Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 109
Filtrar
2.
Transplant Proc ; 39(1): 125-31, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17275488

RESUMO

INTRODUCTION: In dialysis patients, the parathyroid glands (PTGs) may increase progressively, producing abnormal bone metabolism. Changes in PTG volume among patients with hyperplastic PTGs are not well known after kidney transplantation. This study investigated PTG volume by ultrasound (US). METHODS: US of PTG was performed immediately (US-0) and 12 months after (US-12) transplantation to identify glands in all recipients. We calculated the percentage reduction in PTG volume (R%PTG). We declared it significant when it was > or =35%. Bone biochemical markers and renal function were recorded sequentially. RESULTS: Among engrafted patients, parathyroid US-0 was performed in 47 and US-0 and US-12 in 36. Some visible gland was observed upon US-0 in 13 recipients, a group that showed higher pretransplantation parathyroid hormone (PTH) levels than the remaining 34 patients with no visible glands (627 +/- 360.0 vs 280 +/- 240.9 pg/mL; P < .05). Of 36 recipients with US-0 and US-12, the baseline study identified PTGs in 12 patients (p+ group), while the remaining 24 had no identified glands (p- group). In the p+ group, no PTG, at US-12 were visible in four patients, and a significant R%PTG was observed in three at this time, representing a reduction in gland volume after transplantation among 58.3% of p+ patients. There was a progressive reduction in PTH among both groups. Patients with glandular volume reduction displayed better renal function: serum creatinine 1.7 +/- .79 versus 2.9 +/- .74 mg/dL (P < .05). CONCLUSIONS: Transplantation reversed hyperparathyroidism and PTG volume among recipients who achieved nearly normal renal function.


Assuntos
Hiperparatireoidismo Secundário/prevenção & controle , Transplante de Rim/fisiologia , Adulto , Feminino , Humanos , Hiperparatireoidismo Secundário/patologia , Falência Renal Crônica/complicações , Falência Renal Crônica/cirurgia , Masculino , Pessoa de Meia-Idade , Glândulas Paratireoides/patologia , Hormônio Paratireóideo/sangue , Estudos Prospectivos , Doadores de Tecidos/estatística & dados numéricos
3.
Nefrologia ; 24 Suppl 3: 49-55, 2004.
Artigo em Espanhol | MEDLINE | ID: mdl-15219069

RESUMO

The acute renal failure is a grave pathology, of rapid establishment and relatively frequent in the hospital environment. We can describe three etiological groupS, which are responsible for it, amongst which are emphasized the pre-renal reasons. The obstructive pathology, of minor incidence, increases with the age. It is described the case of a 67-yr-old patient who was admitted in the Nephrology Service because of abrupt decline of the renal function. Among the initial symptoms, he presented arterial hypertension (190/90) and preserved diuresis. Blood analysis: urea 199 mg/dl, creatinine 7.7 mg/dl, without proteinuria. Sonography reported a bilateral ureteral hydronephrosis with simple cyst of possible ischemic origin. In view of the absence of previous biochemical data of renal failure, we considered possible reasons which start with an acute pattern. In initial evaluation, pre-renal etiology was not seen (high blood pressure, right cardiac systole function). The absence of prostatic syndrome and sonography discovery did not justify a diagnosis of urinary tract obstruction. Finally, abdominal-pelvic scan showed a periaortic retroperitoneal mass which included both ureters and appeared to trigger the obstruction. Combined efforts were pursued with the Urology Service, which implanted a bilateral "double J" catheter and later operated surgically on the patient, carrying out an alternating ureterolysis of both ureters. The biopsy manifested a retroperitoneal fibrosis, and the renogram showed a residual renal function of 20% in the right kidney and 80% in the left kidney. Due to the failure of the previous measures and as a last therapeutic recourse when one year had passed from the diagnosis, a continuous regimen with tamoxifen (anti-estrogen drug) in dose of 20 mg/dl each 12 hours was started, which began a progressive remission in the size of the observed mass by scan (CT) and magnetic resonance (MR). The treatment was completed during 12 months and in this time, the levels of blood urea nitrogen and creatinine were reduced gradually too. Finally, at the end of the treatment, the magnetic resonance demonstrate the complete disappearance of the fibrosis.


Assuntos
Injúria Renal Aguda/etiologia , Fibrose Retroperitoneal/complicações , Injúria Renal Aguda/sangue , Idoso , Humanos , Hidronefrose/complicações , Hidronefrose/cirurgia , Doenças Renais Císticas/complicações , Imageamento por Ressonância Magnética , Masculino , Fibrose Retroperitoneal/diagnóstico , Fibrose Retroperitoneal/diagnóstico por imagem , Fibrose Retroperitoneal/tratamento farmacológico , Stents , Tamoxifeno/uso terapêutico , Tomografia Computadorizada por Raios X , Ureter/cirurgia , Disfunção Ventricular Esquerda/complicações
4.
Int J Radiat Oncol Biol Phys ; 51(2): 410-8, 2001 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11567815

RESUMO

PURPOSE: Recent studies have suggested relative radioresistance in glioblastoma multiforme (GM) tumors in older patients, consistent with their shorter survival. Two common molecular genetic abnormalities in GM are age related: epidermal growth factor receptor (EGFR) overexpression in older patients and p53 mutations in younger patients. We tested whether these abnormalities correlated with clinical heterogeneity in GM response to radiation treatment. METHODS AND MATERIALS: Radiographically assessed radiation response (5-level scale) was correlated with EGFR immunoreactivity, p53 immunoreactivity, and p53 exon 5-8 mutation status in 170 GM patients treated using 2 prospective clinical protocols. Spearman rank correlation and proportional-odds ordinal regression were used for univariate and multivariate analysis. RESULTS: Positive EGFR immunoreactivity predicted poor radiographically assessed radiation response (p = 0.046). Thirty-three percent of tumors with no EGFR immunoreactivity had good radiation responses (>50% reduction in tumor size by CT or MRI), compared to 18% of tumors with intermediate EGFR staining and 9% of tumors with strong staining. There was no significant relationship between p53 immunoreactivity or mutation status and radiation response. Significant relationships were noted between EGFR score and older age and between p53 score or mutation status and younger age. CONCLUSION: The observed relative radioresistance of some GMs is associated with overexpression of EGFR.


Assuntos
Neoplasias Encefálicas/radioterapia , Receptores ErbB/metabolismo , Genes p53/genética , Glioblastoma/radioterapia , Proteínas de Neoplasias/metabolismo , Neoplasias Supratentoriais/radioterapia , Adolescente , Adulto , Fatores Etários , Idoso , Análise de Variância , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Feminino , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Razão de Chances , Estudos Prospectivos , Dosagem Radioterapêutica , Análise de Regressão , Neoplasias Supratentoriais/genética , Neoplasias Supratentoriais/metabolismo
5.
AJNR Am J Neuroradiol ; 22(4): 613-24, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11290467

RESUMO

BACKGROUND AND PURPOSE: The diagnosis of brain tumors after high-dose radiation therapy is frequently limited by the lack of metabolic discrimination available with conventional imaging methods. The purpose of this study was to use proton MR spectroscopy to investigate serial changes in recurrent malignant gliomas after gamma knife radiosurgery to characterize tissue response to high-dose radiation. METHODS: Eighteen patients with recurrent gliomas were studied with MR imaging and 3D proton MR spectroscopic imaging at the time of radiosurgery and at regular time points thereafter. Choline (Cho) and N-acetyl aspartate levels were calculated on a voxel-by-voxel basis and compared with levels found in normal tissue and with levels observed at previous time points. The results of the spectral analysis were then compared with the radiologic findings. Statistical comparisons were precluded by the small sample sizes involved. RESULTS: Response within the gamma knife target was observed as a reduction of Cho levels and an increase in lactate/lipid levels, typically within 6 months of treatment. Increases in Cho correlated with poor radiologic response and suggested tumor recurrence, confirmed histologically in six cases. The development of a spectral abnormality preceded a coincident increase in contrast enhancement by 1 to 2 months in nine cases. CONCLUSION: Proton MR spectroscopic imaging provided diagnostic and monitoring information before and after radiosurgery. Evaluation of metabolic changes with proton MR spectroscopy and structural changes with MR imaging improved tissue discrimination and provided correlation with histologic findings.


Assuntos
Neoplasias Encefálicas/diagnóstico , Metabolismo Energético/fisiologia , Glioma/diagnóstico , Processamento de Imagem Assistida por Computador , Imageamento por Ressonância Magnética , Espectroscopia de Ressonância Magnética , Recidiva Local de Neoplasia/diagnóstico , Radiocirurgia , Adulto , Ácido Aspártico/análogos & derivados , Ácido Aspártico/metabolismo , Encéfalo/fisiopatologia , Encéfalo/cirurgia , Neoplasias Encefálicas/fisiopatologia , Neoplasias Encefálicas/cirurgia , Colina/metabolismo , Feminino , Seguimentos , Glioma/fisiopatologia , Glioma/cirurgia , Humanos , Ácido Láctico/metabolismo , Metabolismo dos Lipídeos , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/fisiopatologia , Recidiva Local de Neoplasia/cirurgia , Complicações Pós-Operatórias/diagnóstico , Complicações Pós-Operatórias/fisiopatologia , Complicações Pós-Operatórias/cirurgia
6.
Cancer ; 91(2): 417-22, 2001 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-11180089

RESUMO

BACKGROUND: The primary objective of the current study was to determine the response rate of paclitaxel in patients with recurrent malignant glioma by using different doses dependent on the concomitant use of anticonvulsants. Secondary objectives were to determine the time period to treatment failure, to evaluate toxicities, and to obtain pharmacokinetic data. METHODS: Adult patients who had recurrent malignant glioma were treated with paclitaxel. Patients were treated at different doses depending on the concomitant use of anticonvulsants known to induce the p450 hepatic enzyme system. Patients on such agents were treated at a dose of 330 mg/m2, whereas those not on these anticonvulsants were treated at a dose of 210 mg/m2. Tumor response was assessed at 6-week intervals. Treatment was continued until documented tumor progression or unacceptable toxicity occurred, or a total of 12 paclitaxel infusions was completed. RESULTS: From January 1997 to June 1997, 23 patients were treated with paclitaxel. Four patients were ineligible for the current study. Of the 19 eligible patients, there were no responses seen. Four (21%) had stabilization of disease. Median time to treatment failure was 1 month (95% confidence interval [CI], 1-2 mos) and median survival was 7 months (95% CI, 6-10 mos). Three patients were removed from the current study because they had toxicity. Pharmacokinetic studies demonstrated that drug levels and clearance values were consistent with previously reported findings. CONCLUSION: Even though higher doses were administered to patients who had recurrent malignant glioma and who were on concomitant anticonvulsants, there were no objective responses to paclitaxel. Time to tumor progression was 1 month. Further testing of paclitaxel at this dose schedule does not appear to be warranted in this patient population.


Assuntos
Anticonvulsivantes/administração & dosagem , Antineoplásicos Fitogênicos/administração & dosagem , Neoplasias Encefálicas/tratamento farmacológico , Glioma/tratamento farmacológico , Paclitaxel/administração & dosagem , Adolescente , Adulto , Antineoplásicos Fitogênicos/efeitos adversos , Antineoplásicos Fitogênicos/farmacocinética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidade , Intervalos de Confiança , Feminino , Glioma/metabolismo , Glioma/mortalidade , Humanos , Injeções Intravenosas , Masculino , Pessoa de Meia-Idade , Paclitaxel/efeitos adversos , Paclitaxel/farmacocinética , Falha de Tratamento
7.
Int J Radiat Oncol Biol Phys ; 49(1): 71-7, 2001 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-11163499

RESUMO

PURPOSE: To report the results of a prospective Phase III trial for patients with newly diagnosed glioblastoma multiforme (GBM), treated with either accelerated hyperfractionated irradiation with or without difluromethylornithine (DFMO) or standard fractionated irradiation with or without DFMO. METHODS AND MATERIALS: Adult patients with newly diagnosed GBM were registered and randomized following surgery to one of 4 treatment arms: Arm A, accelerated hyperfractionation alone using 2 fractions a day of 1.6 Gy to a total dose of 70.4 Gy in 44 fractions; Arm B, accelerated hyperfractionation as above plus DFMO 1.8 gm/m2 by mouth every 8 h beginning one week before radiation until the last fraction was given; Arm C, single-fraction irradiation of 1.8 Gy/day to 59.4 Gy; Arm D, single-fraction irradiation as in Arm C plus DFMO given as in Arm B. Patients were followed for progression-free survival (PFS) and overall survival (OS), as well as for toxicity. Eligibility required histologically proven GBM, age > or =18, Karnofsky performance status (KPS) > or =60, and no prior chemotherapy or radiotherapy. Adjuvant chemotherapy was not used in this protocol. RESULTS: A total of 231 eligible patients were enrolled. There were 95 men and 136 women with a median age of 57 years, and median KPS of 90. Extent of resection was total in 23, subtotal in 152, and biopsy only in 56 patients. The 4 arms were balanced with respect to age, KPS, and extent of resection. Times to event measurements are from date of diagnosis. Median OS and PFS were 40 and 19 weeks for Arm A; 42 and 22 weeks for Arm B; 37 and 16 weeks for Arm C; and 44 and 19 weeks for Arm D (p = 0.48 for survival; p = 0.32 for PFS). Comparison of the 2 arms treated with DFMO to the 2 arms without DFMO revealed no difference in OS (37 weeks vs. 42 weeks, p = 0.12) or PFS and thus no benefit to the use of DFMO. Comparison of the 2 standard fractionation arms to the 2 accelerated hyperfractionation arms also resulted in no difference in OS (42 weeks vs. 41 weeks, p = 0.75) or PFS, showing no benefit to accelerated hyperfractionated irradiation. CONCLUSION: In this prospective Phase III study, no survival or PFS benefit was seen with accelerated hyperfractionated irradiation to 70.4 Gy, nor was any benefit seen with DFMO as a radiosensitizer. Standard fractionated irradiation to 59.4 Gy remains the treatment of choice for newly diagnosed patients with glioblastoma multiforme.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/radioterapia , Eflornitina/uso terapêutico , Glioblastoma/tratamento farmacológico , Glioblastoma/radioterapia , Radiossensibilizantes/uso terapêutico , Adulto , Idoso , Neoplasias Encefálicas/cirurgia , Terapia Combinada , Intervalo Livre de Doença , Glioblastoma/mortalidade , Glioblastoma/cirurgia , Humanos , Avaliação de Estado de Karnofsky , Masculino , Pessoa de Meia-Idade , Modelos de Riscos Proporcionais , Estudos Prospectivos , Dosagem Radioterapêutica
8.
Neurosurgery ; 49(6): 1288-97; discussion 1297-8, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11846927

RESUMO

OBJECTIVE: Advanced age is a strong predictor of shorter survival in patients with glioblastoma multiforme (GM), especially for those who receive multimodality treatment. Radiographically assessed tumor response to external beam radiation therapy is an important prognostic factor in GM. We hypothesized that older GM patients might have more radioresistant tumors. METHODS: We studied radiographically assessed response to external beam radiation treatment (five-level scale) in relation to age and other prognostic factors in a cohort of 301 GM patients treated on two prospective clinical protocols. A total of 223 patients (74%) were assessable for radiographically assessed radiation response. A proportional odds ordinal regression model was used for univariate and multivariate analysis. RESULTS: Younger age (P = 0.006), higher Karnofsky Performance Scale score before radiotherapy (P = 0.027), and more extensive surgical resection (P = 0.028) predicted better radiation response in univariate analyses. Results were similar when clinical criteria were used to classify an additional 61 patients without radiographically assessed radiation response (stable versus progressive disease). In multivariate analyses, age and extent of resection were significant independent predictors of radiation response (P < 0.05); Karnofsky Performance Scale score was of borderline significance (P = 0.07). CONCLUSION: Older GM patients are less likely to have good responses to postoperative external beam radiation therapy. Karnofsky Performance Scale score before radiation treatment and extent of surgical resection are additional predictors of radiographically assessed radiation response in GM.


Assuntos
Neoplasias Encefálicas/radioterapia , Irradiação Craniana , Glioblastoma/radioterapia , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/cirurgia , Estudos de Coortes , Terapia Combinada , Fracionamento da Dose de Radiação , Glioblastoma/diagnóstico por imagem , Glioblastoma/mortalidade , Glioblastoma/cirurgia , Humanos , Razão de Chances , Radiografia , Radioterapia Adjuvante , Taxa de Sobrevida , Resultado do Tratamento
9.
Clin Cancer Res ; 6(10): 3878-84, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11051233

RESUMO

Although the efficacy of the nitrosourea-based combination chemotherapy procarbazine, N-(2-chloroethyl)-N'-cyclohexyl-N-nitrosurea, and vincristine (PCV) has been previously demonstrated in the setting of anaplastic/intermediate-grade gliomas, the benefit for glioblastoma patients remains unproven. In the current study, we sought to determine whether the addition of alpha-difluoromethylornithine (eflornithine), an inhibitor of ornithine decarboxylase, which has shown encouraging results in the setting of recurrent glioma patients, to a nitrosourea-based therapy (PCV) would constitute a more effective adjuvant therapy in the treatment of glioblastoma multiforme patients in the postradiation therapy setting. Following conventional radiation therapy, 272 glioblastoma (GBM) patients were randomized to receive either alpha-difluoromethylornithine-PCV (DFMO-PCV; 134 patients) or PCV alone (138 patients), with survival and time to tumor progression being the primary endpoints. The starting dosage of DFMO was 3.0 g/m2 p.o. q8h for 14 days before and after treatment with N-(2-chloroethyl)-N-cyclohexyl-N-nitrosurea; PCV was administered as previously described1. Clinical and radiological (Gadolinium-enhanced MRI) follow-ups were nominally at the end of each 6 or 8 week cycle (PCV at 6 weeks; DFMO-PCV at 8 weeks). Laboratory evaluations for hematologic and other adverse effects were at 2 week intervals. There was no difference in median survival or median time-to-tumor progression between the two treatment groups, as measured from day of commencement of postradiotherapy chemotherapy [MS (months): DFMO-PCV, 10.5; Overall survival, as measured from time of tumor diagnosis at first surgery, was 13.3 and 14.2 months at the median and 6.2 and 8.7% at 5 years, respectively, for the DFMO-PCV and PCV arms. The treatment effect was unchanged after adjustment for age, performance status (KPS), extent of surgery, and other factors using the multivariate Cox proportional hazard model. Adverse effects associated with DFMO consisted of gastrointestinal (diarrhea nausea/vomiting), cytopenias, and minimal ototoxicity (limited to tinnitus) at the dose range tested. The addition of DFMO to the nitrosourea-based PCV regimen in this phase III study demonstrated no additional benefit in glioblastoma patients, underscoring the resistance of glioblastoma multiforme tumors to alkylating agents. For patients with anaplastic (intermediate grade) gliomas, in which the previously demonstrated benefit of post-radiation chemotherapy is more substantial, the evaluation of DFMO-PCV vs. PCV is still ongoing and hopefully will yield more encouraging results.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/radioterapia , Cisplatino/administração & dosagem , Ciclofosfamida/administração & dosagem , Eflornitina/administração & dosagem , Glioblastoma/tratamento farmacológico , Glioblastoma/radioterapia , Lomustina/administração & dosagem , Procarbazina/administração & dosagem , Vincristina/administração & dosagem , Vindesina/administração & dosagem , Adolescente , Adulto , Idoso , Neoplasias Encefálicas/mortalidade , Intervalo Livre de Doença , Relação Dose-Resposta a Droga , Feminino , Glioblastoma/mortalidade , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Fatores de Tempo
10.
Eur J Cancer ; 36(14): 1788-95, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10974627

RESUMO

One of the objectives of this phase II study was to determine whether temozolomide (TMZ) improved the health-related quality of life (HRQL) of patients with recurrent anaplastic astrocytoma (AA). HRQL was assessed at baseline (pretreatment) and every 4 weeks at each treatment cycle using the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire (EORTC QLQ-C30) (version 2.0) and the Brain Cancer Module (BCM20). Changes from baseline in the scores of seven preselected HRQL domains (role and social functioning, global QL, visual disorder, motor dysfunction, communication deficit and drowsiness) were determined at 6 months as well as prior to, and at the time of, disease progression. The significance of the changes was assessed by calculating statistical significance, effect sizes and the proportions of patients with improvement in their HRQL scores (changes of >/=10 points). After 6 months of treatment, patients who were free of progression of disease reported either an improvement or maintenance of all the preselected HRQL domains scores. Patients with disease progression by 6 months usually experienced improvement in HRQL before progression, but there was a sharp decline in most of the preselected domains at progression. We conclude that treatment of recurrent AA with temozolomide is associated with significant HRQL benefits.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Astrocitoma/tratamento farmacológico , Neoplasias Encefálicas/tratamento farmacológico , Dacarbazina/análogos & derivados , Qualidade de Vida , Adulto , Idoso , Dacarbazina/uso terapêutico , Progressão da Doença , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia , Análise de Sobrevida , Temozolomida , Falha de Tratamento
11.
Br J Cancer ; 83(5): 588-93, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10944597

RESUMO

A randomized, multicentre, open-label, phase II study compared temozolomide (TMZ), an oral second-generation alkylating agent, and procarbazine (PCB) in 225 patients with glioblastoma multiforme at first relapse. Primary objectives were to determine progression-free survival (PFS) at 6 months and safety for TMZ and PCB in adult patients who failed conventional treatment. Secondary objectives were to assess overall survival and health-related quality of life (HRQL). TMZ was given orally at 200 mg/m(2)/day or 150 mg/m(2)/day (prior chemotherapy) for 5 days, repeated every 28 days. PCB was given orally at 150 mg/m(2)/day or 125 mg/m(2)/day (prior chemotherapy) for 28 days, repeated every 56 days. HRQL was assessed using the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire (EORTC QLQ-C30 [+3]) and the Brain Cancer Module 20 (BCM20). The 6-month PFS rate for patients who received TMZ was 21%, which met the protocol objective. The 6-month PFS rate for those who received PCB was 8% (P = 0.008, for the comparison). Overall PFS significantly improved with TMZ, with a median PFS of 12.4 weeks in the TMZ group and 8.32 weeks in the PCB group (P = 0.0063). The 6-month overall survival rate for TMZ patients was 60% vs. 44% for PCB patients (P = 0.019). Freedom from disease progression was associated with maintenance of HRQL, regardless of treatment received. TMZ had an acceptable safety profile; most adverse events were mild or moderate in severity.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Dacarbazina/análogos & derivados , Glioblastoma/tratamento farmacológico , Gliossarcoma/tratamento farmacológico , Procarbazina/uso terapêutico , Adulto , Idoso , Antineoplásicos/efeitos adversos , Antineoplásicos Alquilantes/efeitos adversos , Neoplasias Encefálicas/mortalidade , Dacarbazina/efeitos adversos , Dacarbazina/uso terapêutico , Intervalo Livre de Doença , Feminino , Glioblastoma/mortalidade , Gliossarcoma/mortalidade , Humanos , Masculino , Pessoa de Meia-Idade , Procarbazina/efeitos adversos , Prognóstico , Qualidade de Vida , Recidiva , Temozolomida , Fatores de Tempo
12.
Semin Oncol ; 27(3 Suppl 6): 1-10, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10866344

RESUMO

Malignant gliomas are a heterogeneous group of tumors that are associated with significant morbidity and mortality. The development of the malignant phenotype is the result of a complex series of events that influence gene expression, angiogenesis, and invasion and favor the growth of tumor cells. Currently, the management of malignant glioma consists of symptom control and cytoreduction with a combination of surgery, radiation therapy, and chemotherapy. However, there is no satisfactory regimen available for adjuvant or salvage chemotherapy for these neoplasms. An overview of the biologic mechanisms, grading systems, and treatment options for anaplastic astrocytoma and glioblastoma multiforme is presented.


Assuntos
Neoplasias Encefálicas/terapia , Glioma/terapia , Astrocitoma/patologia , Astrocitoma/terapia , Neoplasias Encefálicas/patologia , Transformação Celular Neoplásica , Terapia Combinada , Regulação Neoplásica da Expressão Gênica , Glioblastoma/patologia , Glioblastoma/terapia , Glioma/patologia , Humanos , Neovascularização Patológica , Prognóstico
13.
Semin Oncol ; 27(3 Suppl 6): 41-6, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10866349

RESUMO

Temozolomide (TMZ) is a new, orally administered, second-generation imidazotetrazine prodrug with essentially 100% oral bioavailability that has demonstrated meaningful efficacy and an acceptable safety profile in the treatment of patients with recurrent glioblastoma multiforme. Because of its unique properties and broad spectrum of anticancer activity, preliminary studies are being conducted to evaluate the efficacy of TMZ in combination with other chemotherapeutic agents, radiation, or immunotherapy. The presence of de novo or acquired resistance to alkylating agents exhibited by malignant gliomas represents a serious impediment in the treatment of these tumors. This review discusses the mechanism of action of TMZ and strategies for overcoming pathways of resistance to this promising agent, including the use of TMZ in combination with other chemotherapeutic agents or radiation therapy, and exploration of alternate dosing schedules. Studies that have evaluated some of these strategies indicate that TMZ is a useful therapeutic option in patients with high-grade gliomas. Alternative approaches, including the use of high-dose TMZ with bone marrow transplantation and in combination with gene therapy, will also be discussed.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Dacarbazina/análogos & derivados , Glioblastoma/tratamento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Disponibilidade Biológica , Transplante de Medula Óssea , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Carmustina/administração & dosagem , Cisplatino/administração & dosagem , Terapia Combinada , Dacarbazina/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Terapia Genética , Glioblastoma/patologia , Glioblastoma/terapia , Humanos , Temozolomida , Topotecan/administração & dosagem
14.
Neurosurgery ; 46(2): 319-26; discussion 326-8, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10690720

RESUMO

OBJECTIVE: The goal of this study was to investigate the use of proton magnetic resonance spectroscopic imaging as a prognostic indicator in gamma knife radiosurgery of recurrent gliomas. METHODS: Thirty-six patients with recurrent gliomas were studied with proton magnetic resonance spectroscopic imaging at the time of radiosurgery, and with conventional magnetic resonance imaging examinations at regular time intervals until the initiation of a new treatment strategy. Patients were categorized on the basis of their initial spectroscopic results, and their performance was assessed in terms of change in contrast-enhancing volume, time to further treatment, and survival. RESULTS: The trends in the overall population were toward more extensive increase in the percent contrast-enhancing volume, a decreased time to further treatment, and a reduced survival time for patients with more extensive initial metabolic abnormalities. Statistical analysis of the subpopulation of patients with glioblastoma multiforme found a significant increase in relative contrast-enhancing volume (P < 0.01, Wilcoxon signed-rank test), a decrease in time to further treatment (P < 0.01, log-rank test), and a reduction in survival time (P < 0.01, log-rank test) for patients with regions containing tumor-suggestive spectra outside the gamma knife target, compared with patients exhibiting spectral abnormalities restricted to the gamma knife target. Further studies are needed to establish statistical significance for patients with lower-grade lesions and to confirm the results observed in this study. CONCLUSION: The pretreatment spectroscopic results provided information that was predictive of outcome for this patient pool, both in local control (change in contrast-enhancing volume) and global outcome (time to further treatment and survival). This modality may have an important role in improving the selection, planning, and treatment process for glioma patients.


Assuntos
Astrocitoma/cirurgia , Glioblastoma/cirurgia , Espectroscopia de Ressonância Magnética , Recidiva Local de Neoplasia/cirurgia , Radiocirurgia , Adulto , Idoso , Ácido Aspártico/análogos & derivados , Ácido Aspártico/metabolismo , Astrocitoma/diagnóstico , Astrocitoma/mortalidade , Colina/metabolismo , Creatina/metabolismo , Feminino , Glioblastoma/diagnóstico , Glioblastoma/mortalidade , Humanos , Ácido Láctico/metabolismo , Metabolismo dos Lipídeos , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/diagnóstico , Recidiva Local de Neoplasia/mortalidade , Prognóstico , Taxa de Sobrevida
15.
Neuro Oncol ; 2(4): 221-8, 2000 10.
Artigo em Inglês | MEDLINE | ID: mdl-11265231

RESUMO

The burden imposed by disease recurrence in patients with high-grade gliomas is not well documented. We studied the frequency of self-report symptoms and the effects on health-related quality of life in patients who had recurrent glioblastoma multiforme or anaplastic astrocytoma and who had a Karnofsky performance score > or = 70. Patients completed the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire-Core 30 Items (QLQ-C30) and the Brain Cancer Module (BCM20) before initiation of treatment for first recurrence of disease. Six symptoms (fatigue, uncertainty about the future, motor difficulties, drowsiness, communication difficulties, and headache) were reported with a frequency > 50% by both groups of patients. An additional two symptoms (visual problems and pain) were also reported with frequencies of > 50% by patients with recurrent glioblastoma multiforme. Most of the symptoms were likely due to recurrence, but previous radiation therapy and on-going corticosteroid treatment may have also been casual factors for fatigue, whereas uncertainty about the future and pain were probably nonspecific for brain cancer. Problems with motor functioning, vision, leg strength, and pain were reported more frequently by patients with recurrent glioblastoma multiforme than by those with recurrent anaplastic astrocytoma. Scores on health-related quality-of-life functioning scales were similar in the two groups. Finally, the scores for patients who had recurrent high-grade gliomas and a Karnofsky performance score > or = 70 were compared with the reported health-related quality of life scores of patients with other cancers. Their scores were similar to those of patients with metastatic cancers and worse than those of patients with localized cancers.


Assuntos
Neoplasias Encefálicas/fisiopatologia , Neoplasias Encefálicas/psicologia , Glioma/fisiopatologia , Glioma/psicologia , Recidiva Local de Neoplasia/fisiopatologia , Recidiva Local de Neoplasia/psicologia , Qualidade de Vida/psicologia , Índice de Gravidade de Doença , Adulto , Idoso , Astrocitoma/patologia , Astrocitoma/fisiopatologia , Astrocitoma/psicologia , Neoplasias Encefálicas/patologia , Feminino , Glioblastoma/patologia , Glioblastoma/fisiopatologia , Glioblastoma/psicologia , Glioma/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/patologia , Inquéritos e Questionários
16.
Curr Treat Options Oncol ; 1(5): 459-68, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12057153

RESUMO

Gliomas are a heterogeneous group of neoplasms that comprise the majority of tumors originating in the central nervous system (CNS). In adults, the most frequently encountered of these are high-grade or malignant neoplasms of astrocytic and oligodendrocytic lineage, ie, anaplastic astrocytoma (AA), glioblastoma multiforme (GBM), and anaplastic oligodendroglioma (AO), respectively. Tumors of mixed lineage are also seen, the most common of which is designated anaplastic oligoastrocytoma (AOA). Standard treatment for these tumors is typically surgery, followed by radiation then chemotherapy. Surgery is required for a definitive histopathologic diagnosis, which in turn will dictate subsequent therapy options. Moreover, aggressive tumor resection improves survival outcomes, and in many cases, the patient's neurologic function. We generally advocate the safest, maximal resection attainable for patients with these tumors as a way to improve prognosis. In almost all cases, surgery is followed by radiation therapy. Postsurgical irradiation is the most effective treatment currently available for improving survival. There is also mounting evidence to suggest that additional radiation, given in the form of brachytherapy or radiosurgery, at initial diagnosis as a "boost" to standard radiation or at tumor recurrence, may provide added improvement in survival outcome. Radiosurgery and brachytherapy are therapies often used to treat eligible patients at this institution. Adjuvant chemotherapy, conventionally given after radiation, may offer a modest survival benefit in some patients with GBM. Bischloroethylnitrosourea (BCNU) is the typical first-line agent used, but chemotherapy seems to be most beneficial in young patients, with little if any impact on survival for patients over 60 years old. At this institution, we often defer treatment with adjuvant chemotherapy for elderly patients with GBM due to lack of efficacy and the attendant risk with chemotherapy. For anaplastic astrocytomas, oligodendrogliomas, and oligoastrocytomas, a commonly accepted standard is adjuvant chemotherapy following irradiation with the three-drug regimen--procarbazine, CCNU, and vincristine (PCV). This is due to an earlier clinical trial that showed a survival advantage in patients treated with adjuvant PCV compared with patients that received BCNU. However, recent data suggest that treatment with either PCV or BCNU may be appropriate. Both regimens now appear to have equal efficacy for anaplastic gliomas in light of a more recent analysis done with larger numbers of patients. AOs are a unique case with respect to tumor chemosensitivity and patient survival. Molecular studies have identified a subpopulation of patients with AO whose tumors have lost chromosomes 1p and 19q. Patients with this molecular pattern have an exceptional responsiveness to PCV chemotherapy and have prolonged survival. Currently, trials are being conducted to confirm this finding and to determine the best treatment regimen for these patients, with particular regard to the timing of radiation and chemotherapy.


Assuntos
Neoplasias Encefálicas/terapia , Glioma/terapia , Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Ensaios Clínicos como Assunto , Feminino , Glioma/mortalidade , Glioma/patologia , Humanos , Radioterapia , Taxa de Sobrevida
17.
Neuro Oncol ; 2(1): 22-8, 2000 01.
Artigo em Inglês | MEDLINE | ID: mdl-11302250

RESUMO

We conducted a single-arm phase II study to evaluate the efficacy and safety of radiotherapy combined with 6-thioguanine, procarbazine, dibromodulcitol, lomustine, and vincristine (TPDCV) chemotherapy for treating malignant astrocytoma in children and anaplastic ependymoma in patients of all ages. Between 1984 and 1992, 42 patients who had malignant astrocytomas (glioblastomas multiforme, anaplastic astrocytomas, or mixed anaplastic oligoastrocytomas) were treated with TPDCV chemotherapy and radiation therapy. Of these patients, 40 were younger than 18 years, but 2 were older (22 and 23 years) when treated. Cranial radiation averaged 58 Gy. TPDCV chemotherapy was given for 1 year or until progression. Between 1989 and 1991, 17 patients with malignant ependymoma were treated with TPDCV chemotherapy and craniospinal radiation. Radiation was given at an average dose of 54 Gy to the tumor, 28 Gy to the whole brain, and 31 Gy to the spinal axis. TPDCV chemotherapy was given for 1 year or until tumor progressed. Of the patients with glioblastoma multiforme, 13 of 17 died; the median time to progression was 49 weeks, and median survival was 85 weeks. The four patients surviving at this writing were followed a median 537 weeks (range 364-635 weeks). Of the patients with nonglioblastoma malignant astrocytoma, 14 of 25 died; the median time to progression was 224 weeks. Median survival was not reached in this group. The median follow-up for those surviving was 494 weeks. For the patients with ependymoma, 11 of 17 died with a median time to progression of 141 weeks. The median follow-up for the eight who survive was 469 weeks. Nine patients died with a median survival of 183 weeks. The combination of TPDCV and radiotherapy has activity against childhood anaplastic astrocytoma, glioblastoma multiforme, and anaplastic ependymoma. The results of this study for children with glioblastoma were comparable to results in the literature, while the results for children with anaplastic astrocytoma appeared better than most reports. The combination of TPDCV chemotherapy and radiation therapy for anaplastic ependymomas appears to be active and at least as good as published reports using radiation therapy alone.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/radioterapia , Glioma/tratamento farmacológico , Glioma/radioterapia , Lomustina/uso terapêutico , Mitolactol/uso terapêutico , Procarbazina/uso terapêutico , Tioguanina/uso terapêutico , Vincristina/uso terapêutico , Adolescente , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Criança , Pré-Escolar , Terapia Combinada , Feminino , Humanos , Lomustina/administração & dosagem , Masculino , Mitolactol/administração & dosagem , Procarbazina/administração & dosagem , Análise de Sobrevida , Tioguanina/administração & dosagem , Resultado do Tratamento , Vincristina/administração & dosagem
18.
Neuro Oncol ; 2(1): 34-9, 2000 01.
Artigo em Inglês | MEDLINE | ID: mdl-11302252

RESUMO

The North American Brain Tumor Consortium conducted a phase I trial of the combination 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) and temozolomide. Eligibility included a patient with a cancer type that was considered refractory to standard therapy. Prior nitrosourea treatments were not permitted. There were parallel dose escalations in two treatment schedules. Forty-five patients were enrolled during an 18-month period. The maximum tolerated doses (MTDs) when temozolomide followed BCNU (Arm A) were temozolomide at 550 mg/m2/p.o. and BCNU at 150 mg/m2/i.v.), whereas the MTD when temozolomide preceded BCNU (Arm B) was temozolomide at 400 mg/m2/p.o. and BCNU at 100 mg/m2/i.v. Toxicity was predominantly hematologic, although there were three instances of pulmonary toxicity, which in one case could have represented potentiation of nitrosourea-induced pulmonary fibrosis. The half-life of temozolomide was 1.86 (+/-0.31) h. There was a moderate relationship between dose and peak concentration and a strong relationship between dose and plasma concentration time curve. Pharmacokinetic parameters of temozolomide were unaffected by the treatment schedule, so the difference in MTD between the schedules is likely due to a biologic rather than a pharmacokinetic sequence interaction. There were 9 partial responses among 43 patients evaluable for response, including 5 of 25 with a histologic diagnosis of glioblastoma. The recommended dose and schedule for phase II trials of this regimen are BCNU 150 mg/m2/i.v. followed in 2 h by temozolomide 550 mg/m2/p.o. repeated every 6 weeks. We are also recommending screening and periodic pulmonary function testing during treatment to assess the possible potentiation of nitrosourea-induced pulmonary fibrosis.


Assuntos
Antineoplásicos Alquilantes/administração & dosagem , Neoplasias Encefálicas/tratamento farmacológico , Carmustina/administração & dosagem , Dacarbazina/análogos & derivados , Dacarbazina/administração & dosagem , Dacarbazina/uso terapêutico , Antineoplásicos Alquilantes/sangue , Neoplasias Encefálicas/sangue , Dacarbazina/efeitos adversos , Dacarbazina/sangue , Relação Dose-Resposta a Droga , Meia-Vida , Humanos , Dose Máxima Tolerável , Temozolomida
19.
Int J Radiat Oncol Biol Phys ; 45(5): 1109-15, 1999 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-10613302

RESUMO

PURPOSE: This study was an open label, randomized Phase 3 trial in newly diagnosed patients with anaplastic glioma comparing radiotherapy plus adjuvant procarbazine, CCNU, and vincristine (PCV) chemotherapy with or without bromodeoxyuridine (BUdR) given as a 96-hour infusion each week of radiotherapy. METHODS AND MATERIALS: Only patients 18 years or older with newly diagnosed anaplastic glioma were eligible; central pathology review was accomplished, but was not mandated prior to registration. The study had initially opened as a Northern California Oncology Group (NCOG) trial in 1991, becoming an Intergroup RTOG, SWOG, and NCCTG study in July 1994. Total accrual of 293 patients was planned as the sample size, using survival and time to tumor progression as the primary endpoints. The experiment arm (RT/BUdR plus PCV) was to be compared to the control arm (RT plus PCV) using an alpha = 0.05, one-tailed, with a power of 85% for detecting an increase in median survival from 160 to 240 weeks, assuming a 3-year follow-up after completion of enrollment. RESULTS: As of July 1996, 281 patients had been randomized; 53 (20%) were ineligible, primarily based upon central pathology review, and another 39 cases were canceled. In total, 30% of cases were excluded from analysis. The treatment arms were well balanced despite this rate of exclusion. The RTOG Data Monitoring Committee recommended suspension of enrollment in July 1996 based upon a stochastic curtailment analysis which strongly suggested that the addition of BUdR would not be associated with increased survival. In February 1997, the study was closed prior to full enrollment. At that time, the 1-year survival estimates were 82% versus 68% for RT plus PCV and RT/BUdR plus PCV respectively (one-sided, p = 0.96). The conditional power analysis indicated that even with an additional 12 months of additional accrual and follow-up the probability of detecting the prespecified difference was less than 0.01%. The differences in the two arms seem to be due to early deaths in the BUdR arm, not related to toxicity of the treatment. CONCLUSIONS: Despite encouraging Phase 2 results with BUdR, it is unlikely that a survival benefit will be seen. A final study analysis will not be done for at least 3 more years.


Assuntos
Astrocitoma/radioterapia , Neoplasias Encefálicas/radioterapia , Bromodesoxiuridina/uso terapêutico , Radiossensibilizantes/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Astrocitoma/tratamento farmacológico , Astrocitoma/mortalidade , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/mortalidade , Quimioterapia Adjuvante , Progressão da Doença , Feminino , Humanos , Lomustina/administração & dosagem , Masculino , Pessoa de Meia-Idade , Procarbazina/administração & dosagem , Modelos de Riscos Proporcionais , Análise de Sobrevida , Vincristina/administração & dosagem
20.
J Clin Oncol ; 17(8): 2572-8, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10561324

RESUMO

PURPOSE: To determine aggregate outcomes and prognostic covariates in patients with recurrent glioma enrolled onto phase II chemotherapy trials. PATIENTS AND METHODS: Patients from eight consecutive phase II trials included 225 with recurrent glioblastoma multiforme (GBM) and 150 with recurrent anaplastic astrocytoma (AA). Their median age was 45 years (range, 15 to 82 years) and their median Karnofsky performance score was 80 (range, 60 to 100). Prognostic covariates were analyzed with respect to tumor response, progression-free survival (PFS), and overall survival (OS) by multivariate logistic and Cox proportional hazards regression analyses. RESULTS: Overall, 34 (9%) had complete or partial response, whereas 80 (21%) were alive and progression-free at 6 months (APF6). The median PFS was 10 weeks and median OS was 30 weeks. Histology was a robust prognostic factor across all outcomes. GBM patients had significantly poorer outcomes than AA patients. The APF6 proportion was 15% for GBM and 31% for AA, whereas the median PFS was 9 weeks for GBM and 13 weeks for AA. Results were also significantly poorer for patients with more than two prior surgeries or chemotherapy regimens. CONCLUSION: Histology is a dominant factor in determining outcome in patients with recurrent glioma enrolled onto phase II trials. Future trials should be designed with separate histology strata.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Astrocitoma/tratamento farmacológico , Astrocitoma/patologia , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Adolescente , Adulto , Astrocitoma/classificação , Neoplasias Encefálicas/classificação , Neoplasias Encefálicas/patologia , Glioblastoma/classificação , Humanos , Avaliação de Estado de Karnofsky , Modelos Logísticos , Pessoa de Meia-Idade , Recidiva Local de Neoplasia , Probabilidade , Prognóstico , Modelos de Riscos Proporcionais , Terapia de Salvação , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...