Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Neuro Oncol ; 25(12): 2239-2249, 2023 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-37402650

RESUMO

BACKGROUND: Chimeric antigen receptor (CAR) T-cells targeting CD19 have been established as a leading engineered T-cell therapy for B-cell lymphomas; however, data for patients with central nervous system (CNS) involvement are limited. METHODS: We retrospectively report on CNS-specific toxicities, management, and CNS response of 45 consecutive CAR T-cell transfusions for patients with active CNS lymphoma at the Massachusetts General Hospital over a 5-year period. RESULTS: Our cohort includes 17 patients with primary CNS lymphoma (PCNSL; 1 patient with 2 CAR T-cell transfusions) and 27 patients with secondary CNS lymphoma (SCNSL). Mild ICANS (grade 1-2) was observed after 19/45 transfusions (42.2%) and severe immune effector cell-associated neurotoxicity syndrome (ICANS) (grade 3-4) after 7/45 transfusions (15.6%). A larger increase in C-reactive protein (CRP) levels and higher rates of ICANS were detected in SCNSL. Early fever and baseline C-reactive protein levels were associated with ICANS occurrence. CNS response was seen in 31 cases (68.9%), including a complete response of CNS disease in 18 cases (40.0%) which lasted for a median of 11.4 ±â€…4.5 months. Dexamethasone dose at time of lymphodepletion (but not at or after CAR T-cell transfusion) was associated with an increased risk for CNS progression (hazard ratios [HR] per mg/d: 1.16, P = .031). If bridging therapy was warranted, the use of ibrutinib translated into favorable CNS-progression-free survival (5 vs. 1 month, HR 0.28, CI 0.1-0.7; P = .010). CONCLUSIONS: CAR T-cells exhibit promising antitumor effects and a favorable safety profile in CNS lymphoma. Further evaluation of the role of bridging regimens and corticosteroids is warranted.


Assuntos
Neoplasias do Sistema Nervoso Central , Linfoma , Síndromes Neurotóxicas , Receptores de Antígenos Quiméricos , Humanos , Imunoterapia Adotiva/efeitos adversos , Proteína C-Reativa , Estudos Retrospectivos , Linfoma/terapia , Neoplasias do Sistema Nervoso Central/terapia , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/terapia , Sistema Nervoso Central , Linfócitos T
2.
Clin Cancer Res ; 29(16): 3017-3025, 2023 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-37327319

RESUMO

PURPOSE: We evaluated the efficacy of bavituximab-a mAb with anti-angiogenic and immunomodulatory properties-in newly diagnosed patients with glioblastoma (GBM) who also received radiotherapy and temozolomide. Perfusion MRI and myeloid-related gene transcription and inflammatory infiltrates in pre-and post-treatment tumor specimens were studied to evaluate on-target effects (NCT03139916). PATIENTS AND METHODS: Thirty-three adults with IDH--wild-type GBM received 6 weeks of concurrent chemoradiotherapy, followed by 6 cycles of temozolomide (C1-C6). Bavituximab was given weekly, starting week 1 of chemoradiotherapy, for at least 18 weeks. The primary endpoint was proportion of patients alive at 12 months (OS-12). The null hypothesis would be rejected if OS-12 was ≥72%. Relative cerebral blood flow (rCBF) and vascular permeability (Ktrans) were calculated from perfusion MRIs. Peripheral blood mononuclear cells and tumor tissue were analyzed pre-treatment and at disease progression using RNA transcriptomics and multispectral immunofluorescence for myeloid-derived suppressor cells (MDSC) and macrophages. RESULTS: The study met its primary endpoint with an OS-12 of 73% (95% confidence interval, 59%-90%). Decreased pre-C1 rCBF (HR, 4.63; P = 0.029) and increased pre-C1 Ktrans were associated with improved overall survival (HR, 0.09; P = 0.005). Pre-treatment overexpression of myeloid-related genes in tumor tissue was associated with longer survival. Post-treatment tumor specimens contained fewer immunosuppressive MDSCs (P = 0.01). CONCLUSIONS: Bavituximab has activity in newly diagnosed GBM and resulted in on-target depletion of intratumoral immunosuppressive MDSCs. Elevated pre-treatment expression of myeloid-related transcripts in GBM may predict response to bavituximab.

3.
JAMA Neurol ; 80(3): 227-228, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36648934

RESUMO

This Viewpoint discusses the challenges and opportunities of including patients with glioma in clinical trials.


Assuntos
Neoplasias Encefálicas , Glioma , Humanos , Neoplasias Encefálicas/terapia , Glioma/terapia , Ensaios Clínicos como Assunto
4.
Nat Commun ; 13(1): 1325, 2022 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-35289329

RESUMO

High-grade meningiomas are associated with neuro-cognitive morbidity and have limited treatments. High-grade meningiomas harbor an immunosuppressive tumor microenvironment (TME) and programmed death-ligand 1 (PD-L1) expression may contribute to their aggressive phenotype. Here, we present the results of a single-arm, open-label phase 2 trial (NCT03279692) evaluating the efficacy of pembrolizumab, a PD-1 inhibitor, in a cohort of 25 evaluable patients with recurrent and progressive grade 2 and 3 meningiomas. The primary endpoint is the proportion of patients alive and progression-free at 6 months (PFS-6). Secondary endpoints include progression-free and overall survival, best intracranial response, and toxicity. Our study has met its primary endpoint and achieved a PFS-6 rate of 0.48 (90% exact CI: 0.31-0.66) and a median PFS of 7.6 months (90% CI: 3.4-12.9 months). Twenty percent of patients have experienced one (or more) grade-3 or higher treatment-related adverse events. These results suggest that pembrolizumab exerts promising efficacy on a subset of these tumors. Further studies are needed to identify the biological facets within the meningioma TME that may drive response to immune-based therapies.


Assuntos
Neoplasias Meníngeas , Meningioma , Anticorpos Monoclonais Humanizados/efeitos adversos , Progressão da Doença , Humanos , Neoplasias Meníngeas/tratamento farmacológico , Meningioma/tratamento farmacológico , Microambiente Tumoral
6.
Artigo em Inglês | MEDLINE | ID: mdl-32923886

RESUMO

PURPOSE: Despite the high frequency of EGFR genetic alterations in glioblastoma (GBM), EGFR-targeted therapies have not had success in this disease. To improve the likelihood of efficacy, we targeted adult patients with recurrent GBM enriched for EGFR gene amplification, which occurs in approximately half of GBM, with dacomitinib, a second-generation, irreversible epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor that penetrates the blood-brain barrier, in a multicenter phase II trial. PATIENTS AND METHODS: We retrospectively explored whether previously described EGFR extracellular domain (ECD)-sensitizing mutations in the context of EGFR gene amplification could predict response to dacomitinib, and in a predefined subset of patients, we measured post-treatment intratumoral dacomitinib levels to verify tumor penetration. RESULTS: We found that dacomitinib effectively penetrates contrast-enhancing GBM tumors. Among all 56 treated patients, 8 (14.3%) had a clinical benefit as defined by a duration of treatment of at least 6 months, of whom 5 (8.9%) remained progression free for at least 1 year. Presence of EGFRvIII or EGFR ECD missense mutation was not associated with clinical benefit. We evaluated the pretreatment transcriptome in circulating extracellular vesicles (EVs) by RNA sequencing in a subset of patients and identified a signature that distinguished patients who had durable benefit versus those with rapid progression. CONCLUSION: While dacomitinib was not effective in most patients with EGFR-amplified GBM, a subset experienced a durable, clinically meaningful benefit. Moreover, EGFRvIII and EGFR ECD mutation status in archival tumors did not predict clinical benefit. RNA signatures in circulating EVs may warrant investigation as biomarkers of dacomitinib efficacy in GBM.

7.
Blood ; 133(20): 2212-2221, 2019 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-30808634

RESUMO

Chimeric antigen receptor (CAR) T cells have emerged as a promising class of cell-based immunotherapy in refractory malignancies. Neurotoxicity represents a common and potentially life-threatening adverse effect of CAR T cells, and clinical experience is limited. Here, we describe the clinical presentation and management of 25 adult patients who presented with neurotoxic syndromes after CAR T-cell therapy at the Massachusetts General Hospital. This cohort includes 24 patients treated with CD19-directed CAR T cells for non-Hodgkin lymphoma (n = 23) and acute lymphoblastic leukemia (n = 1), and 1 patient treated with α-fetoprotein-directed CAR T cells for hepatocellular carcinoma (n = 1). Twelve of the 25 patients (48%) developed grade 1-2 neurotoxicity and 13 patients (52%) presented with grade 3-4 neurotoxicity. We found that lower platelet counts at time of CAR T-cell infusion were associated with more severe neurotoxicity (P = .030). Cytokine release syndrome occurred in 24 of 25 patients (96%). Serum levels of ferritin peaked with onset of neurologic symptoms, and higher ferritin levels were associated with higher neurotoxicity grade. Grade 3-4 neurotoxicity correlated negatively with overall survival (OS) (P = .013). Median OS of the entire cohort was 54.7 weeks. Eight patients (32%) with grade 3-4 neurotoxicity were deceased at database closure, whereas none died with neurotoxicity grade 1-2. High pretreatment lactate dehydrogenase was frequently encountered in lymphoma patients with grade 3-4 neurotoxicity and correlated negatively with progression-free survival (P = .048). We did not find evidence that steroid use ≥7 days altered the patient's outcome when compared with <7 days of steroids. Management of CAR T cell-mediated neurotoxicity warrants evaluation in prospective clinical trials.


Assuntos
Imunoterapia Adotiva/efeitos adversos , Síndromes Neurotóxicas/diagnóstico , Síndromes Neurotóxicas/etiologia , Adulto , Idoso , Biomarcadores/análise , Carcinoma Hepatocelular/terapia , Estudos de Coortes , Gerenciamento Clínico , Feminino , Humanos , Imunoterapia Adotiva/métodos , Neoplasias Hepáticas/terapia , Linfoma não Hodgkin/terapia , Masculino , Pessoa de Meia-Idade , Síndromes Neurotóxicas/terapia , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Resultado do Tratamento , Adulto Jovem
8.
Nat Med ; 25(3): 477-486, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30742122

RESUMO

Glioblastoma is the most common primary malignant brain tumor in adults and is associated with poor survival. The Ivy Foundation Early Phase Clinical Trials Consortium conducted a randomized, multi-institution clinical trial to evaluate immune responses and survival following neoadjuvant and/or adjuvant therapy with pembrolizumab in 35 patients with recurrent, surgically resectable glioblastoma. Patients who were randomized to receive neoadjuvant pembrolizumab, with continued adjuvant therapy following surgery, had significantly extended overall survival compared to patients that were randomized to receive adjuvant, post-surgical programmed cell death protein 1 (PD-1) blockade alone. Neoadjuvant PD-1 blockade was associated with upregulation of T cell- and interferon-γ-related gene expression, but downregulation of cell-cycle-related gene expression within the tumor, which was not seen in patients that received adjuvant therapy alone. Focal induction of programmed death-ligand 1 in the tumor microenvironment, enhanced clonal expansion of T cells, decreased PD-1 expression on peripheral blood T cells and a decreasing monocytic population was observed more frequently in the neoadjuvant group than in patients treated only in the adjuvant setting. These findings suggest that the neoadjuvant administration of PD-1 blockade enhances both the local and systemic antitumor immune response and may represent a more efficacious approach to the treatment of this uniformly lethal brain tumor.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Antineoplásicos Imunológicos/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Recidiva Local de Neoplasia/tratamento farmacológico , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Microambiente Tumoral/imunologia , Adulto , Idoso , Neoplasias Encefálicas/imunologia , Quimioterapia Adjuvante , Feminino , Glioblastoma/imunologia , Humanos , Masculino , Pessoa de Meia-Idade , Terapia Neoadjuvante , Recidiva Local de Neoplasia/imunologia , Procedimentos Neurocirúrgicos , Receptor de Morte Celular Programada 1/imunologia , Taxa de Sobrevida , Linfócitos T/imunologia
9.
Artigo em Inglês | MEDLINE | ID: mdl-32914038

RESUMO

PURPOSE: Adequately prioritizing the numerous therapies and biomarkers available in late-stage testing for patients with glioblastoma (GBM) requires an efficient clinical testing platform. We developed and implemented INSIGhT (Individualized Screening Trial of Innovative Glioblastoma Therapy) as a novel adaptive platform trial (APT) to develop precision medicine approaches in GBM. METHODS: INSIGhT compares experimental arms with a common control of standard concurrent temozolomide and radiation therapy followed by adjuvant temozolomide. The primary end point is overall survival. Patients with newly diagnosed unmethylated GBM who are IDH R132H mutation negative and with genomic data available for biomarker grouping are eligible. At the initiation of INSIGhT, three experimental arms (neratinib, abemaciclib, and CC-115), each with a proposed genomic biomarker, are tested simultaneously. Initial randomization is equal across arms. As the trial progresses, randomization probabilities adapt on the basis of accumulating results using Bayesian estimation of the biomarker-specific probability of treatment impact on progression-free survival. Treatment arms may drop because of low probability of treatment impact on overall survival, and new arms may be added. Detailed information on the statistical model and randomization algorithm is provided to stimulate discussion on trial design choices more generally and provide an example for other investigators developing APTs. CONCLUSION: INSIGhT (NCT02977780) is an ongoing novel biomarker-based, Bayesian APT for patients with newly diagnosed unmethylated GBM. Our goal is to dramatically shorten trial execution timelines while increasing scientific power of results and biomarker discovery using adaptive randomization. We anticipate that trial execution efficiency will also be improved by using the APT format, which allows for the collaborative addition of new experimental arms while retaining the overall trial structure.

10.
Neurol Clin ; 36(3): 627-651, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30072074

RESUMO

Neurologic complications of systemic cancer therapy encompass a range of symptoms that can affect the central nervous system (CNS), peripheral nervous system (PNS), or both. Their incidence will likely increase as novel agents, such as targeted therapy and immunotherapy, become incorporated into standard treatment regimens, and the number of long-term cancer survivors rises. Recognizing the common adverse effects of treatment is important to avoid misdiagnosis of cancer recurrence in the nervous system or paraneoplastic disease. Ongoing has focused on identifying patient-specific risk factors that predispose to drug toxicity and the development of appropriate preventive and therapeutic strategies.


Assuntos
Antineoplásicos/efeitos adversos , Neoplasias/tratamento farmacológico , Síndromes Neurotóxicas , Humanos , Síndromes Neurotóxicas/diagnóstico , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/patologia
11.
Nat Commun ; 9(1): 1474, 2018 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-29662077

RESUMO

Inhibitors of the mutant isocitrate dehydrogenase 1 (IDH1) entered recently in clinical trials for glioma treatment. Mutant IDH1 produces high levels of 2-hydroxyglurate (2HG), thought to initiate oncogenesis through epigenetic modifications of gene expression. In this study, we show the initial evidence of the pharmacodynamics of a new mutant IDH1 inhibitor in glioma patients, using non-invasive 3D MR spectroscopic imaging of 2HG. Our results from a Phase 1 clinical trial indicate a rapid decrease of 2HG levels by 70% (CI 13%, P = 0.019) after 1 week of treatment. Importantly, inhibition of mutant IDH1 may lead to the reprogramming of tumor metabolism, suggested by simultaneous changes in glutathione, glutamine, glutamate, and lactate. An inverse correlation between metabolic changes and diffusion MRI indicates an effect on the tumor-cell density. We demonstrate a feasible radiopharmacodynamics approach to support the rapid clinical translation of rationally designed drugs targeting IDH1/2 mutations for personalized and precision medicine of glioma patients.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Inibidores Enzimáticos/farmacologia , Glioma/tratamento farmacológico , Glutaratos/antagonistas & inibidores , Isocitrato Desidrogenase/antagonistas & inibidores , Espectroscopia de Ressonância Magnética/métodos , Adulto , Antineoplásicos/farmacocinética , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/genética , Esquema de Medicação , Monitoramento de Medicamentos/métodos , Inibidores Enzimáticos/farmacocinética , Feminino , Expressão Gênica , Glioma/diagnóstico por imagem , Glioma/enzimologia , Glioma/genética , Glutaratos/metabolismo , Humanos , Imageamento Tridimensional/métodos , Isocitrato Desidrogenase/genética , Isocitrato Desidrogenase/metabolismo , Masculino , Pessoa de Meia-Idade , Mutação , Gradação de Tumores
12.
Neurooncol Pract ; 5(1): 64-68, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31385986

RESUMO

BACKGROUND: Treatment of aggressive pituitary adenomas typically involves a multimodality approach based on histopathological features and may include pharmacotherapy, surgery, and occasionally radiation therapy. In cases of treatment-refractory tumor progression, chemotherapy may be considered; however, no standard chemotherapeutic regimen has been established. Literature review suggests that temozolomide may have a beneficial role in a subset of cases. To understand the efficacy of temozolomide in progressive pituitary tumors, we reviewed the outcomes of cases at our center. METHODS: We performed a retrospective chart review to report the outcome and unique features of 7 patients with aggressive functioning pituitary adenomas or carcinomas treated with temozolomide. Tumor pathology included somatotroph (n = 1), corticotroph (n = 3), and lactotroph (n = 3) tumors. RESULTS: Four of the 7 patients had at least 2 prior resections, and all had prior radiation and surgery before treatment with temozolomide. Notably, all patients showed response to therapy, defined as either stable disease (43%) or partial response (57%). Median progression-free survival was 1.66 years, and median overall survival was 4 years. CONCLUSION: Our data suggest that temozolomide has an important role in the management of aggressive functioning pituitary tumors that are resistant to standard therapies, and that optimization of therapy with temozolomide may involve individualized regimens. Future prospective clinical trials should be considered.

14.
Continuum (Minneap Minn) ; 18(2): 355-65, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22810132

RESUMO

PURPOSE OF REVIEW: The purpose of this article is to review neurologic complications associated with systemic anticancer therapies. RECENT FINDINGS: Although neurologic complications from traditional chemotherapies are well described, most neurologists are less familiar with complications from agents that target specific pathways or receptors. This article also reviews the most common neurologic adverse effects associated with newer targeted agents. SUMMARY: Patients with cancer are living longer because of earlier diagnoses and remarkable improvements in treatments. Unfortunately, both traditional chemotherapies and newer targeted agents are known to cause neurologic symptoms that can impact quality of life and play a role in limiting potential treatments. Acute, subacute, and chronic syndromes may affect the central or peripheral nervous system. Since treatments for therapy-induced neurotoxicity are limited, awareness of common neurologic complications is important to prevent permanent damage.


Assuntos
Antineoplásicos/efeitos adversos , Neoplasias/tratamento farmacológico , Doenças do Sistema Nervoso/induzido quimicamente , Sistema Nervoso/efeitos dos fármacos , Síndromes Neurotóxicas/etiologia , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
15.
Semin Neurol ; 32(4): 476-86, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23361490

RESUMO

Cancer therapy can cause several neurotoxic syndromes that are associated with distinct findings on cranial imaging. With the use of more aggressive and combined treatment modalities in oncology and prolonged overall patient survival, neurotoxicity has been reported with increasing frequency in patients with brain cancer and malignancies outside the nervous system. Both cranial irradiation and chemotherapy can be harmful to the nervous system, and be associated with acute and chronic nervous system toxicity. Here we discuss features and imaging characteristics of common neurotoxic syndromes, such as cerebrovascular complications, reversible leukoencephalopathy syndrome, progressive white matter injury, and diffuse brain atrophy. Neurologist and oncologists need to be familiar with the pattern, time course, and evolution of both acute and long-term neurologic complications of cancer therapy. Identification of causative agents and appropriate distinction between treatment-related toxicity and tumor-associated complications are critical steps to improve treatment monitoring and overall patient care.


Assuntos
Diagnóstico por Imagem/métodos , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Síndromes Neurotóxicas/diagnóstico , Animais , Antineoplásicos/efeitos adversos , Diagnóstico por Imagem/tendências , Humanos , Neoplasias/epidemiologia , Síndromes Neurotóxicas/epidemiologia
16.
Neuropsychopharmacology ; 35(10): 2101-9, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20555311

RESUMO

Modafinil differs from other arousal-enhancing agents in chemical structure, neurochemical profile, and behavioral effects. Most functional neuroimaging studies to date examined the effect of modafinil only on information processing underlying executive cognition, but cognitive enhancers in general have been shown to have pronounced effects on emotional behavior, too. We examined the effect of modafinil on neural circuits underlying affective processing and cognitive functions. Healthy volunteers were enrolled in this double-blinded placebo-controlled trial (100 mg/day for 7 days). They underwent BOLD fMRI while performing an emotion information-processing task that activates the amygdala and two prefrontally dependent cognitive tasks-a working memory (WM) task and a variable attentional control (VAC) task. A clinical assessment that included measurement of blood pressure, heart rate, the Hamilton anxiety scale, and the profile of mood state (POMS) questionnaire was also performed on each test day. BOLD fMRI revealed significantly decreased amygdala reactivity to fearful stimuli on modafinil compared with the placebo condition. During executive cognition tasks, a WM task and a VAC task, modafinil reduced BOLD signal in the prefrontal cortex and anterior cingulate. Although not statistically significant, there were trends for reduced anxiety, for decreased fatigue-inertia and increased vigor-activity, as well as decreased anger-hostility on modafinil. Modafinil in low doses has a unique physiologic profile compared with stimulant drugs: it enhances the efficiency of prefrontal cortical cognitive information processing, while dampening reactivity to threatening stimuli in the amygdala, a brain region implicated in anxiety.


Assuntos
Compostos Benzidrílicos/farmacologia , Mapeamento Encefálico , Encéfalo/efeitos dos fármacos , Cognição/efeitos dos fármacos , Emoções/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Adulto , Encéfalo/irrigação sanguínea , Estudos Cross-Over , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Humanos , Processamento de Imagem Assistida por Computador/métodos , Imageamento por Ressonância Magnética/métodos , Masculino , Modafinila , Testes Neuropsicológicos , Oxigênio/sangue , Estatística como Assunto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA