Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Lancet Oncol ; 22(8): 1103-1114, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34214495

RESUMO

BACKGROUND: Malignant glioma is the most common and lethal primary brain tumour, with dismal survival rates and no effective treatment. We examined the safety and activity of NSC-CRAd-S-pk7, an engineered oncolytic adenovirus delivered by neural stem cells (NSCs), in patients with newly diagnosed high-grade glioma. METHODS: This was a first-in-human, open-label, phase 1, dose-escalation trial done to determine the maximal tolerated dose of NSC-CRAd-S-pk7, following a 3 + 3 design. Patients with newly diagnosed, histologically confirmed, high-grade gliomas (WHO grade III or IV) were recruited. After neurosurgical resection, NSC-CRAd-S-pk7 was injected into the walls of the resection cavity. The first patient cohort received a dose starting at 6·25 × 1010 viral particles administered by 5·00 × 107 NSCs, the second cohort a dose of 1·25 × 1011 viral particles administered by 1·00 × 108 NSCs, and the third cohort a dose of 1·875 × 1011 viral particles administered by 1·50 × 108 NSCs. No further dose escalation was planned. Within 10-14 days, treatment with temozolomide and radiotherapy was initiated. Primary endpoints were safety and toxicity profile and the maximum tolerated dose for a future phase 2 trial. All analyses were done in all patients who were included in the trial and received the study treatment and were not excluded from the study. Recruitment is complete and the trial is finished. The trial is registered with ClinicalTrials.gov, NCT03072134. FINDINGS: Between April 24, 2017, and Nov 13, 2019, 12 patients with newly diagnosed, malignant gliomas were recruited and included in the safety analysis. Histopathological evaluation identified 11 (92%) of 12 patients with glioblastoma and one (8%) of 12 patients with anaplastic astrocytoma. The median follow-up was 18 months (IQR 14-22). One patient receiving 1·50 × 108 NSCs loading 1·875 × 1011 viral particles developed viral meningitis (grade 3) due to the inadvertent injection of NSC-CRAd-S-pk7 into the lateral ventricle. Otherwise, treatment was safe as no formal dose-limiting toxicity was reached, so 1·50 × 108 NSCs loading 1·875 × 1011 viral particles was recommended as a phase 2 trial dose. There were no treatment-related deaths. The median progression-free survival was 9·1 months (95% CI 8·5-not reached) and median overall survival was 18·4 months (15·7-not reached). INTERPRETATION: NSC-CRAd-S-pk7 treatment was feasible and safe. Our immunological and histopathological findings support continued investigation of NSC-CRAd-S-pk7 in a phase 2/3 clinical trial. FUNDING: US National Institutes of Health.


Assuntos
Neoplasias Encefálicas/terapia , Glioma/terapia , Células-Tronco Neurais/transplante , Terapia Viral Oncolítica/métodos , Adenoviridae , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Vírus Oncolíticos
2.
J Neurooncol ; 129(1): 139-46, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27300524

RESUMO

One resistance mechanism in malignant gliomas (MG) involves nuclear factor-κB (NF-κB) activation. Bortezomib prevents proteasomal degradation of NF-κB inhibitor α (NFKBIA), an endogenous regulator of NF-κB signaling, thereby limiting the effects of NF-κB on tumor survival and resistance. A presurgical phase II trial of bortezomib in recurrent MG was performed to determine drug concentration in tumor tissue and effects on NFKBIA. Patients were enrolled after signing an IRB approved informed consent. Treatment was bortezomib 1.7 mg/m(2) IV on days 1, 4 and 8 and then surgery on day 8 or 9. Post-operatively, treatment was Temozolomide (TMZ) 75 mg/m(2) PO on days 1-7 and 14-21 and bortezomib 1.7 mg/m(2) on days 7 and 21 [1 cycle was (1) month]. Ten patients were enrolled (8 M and 2 F) with 9 having surgery. Median age and KPS were 50 (42-64) and 90 % (70-100). The median cycles post-operatively was 2 (0-4). The trial was stopped as no patient had a PFS-6. All patients are deceased. Paired plasma and tumor bortezomib concentration measurements revealed higher drug concentrations in tumor than in plasma; NFKBIA protein levels were similar in drug-treated vs. drug-naïve tumor specimens. Nuclear 20S proteasome was less in postoperative samples. Postoperative treatment with TMZ and bortezomib did not show clinical activity. Bortezomib appears to sequester in tumor but pharmacological effects on NFKBIA were not seen, possibly obscured due to downregulation of NFKBIA during tumor progression. Changes in nuclear 20S could be marker of bortezomib effect on tumor.


Assuntos
Antineoplásicos/uso terapêutico , Bortezomib/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Recidiva Local de Neoplasia/tratamento farmacológico , Adulto , Antineoplásicos/farmacocinética , Bortezomib/sangue , Bortezomib/farmacocinética , Neoplasias Encefálicas/metabolismo , Terapia Combinada , Dacarbazina/análogos & derivados , Dacarbazina/uso terapêutico , Quimioterapia Combinada , Feminino , Glioblastoma/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Inibidor de NF-kappaB alfa/metabolismo , Recidiva Local de Neoplasia/metabolismo , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Temozolomida , Resultado do Tratamento
3.
J Neurooncol ; 126(2): 309-16, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26508094

RESUMO

This study tested the hypothesis that ABT-888 (velparib), a poly (ADP-ribose) polymerase (PARP) inhibitor, can modulate temozolomide (TMZ) resistance in recurrent TMZ refractory glioblastoma patients. The combination regimen (TMZ/ABT-888) was tested using two randomized schedules (5 vs. 21 days), with 6-month progression free survival (PFS6) as the primary endpoint. The maximum tolerated dose (MTD) for TMZ using the 21 day of 28 TMZ schedule, in concert with 40 mg BID ABT-888 was determined in a phase I portion of this study, and previously reported to be 75 mg/m(2) (arm1). The MTD for ABT-888 (40 mg BID) and the 5 of 28 day TMZ (150-200 mg/m(2)) schedule was known from prior trials (arm2). Two cohorts were studied: bevacizumab (BEV) naïve (n = 151), and BEV refractory (n = 74). Overall ten patients were ineligible. The incidence rate of grade 3/4 myelosuppression over all was 20.0 %. For the BEV refractory cohort, the PFS 6 was 4.4 %; for the BEV naïve cohort, PFS6 was 17 %. Overall survival was similar for both arms in both the BEV naïve [median survival time (MST) 10.3 M; 95 % CI 8.4-12] and BEV refractory cohort (MST 4.7 M; 95 %CI 3.5-5.6). The median PFS was essentially the same for both arms and both cohorts at ~2.0 M (95 % CI 1.9-2.1).


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Benzimidazóis/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Dacarbazina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico , Adulto , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos Alquilantes/efeitos adversos , Benzimidazóis/efeitos adversos , Bevacizumab/uso terapêutico , Dacarbazina/efeitos adversos , Dacarbazina/uso terapêutico , Intervalo Livre de Doença , Quimioterapia Combinada , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Inibidores de Poli(ADP-Ribose) Polimerases/efeitos adversos , Análise de Sobrevida , Temozolomida , Resultado do Tratamento , Adulto Jovem
4.
J Neurooncol ; 118(1): 147-53, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24574050

RESUMO

Bevacizumab has been reported to cause diffusion restriction in the tumor bed of patients with malignant gliomas. This study evaluated prolonged diffusion restriction, in the corpus callosum (CC), of patients with malignant brain tumors treated with bevacizumab. We retrospectively reviewed our database of patients treated with bevacizumab for malignant brain tumors looking for those with restricted diffusion in the CC. CC ADC ratio measurements were obtained prior to and following treatment. Correlation was made with biopsy (n = 3) and MR perfusion (n = 7) and PET (n = 4). The temporal evolution of these changes relative to therapy was examined with mixed effects regression analysis. Nine patients (eight malignant gliomas, one malignant meningioma) out of 146 patients were found to have developed areas of diffusion restriction in the CC. These areas tended to enlarge and coalesce over serial MRIs and persisted for up to 22 months. Hypoperfusion was demonstrated in MR perfusion in 7/7. PET was hypometabolic in all 4. Biopsy of the CC showed no tumor in 3/3. ADC ratio measurements indicated a significant overall effect of time (F(16,60) = 11.2; p < 0.0001), consistent with persistent diffusion restriction over the measured time periods. Bevacizumab causes prolonged diffusion restriction in the CC. The negative MR perfusion, FDG PET and histopathology suggest this is a toxicity of bevacizumab and not active tumor. Awareness of these changes can assist in patient care.


Assuntos
Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Corpo Caloso/patologia , Idoso , Inibidores da Angiogênese/efeitos adversos , Inibidores da Angiogênese/uso terapêutico , Bevacizumab , Neoplasias Encefálicas/patologia , Imagem de Difusão por Ressonância Magnética , Feminino , Humanos , Processamento de Imagem Assistida por Computador , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos
5.
J Neurooncol ; 117(1): 93-101, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24449400

RESUMO

When surgery and radiation are no longer treatment options, salvage systemic therapy has been used for recurrent meningiomas with little compelling evidence to suggest effectiveness. Patients with surgery and radiation refractory recurrent meningiomas were treated with the oral multifunctional tyrosine kinase inhibitor PTK787/ZK 222584 (PTK787) at a dose of 500 mg twice a day. Each treatment cycle was 4 weeks with MRI done every 8 weeks. Twenty-five patients (14 men; 11 women) with a median age of 59 years and KPS of 80 were treated. Meningioma WHO Grade was I in 2 patients, II in 14 patients and III in 8 patients; 1 patient had a hemangiopericytoma. All patients had prior surgery, external beam radiation therapy or radiosurgery and 11 patients prior systemic chemotherapy. Median number of cycles of PTK 787 administered was 4 (range <1-22). Best response in the 22 evaluable patients was stable disease in 15 (68.2 %). Predominant PTK787 related toxicities included fatigue (60 %), hypertension (24 %) and elevated transaminases (24 %). Grade II patients had a progression free survival (PFS)-6 of 64.3 %, a median PFS of 6.5 months and an overall survival (OS) of 26.0 months; grade III patients had a PFS-6 of 37.5 %, median PFS of 3.6 months and OS 23 months. PTK787 was modestly toxic at the dose of 500 mg administered twice per day. Activity as determined by PFS-6 suggests that targeting PDGF/VEGF pathway warrants further investigation.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias Encefálicas/tratamento farmacológico , Meningioma/tratamento farmacológico , Ftalazinas/administração & dosagem , Piridinas/administração & dosagem , Terapia de Salvação , Administração Oral , Adulto , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos/efeitos adversos , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Encéfalo/efeitos da radiação , Encéfalo/cirurgia , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/radioterapia , Neoplasias Encefálicas/cirurgia , Intervalo Livre de Doença , Feminino , Humanos , Estimativa de Kaplan-Meier , Imageamento por Ressonância Magnética , Masculino , Meningioma/patologia , Meningioma/radioterapia , Meningioma/cirurgia , Pessoa de Meia-Idade , Gradação de Tumores , Recidiva Local de Neoplasia/tratamento farmacológico , Ftalazinas/efeitos adversos , Piridinas/efeitos adversos , Fatores de Tempo
6.
J Neurooncol ; 112(2): 247-55, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23354655

RESUMO

Brain metastases (BM) and leptomeningeal metastases (LM) are devastating neurologic complications. Pemetrexed is a multi-targeted anti-folate agent approved for treatment of nonsquamous non-small cell lung cancer but has anti-tumor activity in other solid tumors. We performed two trials using pemetrexed in patients with BM and LM to assess CSF penetration and anti-tumor activity. Patients were treated with intravenous pemetrexed at doses of 500 (n = 3), 750 (n = 3), 900 (n = 12) or 1,050 mg/m(2) (n = 3) every 3 weeks. Neuro-imaging was done every 6 weeks. Matched CSF and plasma samples were obtained serially from three patients with Ommaya reservoirs; the remaining patients had a single paired collection. Twenty-one patients (15 women and six men) with median age of 50 years and median KPS of 90 were treated. Primary tumors included breast (13), lung (4), colorectal (1), endometrial (1), esophageal (1) and pinealoblastoma (1). Nine patients had prior whole brain RT and median number of prior chemotherapies was two including prior methotrexate in four patients. Median pemetrexed doses administered was three (range 1-14). Responses included one partial response, ten stable disease and ten progressive disease. Median time to progression and survival was 2.7 and 7.3 months; PFS six was 22 %. No major toxicities were seen. Pemetrexed distributed from the plasma to the CSF within 1-4 h with the resulting CSF concentrations < 5 % of plasma. Pemetrexed was tolerated in solid tumor patients with CNS metastases. Limited anti-tumor activity was seen, which might have been due to low CSF concentrations, although some patients displayed prolonged benefit.


Assuntos
Antimetabólitos Antineoplásicos/farmacocinética , Neoplasias Encefálicas/tratamento farmacológico , Glutamatos/farmacocinética , Guanina/análogos & derivados , Neoplasias Meníngeas/tratamento farmacológico , Recidiva Local de Neoplasia/tratamento farmacológico , Neoplasias/tratamento farmacológico , Adulto , Idoso , Antimetabólitos Antineoplásicos/sangue , Antimetabólitos Antineoplásicos/líquido cefalorraquidiano , Antimetabólitos Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/secundário , Progressão da Doença , Feminino , Seguimentos , Glutamatos/sangue , Glutamatos/líquido cefalorraquidiano , Glutamatos/uso terapêutico , Guanina/sangue , Guanina/líquido cefalorraquidiano , Guanina/farmacocinética , Guanina/uso terapêutico , Humanos , Neoplasias Meníngeas/mortalidade , Neoplasias Meníngeas/secundário , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/mortalidade , Recidiva Local de Neoplasia/patologia , Estadiamento de Neoplasias , Neoplasias/mortalidade , Neoplasias/patologia , Pemetrexede , Projetos Piloto , Prognóstico , Taxa de Sobrevida , Distribuição Tecidual
7.
Curr Neurol Neurosci Rep ; 13(5): 346, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23512689

RESUMO

Brainstem gliomas (BGs) are a heterogenous group of gliomas that occur predominately in children. They can be separated into groups on the basis of anatomy and clinical behavior: diffuse intrinsic pontine glioma (DIPG), exophytic medullary glioma, and tectal glioma. DIPG is the commonest BG. Median age at onset is 6.5 years and median survival is less than 1 year. Adults with DIPG survive longer, suggesting a less aggressive and biologically different tumor from that in children. Patients present with cranial nerve dysfunction, long tract signs, or ataxia, either in isolation or in combination. Magnetic resonance imaging shows an infiltrative lesion occupying most of the pons and contrast enhancement is usually not prominent. Standard treatment is fractionated radiotherapy. Platelet-derived growth factor receptor alpha and epidermal growth factor receptor mutations have been identified. Inhibitors of these growth factor receptors are being evaluated in clinical trials. Exophytic medullary and tectal gliomas are relatively indolent tumors that can often be followed closely without treatment.


Assuntos
Neoplasias do Tronco Encefálico , Glioma , Neoplasias do Tronco Encefálico/diagnóstico , Neoplasias do Tronco Encefálico/epidemiologia , Neoplasias do Tronco Encefálico/terapia , Glioma/diagnóstico , Glioma/epidemiologia , Glioma/terapia , Humanos
8.
Neuro Oncol ; 25(3): 557-565, 2023 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-35948282

RESUMO

BACKGROUND: Patients with human epidermal growth factor receptor 2-positive (HER2-positive) cancers have a high incidence of central nervous system (CNS) spread, but unfortunately systemic trastuzumab which targets the HER2 receptor has little CNS penetration. The purpose of this study was to determine the maximum-tolerated dose of intrathecal trastuzumab and its efficacy in patients with HER2-positive leptomeningeal disease (LMD). METHODS: This multicenter study enrolled 34 LMD patients in a combined phase I/II study in treating patients with intrathecal trastuzumab. Any HER2-positive histology was allowed in the phase I; the phase II was limited to HER2-positive breast cancer. RESULTS: Intrathecal trastuzumab was well-tolerated, with one dose limiting toxicity of grade 4 (arachnoiditis) occurring at the 80 mg twice weekly dose. The recommended phase II dose was 80 mg intrathecally twice weekly. Twenty-six patients at dose level 80 mg were included in evaluation for efficacy: partial response was seen in 5 (19.2%) patients, stable disease was observed in 13 (50.0%), and 8 (30.8%) of the patients had progressive disease. Median overall survival (OS) for phase II dose treated patients was 8.3 months (95% CI 5.2-19.6). The phase II HER2-positive breast cancer patients median OS was 10.5 months (95% CI 5.2-20.9). Pharmacokinetic (PK) studies were limited in the setting of concurrent systemic trastuzumab administration, however, did show stable cerebrospinal fluid (CSF) concentrations with repeated dosing suggest that trastuzumab does not accumulate in the CSF in toxic concentrations. CONCLUSION: This study suggests promise for potentially improved outcomes of HER-positive LMD patients when treated with intrathecal trastuzumab while remaining safe and well-tolerated for patients.


Assuntos
Neoplasias da Mama , Carcinomatose Meníngea , Humanos , Feminino , Trastuzumab/efeitos adversos , Receptor ErbB-2/metabolismo , Neoplasias da Mama/patologia , Carcinomatose Meníngea/tratamento farmacológico , Carcinomatose Meníngea/patologia , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos
9.
Cancer ; 118(15): 3743-8, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22179954

RESUMO

BACKGROUND: Despite initial treatment with high-dose methotrexate-based regimens, many patients with primary central nervous system lymphoma (PCNSL) relapse and die from their disease. No standard of care exists at progression or relapse, but chemotherapy and in some cases radiation are usually used. Pemetrexed is a multitargeted antifolate, similar to methotrexate, but with a broader spectrum of activity. Because methotrexate is an integral part of PCSNL treatment, the authors assessed the antitumor activity and safety of pemetrexed in recurrent PCNSL. METHODS: Patients with relapsed/refractory PCNSL were enrolled in this trial. Treatment consisted of pemetrexed 900 mg/m(2) given every 3 weeks with low-dose dexamethasone, folate, and B12 supplementation. Each cycle was 6 weeks, and follow-up imaging was done before each new cycle. Treatment was continued until complete remission, progression, or toxicity. RESULTS: Eleven patients were treated, with a median age of 69.8 years and Karnofsky performance status of 70%; 10 of 11 patients had failed prior high-dose methotrexate. The median number of pemetrexed cycles given was 5, with an associated overall response rate of 55% and disease control rate of 91%. The 6-month progression-free survival (PFS) was 45%, median PFS was 5.7 months, and median overall survival was 10.1 months. Toxicities were primarily hematologic and infectious. CONCLUSIONS: Pemetrexed has single-agent activity in relapsed/refractory PCNSL. Toxicities were seen likely because of the higher than standard dose used. Further investigation of this agent or other multitargeted antifolates in PCNSL is warranted to determine optimal dose and efficacy in a more homogeneous population.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias do Sistema Nervoso Central/tratamento farmacológico , Glutamatos/administração & dosagem , Guanina/análogos & derivados , Linfoma/tratamento farmacológico , Adulto , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica , Intervalo Livre de Doença , Esquema de Medicação , Resistencia a Medicamentos Antineoplásicos , Feminino , Glutamatos/efeitos adversos , Guanina/administração & dosagem , Guanina/efeitos adversos , Humanos , Masculino , Pessoa de Meia-Idade , Pemetrexede , Recidiva , Terapia de Salvação
10.
J Neurooncol ; 107(1): 21-7, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21964740

RESUMO

Extracranial metastases from glioblastoma (GBM) are uncommon with an estimated incidence of less than 2%. We report two cases of metastatic GBM seen within an 8-week period followed by a literature review. We attempted to identify common factors or a causative mechanism. Factors that predominated among the reviewed cases included male gender, tumor location, and younger age. Causative mechanisms were not apparent. While metastatic disease remains rare, it might be occurring with increasing frequency. This trend might be due to increased diagnosis, better imaging, a more extensive physician workup, or an increase in survival. Metastatic GBM can present and progress quite rapidly, and repeat evaluations of persistent or worsening complaints among GBM patients are warranted. Early diagnosis of metastatic disease spread can help to expedite alleviation of patients' discomfort, in an already aggressive disease process.


Assuntos
Neoplasias Ósseas/secundário , Glioblastoma/patologia , Adulto , Idoso , Neoplasias Ósseas/terapia , Feminino , Glioblastoma/terapia , Humanos , Masculino , Literatura de Revisão como Assunto
11.
J Neurooncol ; 110(2): 237-43, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22875709

RESUMO

To evaluate the toxicity and maximum tolerated dose (MTD) of arsenic trioxide (ATO) in combination with temozolomide (TMZ) and radiation therapy (RT) in malignant gliomas. A 3 + 3 dose escalation study was performed in patients with newly diagnosed glioblastoma, anaplastic astrocytoma (AA), and anaplastic oligoastrocytoma (AOA). All patients received RT 59-61 Gy in 28-33 fractions, TMZ for 42 days, and ATO 1-2 h prior to RT for 5 days during the first week, then twice weekly until completing RT. Dose levels (DL) were: (1) TMZ 60 mg/m(2)/ATO 0.2 mg/kg; (2) TMZ 75 mg/m(2)/ATO 0.2 mg/kg; (3) TMZ 75 mg/m(2)/ATO 0.25 mg/kg. Dose-limiting toxicity (DLT) was defined as grade 3 non-hematologic toxicity or grade 4 toxicity of any type from enrollment until 3 weeks after finishing RT. 17 patients (13 glioblastoma, 4 AA/AOA) were accrued. Median age was 52 (range 25-80). Median KPS was 90 %. DLT's occurred at DL 2 (grade 4 transaminase elevation) and DL 3 (grade 4 neutropenia and grade 3 QTc prolongation). The MTD of TMZ 75 mg/m(2)/ATO 0.2 mg/kg was safe and well tolerated. A phase II study evaluating the efficacy of this combination is underway.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Encefálicas/terapia , Quimiorradioterapia , Glioma/terapia , Adulto , Idoso , Idoso de 80 Anos ou mais , Trióxido de Arsênio , Arsenicais/administração & dosagem , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Dacarbazina/administração & dosagem , Dacarbazina/análogos & derivados , Fracionamento da Dose de Radiação , Feminino , Glioma/mortalidade , Glioma/patologia , Humanos , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Óxidos/administração & dosagem , Prognóstico , Dosagem Radioterapêutica , Taxa de Sobrevida , Temozolomida
12.
Curr Oncol Rep ; 14(1): 85-90, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22161303

RESUMO

Although systemic therapy is the primary therapeutic modality for disseminated cancer, it plays a limited role in the treatment of brain metastases (BM). This review discusses the blood-brain barrier (BBB), interactions of systemic therapy with supportive care agents used in BM patients, the role of primary tumor sensitivity in the treatment of BM, and unique issues related to the specific primary tumor histologies. The specialized physiology of the brain vasculature that forms the BBB may preclude large and/or water-soluble systemic agents from reaching BM. Once metastases grow larger than 1-2 mm, there is preclinical and clinical evidence that the BBB is at least partially disrupted. Thus, the best treatment strategy in established BM may be to use an agent that is effective against the primary tumor regardless of its apparent BBB permeability. The use of anticonvulsants and corticosteroids must be carefully considered as they can decrease the effectiveness of systemic anti-tumor therapy. Despite the absence of level I data to routinely recommend the use of systemic therapy for solid tumor BM, these treatments should be considered in patients with good performance status and multiple, small metastases, especially if the primary tumor is chemosensitive. The systemic treatment of BM will continue to evolve as effective small-molecule inhibitors are developed and treatment regimens for each specific primary tumor are optimized.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/secundário , Corticosteroides/uso terapêutico , Anticonvulsivantes/uso terapêutico , Barreira Hematoencefálica/fisiologia , Neoplasias Encefálicas/fisiopatologia , Interações Medicamentosas , Humanos
13.
Radiology ; 255(2): 622-8, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20413772

RESUMO

PURPOSE: To derive a magnetic resonance (MR)-based imaging metric that reflects local perfusion changes resulting from the administration of angiogenic-inhibiting chemotherapy in patients with recurrent glioblastoma multiforme (GBM). MATERIALS AND METHODS: In this retrospective Institutional Review Board-approved HIPAA-compliant study, 16 patients (12 men, four women; mean age, 51.8 years + or - 15.1 [standard deviation]) with recurrent GBM received bevacizumab every 3 weeks (15 mg per kilogram of body weight) as part of a clinical trial. Baseline MR images were acquired, and follow-up images were acquired every 6 weeks thereafter until tumor progression or death. Imaging included perfusion and T1-weighted contrast material-enhanced MR imaging. Perfusion images were analyzed both with and without correction for contrast material leakage. The volumes of interest were selected as enhancing voxels on T1-weighted contrast-enhanced MR images. Relative cerebral blood volume (rCBV) maps were created from analysis of MR perfusion images. The volumes of interest were used to calculate the following parameters: size, mean rCBV, mean leakage coefficient K(2), and hyperperfusion volume (HPV), which is the fraction of the tumor with an rCBV higher than a predetermined threshold. Percent change in each parameter from baseline to first follow-up was compared with time to progression (TTP) by using a Cox proportional hazards model with calculation of hazard ratios. RESULTS: The most significant hazard ratio was seen with a DeltaHPV cutoff of rCBV greater than 1.00 (hazard ratio, 1.077; 95% confidence interval: 1.026, 1.130; P = .002). The only significant ratios greater than one were those that resulted from perfusion calculated as mean rCBV and DeltaHPV. The ratios were also higher after correction for leakage. CONCLUSION: This pilot study derived an imaging metric (HPV) that reflects local perfusion changes in GBMs. This metric was found to show a significantly improved correlation to TTP as compared with more commonly used metrics.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Angiografia por Ressonância Magnética/métodos , Neovascularização Patológica/tratamento farmacológico , Inibidores da Angiogênese/administração & dosagem , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais Humanizados , Bevacizumab , Neoplasias Encefálicas/irrigação sanguínea , Meios de Contraste , Progressão da Doença , Esquema de Medicação , Feminino , Glioblastoma/irrigação sanguínea , Humanos , Processamento de Imagem Assistida por Computador , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Valor Preditivo dos Testes , Modelos de Riscos Proporcionais , Estudos Retrospectivos
14.
J Clin Neurosci ; 64: 18-21, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30948314

RESUMO

Survival outcomes for patients with glioblastoma (GBM) are universally poor with only a small percentage of patients surviving five years beyond initial diagnosis. Activation of the immune system against tumor cells is the basis of immunotherapy and aims to facilitate long-term immune surveillance and tumor suppression. Cytomegalovirus (CMV) has emerged as an immunologic target in GBM given that tumor cells have been shown to express the CMV-associated proteins IE1 and pp65. Moreover, vaccine therapy targeting CMV antigens has promoted improved survival outcomes with long-term survivors. In this report, we present the case of a 69 year-old woman with GBM who survived seven years post-diagnosis. Following tumor resection, the patient underwent concomitant radiation and temozolomide therapy that was complicated by CMV colitis and abdominal abscesses. Despite not receiving adjuvant temozolomide, the patient demonstrated a five year progression-free survival before requiring re-resection for radiation necrosis. Following re-resection, the patient survived for two additional years. As the patient's tumor stained positive for CMV antigens IE1 and pp65, it is hypothesized that she developed an immune response against CMV during recovery that contributed to anti-tumor surveillance and prolonged survival. Overall, this case supports further investigation into the role of CMV and immunotherapy in GBM.


Assuntos
Neoplasias Encefálicas/imunologia , Colite/virologia , Infecções por Citomegalovirus/imunologia , Glioblastoma/imunologia , Hospedeiro Imunocomprometido , Idoso , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/virologia , Quimiorradioterapia/métodos , Colite/complicações , Feminino , Glioblastoma/patologia , Glioblastoma/virologia , Humanos , Proteínas Imediatamente Precoces/imunologia , Fosfoproteínas/imunologia , Temozolomida/uso terapêutico , Proteínas da Matriz Viral/imunologia , Ativação Viral/imunologia
16.
CNS Oncol ; 5(3): 145-57, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27230974

RESUMO

Anaplastic astrocytoma (AA) is a diffusely infiltrating, malignant, astrocytic, primary brain tumor. AA is currently defined by histology although future classification schemes will include molecular alterations. AA can be separated into subgroups, which share similar molecular profiles, age at diagnosis and median survival, based on 1p/19q co-deletion status and IDH mutation status. AA with co-deletion of chromosomes 1p and 19q and IDH mutation have the best prognosis. AA with IDH mutation and no 1p/19q co-deletion have intermediate prognosis and AA with wild-type IDH have the worst prognosis and share many molecular alterations with glioblastoma. Treatment of noncodeleted AA based on preliminary results from the CATNON clinical trial consists of maximal safe resection followed by radiotherapy with post-radiotherapy temozolomide (TMZ) chemotherapy. The role of concurrent TMZ and whether IDH1 subgroups benefit from TMZ is currently being evaluated in the recently completed randomized, prospective Phase III clinical trial, CATNON.


Assuntos
Astrocitoma , Neoplasias Encefálicas , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Astrocitoma/diagnóstico por imagem , Astrocitoma/genética , Astrocitoma/terapia , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Deleção Cromossômica , Cromossomos Humanos Par 1/genética , Humanos , Isocitrato Desidrogenase/genética , Mutação/genética
17.
CNS Oncol ; 4(5): 333-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26509217

RESUMO

Oligodendroglioma (WHO Grade 2) and anaplastic oligodendroglioma (WHO Grade 3) are glial tumors composed of neoplastic cellular elements that resemble oligodendrocytes. The treatment of recurrent, alkylator refractory oligodendroglial tumors is challenging given the paucity of effective treatment and lack of randomized controlled trials on which to base therapy. Notwithstanding the lack of prospective, randomized data, treatment of oligodendroglial tumors with bevacizumab can be recommended tentatively recognizing that preliminary studies suggest efficacy. Somatic mutations of the isocitrate dehydrogenase enzymes (IDH1 and IDH2) appear to play a critical role in the pathogenesis of most oligodendroglial tumors and agents that target these mutations are a potential therapeutic option. Additionally, reversal of CpG island hypermethylated phenotype status through inhibition of DNA methyltransferase with an inhibitor such as decitabine may provide a target for future studies.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Bevacizumab/uso terapêutico , Neoplasias Encefálicas/terapia , Recidiva Local de Neoplasia/terapia , Oligodendroglioma/terapia , Protocolos de Quimioterapia Combinada Antineoplásica , Neoplasias Encefálicas/genética , Humanos , Isocitrato Desidrogenase/genética , Recidiva Local de Neoplasia/genética , Oligodendroglioma/genética , Fator A de Crescimento do Endotélio Vascular/imunologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
18.
J Oncol Pract ; 11(1): e59-65, 2015 01.
Artigo em Inglês | MEDLINE | ID: mdl-25466707

RESUMO

PURPOSE: Approximately 18,500 persons are diagnosed with malignant glioma in the United States annually. Few studies have investigated the comprehensive economic costs. We reviewed the literature to examine costs to patients with malignant glioma and their families, payers, and society. METHODS: A total of 18 fully extracted studies were included. Data were collected on direct and indirect costs, and cost estimates were converted to US dollars using the conversion rate calculated from the study's publication date, and updated to 2011 values after adjustment for inflation. A standardized data abstraction form was used. Data were extracted by one reviewer and checked by another. RESULTS: Before approval of effective chemotherapeutic agents for malignant gliomas, estimated total direct medical costs in the United States for surgery and radiation therapy per patient ranged from $50,600 to $92,700. The addition of temozolomide (TMZ) and bevacizumab to glioblastoma treatment regimens has resulted in increased overall costs for glioma care. Although health care costs are now less front-loaded, they have increased over the course of illness. Analysis using a willingness-to-pay threshold of $50,000 per quality-adjusted life-year suggests that the benefits of TMZ fall on the edge of acceptable therapies. Furthermore, indirect medical costs, such as productivity losses, are not trivial. CONCLUSION: With increased chemotherapy use for malignant glioma, the paradigm for treatment and associated out-of-pocket and total medical costs continue to evolve. Larger out-of-pocket costs may influence the choice of chemotherapeutic agents, the economic implications of which should be evaluated prospectively.


Assuntos
Neoplasias Encefálicas/economia , Glioma/economia , Neoplasias Encefálicas/terapia , Canadá , Efeitos Psicossociais da Doença , Custos e Análise de Custo , Dacarbazina/análogos & derivados , Dacarbazina/economia , Dacarbazina/uso terapêutico , Tratamento Farmacológico/economia , Europa (Continente) , Glioma/terapia , Humanos , Radioterapia/economia , Temozolomida , Estados Unidos
19.
Neuro Oncol ; 17(6): 862-7, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25572329

RESUMO

BACKGROUND: Panobinostat is a histone deacetylase inhibitor with antineoplastic and antiangiogenic effects in glioma that may work synergistically with bevacizumab. We conducted a multicenter phase II trial of panobinostat combined with bevacizumab in patients with recurrent high-grade glioma (HGG). METHODS: Patients with recurrent HGG were treated with oral panobinostat 30 mg 3 times per week, every other week, in combination with bevacizumab 10 mg/kg every other week. The primary endpoint was a 6-month progression-fee survival (PFS6) rate for participants with recurrent glioblastoma (GBM). Patients with recurrent anaplastic glioma (AG) were evaluated as an exploratory arm of the study. RESULTS: At interim analysis, the GBM arm did not meet criteria for continued accrual, and the GBM arm was closed. A total of 24 patients with GBM were accrued prior to closure. The PFS6 rate was 30.4% (95%, CI 12.4%-50.7%), median PFS was 5 months (range, 3-9 months), and median overall survival (OS) was 9 months (range, 6-19 months). Accrual in the AG arm continued to completion, and a total of 15 patients were enrolled. The PFS6 rate was 46.7% (range, 21%-73%), median PFS was 7 months (range, 2-10 months), and median OS was 17 months (range, 5 months-27 months). CONCLUSIONS: This phase II study of panobinostat and bevacizumab in participants with recurrent GBM did not meet criteria for continued accrual, and the GBM cohort of the study was closed. Although it was reasonably well tolerated, the addition of panobinostat to bevacizumab did not significantly improve PFS6 compared with historical controls of bevacizumab monotherapy in either cohort.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Antineoplásicos/uso terapêutico , Bevacizumab/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Ácidos Hidroxâmicos/uso terapêutico , Indóis/uso terapêutico , Adulto , Idoso , Neoplasias Encefálicas/mortalidade , Intervalo Livre de Doença , Quimioterapia Combinada , Feminino , Glioblastoma/mortalidade , Inibidores de Histona Desacetilases/uso terapêutico , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Panobinostat , Resultado do Tratamento , Adulto Jovem
20.
Neurooncol Pract ; 1(2): 71-76, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26034619

RESUMO

BACKGROUND: Patients undergoing treatment for malignant gliomas (MGs) can encounter medical costs beyond what their insurance covers. The magnitude and type of costs experienced by patients are unknown. The purpose of this study was to have patients or their families report on the medical costs incurred during the patients MG treatment. METHODS: Patients with MG were eligible if they were within 6 months of diagnosis or tumor recurrence. Patients had to be ≥18 years of age, fluent in English, and not aphasic. Weekly logbooks were issued to patients for recording associated costs for ∼6 months or until tumor progression. "Out-of-pocket" (OOP) costs included medical and nonmedical expenses that were not reimbursed by insurance. Direct medical costs included hospital and physician bills. Direct nonmedical costs included transportation, parking, and other related items. Indirect medical costs included lost wages. Costs were analyzed to provide mean and medians with range of expenses. RESULTS: Forty-three patients provided cost data for a median of 12 weeks. There were 25 men and 18 women with a median age of 57 years (range, 24y-73y); 79% were married, and 49% reported annual income >$75 000. Health insurance coverage was preferred provider organizations for 58% of patients, and median deductible was $1 500. Median monthly OOP cost was $1 342 (mean, $2 451; range, $333.41-$17 267.16). The highest OOP median costs were medication copayments ($710; range, $0-13 611.20), transportation ($327; range, $0-$1 927), and hospital bill copayments ($403; range, $0-$4 000). Median lost wages were $7 500, and median lost days of work were 12.8. CONCLUSIONS: OOP costs for MG patients can be significant and comprise direct and indirect costs across several areas. Informing patients about expected costs could limit additional duress and allow financial support systems to be implemented.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA