Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Int J Radiat Oncol Biol Phys ; 49(1): 71-7, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11163499

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Eflornithine/therapeutic use , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Radiation-Sensitizing Agents/therapeutic use , Adult , Aged , Brain Neoplasms/surgery , Combined Modality Therapy , Disease-Free Survival , Glioblastoma/mortality , Glioblastoma/surgery , Humans , Karnofsky Performance Status , Male , Middle Aged , Proportional Hazards Models , Prospective Studies , Radiotherapy Dosage
2.
Neuro Oncol ; 2(1): 22-8, 2000 01.
Article in English | MEDLINE | ID: mdl-11302250

ABSTRACT

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.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Glioma/drug therapy , Glioma/radiotherapy , Lomustine/therapeutic use , Mitolactol/therapeutic use , Procarbazine/therapeutic use , Thioguanine/therapeutic use , Vincristine/therapeutic use , Adolescent , Adult , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Child , Child, Preschool , Combined Modality Therapy , Female , Humans , Lomustine/administration & dosage , Male , Mitolactol/administration & dosage , Procarbazine/administration & dosage , Survival Analysis , Thioguanine/administration & dosage , Treatment Outcome , Vincristine/administration & dosage
3.
Int J Radiat Oncol Biol Phys ; 43(2): 279-85, 1999 Jan 15.
Article in English | MEDLINE | ID: mdl-10030250

ABSTRACT

PURPOSE: To report the results of a Phase II study of hyperfractionated craniospinal radiation therapy, with and without adjuvant chemotherapy for primitive neuroectodermal brain tumors (PNETs) and malignant ependymomas. METHODS AND MATERIALS: Newly diagnosed PNET or malignant ependymomas were treated with hyperfractionated craniospinal radiation therapy. The primary tumor site was treated to a dose of 72 Gy, with 30 Gy given to the rest of the craniospinal axis. The fraction size was 1.0 Gy, given twice a day. Patients with poor risk factors also received adjuvant chemotherapy with CCNU, cisplatin, and vincristine. Patients had follow-up for survival, time to tumor progression, and patterns of relapse. RESULTS: A total of 39 patients (21 males/18 females) were treated between March 12, 1990 and October 29, 1992. The median age was 16 years (range 3-59 years). Tumor types included 25 medulloblastomas, 5 pineoblastomas, 5 cerebral PNETs, 1 spinal cord PNET, and 3 malignant ependymomas. Twenty cases were staged as poor-risk and received adjuvant chemotherapy following radiation. Three-year progression-free survival (PFS) was 60% and 63% for poor-risk and good-risk patients, respectively. Overall 3-year survival for these groups was 70% and 79%, respectively. For the 25 patients with medulloblastoma, there were 16 good-risk and 9 poor-risk patients. Three-year PFSs were 63% and 56%, respectively. The 5-year survival for good-risk medulloblastoma was 69% with 43.7% of these patients having failures outside the primary site. CONCLUSIONS: Survival in patients with good-risk medulloblastoma was no better than that seen in previous studies with single-fraction radiation, and the rate of failure outside the primary site is excessive. Those with poor-risk features had comparable survival to that seen in patients with good risk factors, but these patients were treated with chemotherapy, and the role that hyperfractionated radiation played in their outcome is uncertain.


Subject(s)
Brain Neoplasms/radiotherapy , Cranial Irradiation/methods , Neuroectodermal Tumors, Primitive/radiotherapy , Adolescent , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Chemotherapy, Adjuvant , Child , Child, Preschool , Disease Progression , Dose Fractionation, Radiation , Ependymoma/drug therapy , Ependymoma/radiotherapy , Female , Humans , Male , Medulloblastoma/drug therapy , Medulloblastoma/radiotherapy , Middle Aged , Neoplasm Staging , Neuroectodermal Tumors, Primitive/drug therapy , Pineal Gland , Pinealoma/drug therapy , Pinealoma/radiotherapy , Recurrence
4.
J Neurooncol ; 37(2): 169-76, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9524096

ABSTRACT

Chemotherapeutic regimens in present use for recurrent glioma have substantial toxicity. Activity against recurrent gliomas has been reported for both tamoxifen and interferon alpha, agents that have more acceptable toxicity profiles and that can be administered in an outpatient setting. We tested the efficacy and toxicity of the combination of high-dose tamoxifen and interferon alpha in adults with recurrent glioma in a phase II trial. Eligible patients had radiographically measurable recurrent gliomas of any grade after initial radiation therapy. Interferon-alpha [6 x 10(6) U subcutaneously three times per week] and tamoxifen (240 mg/m2/day orally) were administered continuously. Treatment response was assessed at 6 week intervals using clinical and radiographic criteria. Eighteen patients (11 males and 7 females) were enrolled. Median age was 41 years (range 23-61 years). All patients had gliomas that progressed after radiation therapy and nitrosourea chemotherapy. The histologic diagnosis of the original tumor was glioblastoma multiforme in 8 patients, anaplastic astrocytoma in 5 patients, astrocytoma in 4 patients and mixed malignant glioma in 1 patient. Reversible moderate to severe neurological toxicity manifested by dizziness and unsteady gait was seen at tamoxifen doses of 240 mg/m2/day. Although the initial tamoxifen dose was reduced to 120 mg/m2/day, moderate neurotoxicity was noted at this dose as well and the trial was closed early. The combination of oral tamoxifen (120 to 240 mg/m2/day) and subcutaneous interferon-alpha [6 x 10(6) U three times per week] was associated with significant neurotoxicity in this group of recurrent glioma patients, resulting in early study closure. Of 16 evaluable patients, 12 had progressive disease after one cycle of treatment, 3 had stable disease, and there was one minor response. Gradual dose escalation may be required if similar patients are to be treated with high dose tamoxifen in conjunction with interferon.


Subject(s)
Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Agents/administration & dosage , Brain Neoplasms/therapy , Glioma/therapy , Interferon-alpha/administration & dosage , Tamoxifen/administration & dosage , Administration, Oral , Adult , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Antineoplastic Agents, Hormonal/adverse effects , Antineoplastic Agents, Hormonal/therapeutic use , Dose-Response Relationship, Drug , Female , Humans , Injections, Subcutaneous , Interferon alpha-2 , Interferon-alpha/adverse effects , Interferon-alpha/therapeutic use , Male , Middle Aged , Neoplasm Recurrence, Local , Recombinant Proteins , Survival Analysis , Tamoxifen/adverse effects , Tamoxifen/therapeutic use , Treatment Outcome
5.
Semin Oncol Nurs ; 14(1): 53-60, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9503515

ABSTRACT

OBJECTIVES: To identify the causes of and discuss nursing management of those complications that are unique to the brain tumor patient population. DATA SOURCES: Published articles and books related to neurological and nonneurological complications in the neuro-oncology patient. CONCLUSIONS: Seizures, cerebral edema, thromboembolism, and endocrine dysfunction are several complications experienced by the neuro-oncology patient. These complications can be caused by the tumor or treatment of this disease. IMPLICATIONS FOR NURSING PRACTICE: Care of the neuro-oncology patient is complex. Knowledge of the potential complications that patients may experience and how to manage these problems serves to enhance their quality of life.


Subject(s)
Brain Neoplasms/nursing , Brain Neoplasms/complications , Humans , Nursing Assessment , Oncology Nursing , Patient Care Management , Seizures/nursing
6.
Int J Radiat Oncol Biol Phys ; 40(1): 57-63, 1998 Jan 01.
Article in English | MEDLINE | ID: mdl-9422558

ABSTRACT

PURPOSE: This study was designed to evaluate a combined modality treatment for malignant gliomas using radiation therapy with a radiosensitizer and an adjuvant chemotherapy regimen designed to modify resistance to BNCU. METHODS AND MATERIALS: Patients were eligible if they were 15 years of age or older, and had newly diagnosed glioblastoma multiforme (GBM), or anaplastic glioma (AG). Treatment consisted of external beam radiotherapy given to a dose of 60 Gy using a single daily fraction Monday to Friday. Concurrent hydroxyurea at a dose of 300 mg/m2 every 6 h every other day was given during radiation. Following radiotherapy, patients were then treated with BCNU and 6-Thioguanine (6TG). The 6-TG was given by mouth every 6 h for 12 doses prior to BCNU. Patients were initially treated with 60 mg/m2/dose of 6TG, with escalation to a maximum dose of 100 mg/m2/dose. The primary study end points were time to tumor progression and survival. RESULTS: A total of 245 eligible patients were enrolled from 1/18/88 to 12/26/91. The histologic subtypes included 135 GBM, and 110 with AG (103 with anaplastic astrocytoma, 7 with high-grade mixed oligoastrocytoma). For the GBM group, the median time to tumor progression (TTP) and median survival were 33 (95% CI 26, 39) and 56 (95% CI 49, 69) weeks, respectively. For the AG group the median TTP was 282 weeks (95% lower confidence bound = 155 weeks). Median survival for this group has not been reached (95% lower confidence bound = 284 weeks) with a median follow-up for surviving patients of 298 weeks. A proportional hazards model was used to look at potential prognostic factors for survival, including initial Karnofsky Performance Scale (KPS), age, and extent of surgery, as well as dose of 6TG. Higher KPS, and lower age, predicted for longer survival (p < 0.01, < 0.001) in GBM patients; lower age was significant (p = 0.05) for AG cases. A higher (greater than 95 mg/m2) or lower dose of 6TG was not statistically significant in this model. CONCLUSIONS: This therapy was no more effective in patients with GBM than other reported series. In patients with malignant gliomas other than GBM, prolonged progression-free and overall survival is noted, without a median survival reached at the time of this report. In this subset of AG patients, survival is comparable to recent studies using halogenated prymidines during radiation and Procarbazine, CCNU, and Vincristine (PCV) as adjuvant chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Glioma/drug therapy , Glioma/radiotherapy , Adolescent , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carmustine/administration & dosage , Combined Modality Therapy , Disease Progression , Female , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Humans , Hydroxyurea/administration & dosage , Male , Middle Aged , Proportional Hazards Models , Prospective Studies , Radiotherapy Dosage , Survival Analysis , Thioguanine/administration & dosage
7.
J Neurooncol ; 32(3): 235-41, 1997 May.
Article in English | MEDLINE | ID: mdl-9049885

ABSTRACT

Between March 9, 1984 and January 29, 1992, 42 children with newly diagnosed symptomatic or previously diagnosed progressive low-grade gliomas received outpatient chemotherapy as their primary treatment. This study was a single arm, phase II trial designed to estimate the time to tumor progression and toxicity of this regimen. Procarbazine, 6-thioguanine, and dibromodulcitol were given before lomustine (CCNU) and vincristine was given 1 and 3 weeks after CCNU. Patients were treated for six treatment cycles or until the tumor progressed, whichever came first. Twenty-three patients had juvenile pilocytic astrocytomas, 11 had astrocytomas, one had oligodendroglioma, one had ganglioglioma, and six had radiographically diagnosed low-grade gliomas. The mean age of the patients was 5 years (median, 3 years). The median time to treatment failure was 132 weeks (95% confidence interval: 106, 186 weeks). Only eight patients have died the estimated 5-year survival rate is 78% (95% confidence interval, 60% 87%). There were two episodes of grade 4 neutropenia, and three episodes of grade 4 thrombocytopenia. This regimen was safe, able to be delivered in the outpatient setting, and produced prolonged periods of disease stabilization in children with low-grade gliomas.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Glioma/drug therapy , Adolescent , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Brain Neoplasms/mortality , Child , Child, Preschool , Disease Progression , Female , Glioma/mortality , Humans , Infant , Lomustine/administration & dosage , Lomustine/adverse effects , Male , Mitolactol/administration & dosage , Mitolactol/adverse effects , Procarbazine/administration & dosage , Procarbazine/adverse effects , Thioguanine/administration & dosage , Thioguanine/adverse effects , Treatment Outcome , Vincristine/administration & dosage , Vincristine/adverse effects
8.
Am J Clin Oncol ; 19(6): 609-12, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8931682

ABSTRACT

In a Phase II trial, 63 evaluable patients with recurrent glioma received i.v. infusions of carboplatin every 3 weeks beginning at a dose of 400 mg/m2. The dose was increased by 50 mg/m2 at each subsequent infusion until the maximum tolerated dose reached, as defined by a platelet count < 25,000/mm3 or an absolute neutrophil count (ANC) < 500/mm3. Treatment was then resumed at the previous dose level and continued until tumor progression occurred. There were 43 men and 20 women studied (mean age, 41 years; range, 6 months to 70.6 years). The combined response and stabilization rate was 29% for 31 patients with glioblastoma and 71.9% for 32 patients with other tumors; median time to tumor progression was 8.2 and 20.3 weeks and median survival was 25.9 and 58.3 weeks, respectively. Twenty patients had level 4 platelet toxicity and nine had level 4 ANC toxicity. Most tumors progressed before the maximum tolerated dose was reached. These results were not better than those from a previous trial of carboplatin at an initial dose of 350 mg/m2, which was escalated by 25 mg/ m2 after every two infusions. Therefore, an optimal dosing schedule was not achieved in this trial.


Subject(s)
Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Carboplatin/administration & dosage , Glioma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Adolescent , Adult , Aged , Child , Child, Preschool , Disease Progression , Drug Administration Schedule , Female , Glioblastoma/drug therapy , Humans , Infant , Infusions, Intravenous , Male , Middle Aged , Neutropenia/chemically induced , Remission Induction , Survival Rate , Thrombocytopenia/chemically induced
9.
Crit Care Nurs Clin North Am ; 7(1): 143-9, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7766368

ABSTRACT

Because patients with systemic cancer are surviving longer, the incidence of metastatic brain tumors has increased. Currently, studies are investigating alternative radiation regimens. Treatment options of surgery, radiosurgery, brachytherapy, and chemotherapy all provide therapeutic strategies for this patient population.


Subject(s)
Brain Neoplasms/secondary , Brain Neoplasms/diagnosis , Brain Neoplasms/mortality , Brain Neoplasms/therapy , Combined Modality Therapy , Humans , Survival Rate
10.
J Neurooncol ; 19(1): 69-74, 1994.
Article in English | MEDLINE | ID: mdl-7815106

ABSTRACT

Thirty-two patients with recurrent glioma who had previously received radiation therapy and chemotherapy with nitrosoureas were treated with intravenous carboplatin every 3 weeks, starting at a dose of 350 mg/m2, with a dose escalation of 25 mg/m2 every 6 weeks until a level 4 hematologic toxicity was reached. Of the 28 patients who could be evaluated for a response, 50% demonstrated a response or had stabilization of their disease after two infusions of carboplatin. Their median time to tumor progression and median duration of survival were 19 weeks and 38 weeks. Thrombocytopenia was the major toxicity and was severe in one-third of the patients. No neurologic or renal toxicities were noted. Carboplatin has demonstrated activity against recurrent gliomas in patients who have already had extensive chemotherapy. Increasing the dose of carboplatin may improve the rate of response and the duration of progression-free survival in patients with recurrent glioma.


Subject(s)
Brain Neoplasms/drug therapy , Carboplatin/therapeutic use , Glioma/drug therapy , Adult , Aged , Carboplatin/administration & dosage , Carboplatin/adverse effects , Disease Progression , Female , Humans , Infusions, Intravenous , Magnetic Resonance Imaging , Male , Middle Aged , Neoplasm Recurrence, Local , Tomography, X-Ray Computed
11.
J Neurosurg ; 74(5): 701-8, 1991 May.
Article in English | MEDLINE | ID: mdl-1901597

ABSTRACT

Between March, 1983, and February, 1989, 19 infants or children with chiasmal/hypothalamic gliomas were treated with chemotherapy after either surgical or radiological diagnosis. The patients ranged in age from 15 weeks to 15.6 years (median 3.2 years) at the start of therapy. Twelve patients were treated immediately after diagnosis because of progressive symptoms, and seven received chemotherapy after either radiographic progression or clinical deterioration, including progressive visual loss or intracranial hypertension. Based on biopsy results, seven of these tumors were classified as juvenile pilocytic astrocytomas, two as astrocytomas, two as highly anaplastic astrocytomas, and one as a subependymal giant-cell astrocytoma. There was associated neurofibromatosis in four patients. The two initial patients were treated with either actinomycin D and vincristine or 5-fluorouracil, hydroxyurea, and 6-thioguanine. The remaining patients received nitrosourea-based therapy; 15 evaluable patients were treated with a five-drug regimen that included 6-thioguanine, procarbazine, dibromodulcitol, 1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea (CCNU), and vincristine and one received 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) and 5-fluorouracil. Fifteen of the 18 evaluable patients initially managed with chemotherapy either responded to therapy or their condition stabilized. Median time to tumor progression has not been reached at a median follow-up period of 79 weeks (range 6.6 to 303 weeks), and no tumor-related death has occurred with a median follow-up period of 79 weeks (range 18 to 322 weeks) from the initiation of therapy. The four patients who failed therapy or whose disease progressed after chemotherapy were treated satisfactorily with radiation therapy. Initial improvement or stabilization of visual function was obtained in 16 patients. Endocrine function remained stable in all patients during treatment, although three patients required pharmacological treatment for endocrinopathy that was present at diagnosis. These preliminary results suggest that nitrosourea-based cytotoxic regimens are useful for the initial treatment of children with chiasmal/hypothalamic gliomas, and allow potentially harmful radiation therapy to be deferred until progression of disease.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Astrocytoma/drug therapy , Cranial Nerve Neoplasms/drug therapy , Glioma/drug therapy , Hypothalamic Neoplasms/drug therapy , Optic Chiasm , Adolescent , Astrocytoma/complications , Child , Child, Preschool , Combined Modality Therapy , Cranial Nerve Neoplasms/complications , Female , Glioma/complications , Humans , Hypothalamic Neoplasms/complications , Infant , Male , Neoplasm Recurrence, Local/drug therapy , Neurofibromatosis 1/complications , Nitrosourea Compounds/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...