Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Clin Cancer Res ; 30(2): 379-388, 2024 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-37939133

RESUMO

PURPOSE: Evidence suggests that MAPK pathway activation, as measured by ERK1/2 phosphorylation (p-ERK), predicts overall survival (OS) in patients with recurrent glioblastoma receiving anti-PD-1 therapy. We aimed to validate these findings in independent cohorts. EXPERIMENTAL DESIGN: In a 24-patient clinical trial on recurrent glioblastoma and high-grade gliomas, we examined the link between p-ERK levels and OS. Patients received intravenous nivolumab, followed by maximal safe resection and an intracerebral injection of either ipilimumab alone or combined with nivolumab. Biweekly adjuvant nivolumab was then administered up to five times (NCT03233152). Using REporting recommendations for tumor MARKER prognostic studies (REMARK) criteria, we conducted independent analyses for p-ERK quantification and statistical evaluations. Additional comparative analysis included prior cohorts, totaling 65 patients. Cox proportional hazards models and meta-analysis were employed to assess p-ERK as a predictive biomarker after immunotherapy. RESULTS: Lower median p-ERK+ cell density was observed compared with prior studies, likely due to variable tissue processing across cohorts. Nonetheless, high p-ERK was associated with prolonged OS, particularly in isocitrate dehydrogenase wild-type glioblastomas (P = 0.036). Median OS for high and low p-ERK patients were 55.6 and 30 weeks, respectively. Multivariable analysis reinforced p-ERK's significance in survival prediction (P = 0.011). Upon p-ERK normalization across cohorts (n = 65), meta-analysis supported the survival benefit of elevated tumor p-ERK levels (P = 0.0424). CONCLUSIONS: This study strengthens the role of p-ERK as a predictive biomarker for OS in patients with glioblastoma on immune checkpoint blockade. Future research should focus on further validation in prospective trials and the standardization of preanalytical variables influencing p-ERK quantification.


Assuntos
Glioblastoma , Humanos , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Antígeno CTLA-4 , Nivolumabe/uso terapêutico , Receptor de Morte Celular Programada 1 , Fosforilação , Sistema de Sinalização das MAP Quinases , Estudos Prospectivos , Recidiva Local de Neoplasia/tratamento farmacológico , Ipilimumab/uso terapêutico , Adjuvantes Imunológicos/uso terapêutico , Imunoterapia
2.
Neuro Oncol ; 2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-39028616

RESUMO

BACKGROUND: Glioblastoma is a highly aggressive brain cancer that is resistant to conventional immunotherapy strategies. Botensilimab, an Fc-enhanced anti-CTLA-4 antibody (FcE-aCTLA-4), has shown durable activity in "cold" and immunotherapy-refractory cancers. METHOD: We evaluated the efficacy and immune microenvironment phenotype of a mouse analogue of FcE-aCTLA-4 in treatment-refractory preclinical models of glioblastoma, both as a monotherapy and in combination with doxorubicin delivered via low-intensity pulsed ultrasound and microbubbles (LIPU/MB). Additionally, we studied 4 glioblastoma patients treated with doxorubicin, anti-PD-1 with concomitant LIPU/MB to investigate the novel effect of doxorubicin modulating FcγR expressions in tumor associated macrophages/microglia (TAMs). RESULTS: FcE-aCTLA-4 demonstrated high-affinity binding to FcγRIV, the mouse ortholog of human FcγRIIIA, which was highly expressed in TAMs in human glioblastoma, most robustly at diagnosis. Notably, FcE-aCTLA-4 mediated selective depletion of intra-tumoral regulatory T cells (Tregs) via TAM-mediated phagocytosis, while sparing peripheral Tregs. Doxorubicin, a chemotherapeutic drug with immunomodulatory functions, was found to upregulate FcγRIIIA on TAMs in glioblastoma patients who received doxorubicin and anti-PD-1 with concomitant LIPU/MB. In murine models of immunotherapy-resistant gliomas, a combinatorial regimen of FcE-aCTLA-4, anti-PD-1, and doxorubicin with LIPU/MB, achieved a 90% cure rate, that was associated robust infiltration of activated CD8+ T cells and establishment of immunological memory as evidenced by rejection upon tumor rechallenge. CONCLUSION: Our findings demonstrate that FcE-aCTLA-4 promotes robust immunomodulatory and anti-tumor effects in murine gliomas and is significantly enhanced when combined with anti-PD-1, doxorubicin, and LIPU/MB. We are currently investigating this combinatory strategy in a clinical trial (clinicaltrials.gov NCT05864534).

3.
Nat Commun ; 15(1): 4698, 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38844770

RESUMO

Given the marginal penetration of most drugs across the blood-brain barrier, the efficacy of various agents remains limited for glioblastoma (GBM). Here we employ low-intensity pulsed ultrasound (LIPU) and intravenously administered microbubbles (MB) to open the blood-brain barrier and increase the concentration of liposomal doxorubicin and PD-1 blocking antibodies (aPD-1). We report results on a cohort of 4 GBM patients and preclinical models treated with this approach. LIPU/MB increases the concentration of doxorubicin by 2-fold and 3.9-fold in the human and murine brains two days after sonication, respectively. Similarly, LIPU/MB-mediated blood-brain barrier disruption leads to a 6-fold and a 2-fold increase in aPD-1 concentrations in murine brains and peritumoral brain regions from GBM patients treated with pembrolizumab, respectively. Doxorubicin and aPD-1 delivered with LIPU/MB upregulate major histocompatibility complex (MHC) class I and II in tumor cells. Increased brain concentrations of doxorubicin achieved by LIPU/MB elicit IFN-γ and MHC class I expression in microglia and macrophages. Doxorubicin and aPD-1 delivered with LIPU/MB results in the long-term survival of most glioma-bearing mice, which rely on myeloid cells and lymphocytes for their efficacy. Overall, this translational study supports the utility of LIPU/MB to potentiate the antitumoral activities of doxorubicin and aPD-1 for GBM.


Assuntos
Barreira Hematoencefálica , Neoplasias Encefálicas , Doxorrubicina , Microbolhas , Receptor de Morte Celular Programada 1 , Doxorrubicina/farmacologia , Doxorrubicina/administração & dosagem , Doxorrubicina/uso terapêutico , Doxorrubicina/análogos & derivados , Animais , Humanos , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/metabolismo , Camundongos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Glioma/tratamento farmacológico , Glioma/imunologia , Glioma/patologia , Encéfalo/metabolismo , Encéfalo/efeitos dos fármacos , Feminino , Sistemas de Liberação de Medicamentos , Ondas Ultrassônicas , Glioblastoma/tratamento farmacológico , Glioblastoma/imunologia , Glioblastoma/patologia , Masculino , Microglia/efeitos dos fármacos , Microglia/metabolismo , Camundongos Endogâmicos C57BL , Anticorpos Monoclonais Humanizados/uso terapêutico , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/farmacologia , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/administração & dosagem , Polietilenoglicóis
4.
Neurosurgery ; 92(4): 795-802, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36512809

RESUMO

BACKGROUND: Transradial access (TRA) recently has gained popularity among neurointerventionalists. However, hesitation to its use for mechanical thrombectomy (MT) remains. OBJECTIVE: To evaluate and describe the evolution of TRA for MT. METHODS: We performed a retrospective analysis of patients undergoing TRA for MT. We performed a chronological ternary analysis to assess the impact of experience. We assessed the impact of a guide catheter designed specifically for TRA. RESULTS: We identified 53 patients who underwent TRA for MT. There was a statistically significant decrease in contrast use (148.9 vs 109.3 vs 115.2 cc), procedure time (62.4 vs 44.7 vs 41.3 minutes), fluoroscopy time (39.2 vs 44.7 vs 41.3 minutes), and puncture-to-recanalization time (40.6 vs 27.3 vs 29.4) over time. There was trend toward improved thrombolysis in cerebral infarction ≥ 2b recanalization rate (72.2% vs 77.8% vs 100%) over time. The introduction of a radial-specific catheter had a statistically significant positive impact on contrast use (133.8 vs 93 cc, P = .043), procedure time (54.2 vs 36.4 minutes, P = .003), fluoroscopy time (33.7 vs 19.8 minutes, P = .004), puncture-to-recanalization time (35.8 vs 25.1 minutes, P = .016), and thrombolysis in cerebral infarction ≥ 2b recanalization rate (71.4% vs 100%, P = .016). CONCLUSION: TRA is a safe and effective route of endovascular access for MT. Experience with this technique improves its efficacy and efficiency. The introduction of a TRA-specific catheter expands the armamentarium of the neurointerventionalist and may facilitate lesion access during MT procedures. Continued development of radial-specific devices may further improve MT outcomes.


Assuntos
Infarto Cerebral , Trombectomia , Humanos , Estudos Retrospectivos , Trombectomia/métodos , Resultado do Tratamento , Artéria Radial/cirurgia
5.
Nat Commun ; 14(1): 1566, 2023 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-36949040

RESUMO

Whereas the contribution of tumor microenvironment to the profound immune suppression of glioblastoma (GBM) is clear, tumor-cell intrinsic mechanisms that regulate resistance to CD8 T cell mediated killing are less understood. Kinases are potentially druggable targets that drive tumor progression and might influence immune response. Here, we perform an in vivo CRISPR screen to identify glioma intrinsic kinases that contribute to evasion of tumor cells from CD8 T cell recognition. The screen reveals checkpoint kinase 2 (Chek2) to be the most important kinase contributing to escape from CD8 T-cell recognition. Genetic depletion or pharmacological inhibition of Chek2 with blood-brain-barrier permeable drugs that are currently being evaluated in clinical trials, in combination with PD-1 or PD-L1 blockade, lead to survival benefit in multiple preclinical glioma models. Mechanistically, loss of Chek2 enhances antigen presentation, STING pathway activation and PD-L1 expression in mouse gliomas. Analysis of human GBMs demonstrates that Chek2 expression is inversely associated with antigen presentation and T-cell activation. Collectively, these results support Chek2 as a promising target for enhancement of response to immune checkpoint blockade therapy in GBM.


Assuntos
Glioblastoma , Glioma , Humanos , Animais , Camundongos , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Antígeno B7-H1 , Quinase 1 do Ponto de Checagem , Glioma/tratamento farmacológico , Glioma/genética , Glioma/patologia , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Linfócitos T CD8-Positivos , Imunidade , Microambiente Tumoral
6.
Nat Med ; 25(12): 1839-1842, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31768065

RESUMO

Histiocytoses are clonal hematopoietic disorders frequently driven by mutations mapping to the BRAF and MEK1 and MEK2 kinases. Currently, however, the developmental origins of histiocytoses in patients are not well understood, and clinically meaningful therapeutic targets outside of BRAF and MEK are undefined. In this study, we uncovered activating mutations in CSF1R and rearrangements in RET and ALK that conferred dramatic responses to selective inhibition of RET (selpercatinib) and crizotinib, respectively, in patients with histiocytosis.


Assuntos
Quinase do Linfoma Anaplásico/genética , Histiocitose/genética , Proteínas Proto-Oncogênicas c-ret/genética , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Adolescente , Adulto , Aminopiridinas/farmacologia , Benzotiazóis/farmacologia , Criança , Pré-Escolar , Feminino , Genoma Humano , Neoplasias Hematológicas/tratamento farmacológico , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/patologia , Histiocitose/tratamento farmacológico , Histiocitose/patologia , Humanos , Lactente , Masculino , Mutação , Ácidos Picolínicos/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Pirazóis/farmacologia , Piridinas/farmacologia , Pirróis/farmacologia , Receptores Proteína Tirosina Quinases/genética , Gêmeos Monozigóticos , Sequenciamento do Exoma , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA