Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 126
Filtrar
1.
Nat Med ; 7(7): 781-7, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11433341

RESUMO

In the search for novel cancer therapies that can be used in conjunction with existing treatments, one promising area of research is the use of viral vectors and whole viruses. This review describes the underlying biological principles and current status of the field, outlines approaches for improving clinical effectiveness and discusses the unique safety and regulatory issues surrounding viral therapies.


Assuntos
Terapia Biológica , Neoplasias/terapia , Replicação Viral , Vírus , Humanos , Regiões Promotoras Genéticas , Gestão de Riscos , Replicação Viral/genética
2.
Nat Med ; 1(9): 938-43, 1995 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-7585221

RESUMO

We have created a double mutant of the herpes simplex virus (HSV) type 1 (termed G207) with favourable properties for treating human malignant brain tumours: replication-competence in glioblastoma cells (and other dividing cells), attenuated neurovirulence, temperature sensitivity, ganciclovir hypersensitivity, and the presence of an easily detectable histochemical marker. G207 has deletions at both gamma 34.5 (RL1) loci and a lacZ gene insertion inactivating the ICP6 gene (UL39). G207 kills human glioma cells in monolayer cultures. In nude mice harbouring subcutaneous or intracerebral U-87MG gliomas, intraneoplastic inoculation with G207 causes decreased tumour growth and/or prolonged survival. G207 is avirulent upon intracerebral inoculation of mice and HSV-sensitive non-human primates. These results suggest that G207 should be considered for clinical evaluation in the treatment of glioblastomas.


Assuntos
Neoplasias Encefálicas/terapia , Vírus Defeituosos/fisiologia , Terapia Genética , Glioblastoma/terapia , Simplexvirus/fisiologia , Animais , Aotidae , Chlorocebus aethiops , Efeito Citopatogênico Viral , Vírus Defeituosos/efeitos dos fármacos , Vírus Defeituosos/genética , Vírus Defeituosos/patogenicidade , Ganciclovir/farmacologia , Genes Reporter , Humanos , Injeções Intralesionais , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Ratos , Segurança , Simplexvirus/efeitos dos fármacos , Simplexvirus/genética , Simplexvirus/patogenicidade , Temperatura , Células Vero , Proteínas Virais/genética , Virulência/genética , Latência Viral , Replicação Viral , beta-Galactosidase/genética
3.
Gene Ther ; 17(6): 805-10, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20220784

RESUMO

Prostate cancer is one of the most prevalent cancers in men. Replication-competent oncolytic herpes simplex virus (oHSV) vectors are a powerful antitumor therapy that can exert at least two effects: direct cytocidal activity that selectively kills cancer cells and induction of antitumor immunity. In addition, oHSV vectors can also function as a platform to deliver transgenes of interest. In these studies, we have examined the expression of a xenogeneic homologue of the prostate cancer antigen, prostatic acid phosphatase (PAP), with the goal of enhancing virotherapy against PAP-expressing tumors. PAP has already been used for cancer vaccination in patients with prostate cancer. Here we show that treatment with oHSV bPDelta6 expressing xenogeneic human PAP (hPAP) significantly reduces tumor growth and increases survival of C57/BL6 mice bearing mouse TRAMP-C2 prostate tumors, whereas expression of syngeneic mouse PAP (mPAP) from the same oHSV vector did not enhance antitumor activity. Treatment of mice bearing metastatic TRAMP-C2 lung tumors with oHSV-expressing hPAP resulted in fewer tumor nodules. To our knowledge, this is the first report of oncolytic viruses being used to express xenoantigens. These data lend support to the concept of combining oncolytic and immunogenic therapies as a way to improve therapy of metastatic prostate cancer.


Assuntos
Terapia Viral Oncolítica/métodos , Neoplasias da Próstata/terapia , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/imunologia , Simplexvirus/genética , Fosfatase Ácida , Animais , Citotoxicidade Imunológica , Técnicas de Transferência de Genes , Imunização , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Vírus Oncolíticos/genética , Transgenes
4.
Gene Ther ; 16(12): 1477-82, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19693098

RESUMO

Oncolytic herpes simplex viruses type 1 (oHSVs) such as G47Delta and G207 are genetically engineered for selective replication competence in cancer cells. Several factors can influence the overall effectiveness of oHSV tropism, including HSV-1 receptor expression, extracellular matrix milieu and cellular permissiveness. We have taken advantage of human prostate organ cultures derived from radical prostatectomies to investigate oHSV tropism. In this study, we show that both G47Delta and G207 specifically replicate in epithelial cells of the prostatic glands but not in the surrounding stroma. In contrast, both the epithelial and stromal cell compartments were readily infected by wild-type HSV-1. Analysis of oHSV replication in prostate surgical specimens 3 days post infection showed that G47Delta generated approximately 30-fold more viral progeny than did G207. This correlated with the enhanced expression of G47Delta-derived glycoprotein gB protein levels as compared with G207. In benign prostate tissues, G207 and G47Delta titers were notably reduced, whereas strain F titers were maintained at similar levels compared with prostate cancer specimens. Overall, our results show that these oncolytic herpes vectors show both target specificity and replication competence in human prostate cancer specimens and point to the utility of using human prostate organ cultures in assessing oHSV tropism and cellular specificity.


Assuntos
Herpesvirus Humano 1/genética , Vírus Oncolíticos/genética , Técnicas de Cultura de Órgãos , Animais , Engenharia Genética , Vetores Genéticos , Humanos , Masculino , Glândulas Mamárias Humanas/virologia , Camundongos , Terapia Viral Oncolítica/métodos , Neoplasias da Próstata/genética , Neoplasias da Próstata/terapia
5.
Science ; 252(5007): 854-6, 1991 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-1851332

RESUMO

Malignant gliomas are the most common malignant brain tumors and are almost always fatal. A thymidine kinase-negative mutant of herpes simplex virus-1 (dlsptk) that is attenuated for neurovirulence was tested as a possible treatment for gliomas. In cell culture, dlsptk killed two long-term human glioma lines and three short-term human glioma cell populations. In nude mice with implanted subcutaneous and subrenal U87 human gliomas, intraneoplastic inoculation of dlsptk caused growth inhibition. In nude mice with intracranial U87 gliomas, intraneoplastic inoculation of dlsptk prolonged survival. Genetically engineered viruses such as dlsptk merit further evaluation as novel antineoplastic agents.


Assuntos
Neoplasias Encefálicas/terapia , Glioma/terapia , Simplexvirus/imunologia , Vacinas Virais/uso terapêutico , Animais , Antivirais/farmacologia , Células Cultivadas , Relação Dose-Resposta a Droga , Foscarnet , Camundongos , Camundongos Nus , Mutagênese , Ácido Fosfonoacéticos/análogos & derivados , Ácido Fosfonoacéticos/farmacologia , Simplexvirus/genética , Timidina Quinase/genética , Vidarabina/farmacologia
6.
Science ; 236(4799): 317-9, 1987 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-3105060

RESUMO

Bilateral acoustic neurofibromatosis (BANF) is a genetic defect associated with multiple tumors of neural crest origin. Specific loss of alleles from chromosome 22 was detected with polymorphic DNA markers in two acoustic neuromas, two neurofibromas, and one meningioma from BANF patients. This indicates a common pathogenetic mechanism for all three tumor types. The two neurofibromas were among three taken from the same patient, and both showed loss of identical alleles demonstrating that the same chromosome suffered deletion in both tumors. The third neurofibroma from this patient showed no detectable loss of heterozygosity, which suggests the possibility of a more subtle mutational event that affects chromosome 22. In the two acoustic neuromas, only a portion of chromosome 22 was deleted, narrowing the possible chromosomal location of the gene that causes BANF to the region distal to the D22S9 locus in band 22q11. The identification of progressively smaller deletions on chromosome 22 in these tumor types may well provide a means to clone and characterize the defect.


Assuntos
Cromossomos Humanos Par 22 , Neurofibromatose 1/genética , Neuroma Acústico/genética , Alelos , Mapeamento Cromossômico , Triagem de Portadores Genéticos , Humanos , Leucócitos/citologia , Neoplasias/genética
7.
Cancer Gene Ther ; 13(3): 253-65, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16179929

RESUMO

Replication competent oncolytic herpes simplex viruses (HSV) with broad-spectrum activity against various cancers, including prostate cancer, exert a dual effect by their direct cytocidal action and by eliciting tumor-specific immunity. These viruses can deliver immunoregulatory molecules to tumors so as to enhance the cumulative antitumor response. This is particularly desirable for prostate cancers, which are usually poorly immunogenic. Initial studies described herein comparing the efficacy of three different oncolytic HSVs (G207, G47Delta, and NV1023) to inhibit the growth of the poorly immunogenic TRAMP-C2 mouse prostate tumors demonstrated that NV1023 was most effective in treating established tumors. The expression of IL-12 on an NV1023 background (NV1042), but not the expression of GM-CSF (NV1034), further enhanced the efficacy of NV1023 in two murine prostate cancer models with highly variable MHC class I levels, Pr14-2 with 91% and TRAMP-C2 with 2% of cells staining. NV1042 also inhibited the growth of distant noninoculated tumors in both prostate cancer models. NV1042 treated tumors exhibited increased immune cell infiltration and decreased levels of angiogenesis. Thus, an IL-12 expressing oncolytic herpes virus, which is capable of direct cytotoxicity and can modulate the otherwise suboptimal immune response through concomitant expression of the cytokine at the site of tumor destruction, could serve as a valuable clinical agent to seek out both overt and occult prostate cancers.


Assuntos
Terapia Genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Herpesvirus Humano 1/imunologia , Interleucina-12/uso terapêutico , Terapia Viral Oncolítica , Neoplasias da Próstata/terapia , Simplexvirus/genética , Animais , Terapia Combinada , Genes MHC Classe I/fisiologia , Vetores Genéticos/uso terapêutico , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Herpesvirus Humano 1/genética , Humanos , Interferon gama/farmacologia , Interleucina-12/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Vírus Oncolíticos/patogenicidade , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/patologia , Células Tumorais Cultivadas , Replicação Viral
9.
Cancer Res ; 54(15): 3963-6, 1994 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-8033122

RESUMO

We have demonstrated that attenuated mutants of herpes simplex virus (HSV) have therapeutic potential for malignant brain tumors. In this report, we tested a ribonucleotide reductase-deficient (RR-) HSV mutant as an experimental treatment for malignant brain tumors. The HSV-RR- mutant hrR3, containing an Escherichia coli lacZ gene insertion in the ICP6 gene that encodes the large subunit of RR, was used in this study. We examined the cytopathic effect of hrR3 (0.1 plaque-forming unit/cell) on the U-87MG human glioblastoma cell line in vitro. Only 0.2% of U-87 cells were alive 67 h postinfection. Drug sensitivity assays demonstrated that hrR3 is hypersensitive to the antiherpetic agent ganciclovir. For in vivo studies, 10 animals harboring U-87MG tumors were randomly divided and treated intraneoplastically with either 5 x 10(6) plaque-forming units of hrR3 or medium alone. The viral treatment group showed significant inhibition of tumor growth (P < 0.01; one-sided Wilcoxon rank test). Expression of the lacZ gene in hrR3, visualized by 5-bromo-4-chrolo-3-indolyl-beta-D-galactopyranoside histochemistry, could be detected in treated tumors. The therapeutic potential of this HSV-RR- mutant for malignant gliomas is discussed.


Assuntos
Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Glioblastoma/terapia , Ribonucleotídeo Redutases/deficiência , Simplexvirus/genética , Animais , Efeito Citopatogênico Viral , Resistência a Medicamentos , Feminino , Ganciclovir/farmacologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mutação/genética , Distribuição Aleatória , Simplexvirus/efeitos dos fármacos , Simplexvirus/enzimologia , Células Tumorais Cultivadas , Células Vero , beta-Galactosidase/análise
10.
Cancer Res ; 50(21): 6783-6, 1990 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-2208143

RESUMO

Meningiomas and schwannomas are two of the most common tumors of the human nervous system. To determine whether these tumors arise from a single cell or from multiple cells, we used molecular genetic techniques to study X chromosome inactivation in meningiomas and schwannomas isolated from females including one who had neurofibromatosis type 2. The tumors were also screened for loss of heterozygosity at several loci on chromosome 22 using polymorphic DNA markers. Among nine meningiomas, at least three of which showed loss of alleles on chromosome 22 and five of which retained heterozygosity for the chromosome 22 alleles examined, all nine tumors were monoclonal. Among eight schwannomas, at least seven of which retained heterozygosity for chromosome 22 loci, seven were monoclonal. We conclude that human meningiomas and schwannomas arise from a single cell.


Assuntos
Neoplasias Meníngeas/genética , Meningioma/genética , Neurilemoma/genética , Cromossomos Humanos Par 22/fisiologia , Clonagem Molecular , DNA/sangue , DNA/genética , Feminino , Heterozigoto , Humanos , Leucócitos/fisiologia , Neoplasias Meníngeas/sangue , Meningioma/sangue , Neurilemoma/sangue
11.
Cancer Res ; 61(1): 153-61, 2001 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-11196154

RESUMO

In vivo delivery of immunomodulatory genes is a promising strategy for solid tumor vaccination. A drawback is that it necessitates induction of a large effect from transgene expression in a small percentage of tumor cells. Although the B7 family is known to be the most potent of the costimulatory molecules, gene transduction of B7 alone has not been effective in inducing antitumor immunity in nonimmunogenic tumors by ex vivo methods, much less in vivo. We have developed a novel approach where a gene encoding soluble B7-1, a fusion protein of the extracellular domain of murine B7-1 and the Fc portion of human IgG1, is delivered to tumor cells in vivo in the context of an oncolytic replication-competent herpes simplex virus, and the gene product is secreted by tumor cells rather than expressed on the cell surface. Defective herpes simplex virus vectors containing the B7-1-immunoglobulin (B7-1-Ig) fusion transgene (dvB7Ig) were generated using G207 as a helper virus and tested in the poorly immunogenic murine neuroblastoma, Neuro2a, in syngeneic A/J mice. Intraneoplastic inoculation of dvB7Ig/G207 at a low titer successfully inhibited the growth of established s.c. tumors, despite the expression of B7-1-Ig being detected in only 1% or fewer of tumor cells at the inoculation site, and prolonged the survival of mice bearing intracerebral tumors. Immunohistochemistry of dvB7Ig/G207-inoculated tumors revealed a significant increase in CD4+ and CD8+ T-cell infiltration compared with control tumors inoculated with defective vector expressing alkaline phosphatase (dvAP/G207). The antitumor effect of dvB7Ig/G207 was not manifested in athymic mice. In vivo depletion of immune cell subsets in A/J mice further revealed that CD8+ T cells, but not CD4+ T cells, were required. Animals cured of their tumors by dvB7Ig/G207 treatment were protected against rechallenge with a lethal dose of Neuro2a cells but not SaI/N cells. The results demonstrate that the use of soluble B7-1 for immune gene therapy is a potent and clinically applicable means of in situ cancer vaccination.


Assuntos
Antígeno B7-1/genética , Antígeno B7-1/imunologia , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Animais , Antígenos de Diferenciação/imunologia , Antígeno B7-1/biossíntese , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/terapia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Divisão Celular/imunologia , Chlorocebus aethiops , Feminino , Terapia Genética/métodos , Vetores Genéticos/genética , Herpesvirus Humano 1/genética , Imunoconjugados/genética , Imunoconjugados/imunologia , Fragmentos Fc das Imunoglobulinas/biossíntese , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/imunologia , Imunoglobulina G/biossíntese , Imunoterapia Ativa/métodos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos BALB C , Camundongos Nus , Neuroblastoma/imunologia , Neuroblastoma/patologia , Neuroblastoma/terapia , Proteínas Recombinantes de Fusão/biossíntese , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/terapia , Solubilidade , Transgenes , Células Vero
12.
Cancer Res ; 55(21): 4752-6, 1995 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-7585498

RESUMO

We have demonstrated that replication-competent attenuated mutants of herpes simplex virus type 1 (HSV-1) have therapeutic potential for malignant gliomas. Moreover, a recently described multiple mutant HSV (termed G207) has properties which may allow human clinical trials. G207 is able to replicate within and kill cells from three human malignant meningiomas in cell culture. In nude mice harboring s.c. human malignant meningioma (F5), G207 can inhibit growth in a dose-dependent fashion. In nude mice harboring intracranial subdural human malignant meningioma (F5), one injection of G207 caused significantly decreased tumor growth and one apparent cure with neither neurological dysfunction nor pathological changes in the surrounding brain. These results suggest that G207 should be considered for therapeutic trials in the treatment of malignant meningioma refractory to currently available therapies.


Assuntos
Herpesvirus Humano 1/fisiologia , Neoplasias Meníngeas/terapia , Meningioma/terapia , Vacinas Virais/uso terapêutico , Animais , Feminino , Herpes Simples/fisiopatologia , Herpesvirus Humano 1/genética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mutação , Transplante de Neoplasias , Transplante Heterólogo , Vacinas Atenuadas/uso terapêutico
13.
Cancer Res ; 53(13): 3125-8, 1993 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-8319220

RESUMO

The ability of interleukin-4 (IL-4) to mediate an antitumor response to human gliomas was studied in vivo in nude mice. To allow the effect of IL-4 to be exerted over a relatively short distance and at an optimal concentration, a transfected tumor cell line expressing a high level of IL-4 was used in mixed tumor transplantation assays. There was a significant inhibition of growth of the U87 human glioma line when the IL-4-secreting cell line, LT-1, was implanted s.c. with the glioma in 5 nude mice when compared to contralateral control tumors consisting of the U87 glioma and IL-4-negative control cells. In addition, there was a prolongation of survival when U87 along with IL-4-secreting cells were implanted intracerebrally in 12 nude mice compared to 12 control nude mice implanted with U87 and IL-4-negative control cells and 11 control animals receiving U87 alone. Histological analysis 4 days after i.c. inoculation revealed the presence of a dramatic eosinophil infiltrate and tumor necrosis. The absence of viable glioma cells as well as resolution of inflammation 19 days after treatment suggests the potential for complete tumor regression without ongoing inflammatory sequelae resulting from cytokine treatment.


Assuntos
Neoplasias Encefálicas/terapia , Glioma/terapia , Imunoterapia , Interleucina-4/metabolismo , Plasmocitoma/metabolismo , Adjuvantes Imunológicos/farmacologia , Animais , Feminino , Humanos , Injeções Intraventriculares , Injeções Subcutâneas , Interleucina-4/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Plasmocitoma/patologia , Ratos , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia
14.
Cancer Res ; 54(21): 5614-7, 1994 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-7923206

RESUMO

We have found that thymidine kinase expression is a major radioresponse determinant in rat glioma cells. Cells that lack thymidine kinase expression are significantly more radiosensitive relative to the wild-type cells. The degree of sensitization is large, particularly at the dose levels used in fractionated radiotherapy. The difference in low dose survival can be accounted for by a marked difference in the ability of the cells to undergo repair of sublethal damage. When herpes thymidine kinase was introduced into the thymidine kinase-deficient mutant cells, radioresistance was partially restored, and sublethal damage repair was also enhanced. All other radiobiological responses, including DNA double-strand break repair, potentially lethal damage repair, G2 arrest, and cell cycle distribution, appeared similar among the cell lines. These data suggest that the thymidine kinase enzyme or its cellular gene may be an excellent therapeutic target to increase radiosensitivity and thereby, to enhance the radiocurability of malignant brain gliomas.


Assuntos
Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/radioterapia , Dano ao DNA , Reparo do DNA , DNA de Neoplasias/efeitos da radiação , Glioma/enzimologia , Glioma/radioterapia , Tolerância a Radiação , Timidina Quinase/metabolismo , Animais , Neoplasias Encefálicas/patologia , Sobrevivência Celular , Fase G2 , Glioma/patologia , Ratos , Células Tumorais Cultivadas
15.
Cancer Res ; 61(7): 3009-15, 2001 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11306480

RESUMO

Autologous hematopoietic stem cell transplantation after myelosuppressive chemotherapy is used for the treatment of high-risk breast cancer and other solid tumors. However, contamination of the autologous graft with tumor cells may adversely affect outcomes. Human hematopoietic bone marrow cells are resistant to herpes simplex virus type 1 (HSV-1) replication, whereas human breast cancer cells are sensitive to HSV-1 cytotoxicity. Therefore, we examined the utility of G207, a safe replication-competent multimutated HSV-1 vector, as a biological purging agent for breast cancer in the setting of stem cell transplantation. G207 infection of human bone marrow cells had no effect on the proportion or clonogenic capacity of CD34+ cells but did enhance the proliferation of bone marrow cells in culture and the proportion of CD14+ and CD38+ cells. On the other hand, G207 at a multiplicity of infection of 0.1 was able to purge bone marrow of contaminating human breast cancer cells. Because G207 also stimulates the proliferation of human hematopoietic cells, it overcomes a limitation of other purging methods that result in delayed reconstitution of hematopoiesis. The efficient infection of human bone marrow cells in the absence of detected toxicity suggests that HSV vectors may also prove useful for gene therapy to hematopoietic progenitor cells.


Assuntos
Células da Medula Óssea/virologia , Purging da Medula Óssea , Neoplasias da Mama/patologia , Neoplasias da Mama/virologia , Transplante de Células-Tronco Hematopoéticas , Herpesvirus Humano 1/fisiologia , Células da Medula Óssea/citologia , Células-Tronco Hematopoéticas/virologia , Herpesvirus Humano 1/genética , Humanos , Células K562/virologia , Replicação Viral
16.
Oncogene ; 20(1): 97-105, 2001 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-11244508

RESUMO

Loss of the tumor suppressor gene NF1 in neurofibromatosis type 1 (NF1) contributes to the development of a variety of tumors, including malignant peripheral nerve sheath tumors (MPNST) and benign neurofibromas. Of the different cell types found in neurofibromas, Schwann cells usually provide between 40 and 80%, and are thought to be critical for tumor growth. Here we describe the identification of growth factors that are upregulated in NF1-/- mouse Schwann cells and are potential regulators of angiogenesis and cell growth. Basic fibroblast growth factor (FGF-2), platelet-derived growth factor (PDGF) and midkine (MK) were found to be induced by loss of neurofibromin and MK was further characterized. MK was induced in human neurofibromas, schwannomas, and various nervous system tumors associated with NF1 or NF2; midkine showed an expression pattern overlapping but distinct from its homolog pleiotrophin (PTN). Immunohistochemistry revealed expression of MK in S-100 positive Schwann cells of dermal and plexiform neurofibromas, and in endothelial cells of tumor blood vessels, but not in normal blood vessels. Furthermore, MK demonstrated potent mitogenic activity for human systemic and brain endothelial cells in vitro and stimulated proliferation and soft agar colony formation of human MPNST derived S100 positive cells and fibroblastoid cells derived from an NF1 neurofibroma. The data support a possible central role for MK as a mediator of angiogenesis and neurofibroma growth in NF1. Oncogene (2001) 20, 97 - 105.


Assuntos
Indutores da Angiogênese/biossíntese , Proteínas de Transporte/biossíntese , Citocinas , Regulação Neoplásica da Expressão Gênica , Genes da Neurofibromatose 1 , Mitógenos/fisiologia , Proteínas do Tecido Nervoso/deficiência , Neurofibroma/genética , Células de Schwann/metabolismo , Adulto , Indutores da Angiogênese/genética , Indutores da Angiogênese/fisiologia , Animais , Encéfalo/irrigação sanguínea , Encéfalo/citologia , Encéfalo/metabolismo , Encéfalo/patologia , Neoplasias Encefálicas/química , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Proteínas de Transporte/genética , Linhagem Celular , Endotélio Vascular/química , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Humanos , Imuno-Histoquímica , Camundongos , Midkina , Proteínas do Tecido Nervoso/genética , Neurofibroma/metabolismo , Neurofibroma/patologia , Neurofibromatose 1/metabolismo , Neurofibromatose 1/patologia , Neurofibromatose 2/metabolismo , Neurofibromina 1 , Células de Schwann/patologia , Células Tumorais Cultivadas
17.
Oncogene ; 14(5): 611-6, 1997 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-9053860

RESUMO

Formation of meningiomas has been associated with the loss of genetic material on chromosome 22. To approach the additional chromosomal events that underlie progression of these tumors to malignancy, we have examined several other chromosomal regions for loss of heterozygosity (LOH) in these tumors. Fifty-eight tumors, comprising 43 benign meningiomas, 11 atypical meningiomas and four malignant meningiomas, were examined. While the loss of chromosome 22 was seen in approximately half of all these tumors, regardless of their malignancy, the most frequent chromosomal losses observed in the malignant and atypical tumors were on the long arm of chromosome 14. Thirty-nine tumors were informative for at least one of the three markers on chromosome 14 that we tested. Of these, 7/14 malignant and atypical tumors showed LOH in contrast to only 1/25 benign tumors. Other loci that showed LOH in malignant tumors, although at a much lower frequency, were on chromosomes 17p and 1p. The high frequency of LOH for loci on chromosome 14q in atypical and malignant tumors suggests the presence of a tumor progression gene at this locus. In one of the malignant meningiomas heterozygosity was lost at D14S13 and D14S16 but retained at the proximal marker D14S43 as well as the more distal marker D14S23. This suggests that an interstitial deletion occurred in this tumor which should be useful for further refining the position of the putative tumor progression locus.


Assuntos
Deleção Cromossômica , Cromossomos Humanos Par 14 , Neoplasias Meníngeas/genética , Meningioma/genética , Mapeamento Cromossômico , Cromossomos Humanos Par 22 , Marcadores Genéticos , Humanos , Neoplasias Meníngeas/patologia , Neoplasias Meníngeas/cirurgia , Meningioma/patologia , Meningioma/cirurgia , Reação em Cadeia da Polimerase , Polimorfismo Genético
18.
Arch Gen Psychiatry ; 48(6): 548-55, 1991 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-2039338

RESUMO

To evaluate the feasibility of cingulotomy as a treatment for patients with intractable obsessive-compulsive disorder, we evaluated the records of all 35 patients with this diagnosis who had undergone one or more such procedures at Massachusetts General Hospital, Boston, during the last 25 years. Retrospectively, all but two of these patients met DSM-III-R criteria for obsessive-compulsive disorder. Six patients were deceased; four by suicide. Questionnaires were sent to the remaining 27 patients with obsessive-compulsive disorder; 17 patients returned the questionnaire and another agreed to an interview without completing the forms. Sixteen of these 18 patients participated in a telephone interview, and patient reports were corroborated by an informant in 10 cases. Despite the presence of some side effects, such as easily controlled seizures (9%) and transient mania (6%), the results of this investigation support the use of cingulotomy as a potentially effective treatment for patients with severe and disabling obsessive-compulsive disorder. With the use of very conservative criteria, we estimated that at least 25% to 30% of the patients benefited substantially from this procedure. Similar results were found in a preliminary prospective study of four patients who recently underwent cingulotomy after state-of-the-art preoperative treatments had failed.


Assuntos
Giro do Cíngulo/cirurgia , Transtorno Obsessivo-Compulsivo/cirurgia , Psicocirurgia , Adulto , Idoso , Feminino , Seguimentos , Humanos , Masculino , Transtornos Mentais/complicações , Pessoa de Meia-Idade , Transtorno Obsessivo-Compulsivo/complicações , Transtorno Obsessivo-Compulsivo/terapia , Escalas de Graduação Psiquiátrica , Psicoterapia , Psicotrópicos/uso terapêutico , Técnicas Estereotáxicas
19.
Hum Gene Ther ; 9(15): 2177-85, 1998 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-9794202

RESUMO

We investigated the therapeutic efficacy of G207, a replication-competent multimutated herpes simplex virus type 1, for the treatment of human malignant mammary tumors metastatic to the brain. In vitro studies demonstrated that G207 efficiently destroyed three of four human malignant breast cancer cell lines. MDA-MB-435 was most susceptible and MDA-MB-231 was least susceptible to G207. In athymic mice harboring subcutaneous or intracerebral MDA-MB-435 cells, intraneoplastic inoculation of G207 caused growth inhibition and/or prolonged survival. In contrast, G207 had minimal effects on MDA-MB-231 subcutaneous tumor growth or survival in the intracerebral tumor model. The efficacy of G207 therapy in vivo correlated well with the susceptibility of the human cancer cells to G207 in vitro. Histological studies indicate that G207 replication is restricted to tumor cells in vivo and does not occur in the surrounding brain tissue. These results suggest that G207 shows particular promise for use as a novel antineoplastic agent for metastatic brain tumors and that in vitro testing may predict which tumors will be most responsive in vivo.


Assuntos
Adenocarcinoma/terapia , Neoplasias Encefálicas/secundário , Neoplasias da Mama/terapia , Terapia Genética , Herpesvirus Humano 1/genética , Adenocarcinoma/secundário , Animais , Neoplasias Encefálicas/patologia , Efeito Citopatogênico Viral , Modelos Animais de Doenças , Feminino , Engenharia Genética , Herpesvirus Humano 1/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Células Tumorais Cultivadas , Replicação Viral
20.
Hum Gene Ther ; 10(17): 2869-78, 1999 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-10584932

RESUMO

A multimutated, conditionally replicating herpes simplex virus type 1, G207, has been developed as an effective means of treating human malignant brain tumors. We have shown that intraneoplastic inoculation of G207 induces a specific and systemic antitumor immune response that plays an important role in the antitumor activity, in addition to the direct oncolytic action of G207. Since a large number of malignant brain tumor patients are treated with corticosteroids, it is important to evaluate whether the therapeutic efficacy of G207 is affected by corticosteroid-induced immunosuppression. For a tumor model, we used G207-permissive N18 murine neuroblastoma cells implanted subcutaneously in syngeneic A/J mice. Intraneoplastic inoculation of G207 (10(7) PFU) induced significant suppression of tumor growth whether or not dexamethasone (5 mg/kg) was given. When dexamethasone was given for an extensive time (16 days starting on day -2), all G207-treated mice showed tumor growth despite initial shrinkage, whereas in the saline group, four of eight of the G207-treated mice were cured. Dexamethasone administration significantly reduced serum neutralizing antibodies against G207 at 14 and 21 days after intraneoplastic G207 inoculation. However, there was no difference between the dexamethasone and saline groups in terms of the amount of infectious G207 isolated from tumors. Dexamethasone administration completely abolished G207-induced cytotoxic T lymphocyte activity against N18 cells. These results indicate that the oncolytic activity of G207 is retained under corticosteroid administration. However, intensive immunosuppression may diminish the long-term efficacy of G207 owing to suppression of tumor-specific cytotoxic T lymphocyte induction.


Assuntos
Antineoplásicos Hormonais/farmacologia , Terapia Biológica , Dexametasona/farmacologia , Neuroblastoma/terapia , Simplexvirus/efeitos dos fármacos , Animais , Antineoplásicos Hormonais/uso terapêutico , Terapia Combinada , Efeito Citopatogênico Viral/efeitos dos fármacos , Dexametasona/uso terapêutico , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Transplante de Neoplasias , Neuroblastoma/imunologia , Simplexvirus/genética , Simplexvirus/fisiologia , Linfócitos T Citotóxicos/imunologia , Células Tumorais Cultivadas , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA