Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Pharmacogenomics J ; 15(4): 347-53, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25446780

RESUMO

Cancer stem cells (CSCs) are thought to promote resistance to chemotherapeutic drugs in glioblastoma, the most common and aggressive primary brain tumor. However, the use of high-throughput drug screens to discover novel small-molecule inhibitors for CSC has been hampered by their instability in long-term cell culture. We asked whether predictive models of drug response could be developed from gene expression signatures of established cell lines and applied to predict drug response in glioblastoma stem cells. Predictions for active compounds were confirmed both for 185 compounds in seven established glioma cell lines and 21 compounds in three glioblastoma stem cells. The use of established cell lines as a surrogate for drug response in CSC lines may enable the large-scale virtual screening of drug candidates that would otherwise be difficult or impossible to test directly in CSCs.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Expressão Gênica/genética , Glioblastoma/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Mapeamento Cromossômico/métodos , Simulação por Computador , Ensaios de Seleção de Medicamentos Antitumorais , Reações Falso-Positivas , Glioblastoma/patologia , Ensaios de Triagem em Larga Escala , Humanos , Valor Preditivo dos Testes , RNA Neoplásico/genética
2.
Acta Neuropathol Commun ; 12(1): 46, 2024 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-38528608

RESUMO

Cerebral organoids co-cultured with patient derived glioma stem cells (GLICOs) are an experimentally tractable research tool useful for investigating the role of the human brain tumor microenvironment in glioblastoma. Here we describe long-term GLICOs, a novel model in which COs are grown from embryonic stem cell cultures containing low levels of GSCs and tumor development is monitored over extended durations (ltGLICOs). Single-cell profiling of ltGLICOs revealed an unexpectedly long latency period prior to GSC expansion, and that normal organoid development was unimpaired by the presence of low numbers of GSCs. However, as organoids age they experience chronic hypoxia and oxidative stress which remodels the tumor microenvironment to promote GSC expansion. Receptor-ligand modelling identified astrocytes, which secreted various pro-tumorigenic ligands including FGF1, as the primary cell type for GSC crosstalk and single-cell multi-omic analysis revealed these astrocytes were under the control of ischemic regulatory networks. Functional validation confirmed hypoxia as a driver of pro-tumorigenic astrocytic ligand secretion and that GSC expansion was accelerated by pharmacological induction of oxidative stress. When controlled for genotype, the close association between glioma aggressiveness and patient age has very few proposed biological explanations. Our findings indicate that age-associated increases in cerebral vascular insufficiency and associated regional chronic cerebral hypoxia may contribute to this phenomenon.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Glioma , Humanos , Neoplasias Encefálicas/patologia , Microambiente Tumoral , Ligantes , Células-Tronco Neoplásicas/metabolismo , Glioma/patologia , Glioblastoma/patologia , Hipóxia/metabolismo , Linhagem Celular Tumoral
3.
Nat Med ; 3(4): 437-42, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9095178

RESUMO

Chronic systemic delivery of therapeutic proteins, such as inhibitors of angiogenesis, present a number of difficult pharmacological challenges. To overcome these problems for one such protein, we constructed retroviral and adenoviral vectors that express a novel, secretable form of the antiangiogenic protein, platelet factor 4 (sPF4). Vector-mediated sPF4 transduction selectively inhibits endothelial cell proliferation in vitro, and results in hypovascular tumors that grow slowly in vivo. Additionally, tumor-associated angiogenesis is inhibited and animal survival is prolonged, following transduction of established intracerebral gliomas by an sPF4-expressing adenoviral vector. These data support the concept that targeted antiangiogenesis, using virally mediated gene transfer, represents a promising strategy for delivering antiangiogenic therapy.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Terapia Genética/métodos , Glioma/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Fator Plaquetário 4/uso terapêutico , Adenoviridae/genética , Animais , Neoplasias Encefálicas/mortalidade , Vetores Genéticos , Glioblastoma/tratamento farmacológico , Glioblastoma/mortalidade , Glioma/mortalidade , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Fator Plaquetário 4/genética , Ratos , Retroviridae/genética , Análise de Sobrevida , Transplante Heterólogo , Células Tumorais Cultivadas
4.
Nat Med ; 3(10): 1145-9, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9334729

RESUMO

Recent data suggest that many tumors, such as malignant gliomas, have disrupted pRB function, either because of RB-1 gene mutations or as a result of mutations affecting upstream regulators of pRB such as cyclin D1 or p16/INK4a/MTS1 (ref. 1-5). Tumor suppression by pRB has been linked to its ability to repress E2F-responsive promoters such as the E2F-1 promoter. Thus, a prediction, which has not yet been demonstrated experimentally in vivo, is that E2F-responsive promoters should be more active in tumor cells relative to normal cells because of an excess of "free" E2F and loss of pRB/E2F repressor complexes. We demonstrate that adenoviral vectors that contain transgenes driven by the E2F-1 promoter can mediate tumor-selective gene expression in vivo, allowing for eradication of established gliomas with significantly less normal tissue toxicity than seen with standard adenoviral vectors. Our data indicate that de-repression of the E2F-1 promoter occurs in cancer cells in vivo, a finding that can be exploited to design viral vectors that mediate tumor-selective gene expression.


Assuntos
Adenovírus Humanos , Astrocitoma/patologia , Neoplasias Encefálicas/patologia , Proteínas de Transporte , Vetores Genéticos , Glioma/patologia , Fatores de Transcrição/biossíntese , Transfecção/métodos , Animais , Encéfalo/patologia , Neoplasias Encefálicas/terapia , Ciclo Celular , Proteínas de Ciclo Celular/biossíntese , Proteínas de Ligação a DNA/biossíntese , Fatores de Transcrição E2F , Fator de Transcrição E2F1 , Terapia Genética , Glioma/terapia , Regiões Promotoras Genéticas , Ratos , Ratos Sprague-Dawley , Proteína 1 de Ligação ao Retinoblastoma , Fator de Transcrição DP1 , Fatores de Transcrição/genética , Transcrição Gênica , Células Tumorais Cultivadas , beta-Galactosidase/biossíntese
5.
Nat Med ; 2(5): 567-73, 1996 May.
Artigo em Inglês | MEDLINE | ID: mdl-8616717

RESUMO

Cytosine arabinoside (ara-C) is a cytidine analog that incorporates into replicating DNA and induces lethal DNA strand breaks. Although ara-C is a potent antitumor agent for hematologic malignancies, it has only minimal activity against most solid tumors. The rate-limiting step in intracellular ara-C activation is phosphorylation of the prodrug by deoxycytidine kinase (dCK). The present results demonstrate that both retroviral and adenoviral vector-mediated transduction of the dCK cDNA results in marked sensitization of glioma cells lines to the cytotoxic effects of ara-C in vitro. We also demonstrate that ara-C treatment of established intradermal and intracerebral gliomas transduced with dCK results in significant antitumor effects in vivo. These data suggest that viral vector transduction of the dCK gene followed by treatment with ara-C represents a new chemosensitization strategy for cancer gene therapy.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Citarabina/farmacologia , Desoxicitidina Quinase/metabolismo , Gliossarcoma/tratamento farmacológico , Pró-Fármacos/farmacologia , Adenoviridae/genética , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Citarabina/metabolismo , Desoxicitidina Quinase/genética , Relação Dose-Resposta a Droga , Vetores Genéticos , Gliossarcoma/genética , Gliossarcoma/metabolismo , Humanos , Masculino , Pró-Fármacos/metabolismo , Ratos , Ratos Endogâmicos F344 , Sarcoma Experimental/tratamento farmacológico , Sarcoma Experimental/genética , Sarcoma Experimental/metabolismo , Análise de Sobrevida , Transfecção
6.
Carcinogenesis ; 28(12): 2543-7, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17916900

RESUMO

A role of immunological factors in glioma etiology is suggested by reports of an inverse relationship with history of allergy or autoimmune disease. To test whether single-nucleotide polymorphisms (SNPs) in cytokine genes were related to risk of adult glioma, we genotyped 11 SNPs in seven cytokine genes within a hospital-based study conducted by the National Cancer Institute and an independent, population-based study by the National Institute for Occupational Safety and Health (overall 756 cases and 1190 controls with blood samples). The IL4 (rs2243248, -1098T>G) and IL6 (rs1800795, -174G>C) polymorphisms were significantly associated with risk of glioma in the pooled analysis (P trend = 0.006 and 0.04, respectively), although these became attenuated after controlling for the false discovery rate (P trend = 0.07 and 0.22, respectively). Our results underscore the importance of pooled analyses in genetic association studies and suggest that SNPs in cytokine genes may influence susceptibility to glioma.


Assuntos
Neoplasias Encefálicas/genética , Citocinas/genética , Predisposição Genética para Doença , Glioma/genética , Polimorfismo de Nucleotídeo Único , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Feminino , Marcadores Genéticos , Humanos , Masculino , Pessoa de Meia-Idade , Risco
7.
J Clin Invest ; 96(6): 2775-82, 1995 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8675647

RESUMO

The high molecular weight mucin-like glycoprotein, DF3 (MUC1), is overexpressed in the majority of human breast cancers. Here we demonstrate that replication defective recombinant adenoviral vectors, containing the DF3 promoter (bp -725 to +31), can be used to express beta-galactosidase (Ad.DF3-betagal) and the herpes simplex virus thymidine kinase (HSV-tk) gene (Ad.Df3-tk) in DF3 positive breast carcinoma cell lines. In vivo experiments using breast tumor implants in nude mice injected with Ad.DF3-betagal demonstrated that expression of the beta-galactosidase gene is limited to DF3-positive breast cancer xenografts. Moreover, in an intraperitoneal breast cancer metastases model, we show that i.p. injection of Ad.DF3-tk followed by GCV treatment results in inhibition of tumor growth. These results demonstrate that utilization of the DF3 promoter in an adenoviral vector can confer selective expression of heterologous genes in breast cancer cells in vitro and in vivo.


Assuntos
Adenovírus Humanos , Neoplasias da Mama/metabolismo , Neoplasias da Mama/terapia , Mucina-1/biossíntese , Mucina-1/genética , Proteínas de Neoplasias/biossíntese , Regiões Promotoras Genéticas , Animais , Sequência de Bases , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular , Primers do DNA , Feminino , Terapia Genética , Vetores Genéticos , Humanos , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Proteínas de Neoplasias/genética , Neoplasias Peritoneais/patologia , Neoplasias Peritoneais/secundário , Neoplasias Peritoneais/terapia , Reação em Cadeia da Polimerase , Proteínas Recombinantes/biossíntese , Simplexvirus/enzimologia , Simplexvirus/genética , Timidina Quinase/biossíntese , Transfecção , Transplante Heterólogo , Células Tumorais Cultivadas , beta-Galactosidase/biossíntese
8.
J Clin Invest ; 98(11): 2539-48, 1996 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-8958216

RESUMO

Tumor contamination of bone marrow (BM) and peripheral blood (PB) may affect the outcome of patients receiving high dose chemotherapy with autologous transplantation of hematopoietic stem cell products. In this report, we demonstrate that replication defective adenoviral vectors containing the cytomegalovirus (CMV) or DF3/MUC1 carcinoma-selective promoter can be used to selectively transduce contaminating carcinoma cells. Adenoviral-mediated reporter gene expression in breast cancer cells was five orders of magnitude higher than that found in BM, PB, and CD34+ cells. Our results demonstrate that CD34+ cells have low to undetectable levels of integrins responsible for adenoviral internalization. We show that adenoviral-mediated transduction of a reporter gene can detect one breast cancer cell in 5 x 10(5) BM or PB cells with a vector containing the DF3/MUC1 promoter. We also show that transduction of the HSV-tk gene for selective killing by ganciclovir can be exploited for purging cancer cells from hematopoietic stem cell populations. The selective expression of TK followed by ganciclovir treatment resulted in the elimination of 6-logs of contaminating cancer cells. By contrast, there was little effect on CFU-GM and BFU-E formulation or on long term culture initiating cells. These results indicate that adenoviral vectors with a tumor-selective promoter provide a highly efficient and effective approach for the detection and purging of carcinoma cells in hematopoietic stem cell preparations.


Assuntos
Medula Óssea/patologia , Células-Tronco Hematopoéticas/citologia , Adenovírus Humanos , Antígenos CD/análise , Antígenos CD34/análise , Células da Medula Óssea , Purging da Medula Óssea , Neoplasias da Mama , Linhagem Celular , Ensaio de Unidades Formadoras de Colônias , Feminino , Citometria de Fluxo , Genes Reporter , Glioblastoma , Humanos , Neoplasias Pulmonares , Masculino , Neoplasias Ovarianas , Neoplasias da Próstata , Células Tumorais Cultivadas
9.
J Natl Cancer Inst ; 87(1): 34-40, 1995 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-7666461

RESUMO

BACKGROUND: The spread of systemic cancer to the brain is a common complication for cancer patients. Conventional radiotherapy offers modest palliation, and surgery is helpful only for the patient with a single metastasis in an accessible location. Stereotactic radiosurgery, a technique that permits the precise delivery of a high dose of radiation to a small intracranial target while sparing the surrounding normal brain, has been used as an alternative treatment for brain metastases. PURPOSE: Our medical center's 7-year experience with radiosurgery for metastases was reviewed to establish the effectiveness of the treatment and to understand the prognoses in patients so treated. METHODS: Retrospective analysis of hospital records, from 248 consecutive patients (421 lesions) that were treated with radiosurgery between May 1986 and May 1993, was performed. Patients were only excluded for a Karnofsky performance score of less than 70, evidence of acute neurologic deterioration, or tumor diameter more than 4 cm. Median follow-up was 26.2 months. Seventy-six percent of patients had recurrent disease, 69% had evidence of systemic disease, 69% had a single metastasis. Treatment was performed using a 6-MeV linear accelerator. The median tumor volume was 3 cm3. The median treatment dose was 1500 cGy. Whole brain radiotherapy was given to all newly diagnosed patients. Patients were followed by neurological examination and neuroimaging at regular intervals. Local control of disease was defined as a lack of progression of solid-contrast enhancement on computed tomography scan or magnetic resonance imaging. RESULTS: Median overall survival from radiosurgery was 9.4 months. The absence of active systemic disease, younger than 60 years of age, two or fewer lesions, and female sex were significantly associated with increased survival (two-sided P < .05). Actuarial local control rates were approximately 85% at 1 year and 65% at 2 years. Factors associated with a significantly decreased local control rate were location below the tentorium, recurrent tumor, and larger tumor volume (two-sided P < .05). Radioresponsive and radioresistant tumor types had similar control rates. The median drop in Karnofsky performance score at 1 year was 10%. CONCLUSIONS: The results of this retrospective analysis show that radiosurgery is an effective, minimally invasive outpatient treatment option for small intracranial metastases. Results of this study also indicate that radiosurgery not only provides local control rates equivalent to those from surgical series but is also effective in treating patients with surgically inaccessible lesions, with multiple lesions, or with tumor types that are resistant to conventional treatment.


Assuntos
Neoplasias Encefálicas/secundário , Neoplasias Encefálicas/cirurgia , Radiocirurgia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Análise de Variância , Neoplasias Encefálicas/radioterapia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Dosagem Radioterapêutica , Radioterapia Adjuvante , Estudos Retrospectivos , Análise de Sobrevida , Resultado do Tratamento
10.
J Natl Cancer Inst ; 82(24): 1918-21, 1990 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-2250312

RESUMO

Recent studies have shown a survival benefit for patients with recurrent glioblastomas treated with stereotactic brachytherapy. On the basis of these encouraging results, we began a prospective study in 1987 to evaluate the use of brachytherapy in patients with newly diagnosed glioblastoma. Patients were considered eligible for this study if they met the following criteria: Karnofsky performance status 70% or greater; tumor size not greater than 5 cm in any dimension; a radiographically well delineated, supratentorial lesion not involving the ependymal surfaces; and pathologically confirmed glioblastoma. We treated 35 such patients between 1987 and 1990 with stereotactic brachytherapy as part of their initial therapy. The treatment protocol involved surgery, partial brain external-beam radiotherapy (59.4 Gy in 33 fractions), and stereotactic brachytherapy with temporary high-activity iodine 125 sources giving an additional 50 Gy to the tumor bed. Chemotherapy was not used in the initial management of these 35 patients. To compare our results with those obtained in a matched control group, we identified 40 patients with glioblastoma treated with surgery and external radiotherapy, with or without chemotherapy, between 1977 and 1986 at our institution. These patients had clinical and radiographic characteristics that would have made them eligible for the brachytherapy protocol. Survival rates at 1 and 2 years after diagnosis were 87% and 57%, respectively, for patients receiving brachytherapy versus 40% and 12.5%, respectively, for the controls (P less than .001). We conclude that stereotactic brachytherapy improves the survival of patients with glioblastoma when it can be incorporated into the initial treatment approach. Unfortunately, only about one in four patients with glioblastoma are suitable candidates for brachytherapy at the time of initial presentation.


Assuntos
Braquiterapia , Glioma/radioterapia , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Feminino , Humanos , Radioisótopos do Iodo/uso terapêutico , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Estudos Retrospectivos , Análise de Sobrevida
11.
Cancer Res ; 58(15): 3362-9, 1998 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-9699667

RESUMO

Despite recent advances in neurosurgery, radiation, and chemotherapy, the prognosis of patients with malignant gliomas remains dismal. Based on the observation that solid tumor growth is angiogenic dependent, and gliomas are among the most angiogenic of all tumors, therapeutic strategies aimed at inhibiting angiogenesis are theoretically attractive. Angiostatin, an internal peptide fragment of plasminogen, has recently been shown to potently inhibit endothelial proliferation in vitro and tumor growth in vivo. Long-term systemic delivery of proteins, however, poses a number of difficult logistic and pharmacological problems and may not be necessary or optimal for treating locally aggressive tumors such as gliomas. We now demonstrate that retroviral and adenoviral vectors that transduce the angiostatin cDNA can be used to inhibit endothelial cell growth in vitro and angiogenesis in vivo. Vector-mediated inhibition of tumor-associated angiogenesis results in increased apoptotic tumor cell death, leading to inhibition of tumor growth. These studies support a potential role of vector-mediated transduction of the cDNA encoding angiostatin as a potential novel therapeutic strategy for the treatment of malignant brain tumors and confirm the antitumor activity of angiostatin and the concept of dormancy therapy.


Assuntos
Neoplasias Encefálicas/irrigação sanguínea , DNA Complementar/genética , Terapia Genética/métodos , Glioblastoma/irrigação sanguínea , Glioma/irrigação sanguínea , Neovascularização Patológica/terapia , Fragmentos de Peptídeos/genética , Plasminogênio/genética , Adenoviridae/genética , Adenoviridae/metabolismo , Angiostatinas , Animais , Northern Blotting , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Divisão Celular/fisiologia , DNA Complementar/metabolismo , Vetores Genéticos , Glioblastoma/patologia , Glioblastoma/terapia , Glioma/patologia , Glioma/terapia , Humanos , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Fragmentos de Peptídeos/biossíntese , Plasminogênio/biossíntese , Ratos , Ratos Endogâmicos F344 , Transfecção
12.
Cancer Res ; 54(20): 5408-13, 1994 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-7923173

RESUMO

A potentially novel therapeutic strategy for breast cancer treatment involves sensitization of tumor cells to chemotherapy through gene transfer. Clinical application of this approach, however, may be limited by the lack of target cell specificity of currently available gene delivery techniques. Development of vectors with tumor-selective gene expression could overcome this problem. The DF3/MUC1 gene encodes a high molecular weight mucin-like glycoprotein which is overexpressed at the transcriptional level in the majority of human breast cancers. To develop a breast tumor-selective enhancer, we cloned the upstream region of the DF3 gene and have identified a 114-base pair enhancer region that can modulate transcription from heterologous promoters. The present studies demonstrate that the DF3 enhancer sequences can direct selective gene expression in DF3-positive breast carcinoma cells. DF3-positive breast carcinoma cell lines transfected with herpes simplex virus thymidine kinase gene expression cassettes modified by the DF3 enhancer were markedly more sensitive to killing by ganciclovir than were the same cells transfected with the expression cassettes lacking the DF3 enhancer. DF3-negative cell lines transfected with the DF3 enhancer constructs, however, were no more sensitive to ganciclovir than were cells treated with the unmodified expression plasmids. Consistent with an innocent bystander effect, nontransfected human breast carcinoma cells were susceptible in a cell density-dependent manner to ganciclovir-induced cell killing when adjacent to transfected cells. The results also demonstrate that the DF3 enhancer sequences can be effectively incorporated into a retroviral vector to mediate selective gene expression following retroviral infection. These findings suggest that the DF3 promoter/enhancer may be useful for incorporation into vectors designed for gene therapy of breast cancer.


Assuntos
Neoplasias da Mama/genética , Elementos Facilitadores Genéticos/fisiologia , Ganciclovir/farmacologia , Regulação Neoplásica da Expressão Gênica , Simplexvirus/genética , Timidina Quinase/genética , Sequência de Bases , Neoplasias da Mama/tratamento farmacológico , Contagem de Células , Ensaios de Seleção de Medicamentos Antitumorais , Vetores Genéticos , Humanos , Dados de Sequência Molecular , Transfecção
13.
J Clin Oncol ; 8(4): 576-82, 1990 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-2179476

RESUMO

Between May 1986 and August 1989, we treated 18 patients with 21 recurrent or persistent brain metastases with stereotactic radiosurgery using a modified linear accelerator. To be eligible for radiosurgery, patients had to have a performance status of greater than or equal to 70% and have no evidence of (or stable) systemic disease. All but one patient had received prior radiotherapy, and were treated with stereotactic radiosurgery at the time of recurrence. Polar lesions were treated only if the patient had undergone and failed previous complete surgical resection (10 patients). Single doses of radiation (900 to 2,500 cGy) were delivered to limited volumes (less than 27 cm3) using a modified 6MV linear accelerator. The most common histology of the metastatic lesion was carcinoma of the lung (seven patients), followed by carcinoma of the breast (four patients), and melanoma (four patients). With median follow-up of 9 months (range, 1 to 39), all tumors have been controlled in the radiosurgery field. Two patients failed in the immediate margin of the treated volume and were subsequently treated with surgery and implantation of 125I to control the disease. Radiographic response was dramatic and rapid in the patients with adenocarcinoma, while slight reduction and stabilization occurred in those patients with melanoma, renal cell carcinoma, and sarcoma. The majority of patients improved neurologically following treatment, and were able to be withdrawn from corticosteroid therapy. Complications were limited and transient in nature and no cases of symptomatic radiation necrosis occurred in any patient despite previous exposure to radiotherapy. Stereotactic radiosurgery is an effective and relatively safe treatment for recurrent solitary metastases and is an appealing technique for the initial management of deep-seated lesions as a boost to whole brain radiotherapy.


Assuntos
Neoplasias Encefálicas/radioterapia , Recidiva Local de Neoplasia/radioterapia , Adenocarcinoma/radioterapia , Adulto , Idoso , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/secundário , Carcinoma/radioterapia , Seguimentos , Humanos , Incidência , Melanoma/radioterapia , Pessoa de Meia-Idade , Lesões por Radiação/epidemiologia , Indução de Remissão , Sarcoma/radioterapia , Técnicas Estereotáxicas , Tomografia Computadorizada por Raios X
14.
J Clin Oncol ; 10(9): 1379-85, 1992 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-1325539

RESUMO

PURPOSE: Between May 1988 and May 1991, 41 patients with malignant gliomas were enrolled onto a prospective study designed to evaluate the role of radiosurgery as a component of initial management. PATIENTS AND METHODS: Thirty-seven patients underwent radiosurgery according to the protocol and were assessable for survival and complications of treatment. Diagnoses included glioblastoma multiforme (GBM) in 23 (62%) cases and anaplastic astrocytoma in 14 (38%) cases. In 20 (54%) cases, surgical resection was attempted initially, whereas 17 (46%) patients underwent biopsy only. Patients in the study group received external-beam radiotherapy that consisted of 5,940 cGy given in 33 fractions to partial brain fields that encompassed the primary tumor with a 3 to 4 cm margin. Radiosurgery, used as a technique for boosting the dose to any residual contrast-enhancing mass lesion, was given 2 to 4 weeks after the completion of conventional radiotherapy. Minimum radiosurgical doses ranged from 1,000 to 2,000 cGy (median, 1,200 cGy), whereas maximum doses ranged from 1,250 to 2,500 cGy (median, 1,500 cGy). The median tumor volume at the time of radiosurgery was 4.8 cm3 (range, 1.2 to 72 cm3). Adjuvant chemotherapy was not given. RESULTS: After a median follow-up of 19 months, only nine of 37 (24%) patients have died. Six patients (all glioblastoma multiforme) died of recurrent tumor, whereas death was attributable to complications of treatment in two cases and intercurrent disease in one case. Four patients with recurrent tumor failed at the margins of the radiosurgical treatment volume, whereas two patients progressed locally. One patient is alive with local and marginal failure. Seven (19%) patients underwent reoperation at a median time of 5 months (range, 1 to 14 months) after radiosurgery. CONCLUSION: We conclude that radiosurgery is a useful adjunct to other modalities in the initial management of patients with small, radiographically well-defined malignant gliomas.


Assuntos
Neoplasias Encefálicas/cirurgia , Glioma/cirurgia , Radiocirurgia , Análise Atuarial , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Astrocitoma/cirurgia , Braquiterapia , Neoplasias Encefálicas/radioterapia , Terapia Combinada , Feminino , Glioblastoma/cirurgia , Glioma/radioterapia , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Análise de Sobrevida , Resultado do Tratamento
15.
J Clin Oncol ; 18(4): 708-15, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10673511

RESUMO

PURPOSE: Little progress has been made in the treatment of adult high-grade gliomas over the last two decades, thus necessitating a search for novel therapeutic strategies. Malignant gliomas are vascular or angiogenic tumors, which leads to the supposition that angiogenesis inhibition may represent a potentially promising strategy in the treatment of these tumors. We present the results of a phase II trial of thalidomide, a putative inhibitor of angiogenesis, in the treatment of adults with previously irradiated, recurrent high-grade gliomas. PATIENTS AND METHODS: Patients with a histologic diagnosis of anaplastic mixed glioma, anaplastic astrocytoma, or glioblastoma multiforme who had radiographic demonstration of tumor progression after standard external-beam radiotherapy with or without chemotherapy were eligible. Patients were initially treated with thalidomide 800 mg/d with increases in dose by 200 mg/d every 2 weeks until a final daily dose of 1,200 mg was achieved. Patients were evaluated every 8 weeks for response by both clinical and radiographic criteria. RESULTS: A total of 39 patients were accrued, with 36 patients being assessable for both toxicity and response. Thalidomide was well tolerated, with constipation and sedation being the major toxicities. One patient developed a grade 2 peripheral neuropathy after treatment with thalidomide for nearly a year. There were two objective radiographic partial responses (6%), two minor responses (6%), and 12 patients with stable disease (33%). Eight patients were alive more than 1 year after starting thalidomide, although almost all with tumor progression. Changes in serum levels of basic fibroblastic growth factor (bFGF) were correlated with time to tumor progression and overall survival. CONCLUSION: Thalidomide is a generally well-tolerated drug that may have antitumor activity in a minority of patients with recurrent high-grade gliomas. Future studies will better define the usefulness of thalidomide in newly diagnosed patients with malignant gliomas and in combination with radiotherapy and chemotherapy. Additionally, studies will be needed to confirm the potential utility of changes in serum bFGF as a marker of antiangiogenic activity and/or glioma growth.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Glioma/tratamento farmacológico , Recidiva Local de Neoplasia/tratamento farmacológico , Neoplasias Supratentoriais/tratamento farmacológico , Talidomida/uso terapêutico , Adulto , Idoso , Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/efeitos adversos , Biomarcadores Tumorais/sangue , Quimioterapia Adjuvante , Terapia Combinada , Constipação Intestinal/induzido quimicamente , Progressão da Doença , Feminino , Fator 2 de Crescimento de Fibroblastos/sangue , Seguimentos , Glioblastoma/tratamento farmacológico , Glioblastoma/radioterapia , Glioma/radioterapia , Humanos , Hipnóticos e Sedativos/efeitos adversos , Masculino , Pessoa de Meia-Idade , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Indução de Remissão , Neoplasias Supratentoriais/radioterapia , Taxa de Sobrevida , Talidomida/administração & dosagem , Talidomida/efeitos adversos
16.
Clin Cancer Res ; 3(3): 381-7, 1997 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9815695

RESUMO

Primary brain tumors represent an important cause of cancer-related morbidity and mortality in the United States. Despite advances in neurosurgery and radiotherapy, the median survival of patients with malignant gliomas remains less than 1 year. A contributing factor to the poor prognoses of these patients is the diffuse, infiltrative nature of these tumors, which limits the effectiveness of focal therapies (i.e., surgery and radiation). Unfortunately, standard chemotherapy has been of limited benefit in the treatment of malignant gliomas, underlying the necessity for new drugs with novel mechanisms of action. On the basis of promising in vitro and clinical data demonstrating significant antiglioma activity of purified IFN-beta and a synthetic IFN-beta (Betaseron), we conducted a Phase I trial of a new, nonmutated, glycosylated recombinant human IFN-beta (BG9015) in patients with recurrent, high-grade astrocytomas. In this trial, we demonstrate that the maximally tolerated dose of BG9015 is 6 million units/m2 delivered by intramuscular injection three times per week. Dose-limiting neurotoxicity was seen in both patients treated at 8 million units/m2. Additionally, we demonstrate that high BG9015 serum levels are associated with a fall in natural killer cell number, radiographic response, and prolonged survival. We conclude that BG9015 has activity in patients with malignant gliomas, although the therapeutic index may be narrow. Future studies will be needed to confirm the observation that natural killer cell number and activity as well as BG9015 serum levels are important markers of antitumor activity.


Assuntos
Astrocitoma/terapia , Neoplasias Encefálicas/terapia , Glioma/terapia , Interferon beta/efeitos adversos , Adulto , Idoso , Astrocitoma/mortalidade , Neoplasias Encefálicas/mortalidade , Intervalo Livre de Doença , Relação Dose-Resposta a Droga , Feminino , Glioblastoma/mortalidade , Glioblastoma/terapia , Glioma/mortalidade , Glicosilação , Humanos , Interferon beta/farmacocinética , Linfócitos/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/farmacocinética , Recidiva , Taxa de Sobrevida
17.
Hum Gene Ther ; 12(14): 1713-29, 2001 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-11560766

RESUMO

Malignant ascites is a common complication of advanced intraabdominal neoplasms for which standard treatments are suboptimal. Evidence suggests that tumor-mediated angiogenesis and enhanced vascular permeability in the peritoneal wall due to high levels of vascular endothelial growth factor play a fundamental role in the pathogenesis of malignant ascites. To explore the advantage of viral vector-mediated "targeted antiangiogenic therapy" in ascites formation, we constructed and administered adenoviral vectors encoding several different antiangiogenic proteins (angiostatin, endostatin, platelet factor 4, and a fusion protein between angiostatin and endostatin) alone or in combination intraperitoneally in mice with peritoneal carcinomatosis from breast cancer (TA3 cells) and ovarian cancer (SKOV-3 i.p. and ES-2 cell lines) to explore the potential of additive or synergistic activity. Our data demonstrated statistically significant downregulation of ascites formation, tumor growth, vascularity, and prolongation of animal survival after intraperitoneal treatment with antiangiogenic adenoviral vectors in three different ascites tumor models. Combined treatment proved to be more effective than treatment with one vector alone. Reduced ascites formation was accompanied by decreased microvascular density in the peritoneal wall and increased apoptosis of tumor cells after administration of antiangiogenic vectors in vivo. Of interest was the observation that AdPF4 caused a significant decrease in the level of VEGF secreted by tumor cells both in vitro and in TA3 ascites tumor-bearing animals in vivo. These data suggest that adenoviral vector-mediated delivery of genes encoding antiangiogenic proteins may represent a potentially new treatment modality for malignant ascites.


Assuntos
Ascite/terapia , Técnicas de Transferência de Genes , Vetores Genéticos , Neovascularização Patológica/genética , Adenoviridae/genética , Angiostatinas , Animais , Apoptose , Northern Blotting , Western Blotting , Neoplasias da Mama/terapia , Divisão Celular , Movimento Celular , Células Cultivadas , Colágeno/genética , Corantes/farmacologia , Regulação para Baixo , Endostatinas , Fatores de Crescimento Endotelial/genética , Endotélio/citologia , Endotélio Vascular/citologia , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Imuno-Histoquímica , Linfocinas/genética , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Fragmentos de Peptídeos/genética , Plasmídeos/metabolismo , Plasminogênio/genética , Fator Plaquetário 4/genética , Proteínas Recombinantes de Fusão/genética , Transdução de Sinais , Sais de Tetrazólio/farmacologia , Tiazóis/farmacologia , Células Tumorais Cultivadas , Veias Umbilicais/citologia , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
18.
Hum Gene Ther ; 7(6): 713-20, 1996 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-8919593

RESUMO

Viral vector-mediated transfer of chemosensitization genes represents a promising new approach to the treatment of cancer. Previous reports have demonstrated that transfection of the bacterial cytosine deaminase (cd) gene into mammalian cells can sensitize them to the otherwise nontoxic nucleoside, 5-fluorocytosine (5-FC). We now report that a replication-deficient adenovirus vector that transduces the cd gene (Ad.CMV-cd) highly sensitizes 9L gliosarcoma cells to 5-FC, and that gene transduction is associated with a potent bystander effect that is not dependent on direct cell-to-cell contact. Stereotactic injection of Ad.CMV-cd into established rat gliomas, followed by systemic administration of 5-FC in vivo, results in prolongation of survival.


Assuntos
Adenovírus Humanos/genética , Vírus Defeituosos/genética , Flucitosina/farmacologia , Vetores Genéticos/genética , Gliossarcoma/terapia , Nucleosídeo Desaminases/metabolismo , Transdução Genética , Adenovírus Humanos/fisiologia , Animais , Divisão Celular , Linhagem Celular Transformada , Citosina Desaminase , Vírus Defeituosos/fisiologia , Gliossarcoma/patologia , Humanos , Masculino , Nucleosídeo Desaminases/genética , Ratos , Ratos Endogâmicos F344 , Replicação Viral
19.
J Neuropathol Exp Neurol ; 59(12): 1044-50, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11138924

RESUMO

Glioblastomas only rarely metastasize to sites outside the central nervous system, for reasons that are poorly understood. We report the clinicopathological and molecular genetic findings in 6 patients with metastatic glioblastoma. Four patients were under the age of 32 and all but 1 patient died within 2 yr of diagnosis. The number of metastases ranged from 1 to 3. At the time of death, 3 patients had apparent tumor control at their primary site. We evaluated DNA from both primary and metastatic glioblastomas for genetic alterations commonly found in glioblastomas: TP53 mutations, CDKN2A/p16 deletions, EGFR amplification, and allelic loss of chromosomes 1p, 10q and 19q. Four of 6 cases had TP53 mutations and only single cases had EGFR amplification, CDKN2A/p16 deletions, or allelic loss of 1p, 10q and 19q; 2 cases had no detectable genetic alterations. In 2 cases, the primary and metastatic tumors had identical genotypes. Remarkably, however, 2 cases had different TP53 alterations in the primary and metastatic lesions, or among the metastatic tumors, which suggests that some metastatic deposits may represent emergence of subclones that were not necessarily dominant in the primary tumor. The present observations and a review of the recent literature demonstrate that metastatic glioblastomas tend to occur in younger adults who do not follow long clinical courses, and may be characterized by TP53 mutations and differential clonal selection.


Assuntos
Glioblastoma/patologia , Glioblastoma/secundário , Adulto , DNA de Neoplasias/genética , Evolução Fatal , Genes erbB-1/genética , Genes p16/genética , Genes p53/genética , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Mutação/genética
20.
Neurology ; 43(6): 1111-4, 1993 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8170553

RESUMO

Venous thromboembolic disease is a frequent complication in patients with intracranial malignancies. Because these patients are often perceived to be at increased risk of intracranial hemorrhage with anticoagulation, inferior vena cava (IVC) filters are frequently used in their treatment. We reviewed the records of 49 patients with intracranial malignancies and venous thromboembolic disease to determine the effectiveness of, and the complications resulting from, treatment. Of the 42 patients receiving IVC filters, a strikingly high percentage (62%) developed complications. Twelve percent developed recurrent pulmonary embolism, while 57% developed either IVC or filter thrombosis, recurrent deep venous thrombosis, or post-phlebitic syndrome. These complications severely reduced the quality of life of the affected patients. Only 15 of our patients were treated with anticoagulation, and seven of these received it because of continued thromboembolic disease. None of these 15 patients had proven hemorrhagic complications. This study suggests that the complication rate of IVC filters in patients with brain tumors is higher than commonly perceived and may outweigh the risk of anticoagulation.


Assuntos
Neoplasias Encefálicas/complicações , Tromboembolia/terapia , Anticoagulantes/uso terapêutico , Hemorragia Cerebral/prevenção & controle , Seguimentos , Humanos , Embolia Pulmonar/etiologia , Qualidade de Vida , Estudos Retrospectivos , Tromboembolia/etiologia , Resultado do Tratamento , Filtros de Veia Cava/efeitos adversos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA