genética , Proteínas/genética , Proteínas Supressoras de Tumor , Sequência de Aminoácidos , Animais , Linhagem Celular , Mapeamento Cromossômico , Cricetinae , Análise Mutacional de DNA , Humanos , Dados de Sequência Molecular , Alinhamento de Sequência
13.
Cancer Res ; 59(1): 172-4, 1999 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-9892203

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/genética , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Melanoma/genética , Proteínas Nucleares/genética , Mapeamento Cromossômico , Análise Mutacional de DNA , Humanos , Transcrição Gênica , Células Tumorais Cultivadas , Proteína Tumoral p73 , Proteínas Supressoras de Tumor
14.
Artigo em Inglês | MEDLINE | ID: mdl-10800498

RESUMO

In melanoma, conventional therapies, especially cytotoxic chemotherapies, have proven unsatisfactory. Although a variety of agents have been tested singly and in combination, recent randomized studies have demonstrated that the response rates observed in single institution phase II trials are not generalizable to multi-institution settings. Studies of cytokine therapy, especially interferon-alpha in the adjuvant situation, and interleukin-2 for advanced disease, have demonstrated some utility to these agents. However, their toxicities remain formidable, and studies refining their use are under way. The limited successes of such immunomodulatory strategies have provided a rationale for continued pursuit of immunotherapy approaches. It is likely that in the near future such active immunization protocols will continue to be actively investigated.


Assuntos
Melanoma/terapia , Quimioterapia Adjuvante , Humanos , Imunoterapia , Interferons/uso terapêutico , Interleucina-12/uso terapêutico , Interleucina-2/uso terapêutico , Estadiamento de Neoplasias
16.
Oncogene ; 16(26): 3397-402, 1998 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-9692547

RESUMO

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.


Assuntos
Genes Supressores de Tumor , Melanoma/genética , Monoéster Fosfórico Hidrolases , Proteínas Tirosina Fosfatases/genética , Proteínas Supressoras de Tumor , Cromossomos Humanos Par 10 , Éxons , Humanos , Perda de Heterozigosidade , Melanoma/etiologia , Família Multigênica , PTEN Fosfo-Hidrolase , Polimorfismo Genético , Polimorfismo Conformacional de Fita Simples , Células Tumorais Cultivadas
17.
Oncogene ; 16(18): 2403-6, 1998 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-9620558

RESUMO

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.


Assuntos
Cromossomos Humanos Par 9 , Monoéster Fosfórico Hidrolases , Proteínas Tirosina Fosfatases/genética , Pseudogenes , Proteínas Supressoras de Tumor , Sequência de Aminoácidos , Sequência de Bases , Bandeamento Cromossômico , Sequência Conservada , Síndrome do Hamartoma Múltiplo , Humanos , Dados de Sequência Molecular , PTEN Fosfo-Hidrolase , Homologia de Sequência de Aminoácidos , Homologia de Sequência do Ácido Nucleico , Especificidade da Espécie , Síndrome
18.
J Clin Oncol ; 16(2): 670-82, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9469357

RESUMO

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.


Assuntos
Melanoma/genética , Neoplasias Cutâneas/genética , Cromossomos Humanos Par 1/genética , Cromossomos Humanos Par 9/genética , Quinases Ciclina-Dependentes/genética , Genes p16/genética , Ligação Genética , Marcadores Genéticos , Predisposição Genética para Doença , Mutação em Linhagem Germinativa , Humanos
19.
Cancer Res ; 58(1): 109-13, 1998 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-9426066

RESUMO

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.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/genética , Quinases Ciclina-Dependentes/genética , Mutação Puntual , Proteínas Proto-Oncogênicas , Sítios de Ligação/genética , Códon/genética , Neoplasias do Colo/genética , Quinase 4 Dependente de Ciclina , Éxons/genética , Mutação em Linhagem Germinativa , Humanos , Melanoma/genética , Neuroblastoma/genética , Polimorfismo Conformacional de Fita Simples , Análise de Sequência de DNA , Células Tumorais Cultivadas
20.
Int J Cancer ; 72(2): 337-9, 1997 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-9219842

RESUMO

A candidate murine tumor-suppressor gene, Mom1, has been identified as the secretory phospholipase A2 (GDB nomenclature: PLA2G2A) gene. Evidence suggests that PLA2G2A functions as a tumor-suppressor because mice lacking PLA2G2A expression demonstrate increased colonic polyposis. The human homologue of PLA2G2A has been mapped to chromosome 1p36, a region frequently implicated in the pathogenesis of neuroblastoma, colon cancer and melanoma. We identified 2 alterations in the PLA2G2A gene in a single neuroblastoma cell line out of 20 examined; however, we found no mutations in 24 melanoma cell lines, 12 lymphoblastoid cell lines from patients having chromosome 1-linked familial melanoma and 10 colon cancer cell lines. Secretory phospholipase A2 is unlikely to play a significant role in the pathogenesis of these tumors.


Assuntos
Neoplasias Encefálicas/genética , Cromossomos Humanos Par 1 , Neoplasias do Colo/genética , Melanoma/genética , Mutação , Neuroblastoma/genética , Fosfolipases A/genética , Animais , Mapeamento Cromossômico , Genes Supressores de Tumor , Humanos , Camundongos , Fosfolipases A2 , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA