Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
J Neurooncol ; 160(2): 345-350, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36355259

RESUMEN

Current standard of care for glioblastoma (GBM) includes concurrent chemoradiation and maintenance temozolomide (TMZ) with Tumor Treating Fields (TTFields). Preclinical studies suggest TTFields and radiation treatment have synergistic effects. We conducted a pilot clinical trial of concurrent chemoradiation with TTFields and report pattern of progression. MATERIALS AND METHODS: This is a single arm pilot study (clinicaltrials.gov Identifier: NCT03477110). Adult patients (age ≥ 18 years) with KPS ≥ 60 with newly diagnosed GBM were eligible. All patients received concurrent scalp-sparing radiation (60 Gy in 30 fractions), standard concurrent TMZ and TTFields. Maintenance therapy included standard TMZ and continuation of TTFields. Radiation treatment was delivered through TTFields arrays. Incidence and location of progression was documented. Distant recurrence was defined as recurrence more than 2 cm from the primary enhancing lesion. RESULTS: Thirty patients were enrolled on the trial. Twenty were male with median age 58 years (19-77 years). Median KPS was 90 (70-100). Median follow-up was 15.2 months (1.7-23.6 months). Ten (33.3%) patients had a methylated promoter status. Twenty-seven patients (90%) had progression, with median PFS of 9.3 months (range 8.5 to 11.6 months). Six patients presented with distant recurrence, with median distance from primary lesion of 5.05 cm (2.26-6.95 cm). One infratentorial progression was noted. CONCLUSIONS: We observed improved local control using concurrent chemoradiation with TTFields for patients with newly diagnosed when compared to historical controls. Further data are needed to validate this finding. TRIAL REGISTRATION: Clinicaltrials.gov Identifier NCT03477110.


Asunto(s)
Neoplasias Encefálicas , Terapia por Estimulación Eléctrica , Glioblastoma , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Antineoplásicos Alquilantes/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Terapia Combinada , Glioblastoma/tratamiento farmacológico , Proyectos Piloto , Temozolomida/uso terapéutico , Adulto Joven , Anciano
2.
JAMA Oncol ; 9(1): 112-121, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36394838

RESUMEN

Importance: Glioblastoma is the most lethal primary brain cancer. Clinical outcomes for glioblastoma remain poor, and new treatments are needed. Objective: To investigate whether adding autologous tumor lysate-loaded dendritic cell vaccine (DCVax-L) to standard of care (SOC) extends survival among patients with glioblastoma. Design, Setting, and Participants: This phase 3, prospective, externally controlled nonrandomized trial compared overall survival (OS) in patients with newly diagnosed glioblastoma (nGBM) and recurrent glioblastoma (rGBM) treated with DCVax-L plus SOC vs contemporaneous matched external control patients treated with SOC. This international, multicenter trial was conducted at 94 sites in 4 countries from August 2007 to November 2015. Data analysis was conducted from October 2020 to September 2021. Interventions: The active treatment was DCVax-L plus SOC temozolomide. The nGBM external control patients received SOC temozolomide and placebo; the rGBM external controls received approved rGBM therapies. Main Outcomes and Measures: The primary and secondary end points compared overall survival (OS) in nGBM and rGBM, respectively, with contemporaneous matched external control populations from the control groups of other formal randomized clinical trials. Results: A total of 331 patients were enrolled in the trial, with 232 randomized to the DCVax-L group and 99 to the placebo group. Median OS (mOS) for the 232 patients with nGBM receiving DCVax-L was 19.3 (95% CI, 17.5-21.3) months from randomization (22.4 months from surgery) vs 16.5 (95% CI, 16.0-17.5) months from randomization in control patients (HR = 0.80; 98% CI, 0.00-0.94; P = .002). Survival at 48 months from randomization was 15.7% vs 9.9%, and at 60 months, it was 13.0% vs 5.7%. For 64 patients with rGBM receiving DCVax-L, mOS was 13.2 (95% CI, 9.7-16.8) months from relapse vs 7.8 (95% CI, 7.2-8.2) months among control patients (HR, 0.58; 98% CI, 0.00-0.76; P < .001). Survival at 24 and 30 months after recurrence was 20.7% vs 9.6% and 11.1% vs 5.1%, respectively. Survival was improved in patients with nGBM with methylated MGMT receiving DCVax-L compared with external control patients (HR, 0.74; 98% CI, 0.55-1.00; P = .03). Conclusions and Relevance: In this study, adding DCVax-L to SOC resulted in clinically meaningful and statistically significant extension of survival for patients with both nGBM and rGBM compared with contemporaneous, matched external controls who received SOC alone. Trial Registration: ClinicalTrials.gov Identifier: NCT00045968.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Temozolomida/uso terapéutico , Estudios Prospectivos , Neoplasias Encefálicas/patología , Recurrencia , Células Dendríticas/patología , Vacunación
3.
Chin Clin Oncol ; 9(6): 75, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32819111

RESUMEN

Management of solid tumors involving the skull base are primarily managed with surgery and radiation, though proximity to important vascular and neuroanatomic structures often limit the extent of resection and permissible radiation dose. Meningiomas are the most common primary brain tumor in adults, and although the majority of skull base meningiomas are low-grade, their location in proximity to critical anatomical structures precludes aggressive surgical resection, and larger tumors are often resistant to radiation treatment. In patients with clinically aggressive, unresectable meningiomas, several molecular biomarkers of angiogenesis, as well as genetic mutations (SMO, AKT1, PIK3CA, KLF4, POLR2, SMARCE1, and TRAF7), have been shown to play a crucial role in the pathophysiology of these tumors. Pituitary adenomas are commonly slow growing tumors that are amenable to surgical resection, but tumors with higher Ki67 proliferative indices are associated with an increased risk of relapse and resistance to standard therapies. Chemotherapeutic agents and checkpoint inhibitors have been trialed, albeit with limited success, to treat these aggressive pituitary adenomas. Craniopharyngiomas are categorized as adamantinomatous and papillary subtypes, each with unique molecular mechanisms that drive pathogenesis of these tumors, and have introduced the possibility that targeted therapies may be developed for improved neurologic and endocrinological outcomes. Skull base tumors that exhibit recurrence despite surgical resection and radiation treatment pose a unique challenge, and systemic agents offer a non-invasive option of treating tumors that are refractory to conventional approaches. Recent insights into the molecular aberrations that elucidate the pathophysiology of these difficult-to-treat tumors have provided potential therapeutic targets for drug delivery. In this review, the authors discuss promising therapies and current knowledge gaps needed for the development of effective targeted agents for meningioma, pituitary adenoma, and craniopharyngioma.


Asunto(s)
Craneofaringioma , Neoplasias Meníngeas , Meningioma , Neoplasias Hipofisarias , Neoplasias de la Base del Cráneo , Adulto , Proteínas Cromosómicas no Histona , Craneofaringioma/genética , Craneofaringioma/terapia , Humanos , Factor 4 Similar a Kruppel , Neoplasias Meníngeas/terapia , Meningioma/genética , Meningioma/terapia , Recurrencia Local de Neoplasia , Neoplasias Hipofisarias/terapia , Base del Cráneo , Neoplasias de la Base del Cráneo/terapia
4.
Asia Pac J Ophthalmol (Phila) ; 9(2): 110-116, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31923035

RESUMEN

PURPOSE: The aim of this study was to identify clinical factors predictive of time to central nervous system (CNS) lymphoma or death in patients with vitreoretinal lymphoma (VRL). DESIGN: Retrospective cohort study. METHODS: Patients with VRL (n = 95 patients) from Januray 1, 1984 to July 30, 2018 were identified at a single ocular oncology center and records were retrospectively reviewed. Outcomes included Kaplan-Meier estimated time to CNS lymphoma and death. RESULTS: There were 95 patients with VRL diagnosed at mean age 67 years, of which 70 patients had follow-up with the ocular oncology service. Mean time to CNS lymphoma in patients with isolated VRL was 56 months and did not differ by age, sex, bilateral ocular involvement, retinal infiltration, subretinal pigment epithelial (sub-RPE) infiltration, or treatment with prophylactic systemic chemotherapy (P > 0.05). Mean time to death was 66 months and did not differ when comparing those with CNS lymphoma diagnosed before VRL versus after VRL versus no CNS lymphoma at any time (67 vs 60 vs 64 months, P > 0.05). Presence of sub-RPE infiltration was associated with shorter mean time to death (46 vs 76 months, P = 0.04, odds ratio 1.9). Older patient age was associated with increased risk of death (odds ratio 1.0, P = 0.02). The mean time to death did not differ by sex, bilateral ocular involvement, retinal infiltration, timing of CNS or systemic lymphoma, or treatment with prophylactic systemic chemotherapy (P > 0.05). CONCLUSIONS: Patients with VRL presenting with sub-RPE infiltration could have shorter mean survival time. Further studies are required to confirm these findings and determine whether sub-RPE infiltration is associated with more aggressive CNS lymphoma.


Asunto(s)
Neoplasias del Ojo/mortalidad , Linfoma Intraocular/mortalidad , Neoplasias de la Retina/mortalidad , Cuerpo Vítreo/patología , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias del Ojo/patología , Femenino , Angiografía con Fluoresceína , Humanos , Linfoma Intraocular/patología , Masculino , Oncología Médica , Persona de Mediana Edad , Fotograbar , Neoplasias de la Retina/patología , Estudios Retrospectivos , Tasa de Supervivencia , Ultrasonografía
5.
Neuro Oncol ; 19(10): 1380-1390, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28472509

RESUMEN

BACKGROUND: Anaplastic oligodendroglioma (AO) and anaplastic oligoastrocytoma (AOA) are chemotherapy-sensitive tumors with prolonged survival after radiochemotherapy. We report a prospective trial using induction temozolomide (TMZ) followed by myeloablative high-dose chemotherapy (HDC) with autologous stem-cell transplant (ASCT) as a potential strategy to defer radiotherapy. METHODS: Patients with AO/AOA received 6 cycles of TMZ (200 mg/m2 × 5/28 day). Responding patients were eligible for HDC (thiotepa 250 mg/m2/day × 3 days, then busulfan 3.2 mg/kg/day × 3 days), followed by ASCT. Genomic characterization was performed using next-generation sequencing. RESULTS: Forty-one patients were enrolled; 85% had 1p/19q codeleted tumors. After induction, 26 patients were eligible for HDC-ASCT and 21 agreed to proceed. There were no unexpected adverse events or toxic deaths. After median follow-up of 66 months, 2-year progression-free survival (PFS) for transplanted patients was 86%, 5-year PFS 60%, and no patient has died. Among all 1p/19q codeleted patients (N = 33), 5-year PFS was 50% and 5-year overall survival (OS) 93%, with median time to radiotherapy not reached. Next-generation sequencing disclosed typical oligodendroglioma-related mutations, including IDH1, TERT, CIC, and FUBP1 mutations in 1p/19q codeleted patients, and glioblastoma-like signatures in 1p/19q intact patients. Aside from IDH1, potentially oncogenic/actionable mutations were variable, depicting wide molecular heterogeneity within oligodendroglial tumors. CONCLUSIONS: TMZ followed by HDC-ASCT can be safely administered to patients with newly diagnosed 1p/19q codeleted AO. This strategy was associated with promising PFS and OS, suggesting that a chemotherapy-based approach may delay the need for radiotherapy and radiation-related toxicities. Raw data for further genomic and meta-analyses are publicly available at http://cbioportal.org/study?id=odg_msk_2017, accessed 6 January 2017. CLINICALTRIALS.GOV REGISTRY: NCT00588523.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Neoplasias Encefálicas/terapia , Dacarbazina/análogos & derivados , Oligodendroglioma/tratamiento farmacológico , Adulto , Anciano , Neoplasias Encefálicas/patología , Quimioradioterapia/métodos , Dacarbazina/uso terapéutico , Supervivencia sin Enfermedad , Femenino , Humanos , Isocitrato Deshidrogenasa/efectos de los fármacos , Masculino , Persona de Mediana Edad , Oligodendroglioma/genética , Oligodendroglioma/terapia , Trasplante de Células Madre , Temozolomida , Trasplante Autólogo
6.
Transl Cancer Res ; 8(Suppl 2): S162-S163, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35117090
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA