Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Ann Oncol ; 30(9): 1521-1530, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31282941

RESUMEN

BACKGROUND: The tumor immune microenvironment (TIME) of lung cancer brain metastasis is largely unexplored. We carried out immune profiling and sequencing analysis of paired resected primary tumors and brain metastases of non-small-cell lung carcinoma (NSCLC). PATIENTS AND METHODS: TIME profiling of archival formalin-fixed and paraffin-embedded specimens of paired primary tumors and brain metastases from 39 patients with surgically resected NSCLCs was carried out using a 770 immune gene expression panel and by T-cell receptor beta repertoire (TCRß) sequencing. Immunohistochemistry was carried out for validation. Targeted sequencing was carried out to catalog hot spot mutations in cancer genes. RESULTS: Somatic hot spot mutations were mostly shared between both tumor sites (28/39 patients; 71%). We identified 161 differentially expressed genes, indicating inhibition of dendritic cell maturation, Th1, and leukocyte extravasation signaling pathways, in brain metastases compared with primary tumors (P < 0.01). The proinflammatory cell adhesion molecule vascular cell adhesion protein 1 was significantly suppressed in brain metastases compared with primary tumors. Brain metastases exhibited lower T cell and elevated macrophage infiltration compared with primary tumors (P < 0.001). T-cell clones were expanded in 64% of brain metastases compared with their corresponding primary tumors. Furthermore, while TCR repertoires were largely shared between paired brain metastases and primary tumors, T-cell densities were sparse in the metastases. CONCLUSION: We present findings that suggest that the TIME in brain metastases from NSCLC is immunosuppressed and comprises immune phenotypes (e.g. immunosuppressive tumor-associated macrophages) that may help guide immunotherapeutic strategies for NSCLC brain metastases.


Asunto(s)
Biomarcadores de Tumor/inmunología , Neoplasias Encefálicas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Proteínas de Neoplasias/inmunología , Microambiente Tumoral/inmunología , Adulto , Anciano , Biomarcadores de Tumor/genética , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/secundario , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/cirugía , Células Dendríticas/inmunología , Femenino , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Mutación/genética , Proteínas de Neoplasias/genética , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Microambiente Tumoral/genética
2.
Oncogene ; 36(47): 6568-6580, 2017 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-28783169

RESUMEN

Glioblastoma (GBM) is a primary brain cancer that contains populations of stem-like cancer cells (GSCs) that home to specialized perivascular niches. GSC interactions with their niche influence self-renewal, differentiation and drug resistance, although the pathways underlying these events remain largely unknown. Here, we report that the integrin αvß8 and its latent transforming growth factor ß1 (TGFß1) protein ligand have central roles in promoting niche co-option and GBM initiation. αvß8 integrin is highly expressed in GSCs and is essential for self-renewal and lineage commitment in vitro. Fractionation of ß8high cells from freshly resected human GBM samples also reveals a requirement for this integrin in tumorigenesis in vivo. Whole-transcriptome sequencing reveals that αvß8 integrin regulates tumor development, in part, by driving TGFß1-induced DNA replication and mitotic checkpoint progression. Collectively, these data identify the αvß8 integrin-TGFß1 signaling axis as crucial for exploitation of the perivascular niche and identify potential therapeutic targets for inhibiting tumor growth and progression in patients with GBM.


Asunto(s)
Neoplasias Encefálicas/patología , Carcinogénesis/metabolismo , Glioblastoma/patología , Integrinas/metabolismo , Células Madre Neoplásicas/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Antígeno AC133/metabolismo , Animales , Biomarcadores de Tumor/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Autorrenovación de las Células , Progresión de la Enfermedad , Humanos , Cadenas beta de Integrinas/metabolismo , Puntos de Control de la Fase M del Ciclo Celular , Ratones , Ratones SCID , Cultivo Primario de Células , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Oncogene ; 36(20): 2813-2823, 2017 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-27991920

RESUMEN

Epidermal growth factor receptor (EGFR) family members play pivotal roles in cell proliferation, differentiation and survival. Overexpression and mutations of EGFRs, or aberrant EGFR signaling are commonly associated with the development of various cancers, where constitutive NF-κB activation is often found to promote the expression of various proteins involved in the proliferation, survival, migration and epithelial-to-mesenchymal transition of cancer cells. However, the mechanism of EGFR-induced NF-κB activation is not fully defined. Here, we used a Bimolecular Fluorescence Complementation-based functional genomics method to perform a high throughput screening and identified TMEM43/LUMA as a critical component in EGFR signaling network, mediating EGFR-induced NF-κB activation. Our data show that EGFR recruits TMEM43 following EGF stimulation. TMEM43 interacts with the scaffold protein CARMA3 and its associating complex to induce downstream NF-κB activation, and plays a critical role in controlling cell survival. TMEM43 deficiency significantly affects colony formation, survival of anoikis-induced cell death, migration and invasion of cancer cells in vitro, as well as tumor progression in vivo. Importantly, higher expression of TMEM43 closely correlates with brain tumor malignancy, and suppression of TMEM43 expression in brain tumor cells inhibited their growth both in vitro and in vivo. Altogether, our studies reveal a crucial link of EGF receptor to NF-κB activation and tumor progression.


Asunto(s)
Receptores ErbB/metabolismo , Proteínas de la Membrana/metabolismo , FN-kappa B/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Transducción de Señal , Animales , Proteínas Adaptadoras de Señalización CARD/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Supervivencia Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Genómica/métodos , Xenoinjertos , Humanos , Proteínas de la Membrana/genética , Ratones , Neoplasias/genética , Neoplasias/mortalidad , Pronóstico , Unión Proteica , Carga Tumoral
4.
Cancer Res ; 61(11): 4375-81, 2001 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-11389063

RESUMEN

Cyclooxygenase (COX)-2, the inducible isoform of prostaglandin H synthase, has been implicated in the growth and progression of a variety of human cancers. Although COX-2 overexpression has been observed in human gliomas, the prognostic or clinical relevance of this overexpression has not been investigated to date. In addition, no study has analyzed the relationship between COX-2 expression and other molecular alterations in gliomas. Consequently, we examined COX-2 expression by immunohistochemistry in tumor specimens from 66 patients with low- and high-grade astrocytomas and correlated the percentage of COX-2 expression with patient survival. We also analyzed the relative importance of COX-2 expression in comparison with other clinicopathological features (age and tumor grade) and other molecular alterations commonly found in gliomas (high MIB-1 level, p53 alteration, loss of retinoblastoma (Rb) protein or p16, and high bcl-2 level). Kaplan-Meier analyses demonstrated that high COX-2 expression (>50% of cells stained positive) correlated with poor survival for the study group as a whole (P < 0.0001) and for those with glioblastoma multiforme in particular (P < 0.03). Cox regression analyses demonstrated that COX-2 expression was the strongest predictor of outcome, independent of all other variables. In addition, high COX-2 expression correlated with increasing histological grade but did not correlate with positive p53 immunostaining, bcl-2 expression, loss of p16 or retinoblastoma protein expression, or high MIB-1 expression. These findings indicate that high COX-2 expression in tumor cells is associated with clinically more aggressive gliomas and is a strong predictor of poor survival.


Asunto(s)
Astrocitoma/enzimología , Neoplasias Encefálicas/enzimología , Glioblastoma/enzimología , Isoenzimas/biosíntesis , Prostaglandina-Endoperóxido Sintasas/biosíntesis , Adolescente , Adulto , Anciano , Antígenos Nucleares , Astrocitoma/mortalidad , Astrocitoma/patología , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Niño , Preescolar , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Ciclooxigenasa 2 , Glioblastoma/mortalidad , Glioblastoma/patología , Humanos , Inmunohistoquímica , Antígeno Ki-67 , Proteínas de la Membrana , Persona de Mediana Edad , Proteínas Nucleares/biosíntesis , Pronóstico , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Estudios Retrospectivos , Tasa de Supervivencia , Proteína p53 Supresora de Tumor/biosíntesis
5.
Cancer Res ; 59(20): 5239-44, 1999 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-10537303

RESUMEN

The expression of activated ras genes has been implicated as a contributing factor to the radioresistance of tumor cells. As a strategy for compromising Ras protein activity and potentially enhancing the radiosensitivity of tumor cells, we have investigated the application of the AV1Y28 adenovirus, which expresses a single-chain antibody fragment directed against p21 Ras proteins. The ability of AV1Y28 transduction to modulate radioresponse was investigated using four human tumor cell lines--U251 glioblastoma, MIA PaCa-2 pancreatic carcinoma, and the colon carcinomas SW620 and HT29. Cultures were exposed to sufficient levels of AV1Y28 to transduce more than 90% of the cells; 24 h later, cultures were exposed to ionizing radiation, and clonogenic cell survival was determined. Tumor cell survival was reduced by 40-50% when the tumor cell lines were exposed to AV1Y28 only. In addition, for each tumor cell line, AV1Y28 exposure enhanced the level of radiation-induced cell killing. Dose enhancement factors at a surviving fraction of 0.1 ranged from 1.3 to 1.5. Furthermore, for each of the cell lines, the surviving fraction at 2 Gy was significantly reduced by AV1Y28 exposure. In contrast to the results seen in tumor cells, the radiosensitivity of a normal human fibroblast cell line was not affected by AV1Y28. These data indicate that this anti-Ras adenovirus enhances the radiosensitivity of tumor cells but does not affect the radiosensitivity of normal cells.


Asunto(s)
Adenoviridae/genética , Fragmentos de Inmunoglobulinas/uso terapéutico , Neoplasias/radioterapia , Tolerancia a Radiación , Proteínas ras/antagonistas & inhibidores , Apoptosis/efectos de la radiación , Ciclo Celular/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Fibroblastos/efectos de la radiación , Humanos , Fragmentos de Inmunoglobulinas/genética , Neoplasias/genética , Neoplasias/patología , Proteínas Recombinantes/uso terapéutico , Células Tumorales Cultivadas
6.
Oncogene ; 9(3): 949-54, 1994 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-8108140

RESUMEN

We analysed 31 non-glioblastoma astrocytomas for alterations in p53 protein expression and for mutations in the p53 gene. Immunohistochemistry detected p53 protein accumulation in 71% (five of seven) of juvenile pilocytic astrocytomas (WHO grade I), 63% (five of eight) of astrocytomas (WHO grade II), and 63% (10 of 16) of anaplastic astrocytomas (WHO grade III). The single strand conformation polymorphism (SSCP) assay of exons 2-11 of the p53 gene and direct DNA sequencing identified p53 mutations in 14% (one of seven) of grade I, 25% (two of eight) of grade II, and 19% (three of 16) of grade III astrocytomas. This is the first report of a p53 mutation in grade I juvenile pilocytic astrocytomas. Immunohistochemistry and SSCP analyses gave concordant results in 55% (17 of the 31) of the tumors. A total of 14 tumors, 60-80% within each grade, showed p53 protein accumulation in the absence of detectable mutations of the p53 gene. No mdm-2 gene amplification was found in these tumors. The similar frequency of p53 alterations in tumors of grades I-III suggests that the p53 gene plays a significant role early in the formation of astrocytomas rather than late in tumor progression to higher grade. The data suggest that mechanisms other than p53 gene inactivation by mutation or mdm-2 complex formation result in the accumulation of P53 protein in > 70% of non-glioblastoma astrocytomas.


Asunto(s)
Astrocitoma/metabolismo , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Mutación , Proteína p53 Supresora de Tumor/metabolismo , Adolescente , Adulto , Anciano , Astrocitoma/genética , Secuencia de Bases , Neoplasias Encefálicas/genética , Niño , Preescolar , ADN de Neoplasias , Femenino , Genes p53 , Glioblastoma/genética , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Proteína p53 Supresora de Tumor/genética
7.
Clin Cancer Res ; 6(6): 2513-20, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10873107

RESUMEN

The antitumor effects of the selective cyclooxygenase (COX)-2 inhibitor SC-236 alone and in combination with radiation were investigated using the human glioma cell line U251 grown in monolayer culture and as tumor xenografts. On the basis of Western and Northern blot analyses, these cells express COX-2 protein and mRNA to levels similar to those in the human colon carcinoma cell line HT29. Treatment of U251 cells in monolayer culture with 50 microM SC-236 resulted in a time-dependent decrease in cell survival as determined by a clonogenic assay. The cell death induced by SC-236 was associated with apoptosis and the detachment of cells from the monolayer. After 2 days of drug treatment, the cells that remained attached were exposed to graded doses of radiation, and the clonogenic assay was performed. Comparison of the survival curves for drug-treated and untreated cultures revealed that SC-236 enhanced radiation-induced cell death. In these combination studies, SC-236 treatment resulted in a dose-enhancement factor of 1.4 at a surviving fraction of 0.1, with the surviving fraction at 2 Gy (SF2) reduced from 0.61 to 0.31. These data indicate that in vitro SC-236 induces U251 apoptotic cell death and enhances the radiosensitivity of the surviving cells. To extend these investigations to an in vivo situation, U251 glioma cells were grown as tumor xenografts in the hind leg of nude mice, and SC-236 was administered in drinking water. SC-236 alone slowed tumor growth rate, and when administered in combination with local irradiation, SC-236 caused a greater than additive increase in tumor growth delay. These in vitro and in vivo results suggest that the selective inhibition of COX-2 combined with radiation has potential as a cancer treatment.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/radioterapia , Glioma/tratamiento farmacológico , Glioma/radioterapia , Pirazoles/farmacología , Tolerancia a Radiación/efectos de los fármacos , Sulfonamidas/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Northern Blotting , Western Blotting , Ciclo Celular/efectos de los fármacos , Ciclo Celular/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Terapia Combinada , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa/farmacología , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Citometría de Flujo , Humanos , Etiquetado Corte-Fin in Situ , Isoenzimas/biosíntesis , Proteínas de la Membrana , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Prostaglandina-Endoperóxido Sintasas/biosíntesis , ARN Mensajero/metabolismo , Fármacos Sensibilizantes a Radiaciones/farmacología , Factores de Tiempo , Células Tumorales Cultivadas
8.
Clin Cancer Res ; 7(5): 1398-409, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11350911

RESUMEN

We used isogenic human tumor cell lines to investigate the specific and direct effects of wild-type (wt) p53 on the expression of O(6)-methylguanine-DNA methyltransferase (MGMT), a DNA repair protein that confers tumor resistance to many anticancer alkylating agents. A p53-null, MGMT-proficient lung tumor cell line (H1299) was engineered to express wt p53 in a tetracycline-regulated system. High levels of p53 induction achieved by tetracycline withdrawal were accompanied by G(1) cell cycle arrest without significant apoptosis in this cell line. p53 accumulation resulted in a gradual and dramatic loss of MGMT mRNA, protein, and enzyme activity, whose levels were undetectable by day 3 of induction. The loss of MGMT protein was, however, not due to its degradation because the ubiquitin-promoted in vitro degradation of MGMT, which mediates the cellular disposal of the repair protein, was not altered by p53. Run-on transcription assays revealed a significant reduction in the rate of MGMT gene transcription. The negative regulation of MGMT expression by wt p53 was confirmed in two other human isogenic cell lines, namely, the GM47.23 glioblastoma, which contains a dexamethasone-inducible wt p53, and the H460 lung cancer cell line, in which wt p53 had been inactivated by the human papillomavirus E6 protein. Furthermore, a panel of four human tumor cell lines, including gliomas with wt p53 status, displayed markedly lower levels of MGMT gene transcripts than those having p53 mutations. Induction of wt p53 in these models led to a 3- and 2-fold increase in sensitivity to 1,3-bis(2-chloroethyl)-1-nitrosourea and temozolomide, respectively, which generate the MGMT-repairable O(6)-alkyl adducts in DNA. These results demonstrate that p53 is a negative regulator of MGMT gene expression and can create a MGMT-depleted state in human tumors similar to that achieved by O(6)-benzylguanine, a potent inhibitor of MGMT currently undergoing clinical trials. Thus, our study exposes an additional benefit associated with p53 gene therapy and provides a strong biochemical rationale for combining the MGMT-directed alkylators with p53 gene transfer to achieve improved antitumor efficacy.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , O(6)-Metilguanina-ADN Metiltransferasa/genética , Proteína p53 Supresora de Tumor/fisiología , Adenosina Trifosfato/metabolismo , Apoptosis/genética , División Celular/efectos de los fármacos , Línea Celular , Ensayos de Selección de Medicamentos Antitumorales , Técnica del Anticuerpo Fluorescente/métodos , Regulación Enzimológica de la Expresión Génica , Silenciador del Gen , Humanos , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Péptido Hidrolasas/metabolismo , ARN Mensajero/metabolismo , Transcripción Genética , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética , Ubiquitinas/metabolismo
9.
Brain Pathol ; 3(3): 229-35, 1993 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-8293182

RESUMEN

The p53 tumor suppressor gene is frequently mutated in glioblastomas. Mutations within the p53 gene often result in aberrant expression of the p53 protein leading to protein accumulation within the nucleus of the cells which can be detected by immunochemistry. Many studies have correlated alterations of p53 protein expression with p53 gene mutations. Positive staining of tumor cells for p53 protein has been widely assumed, perhaps incorrectly, to signify the presence of p53 gene mutations. This study compared the immunostaining patterns for p53 protein in 37 glioblastomas with the molecular genetic data obtained by the single strand conformation polymorphism assay. p53 gene mutations were detected in 46% (17 of 37) of glioblastomas, while 65% (24 of 37) of glioblastomas were positive for protein accumulation by immunohistochemistry. Although 30 of 37 glioblastomas analyzed showed concordance for p53 protein expression and p53 gene mutations, a subset of seven glioblastomas showed discordant accumulation of the p53 protein in the absence of any detectable p53 gene mutations. The mdm-2 gene was assessed in 17 glioblastomas for gene rearrangements or amplification, but none were found. This result suggests that a mechanism other than p53 gene mutation can result in altered p53 protein expression.


Asunto(s)
Neoplasias Encefálicas/genética , Genes p53 , Glioblastoma/genética , Mutación , Proteína p53 Supresora de Tumor/genética , Anticuerpos Monoclonales/inmunología , Secuencia de Bases , Southern Blotting , Neoplasias Encefálicas/metabolismo , Exones , Amplificación de Genes , Glioblastoma/metabolismo , Humanos , Inmunohistoquímica , Datos de Secuencia Molecular , Polimorfismo Genético , Proteína p53 Supresora de Tumor/metabolismo
10.
Neuro Oncol ; 3(3): 193-200, 2001 07.
Artículo en Inglés | MEDLINE | ID: mdl-11465400

RESUMEN

Stereotactic biopsy is often performed for diagnostic purposes before treating patients whose imaging studies highly suggest glioma. Indications cited for biopsy include diagnosis and/or the "inoperability" of the tumor. This study questions the routine use of stereotactic biopsy in the initial management of gliomas. At The University of Texas M. D. Anderson Cancer Center, we retrospectively reviewed a consecutive series of 81 patients whose imaging studies suggested glioma and who underwent stereotactic biopsy followed by craniotomy/resection (within 60 days) between 1993 and 1998. All relevant clinical and imaging information was reviewed, including computerized volumetric analysis of the tumors based on pre- and postoperative MRI. Stereotactic biopsy was performed at institutions other than M. D. Anderson in 78 (96%) of 81 patients. The majority of tumors were located either in eloquent brain (36 of 81 = 44%) or near-eloquent brain (41 of 81 = 51%), and this frequently was the rationale cited for performing stereotactic biopsy. Gross total resection (>95%) was achieved in 46 (57%) of 81 patients, with a median extent of resection of 96% for this series. Diagnoses based on biopsy or resection in the same patient differed in 40 (49%) of 82 cases. This discrepancy was reduced to 30 (38%) of 80 cases when the biopsy slides were reviewed preoperatively by each of three neuropathologists at M. D. Anderson. Major neurologic complications occurred in 10 (12.3%) of 81 surgical patients and 3 (3.7%) of 81 patients undergoing biopsy. Surgical morbidity was probably higher in our series than it would be for glioma patients in general because our patients represent a highly selected subset of glioma patients whose tumors present a technical challenge to remove. Stereotactic biopsy is frequently inaccurate in providing a correct diagnosis and is associated with additional risk and cost. If stereotactic biopsy is performed, expert neuropathology consultation should be sought.


Asunto(s)
Neoplasias Encefálicas/patología , Glioma/patología , Técnicas Estereotáxicas , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Biopsia/métodos , Neoplasias Encefálicas/cirugía , Femenino , Glioma/cirugía , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Estudios Retrospectivos , Factores de Riesgo , Sensibilidad y Especificidad , Resultado del Tratamiento
11.
Biotechniques ; 31(1): 196-202, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11464512

RESUMEN

In an initial effort to determine the effect of expressing potentially therapeutic gene products on the growth properties of glioma tumor xenografts, we describe the development of cell lines that can conditionally express beta-galactosidase (beta-gal). To achieve this, we generated stable cell lines that express the modified tetracycline repressor molecule (rtTA) and the beta-gal gene under control of tetracycline-responsive cis-elements. The resulting cell lines express functional beta-gal following treatment with the tetracycline analog doxycyclin (Dox). These cells were then used to form intracranial tumors after injection into the brain using an implantable guide-screw system. The xenografts were found to express beta-gal when the animals were fed drinking water containing Dox. From these studies, we conclude that the expression of a target gene in a human xenograft growing in the brain of a living mouse can be conditionally regulated.


Asunto(s)
Neoplasias Encefálicas/genética , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Animales , Antibacterianos , Doxiciclina , Genes Reporteros , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Trasplante Heterólogo , Células Tumorales Cultivadas , beta-Galactosidasa/genética
12.
Neurosurgery ; 45(5): 1093-104, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10549925

RESUMEN

OBJECTIVE: The rationale and current evidence for using p53 gene replacement as a potential treatment for human gliomas are reviewed. The possible benefits of and obstacles to this approach are delineated. METHODS: One approach to overcoming the poor outcomes associated with conventional glioma therapies involves the replacement of tumor suppressor genes that are fundamental to glioma development. The p53 gene is one of the most frequently mutated genes in human gliomas, and loss of p53 function is critical to the development of glial neoplasms. Consequently, replacement of the p53 gene using viral vectors may be a potential treatment for human gliomas. RESULTS: In vitro studies demonstrate that adenovirus-mediated p53 gene transfer into gliomas with mutant p53 results in massive apoptosis. Similarly, transfer of p53 inhibits tumor growth in vivo. In contrast to mutant p53 gliomas, wild-type p53 glioma cells are resistant to the apoptotic effects of p53 transfer, but this resistance can be overcome by the addition of deoxyribonucleic acid-damaging agents such as ionizing radiation or chemotherapy. The main obstacle to p53 gene therapy involves the limitations associated with current modes of delivery. CONCLUSION: Preclinical data strongly support the use of p53 gene transfer as a potential treatment for human gliomas.


Asunto(s)
Neoplasias Encefálicas/terapia , Técnicas de Transferencia de Gen , Terapia Genética , Glioma/terapia , Proteína p53 Supresora de Tumor/genética , Adenoviridae/genética , Apoptosis/genética , Neoplasias Encefálicas/genética , Terapia Combinada , Vectores Genéticos/genética , Glioma/genética , Humanos , Resultado del Tratamiento
13.
Neurosurgery ; 34(5): 847-53; discussion 853, 1994 May.
Artículo en Inglés | MEDLINE | ID: mdl-8052381

RESUMEN

Many neurosurgeons have made a practice of sectioning the S2 dorsal roots during selective posterior rhizotomy for the treatment of spasticity in children with cerebral palsy, but the efficacy of this treatment has not previously been proven. S2 afferents are involved in reflex arcs of the plantar flexors (PFs), so that S2 lesioning should in theory reduce PF spasticity. To test this assumption, we determined the frequency of postoperative residual spasticity in the PFs when S2 lesioning was or was not performed. We assessed 85 children for whom 6-month follow-up was available. Functional rhizotomy from L2-S1 was performed on 13 of them (26 legs with PF spasticity) and from L2-S2 on 72 (141 legs with PF spasticity). Rootlets were lesioned if there was an abnormal response to intraoperative electrical stimulation. In 20 patients, lesioning of the S2 rootlets was assisted by the "pudendal neurogram," a technique previously shown to prevent bladder dysfunction during sectioning of the sacral roots. When S2 roots were excluded from the lesioning process, residual PF spasticity was detected in 35% of the legs that had it preoperatively, leaving 5 (38%) of 13 children with functionally impairing spasticity. When S2 roots were included, 6% of legs that had PF spasticity retained it postoperatively (P < 0.001), leaving 8 (11%) of 72 patients with functionally limiting spasticity (P < 0.05). Thus, the addition of the S2 roots to the procedure resulted in an 81% reduction in the number of legs with residual PF spasticity and a 71% reduction in the number of patients with residual PF spasticity.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Parálisis Cerebral/cirugía , Ganglios Espinales/cirugía , Espasticidad Muscular/cirugía , Raíces Nerviosas Espinales/cirugía , Vías Aferentes/fisiopatología , Parálisis Cerebral/fisiopatología , Niño , Preescolar , Electromiografía , Femenino , Estudios de Seguimiento , Ganglios Espinales/fisiopatología , Humanos , Pierna/inervación , Masculino , Neuronas Motoras/fisiología , Contracción Muscular , Espasticidad Muscular/fisiopatología , Músculos/inervación , Examen Neurológico , Modalidades de Fisioterapia , Complicaciones Posoperatorias/diagnóstico , Complicaciones Posoperatorias/fisiopatología , Rango del Movimiento Articular/fisiología , Reflejo Anormal/fisiología , Estudios Retrospectivos , Raíces Nerviosas Espinales/fisiopatología , Vejiga Urinaria/inervación
14.
Neurosurgery ; 48(4): 745-54; discussion 754-5, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11322434

RESUMEN

OBJECTIVE: Few reports have addressed the surgical management of cranial metastases that overlie or invade the dural venous sinuses. To examine the role of surgery in the treatment of dural sinus calvarial metastases, we reviewed retrospectively 13 patients who were treated with surgery at the University of Texas M.D. Anderson Cancer Center between 1993 and 1999. We compared them with 14 patients who had calvarial metastases that did not involve a venous sinus. METHODS: Clinical charts, radiological studies, pathological findings, and operative reports were analyzed retrospectively. RESULTS: The median age of patients with dural sinus calvarial metastases was 54 years. Nine patients were men and four were women. Renal cell carcinoma and sarcoma were the most common primary tumors. Similar features were noted in the 14 patients with nonsinus calvarial metastases. Of the 13 dural sinus calvarial metastases, 11 involved the superior sagittal sinus, and 2 involved the transverse sinus. In nine patients, the involved sinus was resected, and in four patients, the sinus was reconstituted after tumor removal. Nine patients in the dural sinus calvarial metastases group received en bloc resection, and four received piecemeal resection. No operative deaths occurred. The overall median actuarial survival was 16.5 months. The survival times of the two groups were comparable. In the group with dural sinus calvarial metastases, transient postoperative neurological deficits occurred in two patients (15%), and a permanent deficit occurred in one patient (8%). No patients in the group with nonsinus calvarial metastases experienced deficits after resection. Compared with piecemeal resection, en bloc resection was associated with significantly less blood loss. CONCLUSION: Complete extirpation of calvarial metastases that overlie or invade a dural sinus can be achieved with only slightly more morbidity than complete removal of calvarial metastases that are located away from the sinuses. En bloc resection is as safe as piecemeal resection and is more effective in limiting operative blood loss. The overall recurrence and survival rates of patients with dural sinus calvarial metastases are similar to those of patients with calvarial metastases that do not involve the sinuses. Therefore, involvement of a dural venous sinus should not discourage resection of calvarial metastases. In carefully selected cancer patients, surgery provides effective palliation of symptomatic calvarial metastases that overlie or invade the venous sinuses.


Asunto(s)
Senos Craneales/cirugía , Duramadre/cirugía , Neoplasias Craneales/secundario , Adulto , Anciano , Carcinoma de Células Renales/mortalidad , Carcinoma de Células Renales/patología , Carcinoma de Células Renales/cirugía , Senos Craneales/patología , Duramadre/patología , Femenino , Estudios de Seguimiento , Humanos , Neoplasias Renales/mortalidad , Neoplasias Renales/patología , Neoplasias Renales/cirugía , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Complicaciones Posoperatorias/etiología , Complicaciones Posoperatorias/mortalidad , Sarcoma/mortalidad , Sarcoma/patología , Sarcoma/secundario , Sarcoma/cirugía , Neoplasias Craneales/mortalidad , Neoplasias Craneales/patología , Neoplasias Craneales/cirugía , Tasa de Supervivencia
15.
J Neurosurg ; 93(6): 940-50, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11117866

RESUMEN

OBJECT: Primary meningiomas arising outside the intracranial compartment (primary extradural meningiomas [PEMs]) are rare tumors. To develop a better understanding of these tumors and to establish a comprehensive classification scheme for them, the authors analyzed a series of patients treated at the M. D. Anderson Cancer Center (MDACC) and reviewed all cases reported in the English-language literature since the inception of the use of computerized tomography (CT) scanning. METHODS: Clinical records, results of radiographic studies, and histological slides were reviewed for all cases of PEM at MDACC. Demographic features, symptoms, tumor location, histological grade, and patient outcome were assessed in all cases. A comprehensive literature search identified 168 PEMs in 142 patients reported during the CT era. These reports were also analyzed for common features. Tumors for both data sets were classified as purely extracalvarial (Type I), purely calvarial (Type II), and calvarial with extracalvarial extension (Type III). Type II and Type III tumors were further categorized as convexity (C) or skull base (B) lesions. The incidence of PEMs at MDACC was 1.6%, which was consistent with the rate reported in the literature. In both data sets, the male/female ratio was nearly 1:1. The most common presenting symptom was a gradually expanding mass. The age of patients at diagnosis of PEM was bimodal, peaking during the second decade and during the fifth to seventh decades. In all MDACC cases and in 90% of those reported in the literature the PEMs were located in the head and neck. The majority of tumors originated in the skull (70%). In the MDACC series and in the literature review, the majority (67% and 89%, respectively) of tumors were histologically benign. Although fewer PEMs were malignant or atypical (33% at MDACC and 11% in the literature), their incidence was higher than that observed for primary intracranial meningiomas. Distant metastasis was not a common feature reported for patients with PEMs (6% in the literature). Outcome data were available in 96 of the cases culled from the CT-era literature. The combination of the MDACC data and the data obtained from the literature demonstrated that patients with benign Type IIB or Type IIIB lesions were more likely to experience recurrence than patients with benign Type IIC or Type IIIC tumors (26% compared with 0%, p < 0.05). The more aggressive atypical and malignant tumors were associated with a statistically significant higher death rate (29%) relative to benign tumors (4.8% death rate, p < 0.004). CONCLUSIONS: Defining a tumor as a PEM is dependent on the tumor's relation to the dura mater and the extent and direction of its growth. Classification of PEMs as calvarial or extracalvarial and as convexity or skull base lesions correlates well with clinical outcome.


Asunto(s)
Neoplasias Meníngeas/cirugía , Meningioma/cirugía , Tomografía Computarizada por Rayos X , Adolescente , Adulto , Anciano , Craneotomía , Espacio Epidural , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Neoplasias Meníngeas/clasificación , Neoplasias Meníngeas/diagnóstico , Meningioma/clasificación , Meningioma/diagnóstico , Persona de Mediana Edad , Estudios Retrospectivos , Resultado del Tratamiento
16.
J Neurosurg ; 81(3): 427-36, 1994 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-8057151

RESUMEN

To characterize some of the genetic events underlying the development of glioblastoma multiforme, the authors analyzed 65 astrocytic tumors (seven pilocytic astrocytomas, eight astrocytomas, 16 anaplastic astrocytomas, and 34 glioblastomas multiforme) for loss of heterozygosity for chromosome 17p, loss of heterozygosity for chromosomes 10p and 10q, amplification of the epidermal growth factor receptor (EGFR) gene, and amplification of the oncogenes N-myc, c-myc, and N-ras using Southern blot analysis. Alterations of the p53 gene (positive immunostaining for p53 protein in tumors with or without p53 gene mutations) in these 65 tumors were analyzed previously. None of the 65 tumors showed amplification or rearrangement of N-myc, c-myc, or N-ras oncogenes. The molecular analysis presented here demonstrates distinct variants of astrocytic tumors, with at least three genetic pathways leading to glioblastoma multiforme. One pathway was characterized by 43 astrocytomas with alterations in p53. Glioblastomas with p53 alterations may represent tumors that progress from lower-grade astrocytomas. This variant was more likely to show loss of chromosome 17p than tumors without p53 alterations (p < 0.04). Seventy-five percent of tumors with loss of one 17p allele demonstrated mutations in the p53 gene. Loss of chromosome 10 was associated with progression from anaplastic astrocytoma (13%) to glioblastoma (38%) (p < 0.04). Amplification of the EGFR gene was a rare (7%) but late event in tumor progression (p < 0.03). A second pathway was characterized by six astrocytomas without p53 alterations and may represent clinically de novo high-grade tumors. These tumors were more likely to show amplification of the EGFR gene (83%) than tumors with p53 alterations. Sixty percent of tumors with EGFR amplification also showed loss of chromosome 10; loss of chromosome 17p was infrequent in this variant. One or more alternative pathways were characterized by 16 astrocytomas without p53 alterations and with none of the genetic changes analyzed in this study. Glioblastomas are a heterogeneous group of tumors that may arise via multiple genetic pathways.


Asunto(s)
Astrocitoma/genética , Neoplasias Encefálicas/genética , Glioblastoma/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Preescolar , Deleción Cromosómica , Cromosomas Humanos Par 10 , Cromosomas Humanos Par 17 , ADN de Neoplasias/análisis , Receptores ErbB/genética , Femenino , Amplificación de Genes , Genes myc/genética , Genes p53/genética , Genes ras/genética , Heterocigoto , Humanos , Masculino , Persona de Mediana Edad , Mutación
17.
J Neurosurg ; 79(6): 859-66, 1993 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-8246054

RESUMEN

Histopathological features that suggest the diagnosis of ganglioglioma require, in most cases, confirmation by special stains to distinguish these tumors from other gliomas. For this purpose, immunostaining for synaptophysin, which has previously been shown to selectively label the cell surface of neoplastic ganglion cells, was used to retrospectively examine glioma tumor specimens. Sixty-three cases of ganglioglioma were identified. The files of the Division of Neuropathology of New York University Medical Center contained 45 tumors that had been diagnosed as ganglioglioma, of which 42 were verified by synaptophysin; three cases were reclassified, two as astrocytomas and one as a gangliocytic paraganglioma. Thus, a tumor identified as ganglioglioma based on other criteria was likely to be a ganglioglioma. The other 21 cases of gangliogliomas were originally diagnosed as astrocytoma or mixed glioma, but were shown by synaptophysin staining to be gangliogliomas. In some cases the ultimate diagnosis was obtained after radical surgery provided relatively abundant amounts of tissue, thereby limiting sampling errors, in contrast to the biopsies from which the original diagnoses were made. Histopathological review of these cases demonstrated that four features represent important clues to the correct diagnosis: 1) clusters of large cells potentially representing neurons (without such cells the tumor cannot be classified as a ganglioglioma); 2) no perineuronal clustering of the glial cells around the alleged neoplastic neurons; 3) fibrosis (desmoplasia); and 4) calcification. Binucleate neurons, previously suggested to be common in gangliogliomas, were not frequently found in this series, and lymphocytic infiltrates, while common, are so often found in other tumors that they gave no specific hint that any single neoplasm was a ganglioglioma. The glial elements were astrocytic in all cases, except that one tumor also had oligodendroglial and ependymal patterns. Four tumors also had small mature neurons, as seen in neurocytomas. Cells from one tumor were successfully grown in short-term tissue culture; the culture contained large dividing neurons with synaptophysin immunoreactivity as well as smaller dividing cells, demonstrating that the neuronal cells are a proliferating element in gangliogliomas.


Asunto(s)
Neoplasias del Sistema Nervioso Central/patología , Ganglioglioma/patología , Adulto , Núcleo Celular/ultraestructura , Niño , Preescolar , Citoplasma/ultraestructura , Errores Diagnósticos , Humanos , Masculino , Neuritas/ultraestructura , Estudios Retrospectivos , Células Tumorales Cultivadas
18.
J Neurosurg ; 92(2): 326-33, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10659021

RESUMEN

OBJECT: To overcome the problems associated with using stereotactic techniques to establish intracranial xenografts in nude mice and to treat engrafted tumors with intratumoral therapies (such as gene or viral therapies), the authors developed an implantable guide-screw system. In this study, they describe the guide-screw system, its method of implantation, and their experience with establishing xenografts and delivering intratumoral therapy. METHODS: The system consists of a 2.6-mm guide screw with a central 0.5-mm diameter hole that accepts the 26-gauge needle of a Hamilton syringe. The screw is implanted into a small drill hole made 2.5 mm lateral and 1 mm anterior to the bregma. A stylet is used to cap the screw between treatments. Tumor cells or therapeutic agents are injected in a freehand fashion by using a Hamilton syringe and a 26-gauge needle fitted with a cuff to determine the depth of injection. To test this system, guide screws were successfully implanted in 44 (98%) of 45 nude mice. After 1 to 2 weeks of recovery, 38 mice were inoculated with U87MG cells and killed 5 days later. On histological studies in 37 (97%) of these animals, xenografts were evident within the caudate nucleus (mean diameter 2.5 mm). To determine whether injections into the center of an established xenograft could be reproducibly achieved with the guide-screw system, an adenovirus vector containing the beta-galactosidase gene was injected 3 days after cell implantation in 15 of the mice. All of these animals demonstrated transduced cells within the tumor. To demonstrate that engrafted animals have a uniform survival time that is indicative of reproducible tumor growth, the survival of six mice was assessed after engraftment with U87MG cells. All six animals died within 28 to 35 days. CONCLUSIONS: The guide-screw system allows a large number of animals to be rapidly and reproducibly engrafted and for intratumoral treatments to be accurately delivered into established xenografts.


Asunto(s)
Tornillos Óseos , Neoplasias Encefálicas/patología , Glioma/patología , Trasplante de Neoplasias/instrumentación , Técnicas Estereotáxicas/instrumentación , Animales , Encéfalo/patología , Neoplasias Encefálicas/terapia , Femenino , Terapia Genética , Glioma/terapia , Ratones , Ratones Desnudos , Células Tumorales Cultivadas
19.
J Neurosurg ; 89(1): 125-32, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9647183

RESUMEN

OBJECT: The authors sought to determine whether combining p53 gene transfer with radiation therapy would enhance the therapeutic killing of p53 wild-type glioma cells. It has been shown in several reports that adenovirus-mediated delivery of the p53 gene into p53 mutant gliomas results in dramatic apoptosis, but has little effect on gliomas containing wild-type p53 alleles. Therefore, p53 gene therapy alone may not be a clinically effective treatment for gliomas because most gliomas are composed of both p53 mutant and wild-type cell populations. One potential approach to overcome this problem is to exploit the role p53 plays as an important determinant in the cellular response to ionizing radiation. METHODS: In vitro experiments were performed using the glioma cell line U87MG, which contains wild-type p53. Comparisons were made to the glioma cell line U251MG, which contains a mutant p53 allele. Monolayer cultures were infected with an adenovirus containing wild-type p53 (Ad5CMV-p53), a control vector (dl312), or Dulbecco's modified Eagle's medium (DMEM). Two days later, cultures were irradiated and colony-forming efficiency was determined. Transfection with p53 had only a minor effect on the plating efficiency of nonirradiated U87MG cells, reducing the plating efficiency from 0.23 +/- 0.01 in DMEM to 0.22 +/- 0.04 after addition of Ad5CMV-p53. However, p53 transfection significantly enhanced the radiosensitivity of these cells. The dose enhancement factor at a surviving fraction of 0.10 was 1.5, and the surviving fraction at 2 Gy was reduced from 0.61 in untransfected controls to 0.38 in p53-transfected cells. Transfection of the viral vector control (dl312) had no effect on U87MG radiosensitivity. In comparison, transfection of Ad5CMV-p53 into the p53 mutant cell line U251 MG resulted in a significant decrease in the surviving fraction of these cells compared with controls, and no radiosensitization was detected. To determine whether Ad5CMV-p53-mediated radiosensitization of U87MG cells involved an increase in the propensity of these cells to undergo apoptosis, flow cytometric analysis of terminal deoxynucleotidyl transferase-mediated biotinylated-deoxyuridinetriphosphate nick-end labeling-stained cells was performed. Whereas the amount of radiation-induced apoptosis in uninfected and dl312-infected control cells was relatively small (2.1 +/- 0.05% and 3.7 +/- 0.5%, respectively), the combination of Ad5CMV-p53 infection and radiation treatment significantly increased the apoptotic frequency (18.6 +/- 1.4%). To determine whether infection with Ad5CMV-p53 resulted in increased expression of functional exogenous p53 protein, Western blot analysis of p53 was performed on U87MG cells that were exposed to 9 Gy of radiation 2 days after exposure to Ad5CMV-p53, dl312, or DMEM. Infection with Ad5CMV-p53 alone increased p53 levels compared with DMEM- or dl312-treated cells. Irradiation of AdSCMV-p53-infected cells resulted in a further increase in p53 that reached a maximum at 2 hours postirradiation. To determine whether exogenous p53 provided by Ad5CMV-p53 had transactivating activity, U87MG cells were treated as described earlier and p21 messenger RNA levels were determined. Infection of U87MG cells with Ad5CMV-p53 only resulted in an increase in p21 compared with DMEM- and dl312-treated cells. Irradiation of AdSCMV-p53-infected cells resulted in an additional time-dependent increase in p21 expression. CONCLUSIONS: These data indicate that adenovirus-mediated delivery of p53 may enhance the radioresponse of brain tumor cells containing wild-type p53 and that this radiosensitization may involve converting from a clonogenic to the more sensitive apoptotic form of cell death. Although the mechanism underlying this enhanced apoptotic susceptibility is unknown, the AdSCMV-p53-infected cells have a higher level of p53 protein, which increases further after irradiation, and this exogenous p53 is transcriptionally active. (ABSTRACT TRUNCATE


Asunto(s)
Adenoviridae/genética , Neoplasias Encefálicas/genética , Técnicas de Transferencia de Gen , Genes p53/efectos de la radiación , Vectores Genéticos , Glioma/genética , Tolerancia a Radiación , Alelos , Apoptosis/genética , Apoptosis/efectos de la radiación , Western Blotting , Neoplasias Encefálicas/radioterapia , División Celular/genética , Supervivencia Celular , Colorantes , Medios de Cultivo , Relación Dosis-Respuesta en la Radiación , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Glioma/radioterapia , Humanos , Mutación/genética , Células Madre Neoplásicas/efectos de la radiación , Proteínas Proto-Oncogénicas p21(ras)/análisis , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/efectos de la radiación , ARN Mensajero/análisis , ARN Mensajero/genética , ARN Mensajero/efectos de la radiación , Dosificación Radioterapéutica , Transcripción Genética/genética , Transcripción Genética/efectos de la radiación , Transfección/genética , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/análisis , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/efectos de la radiación
20.
J Neurosurg ; 94(1 Suppl): 25-37, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11147865

RESUMEN

OBJECT: Few reports are available on the use of pedicle screw fixation for cancer-related spinal instability. The authors present their experience with pedicle screw fixation in the management of malignant spinal column tumors. METHODS: Records for patients with malignant spinal tumors who underwent pedicle screw fixation at the authors' institution between September 1994 and December 1999 were retrospectively reviewed. RESULTS: Ninety-five patients with malignant spinal tumors underwent 100 surgeries involving pedicle screw fixation: metastatic spinal disease was present in 81 patients, and locally invasive tumors were demonstrated in 14 patients. Indications for surgery were pain (98%) and/or neurological dysfunction (80%). A posterior (48%) or a combined anterior-posterior (52%) approach was performed depending on the extent of tumor and the patient's condition. At the mean follow up of 8.2 months, 43 patients (45%) had died; median survival, as determined by Kaplan-Meier analysis, was 14.8 months. At I month postsurgery, self-reported pain had improved in 87% of cases (p < 0.001), which is a finding substantiated by reductions in analgesic use, and 29 (47%) of 62 patients with preoperative neurological impairments were functionally improved (p < 0.001). Postoperative complications were associated only with preoperative radiation therapy (p = 0.002) and with preexisting serious medical conditions (p = 0.04). In two patients asymptomatic violation of the lateral wall of the pedicle was revealed on postoperative radiography. The 30-day mortality rate was 1%. CONCLUSIONS: For selected patients with malignant spinal tumors, pedicle screw fixation after tumor resection may provide considerable pain relief and restore or preserve ambulation with acceptable rates of morbidity and mortality.


Asunto(s)
Tornillos Óseos , Neoplasias de la Columna Vertebral/cirugía , Adolescente , Adulto , Anciano , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Sistema Nervioso/fisiopatología , Dolor/fisiopatología , Complicaciones Posoperatorias , Periodo Posoperatorio , Cuidados Preoperatorios/efectos adversos , Radioterapia/efectos adversos , Estudios Retrospectivos , Neoplasias de la Columna Vertebral/diagnóstico , Neoplasias de la Columna Vertebral/fisiopatología , Neoplasias de la Columna Vertebral/secundario , Análisis de Supervivencia , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA