Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Immunol ; 193(12): 6192-206, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25381437

RESUMEN

Glioblastomas (GBMs) are lethal brain cancers that are resistant to current therapies. We investigated the cytotoxicity of human allogeneic NK cells against patient-derived GBM in vitro and in vivo, as well as mechanisms mediating their efficacy. We demonstrate that KIR2DS2 immunogenotype NK cells were more potent killers, notwithstanding the absence of inhibitory killer Ig-like receptor (KIR)-HLA ligand mismatch. FACS-sorted and enriched KIR2DS2(+) NK cell subpopulations retained significantly high levels of CD69 and CD16 when in contact with GBM cells at a 1:1 ratio and highly expressed CD107a and secreted more soluble CD137 and granzyme A. In contrast, KIR2DS2(-) immunogenotype donor NK cells were less cytotoxic against GBM and K562, and, similar to FACS-sorted or gated KIR2DS2(-) NK cells, significantly diminished CD16, CD107a, granzyme A, and CD69 when in contact with GBM cells. Furthermore, NK cell-mediated GBM killing in vitro depended upon the expression of ligands for the activating receptor NKG2D and was partially abrogated by Ab blockade. Treatment of GBM xenografts in NOD/SCID mice with NK cells from a KIR2DS2(+) donor lacking inhibitory KIR-HLA ligand mismatch significantly prolonged the median survival to 163 d compared with vehicle controls (log-rank test, p = 0.0001), in contrast to 117.5 d (log-rank test, p = 0.0005) for NK cells with several inhibitory KIR-HLA ligand mismatches but lacking KIR2DS2 genotype. Significantly more CD56(+)CD16(+) NK cells from a KIR2DS2(+) donor survived in nontumor-bearing brains 3 wk after infusion compared with KIR2DS2(-) NK cells, independent of their proliferative capacity. In conclusion, KIR2DS2 identifies potent alloreactive NK cells against GBM that are mediated by commensurate, but dominant, activating signals.


Asunto(s)
Citotoxicidad Inmunológica/genética , Glioblastoma/genética , Glioblastoma/inmunología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Receptores KIR/genética , Traslado Adoptivo , Animales , Biomarcadores/metabolismo , Línea Celular Tumoral , Expresión Génica , Genotipo , Glioblastoma/mortalidad , Glioblastoma/patología , Gliosarcoma/inmunología , Granzimas/genética , Granzimas/metabolismo , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Inmunofenotipificación , Ligandos , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Microglía/inmunología , Microglía/metabolismo , Microglía/patología , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Células Madre Neoplásicas/metabolismo , Nestina/genética , Nestina/metabolismo , Pronóstico , Unión Proteica , Receptores KIR/metabolismo , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo
2.
Br J Neurosurg ; 25(2): 286-8, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21158519

RESUMEN

Extracranial metastasis of malignant glioma is an extremely rare event. We report on a 67-year-old patient with a primary gliosarcoma that was treated by open resection. The concomitant radio-chemotherapy which followed induced an unusually severe and early leucocytopenia. Ten months after diagnosis, the patient presented with multiple metastases in the lung and the skeletal system. The clinical, radiological and neuropathological findings are described. In addition, we discuss the possible role of a compromised immune system in the development of extracranial glioma metastasis.


Asunto(s)
Neoplasias Encefálicas/patología , Gliosarcoma/secundario , Neoplasias Pulmonares/secundario , Anciano , Neoplasias Encefálicas/inmunología , Resultado Fatal , Gliosarcoma/inmunología , Humanos , Neoplasias Pulmonares/inmunología , Masculino
3.
Cancer Immunol Immunother ; 59(4): 541-51, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19798500

RESUMEN

The immunosuppressive environment of malignant gliomas is likely to suppress the anti-tumor activity of infiltrating microglial cells and lymphocytes. Macrophages and microglial cells may be activated by oligonucleotides containing unmethylated CpG-motifs, although their value in cancer immunotherapy has remained controversial. Following injection of CpG-containing oligonucleotides (ODN) into normal rat brain, we observed a local inflammatory response with CD8+ T cell infiltration, upregulation of MHC 2, and ED1 expression proving the immunogenic capacity of the CpG-ODN used. This was not observed with a control ODN mutated in the immunostimulatory sequence (m-CpG). To study their effect in a syngeneic tumor model, we implanted rat 9L gliosarcoma cells into the striatum of Fisher 344 rats. After 3 days, immunostimulatory CpG-ODN, control m-CpG-ODN, or saline was injected stereotactically into the tumors (day 3 group). In another group of animals (day 0 group), CpG-ODN were mixed with 9L cells prior to implantation without further treatment on day 3. After 3 weeks, the animals were killed and the brains and spleens were removed. Rather unexpectedly, the tumors in several of the animals treated with CpG-ODN (both day 0 and day 3 group) were larger than in saline or m-CpG-ODN treated control animals. The tumor size in CpG-ODN-treated animals was more variable than in both control groups. This was associated with inflammatory responses and necrosis which was observed in most tumors following CpG treatment. This, however, did not prevent excessive growth of solid tumor masses in the CpG-treated animals similar to the control-treated animals. Dense infiltration with microglial cells resembling ramified microglia was observed within the solid tumor masses of control- and CpG-treated animals. In necrotic areas (phagocytic), activation of microglial cells was suggested by ED1 expression and a more macrophage-like morphology. Dense lymphocytic infiltrates consisting predominantly of CD8+ T cells and fewer NK cells were detected in all tumors including the control-treated animals. Expression of perforin serving as a marker for T cell or NK cell activation was detected only on isolated cells in all treatment groups. Tumors of all treatment groups revealed CD25 expression indicating T cells presumed to maintain peripheral tolerance to self-antigens. Cytotoxic T cell assays with in vitro restimulated lymphocytes ((51)chromium release assay) as well as interferon-gamma production by fresh splenocytes (Elispot assay) revealed specific responses to 9L cells but not another syngeneic cell line (MADB 106 adenocarcinoma). Surprisingly, the lysis rates with lymphocytes from CpG-ODN-treated animals were lower compared to control-treated animals. The tumor size of individual animals did not correlate with the response in both immune assays. Taken together, our data support the immunostimulatory capacity of CpG-ODN in normal brain. However, intratumoral application proved ineffective in a rat glioma model. CpG-ODN treatment may not yield beneficial effects in glioma patients.


Asunto(s)
Neoplasias Encefálicas/patología , Gliosarcoma/patología , Oligodesoxirribonucleótidos/administración & dosificación , Adyuvantes Inmunológicos/uso terapéutico , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Antivirales/farmacología , Neoplasias Encefálicas/inmunología , Linfocitos T CD8-positivos/inmunología , Movimiento Celular , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Gliosarcoma/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Inyecciones Intralesiones , Interferón gamma/farmacología , Linfocitos Infiltrantes de Tumor , Masculino , Ratas , Ratas Endogámicas F344 , Bazo/citología , Bazo/inmunología , Linfocitos T Citotóxicos/inmunología , Células Tumorales Cultivadas
4.
Cancer Immunol Immunother ; 57(12): 1745-56, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18369621

RESUMEN

Despite the increasing number of immunotherapeutic strategies for the treatment of cancer, most approaches have failed to correlate the induction of an anti-tumor immune response with therapeutic efficacy. We therefore took advantage of a successful vaccination strategy-combining dendritic cells and irradiated GM-CSF secreting tumor cells-to compare the immune response induced against 9L gliosarcoma tumors in cured rats versus those with progressively growing tumors. At the systemic level, the tumor specific cytotoxic responses were quite heterogeneous in uncured vaccinated rats, and were surprisingly often high in animals with rapidly-growing tumors. IFN-gamma secretion by activated splenic T cells was more discriminative as the CD4+ T cell-mediated production was weak in uncured rats whereas high in cured ones. At the tumor level, regressing tumors were strongly infiltrated by CD8+ T cells, which demonstrated lytic capacities as high as their splenic counterparts. In contrast, progressing tumors were weakly infiltrated by T cells showing impaired cytotoxic activities. Proportionately to the T cell infiltrate, the expression of Foxp3 was increased in progressive tumors suggesting inhibition by regulatory T cells. In conclusion, the main difference between cured and uncured vaccinated animals does not depend directly upon the induction of systemic cytotoxic responses. Rather the persistence of higher CD4+ Th1 responses, a high intratumoral recruitment of functional CD8+ T cells, and a low proportion of regulatory T cells correlate with tumor rejection.


Asunto(s)
Neoplasias Encefálicas/terapia , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Células Dendríticas/trasplante , Gliosarcoma/terapia , Animales , Neoplasias Encefálicas/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Citotoxicidad Inmunológica , Gliosarcoma/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Ratas , Ratas Endogámicas F344 , Linfocitos T Reguladores/inmunología , Células TH1/inmunología
5.
J Neurooncol ; 78(2): 135-43, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16598429

RESUMEN

PURPOSE: Microbeam radiation therapy (MRT), a novel experimental radiosurgery that largely spares the developing CNS and other normal tissues, is tolerated well by developing animals and palliates advanced 9LGS tumors. This report, to our knowledge, is the first demonstration that gene-mediated immunotherapy (GMIMPR) enhances the efficacy of MRT for advanced 9LGS tumors. METHODS: Seventy-six male Fischer 344 rats were implanted ic with 10(4)9LGS cells on d0. By d14, the cells had generated approximately approximately 40 mm3 ic 9LGS tumours, experimental models for therapy of moderately aggressive human malignant astrocytomas. Each of the 14 untreated (control) rats died from a large (>100 mg) ic tumor before d29 (median, d21). On d14, the remaining 62 rats were given deliberately suboptimal microbeam radiation therapy (MRT) by a single lateral exposure of the tumor-bearing zone of the head to a 10.1 mm-wide, approximately approximately 11 mm-high array of 20-39 microm-wide, nearly parallel beams of synchrotron wiggler-generated radiation (mainly approximately 50-150 keV X-rays) that delivered 625 Gy peak skin doses at approximately approximately 211 microm ctc intervals in approximately approximately 300 ms either without additional treatments (MRT-only, 25 rats), with post-MRT GMIMPR (MRT+GMIMPR, 23 rats: multiple sc injections of irradiated (clonogenically-disabled) GM-CSF gene-transfected 9LGS cells), or with post-MRT IMPR (MRT+IMPR, 14 rats: multiple sc injections of irradiated (clonogenically-disabled) 9LGS cells. RESULTS: The median post-implantation survivals of rats in the MRT-only, MRT+GMIMPR and MRT+IMPR groups were over twice that of controls; further, approximately approximately 20% of rats in MRT-only and MRT+IMPR groups survived >1 yr with no obvious disabilities. Moreover, over 40% of MRT+GMIMPR rats survived >1 yr with no obvious disabilities, a significant (P<0.04) increase over the MRT-only and MRT+IMPR groups. SIGNIFICANCE: These data suggest that the combination of MRT+GMIMPR might be better than MRT only for unifocal CNS tumors, particularly in infants and young children.


Asunto(s)
Neoplasias Encefálicas/terapia , Gliosarcoma/terapia , Inmunoterapia/métodos , Radiocirugia/métodos , Factores de Edad , Animales , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/cirugía , Terapia Combinada/métodos , Terapia Genética , Gliosarcoma/inmunología , Gliosarcoma/cirugía , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Inmunización , Masculino , Dosificación Radioterapéutica , Ratas , Ratas Endogámicas F344 , Estadísticas no Paramétricas , Análisis de Supervivencia , Transfección
7.
Cancer Res ; 64(22): 8435-42, 2004 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-15548715

RESUMEN

In an attempt to induce potent immune antitumor activities, we investigated, within the rat 9L gliosarcoma model, distal therapeutic vaccinations associating three therapies: dendritic cell vaccination, intratumoral granulocyte macrophage colony-stimulating factor (GM-CSF) gene transfer, and tumor apoptosis induction. Vaccines of dendritic cells coinjected with processed GM-CSF secreting 9L cells induced systemic responses, resulting in the complete regression of distant preimplanted 9L tumor masses in, with the best strategy, 94% of male rats. All of the cured rats developed a long-term resistance to a rechallenge with parental cells. The curative responses were correlated with the detection of elevated specific cytotoxic activities and a CD4+, CD8+ T cell-, and natural killer (NK) cell-mediated IFN-gamma production. The survival rate of the rat seemed more directly linked to the amount of GM-CSF secreted by the transduced tumor cells, which in turn depended on the toxicity of the apoptosis-inducing treatment, than to the level of apoptosis induced. Unexpectedly, alive GM-CSF secreting 9L cells became apoptotic when injected in vivo. Thus we documented the positive role of apoptosis in the induction of therapeutic antitumor responses by comparing, at equal GM-CSF exogenous supply, the effects of dendritic cells coinjected with apoptotic or necrotic 9L cells. The data showed the superior therapeutic efficiency of combined vaccines containing apoptotic tumor cells. In conclusion, vaccinations with dendritic cells associated with apoptotic tumor cells secreting GM-CSF show a very high therapeutic potency that should show promise for the treatment of human cancer.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Células Dendríticas/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Animales , Apoptosis , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Gliosarcoma/inmunología , Gliosarcoma/metabolismo , Gliosarcoma/patología , Masculino , Ratas , Ratas Endogámicas F344
8.
J Neurooncol ; 66(1-2): 39-49, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15015768

RESUMEN

Cytokines play a major role in the regulation of the immune system. Granulocyte-macrophage colony-stimulating factor (GM-CSF) has been shown to be useful for immunotherapy against glioma because it can stimulate dendritic cells to present tumor antigen. Interleukin-2 (IL-2) is involved in T-cell expansion, and interleukin-12 (IL-12) drives the T-helper cell type I response. Previous studies have shown that each of these cytokines alone can induce the regression of tumor cells. In the present study we postulated that peripheral infusion of GM-CSF along with either IL-2 or IL-12 and irradiated tumor cells can lead to increased survival from 9L brain tumors. 9L gliosarcoma cells (10(6)) were implanted in the brains of syngeneic Fischer 344 rats. Osmotic minipumps were utilized for subcutaneous, continuous delivery of GM-CSF, either alone or with IL-2 or IL-12. Irradiated 9L cells were injected subcutaneously at various time points during treatment. Delayed-type hypersensitivity (DTH) and immunohistological analysis were used to further characterize the anti-tumor response. Treatment with GM-CSF and irradiated tumor cells led to an increase in survival rate in rats with intracranial 9L tumors when compared to untreated animals. The addition of IL-2 or IL-12 to the GM-CSF/tumor cell therapy further increased the survival rate up to 90%. The anti-tumor response was associated with vigorous DTH against 9L cells and increased infiltration of CD4+ and CD8+ lymphocytes into the tumor. These results suggest that the combined infusion of GM-CSF and other cytokines may be effective adjuvants in treating brain tumors.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Gliosarcoma/tratamiento farmacológico , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Inmunoterapia , Interleucina-12/uso terapéutico , Interleucina-2/uso terapéutico , Animales , Antígenos de Diferenciación/metabolismo , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Células Dendríticas/metabolismo , Células Dendríticas/patología , Quimioterapia Combinada , Femenino , Gliosarcoma/inmunología , Gliosarcoma/patología , Hipersensibilidad Tardía/fisiopatología , Inmunohistoquímica , Memoria Inmunológica , Estadificación de Neoplasias , Ratas , Ratas Endogámicas F344 , Análisis de Supervivencia
9.
Mol Ther ; 8(4): 530-42, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14529825

RESUMEN

Our laboratory has employed replication-defective herpes simplex virus type 1 gene transfer vectors for treatment of animal models of human malignant glioblastoma. The base vectors were defective for the immediate early (IE) genes ICP4, ICP27, and ICP22 but expressed the IE gene ICP0, which can arrest tumor cell division, and an IE thymidine kinase (alpha-tk) gene construct that mediates suicide gene therapy (SGT) in the presence of ganciclovir (GCV). Previously, we reported that SGT using ICP0/alpha-tk vectors in nude mouse models of glioblastoma was improved by coexpression of the gap-junction-forming protein connexin43 (Cx43) or human tumor necrosis factor alpha (TNF alpha). We also showed that further gains in therapeutic outcome could be achieved by combining TNF alpha-enhanced SGT with gamma-knife radiosurgery (GKR). To expand these observations, we have first repeated these studies in immunocompetent rats with brain tumors derived from implanted 9L gliosarcoma cells and second compared the most efficient vector from this study with a new recombinant vector, NUREL-C2, which expressed both TNF alpha and Cx43 along with ICP0 and alpha-tk. Results from the first part indicated that our ICP0/alpha-tk/TNF alpha vector in combination with GKR provides an effective therapy although this treatment was not statistically better than GKR combined with the ICP0/alpha-tk/Cx43 vector. Our observations in the second part suggested that NUREL-C2 may be more effective than the ICP0/alpha-tk/TNF alpha vector in combination treatments with GCV (P = 0.08) or GCV plus GKR (P = 0.10). GKR significantly enhanced the efficacy of NUREL-C2/GCV treatment (P = 0.02) as well as other virus/GCV treatments (P < or = 0.05). Conversely, the efficacy of GKR was significantly improved by both the ICP0/alpha-tk/TNF alpha vector and NUREL-C2 in combination with GCV (P = 0.02 and P < 0.01, respectively). Together these results indicate that NUREL-C2 may be an attractive candidate for Phase I gene-therapy safety studies in patients with recurrent malignant glioblastoma.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/radioterapia , Terapia Genética , Vectores Genéticos , Gliosarcoma/tratamiento farmacológico , Gliosarcoma/radioterapia , Simplexvirus , Animales , Neoplasias Encefálicas/inmunología , Conexina 43/genética , Conexina 43/metabolismo , Gliosarcoma/inmunología , Ratas , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
10.
J Neurooncol ; 64(1-2): 63-9, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12952287

RESUMEN

Immunotherapy of gliomas has been forwarded as an attractive alternative to standard therapeutic modalities. Numerous observations indicate some therapeutic efficacy with this approach, but it is not curative in most reports. It is well established that gliomas suppress immune reactivity via a number of mechanisms, including expression CD95 ligand (CD95L), which induces apoptosis of immune effector cells, and secretion of immunosuppressive factors such as transforming growth factor-beta (TGFbeta). It has been hypothesized that abrogation of production or function of TGFbeta would improve immune reactivity to gliomas. To investigate this in a fashion that is translatable into clinical practice, we utilized a retroviral vector encoding a truncated, soluble form of the Type II receptor for TGFbeta (TFGbeta sr) and expressed it in the rat 9L gliosarcoma line (9L-TGFbeta sr). We then determined whether expression of TGFbeta sr affected in vitro sensitivity of 9L to lysis by immune effector cells, whether expression of TGFbeta sr affected tumorigenesis of 9L in vivo, and whether TGFbeta sr affected expression of immunity to 9L. In these experiments, we determined that 9L-TGFbeta sr was more susceptible than sham transfected 9L (9L-neo) to lysis by natural killer (NK) cells. We also determined that subcutaneously implanted 9L-TGFbeta sr was less tumorigenic than 9L-neo in syngeneic rats. Similarly, survival was extended by approximately 40% in rats given intracranial 9L-TGFbeta sr compared to 9L-neo. Finally, we determined that elimination of CD161+ cells resulted in comparable growth of 9L-neo and 9L-TGFbeta sr in vivo, indicating that NK or NK-like cells were responsible for the anti-tumor effects in this model.


Asunto(s)
Citotoxicidad Inmunológica , Gliosarcoma/metabolismo , Gliosarcoma/patología , Células Asesinas Naturales/inmunología , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Antígenos de Superficie/inmunología , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Citotoxicidad Inmunológica/efectos de los fármacos , Gliosarcoma/inmunología , Gliosarcoma/prevención & control , Células Asesinas Naturales/efectos de los fármacos , Lectinas Tipo C/inmunología , Subfamilia B de Receptores Similares a Lectina de Células NK , Ratas , Ratas Endogámicas F344 , Receptores de Factores de Crecimiento Transformadores beta/química , Neoplasias Cutáneas/patología , Solubilidad , Análisis de Supervivencia , Factor de Crecimiento Transformador beta/farmacología , Factor de Crecimiento Transformador beta2
11.
Gene Ther ; 10(2): 188-92, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12571648

RESUMEN

Retroviral-mediated gene transfer of the IL-4 gene into experimental gliomas can cause tumor rejection, supporting the clinical use of this form of gene therapy for glioblastomas (GBM). In a clinical setting, the administration of dexamethasone (dex) is a standard procedure for GBM patients. This led us to examine the effects of dex on IL-4 gene therapy. We injected intracranially Fischer 344 rats with phosphate-buffered saline, 9L gliosarcoma cells mixed with E86.L4SN(200) cells (retroviral producer cells, RPC, transducing IL-4 cDNA) and 9L cells mixed with PA317.STK.SBA cells (control RPC expressing the HSV-tk gene). The rats from each group were treated with 0, 50, 100 or 250 microg dex/kg/day released by osmotic pumps implanted subcutaneously. While 80-100% of rats receiving 9L cells mixed with IL-4 RPC and not treated by dex survived for at least 2 months following tumor injection, only 50% and 17% of rats receiving 50 or 100 microg/kg/day of dex, respectively, reached this time point. These results indicate that dex significantly diminished the anti-tumor effect of IL-4. Thus, in a clinical setting, IL-4 gene transfer should be performed when low levels of dex are administered or in the absence of dex.


Asunto(s)
Antineoplásicos Hormonales , Neoplasias Encefálicas/terapia , Dexametasona , Gliosarcoma/terapia , Interleucina-4/genética , Animales , Antineoplásicos Hormonales/farmacología , Neoplasias Encefálicas/inmunología , Contraindicaciones , Dexametasona/farmacología , Implantes de Medicamentos , Vectores Genéticos/administración & dosificación , Gliosarcoma/inmunología , Interleucina-4/inmunología , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/terapia , Ratas , Ratas Endogámicas F344 , Retroviridae/genética
12.
Cancer Res ; 62(9): 2583-91, 2002 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-11980653

RESUMEN

As a means of enhancing immunity to gliomas, we investigated local delivery of rat, bone marrow-derived dendritic cells (DCs) into rat 9L gliosarcoma tumors and into 9L tumors induced to undergo apoptosis by gamma knife radiosurgery. Contrary to other tumors, local delivery of DCs had no therapeutic effect on 9L gliomas, even when tumor apoptosis was induced via radiosurgery, which leads to efficient "loading" of the DCs with tumor antigen. To determine whether antigen-presenting cells, such as DCs, were viable in tumors, we carried out multiparametric staining of 9L tumors, using phycoerythrin-conjugated OX6 (MHC class II) or OX62 (DC specific) and FITC-labeled Val-Ala-Asp-fluoromethyl ketone (FITC-VAD-FMK; activated caspases). It was determined that DCs were undergoing apoptosis in these tumors. We therefore sought to determine which glioma cell surface receptors or components of the extracellular matrix in gliomas influenced DC viability. Hyaluronan (HA) is a major component of glioma extracellular matrix and has been found to support tumor cell migration and metastasis. However, its influence on the immune system, and particularly on DCs, via its receptor CD44 is not well documented. Using reverse transcription-PCR, Northern blot, and Western blot analyses, we determined that HA stimulated production of inducible nitric oxide synthase (iNOS) in DCs. NO production by HA-stimulated DCs was then verified biochemically. NO production was dependent on the size of HA; intermediate HA fragments had the greatest capacity to induce NO production in DC, whereas completely digested HA oligosaccharides failed to induce NO. Furthermore, N-monomethyl-L-arginine, an inhibitor of iNOS, completely blocked HA-induced NO production by DCs. Because induction of NO results in the induction of apoptosis in macrophages as well as other cells, DCs treated with HA were examined for apoptosis in terminal deoxynucleotidyl transferase (TdT)-mediated dUTP biotin nick-end labeling assays. It was demonstrated that HA induced apoptosis in DCs and that induction of apoptosis was dependent on the production of NO because it was entirely inhibited by N-monomethyl-L-arginine. Using flow cytometric analyses with FITC-VAD-FMK, which is specific for activated caspases, we also determined that induction of apoptosis in DCs with HA could be titrated. Coincubation of 9L tumor cells with DCs was found to induce apoptosis in DCs as indicated by fluorescent staining with FITC-VAD-FMK. Specificity of this reaction for CD44-HA interactions was determined by pretreatment of DCs with anti-CD44 or pretreatment of 9L tumor cells with hyaluronidase, which blocked the induction of apoptosis in DCs. These data indicate that HA expressed by gliomas may contribute to their immunosuppressive effects by promoting apoptosis among professional antigen-presenting cells such as DCs via iNOS induction after CD44-HA interactions.


Asunto(s)
Apoptosis/efectos de los fármacos , Células Dendríticas/inmunología , Gliosarcoma/terapia , Ácido Hialurónico/farmacología , Inmunoterapia Adoptiva/métodos , Óxido Nítrico Sintasa/biosíntesis , Animales , Apoptosis/inmunología , Células Dendríticas/enzimología , Células Dendríticas/patología , Gliosarcoma/inmunología , Gliosarcoma/patología , Gliosarcoma/cirugía , Receptores de Hialuranos/inmunología , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/inmunología , Ácido Hialurónico/metabolismo , Masculino , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa de Tipo II , Radiocirugia , Ratas , Ratas Endogámicas F344
13.
Cancer Res ; 62(8): 2287-93, 2002 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-11956085

RESUMEN

Tumors arising within the central nervous system (CNS) present the immune system with a challenging target, given the heterogeneous nature of these neoplasms and their location within an "immunologically privileged" site. We used the lymphocytic choriomeningitis virus nucleoprotein (LCMV-NP) as a pseudotumor antigen to investigate recombinant Listeria monocytogenes as a tumor vaccine against s.c. and intracerebral challenges with a NP-expressing glioma, 9L-NP. Using Fischer 344 rats, we demonstrate that vaccination with recombinant L. monocytogenes-NP stimulates protection against s.c., but not intracerebral, 9L-NP tumor challenge in an antigen-specific, CD8(+) T-cell-dependent manner. After s.c. tumor rejection, enhanced antitumor immunity is achieved via epitope spreading that permits complete resistance against lethal intracerebral challenge with 9L-NP and with the untransfected parental 9L tumor. Unlike the CD8(+)-dependent immune responses against s.c. 9L-NP tumors, this expanded intracerebral immunity against endogenous tumor-associated antigens is dependent on both CD4(+) and CD8(+) T cells. Taken together, these results demonstrate that the mechanisms of tumor immunity within the brain are different from those elicited against non-CNS tumors. Furthermore, vaccination approaches exploiting the concept of epitope spreading may enhance the efficacy of antitumor immune responses within the immunologically privileged CNS, potentially mediating tumor cell killing through both CD4(+)- and CD8(+)-dependent effector pathways.


Asunto(s)
Neoplasias Encefálicas/inmunología , Vacunas contra el Cáncer/inmunología , Gliosarcoma/inmunología , Listeria monocytogenes/inmunología , Animales , Antígenos Virales/inmunología , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/prevención & control , Neoplasias Encefálicas/terapia , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Epítopos/inmunología , Femenino , Gliosarcoma/patología , Gliosarcoma/prevención & control , Gliosarcoma/terapia , Activación de Linfocitos/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Nucleoproteínas/inmunología , Ratas , Ratas Endogámicas F344 , Linfocitos T Citotóxicos/inmunología , Vacunas Sintéticas/inmunología
14.
Cancer Res ; 61(24): 8769-74, 2001 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-11751397

RESUMEN

Cytokine gene therapy for the induction of potent immune responses against central nervous system tumors has proven to have significant potential. However, this strategy needs improvement in the process of antigen presentation and/or insufficient recruitment of immunocompetent cells to achieve successful eradication of established brain tumors. We investigated the therapeutic potential of induced systemic immunity in peripheral tissues combined with interleukin-2 (IL-2) production in the vicinity of brain tumors to treat established brain tumors. Sequential magnetic resonance image monitoring showed that the combinatory therapy consisting of intracerebral (i.c.) transplantation of IL-2-producing rat gliosarcoma 9L (9L/IL-2) cells and s.c. vaccination using irradiated 9L or 9L/IL-2 cells could cure 9L-bearing rats, whereas either the i.c. injection of 9L/IL-2 cells or the s.c. vaccination produced little or marginal antitumor effects, respectively. Xenogeneic murine neuroblastoma cells secreting IL-2 could substitute for 9L/IL-2 cells, producing significant antitumor effects in the vaccinated rats. Tumor-specific cytotoxic activity was induced in the vaccinated rats but not fully in the rats treated only with i.c. injection of 9L/IL-2 cells. Immunohistochemical analysis revealed that a number of CD4(+) and CD8(+) T cells infiltrated into the brain tumors which were treated with the combinatory therapy. The level of cell infiltration was similar to that found in s.c. 9L/IL-2 tumors which were subsequently rejected. In contrast, the brain tumors treated with either i.c. transplantation of 9L/IL-2 cells or the s.c. vaccination showed only moderate infiltration of T cells. The combinatory strategy, i.c. grafting of IL-2-producing cells, and s.c. immunization of irradiated whole tumor cell vaccine, is, thus, effective for recruiting activated T cells into the brain tumor site and could be a potential therapy for brain tumors.


Asunto(s)
Neoplasias Encefálicas/terapia , Vacunas contra el Cáncer/inmunología , Terapia Genética/métodos , Gliosarcoma/terapia , Inmunoterapia Activa/métodos , Interleucina-2/inmunología , Trasplante de Neoplasias/inmunología , Animales , Presentación de Antígeno/inmunología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/genética , Trasplante de Células , Terapia Combinada , Glioblastoma/genética , Glioblastoma/inmunología , Glioblastoma/metabolismo , Glioblastoma/terapia , Gliosarcoma/genética , Gliosarcoma/inmunología , Gliosarcoma/metabolismo , Inmunohistoquímica , Interleucina-2/biosíntesis , Interleucina-2/genética , Interleucina-2/metabolismo , Activación de Linfocitos/inmunología , Masculino , Neuroblastoma/genética , Neuroblastoma/inmunología , Neuroblastoma/metabolismo , Neuroblastoma/terapia , Ratas , Ratas Endogámicas F344 , Linfocitos T Citotóxicos/inmunología
15.
Neurosurgery ; 49(3): 681-8; discussion 688-9, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11523680

RESUMEN

OBJECTIVE: Acquired immunity has been demonstrated in Fischer rats bearing syngeneic 9L tumors after herpes simplex virus (HSV) thymidine kinase (TK) gene transfection and ganciclovir treatment. The nature of this immunity in rats and its relevance to the HSV TK/ganciclovir protocol for human subjects remain to be determined. In this study, levels of major histocompatibility complex (MHC) Class I and II antigen expression were measured before and after HSV TK transfection, in an effort to document immunomodulatory changes caused by gene therapy. METHODS: Tumor cells from the 9L gliosarcoma cell line, three primary human glioma cultures, and the human glioma cell line U87 MG were transduced with HSV TK vector-containing supernatant from fibroblast-producing cells (titer of 5 x 10(6) colony-forming units/ml) and selected in G418 medium for neomycin resistance. Clones were pooled or individually selected for cell-killing assays with ganciclovir, to confirm TK expression (10(3) cells/well in a 96-well dish). Northern analyses using MHC Class I and Class II complementary deoxyribonucleic acid probes were performed on blots containing total ribonucleic acid from wild-type tumor cells and HSV TK transfectants. A beta-actin complementary deoxyribonucleic acid probe served as an internal control. Cell surface expression was confirmed with flow cytometry. The induction of MHC Class I was tested for cycloheximide and genistein sensitivity. RESULTS: All cell cultures exhibited increases in MHC Class I but not MHC Class II expression, as determined by Northern analysis densitometry and flow cytometry. Cycloheximide treatment did not diminish the up-regulation of MHC Class I after retroviral transfection, implicating a signal transduction pathway that does not require ongoing protein synthesis. Genistein pretreatment of cell cultures did diminish the up-regulation of MHC Class I, implicating a tyrosine kinase in the signaling cascade. CONCLUSION: Induction of MHC Class I in rat and human glioma cells after HSV TK retroviral gene therapy is a primary effect that is dependent on tyrosine kinase activity. Specific immune responses generated after transfection may represent an important general side effect of gene therapy protocols. Elucidation of the mechanism of immunomodulation after gene therapy will likely yield safer and more effective clinical protocols.


Asunto(s)
Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Gliosarcoma/inmunología , Gliosarcoma/terapia , Complejo Mayor de Histocompatibilidad/genética , Complejo Mayor de Histocompatibilidad/inmunología , Neuroinmunomodulación/fisiología , Animales , Modulación Antigénica/genética , Modulación Antigénica/inmunología , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Antígenos de Superficie/genética , Antígenos de Superficie/inmunología , Northern Blotting , Citometría de Flujo , Expresión Génica/genética , Técnicas de Transferencia de Gen , Vectores Genéticos , Técnicas In Vitro , Ratas , Ratas Endogámicas F344 , Simplexvirus/enzimología , Simplexvirus/genética , Simplexvirus/inmunología , Timidina Quinasa/genética , Timidina Quinasa/inmunología , Timidina Quinasa/metabolismo , Transfección/métodos , Regulación hacia Arriba
16.
Cancer Res ; 61(6): 2625-31, 2001 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-11289140

RESUMEN

We have reported previously that s.c. immunization of rats with IL-4 transduced 9L gliosarcoma cells (9L-IL-4) induced a potent antitumor immunity against intracranial, parental 9L tumors. Subcutaneous implantation of 9L-IL-4 influenced the systemic humoral response, which was demonstrated by Th2-type isotype-switching and the induction of cellular immune responses, which played a critical role in the rejection of tumors. Serological analyses of recombinant cDNA expression libraries (SEREX), has recently emerged as a powerful method for serological identification of tumor-associated antigens (TAAs) and/or tumor rejection antigens (TRAs). Because IL-4 is known to activate B cells and to promote humoral responses, and inasmuch as induction of humoral responses by central nervous system tumors has been reported to be minimal, we investigated whether the induction of a potent humoral immune response against 9L TAAs or TRAs in rats immunized s.c. with 9L-IL4 could be demonstrated. Screening of 5 x 10(5) independent clones of 9L-expression cDNA library for the presence of reactive antibodies in the serum from a 91-IL-4 immunized rat led to the identification of three different TAAs. One 9L TAA (clone 29) was demonstrated to be calcyclin, a member of the S-100 family of calcium-binding proteins. The second 9L TAA (clone 37) was demonstrated to be the rat homologue of the J6B7 mouse immunomodulatory molecule. The third TAA (clones 158 and 171) was determined to be the rat homologue of the mouse Id-associated protein 1 (MIDA1), a DNA-binding, protein-associated protein. Northern blotting demonstrated that message for calcyclin was overexpressed in 9L cells. Message encoding MIDA1 was highly expressed in parental 9L cells and thymus and, to a lesser degree, in testis, suggesting that MIDA1 was comparable with the cancer/testis category of TAAs. Sera obtained from animals bearing 9L-IL-4 were found to have a higher a frequency and titer of antibodies to these antigens when compared with sera obtained from rats bearing sham-transduced 9L (9L-neo) cells. To determine whether immunization with these TAAs induced antitumor immunity, animals were immunized by intradermal injection with expression plasmids encoding calcyclin or MIDA1. Subsequent challenge of rats with parental 9L resulted in significant suppression of tumor growth in animals immunized with MIDA1, but not with calcyclin. These results indicate that MIDA1 is an effective 9L TRA and will be useful for the investigation of specific antitumor immunity in this glioma model. Furthermore, these results suggest that this approach, termed "cytokine-assisted SEREX (CAS)," may serve as an effective strategy for identification of TRAs for in animal-glioma models of cytokine gene therapy, and potentially in humans undergoing cytokine gene therapy protocols as well.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Proteínas de Ciclo Celular , Gliosarcoma/inmunología , Pruebas Serológicas/métodos , Vacunas de ADN/inmunología , Animales , Anticuerpos Antineoplásicos/biosíntesis , Anticuerpos Antineoplásicos/inmunología , Antígenos de Neoplasias/sangre , Antígenos de Neoplasias/aislamiento & purificación , Secuencia de Bases , Vacunas contra el Cáncer/genética , División Celular/inmunología , ADN Complementario/administración & dosificación , ADN Complementario/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Gliosarcoma/patología , Isotipos de Inmunoglobulinas/inmunología , Región de Cambio de la Inmunoglobulina/inmunología , Interleucina-4/genética , Interleucina-4/inmunología , Masculino , Ratones , Datos de Secuencia Molecular , Ratas , Ratas Endogámicas F344 , Proteína A6 de Unión a Calcio de la Familia S100 , Proteínas S100/genética , Proteínas S100/inmunología , Sensibilidad y Especificidad , Células Th2/inmunología , Células Tumorales Cultivadas , Vacunas de ADN/genética
17.
Neoplasia ; 3(6): 489-99, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11774031

RESUMEN

Microglia, as intrinsic immunoeffector cells of the central nervous system (CNS), play a very sensitive, crucial role in the response to almost any brain pathology where they are activated to a phagocytic state. Based on the characteristic features of activated microglia, we investigated whether these cells can be visualized with magnetic resonance imaging (MRI) using ultrasmall superparamagnetic iron oxides (USPIOs). The hypothesis of this study was that MR microglia visualization could not only reveal the extent of the tumor, but also allow for assessing the status of immunologic defense. Using USPIOs in cell culture experiments and in a rat glioma model, we showed that microglia can be labeled magnetically. Labeled microglia are detected by confocal microscopy within and around tumors in a typical border-like pattern. Quantitative in vitro studies revealed that microglia internalize amounts of USPIOs that are significantly higher than those incorporated by tumor cells and astrocytes. Labeled microglia can be detected and quantified with MRI in cell phantoms, and the extent of the tumor can be seen in glioma-bearing rats in vivo. We conclude that magnetic labeling of microglia provides a potential tool for MRI of gliomas, which reflects tumor morphology precisely. Furthermore, the results suggest that MRI may yield functional data on the immunologic reaction of the CNS.


Asunto(s)
Neoplasias Encefálicas/patología , Medios de Contraste , Glioma/patología , Hierro , Imagen por Resonancia Magnética , Microglía/inmunología , Óxidos , Animales , Neoplasias Encefálicas/inmunología , Células Cultivadas/citología , Células Cultivadas/inmunología , Medios de Contraste/farmacocinética , Dextranos , Óxido Ferrosoférrico , Colorantes Fluorescentes , Glioma/inmunología , Gliosarcoma/inmunología , Gliosarcoma/patología , Hierro/farmacocinética , Macrófagos Peritoneales/inmunología , Magnetismo , Nanopartículas de Magnetita , Masculino , Microscopía Confocal , Trasplante de Neoplasias , Óxidos/farmacocinética , Fagocitosis , Fantasmas de Imagen , Ratas , Ratas Endogámicas F344 , Coloración y Etiquetado , Células Tumorales Cultivadas/inmunología , Células Tumorales Cultivadas/patología , Xantenos
18.
Cancer Res ; 60(20): 5731-9, 2000 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-11059767

RESUMEN

Preclinical studies with the human MHC nonrestricted cytotoxic T-cell leukemic line, TALL-104, were performed in anticipation of its use in cellular immunotherapy trials for primary malignant brain tumors. In this study, we have: (a) quantitated the in vitro brain tumor cell lysis; (b) measured the cytokine secretion upon coincubation of TALL-104 cells with brain tumor cells; (c) investigated the effect of dexamethasone on brain tumor cell cytolysis by TALL-104 cells; (d) explored the effects of lethal irradiation and cryopreservation on TALL-104 cell viability and lytic efficacy; and (e) estimated the damage TALL-104 cells induce to murine normal and tumor brain cells and their trafficking patterns in both normal and tumor-bearing rat brain upon intracranial infusion. In vitro coincubation of TALL-104 cells with human brain tumor cells, explants, and cell lines resulted in significant lysis of them, but normal brain cells were spared. Lysis of tumor at 4 h was unaffected by dexamethasone or lethal irradiation. Secretion of tumor necrosis factor-alpha, tumor necrosis factor-beta, IFN-gamma, or granulocyte/macrophage-colony stimulating factor upon TALL-104 cell coincubation with brain tumor cells variably occurred without always correlating with lysis. In vivo experiments using irradiated TALL-104 cells, placed at multiple times into normal cannulated rat brain, produced focal sterile abscesses at the instillation site but no widespread allergic encephalitic reaction. Cells morphologically consistent with TALL-104 cells specifically trafficked from the site of instillation through the neuropil, occasionally into the contralateral brain, and egressed at perivascular and leptomeningeal spaces. In vivo experiments with cannulated rats bearing 9L gliosarcoma showed a preferential localization of the TALL-104 cells in tumor compared with normal brain. Taken together, these data support the concept that TALL-104 cells can be used as a novel nontoxic and efficacious paradigm for cellular immunotherapy trials in human primary malignant brain tumors.


Asunto(s)
Neoplasias Encefálicas/terapia , Inmunoterapia Adoptiva/métodos , Linfocitos T Citotóxicos/inmunología , Animales , Antineoplásicos Hormonales/farmacología , Astrocitos/citología , Encéfalo/citología , Encéfalo/inmunología , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Cateterismo , Movimiento Celular/fisiología , Supervivencia Celular/efectos de la radiación , Técnicas de Cocultivo , Criopreservación , Citocinas/metabolismo , Citotoxicidad Inmunológica , Dexametasona/farmacología , Glioblastoma/inmunología , Glioblastoma/patología , Glioblastoma/terapia , Gliosarcoma/inmunología , Gliosarcoma/patología , Gliosarcoma/terapia , Humanos , Leucemia de Células T , Masculino , Ratones , Ratas , Ratas Endogámicas F344 , Ratas Sprague-Dawley , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/efectos de la radiación , Células Tumorales Cultivadas
19.
J Neurooncol ; 46(3): 193-203, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10902851

RESUMEN

Glioblastoma multiforme (GBM) is the most common primary human brain tumor. About 7000 new cases are diagnosed yearly in the USA. Despite current neurosurgical and postoperative radiotherapeutic tumor cytoreduction methods, in most cases occult foci of tumor cells infiltrate surrounding edematous brain tissues and cause recurrent disease within one year. GBM is almost invariably fatal within a few years after it is diagnosed. Our goal is to achieve long-term control of GBM by combining immunoprophylaxis with a radiation-based technique, such as boron neutron-capture therapy (BNCT), potentially capable of specifically targeting the infiltrating tumor cells while sparing the surrounding normal brain tissue. It has long been known that the subcutaneous (sc) injection of irradiated cells or untreated cultured cells (and the removal of the resulting tumors) derived from the well characterized, highly immunogenic 9L gliosarcoma (9LGS) rat model into young isogenic rats can prevent tumor growth after subsequent sc or intracranial (ic) injection of untreated, otherwise lethal 9LGS cells. In this study we have confirmed, quantified and extended those findings to study the efficacy of such immunological memory in normal aging rats and in aging rats previously treated for ic 9LGS tumors by BNCT. (1) The sc injection of 5,000,000 untreated 9LGS cells and the surgical removal of the resulting tumors (method A) protected 80% of normal young rats from an ic challenge with 10,000 untreated 9LGS cells, and a single sc injection of 5,000,000 lethally X-irradiated 9LGS cells (method B) protected 66% of them, but multiple sc injections with a crude particulate fraction prepared from 9LGS cells were not protective. Protection is long-lasting since contralateral ic rechallenge of six-month survivors with an injection of 10,000 viable 9LGS cells resulted in 100% survival. (2) Normal one-year-old rats were only slightly less protected than were normal young rats, approximately 70% rather than approximately 80% (method A) and approximately 60% rather than approximately 66% (method B). (3) BNCT treatment alone resulted in partial immunological protection, as 30% of one-year post-BNCT survivors of ic 9LGS tumors prevailed after contralateral ic rechallenge with 10,000 viable 9LGS cells. Moreover a single sc immunization with 5,000,000 untreated 9LGS cells prior to ic rechallenge boosted survival from 30% to 100%. The relevance of these observations to strategies of preclinical experimentation for immunoprophylaxis of malignant gliomas is discussed.


Asunto(s)
Terapia por Captura de Neutrón de Boro , Neoplasias Encefálicas/terapia , Vacunas contra el Cáncer/uso terapéutico , Gliosarcoma/terapia , Memoria Inmunológica , Inmunoterapia Activa , Vacunación , Animales , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/prevención & control , Neoplasias Encefálicas/radioterapia , Neoplasias Encefálicas/cirugía , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Terapia Combinada , Gliosarcoma/inmunología , Gliosarcoma/prevención & control , Gliosarcoma/radioterapia , Gliosarcoma/cirugía , Inmunidad Innata , Inyecciones Subcutáneas , Masculino , Trasplante de Neoplasias/inmunología , Ratas , Ratas Endogámicas F344 , Células Tumorales Cultivadas/inmunología , Células Tumorales Cultivadas/efectos de la radiación , Células Tumorales Cultivadas/trasplante
20.
J Neurooncol ; 46(3): 231-40, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10902854

RESUMEN

Glioblastoma multiforme (GBM) is the most common primary human brain tumor. About 7000 new cases are diagnosed yearly in the USA and GBM is almost invariably fatal within a few years after it is diagnosed. Despite current neurosurgical and radiotherapeutic tumor cytoreduction methods, in most cases occult foci of tumor cells infiltrate surrounding brain tissues and cause recurrent disease. Therefore the combination of neurosurgical and radiotherapeutic debulking methods with therapies to inhibit occult GBM cells should improve prognosis. In this study we have combined boron neutron-capture therapy (BNCT), a novel binary radiotherapeutic treatment modality that selectively irradiates tumor tissue and largely spares normal brain tissue, with immunoprophylaxis, a form of active immunization initiated soon after BNCT treatment, to treat advanced, clinically relevantly-sized brain tumors in rats. Using a malignant rat glioma model of high immunogenicity, the 9L gliosarcoma, we have shown that about half of the rats that would have died after receiving BNCT debulking alone, survived after receiving BNCT plus immunoprophylaxis. Further, most of the surviving rats display immunological-based resistance to recurrent 9LGS growth six months or more after treatment. To our knowledge this study represents the first time BNCT and immunoprophylaxis have been combined to treat advanced brain tumors in rats.


Asunto(s)
Terapia por Captura de Neutrón de Boro , Neoplasias Encefálicas/terapia , Vacunas contra el Cáncer/uso terapéutico , Gliosarcoma/terapia , Inmunoterapia Activa , Animales , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/radioterapia , Neoplasias Encefálicas/cirugía , Vacunas contra el Cáncer/administración & dosificación , Terapia Combinada , Gliosarcoma/inmunología , Gliosarcoma/radioterapia , Gliosarcoma/cirugía , Masculino , Recurrencia Local de Neoplasia , Ratas , Ratas Endogámicas F344 , Organismos Libres de Patógenos Específicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA