Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Nature ; 603(7903): 934-941, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35130560

RESUMEN

Diffuse intrinsic pontine glioma (DIPG) and other H3K27M-mutated diffuse midline gliomas (DMGs) are universally lethal paediatric tumours of the central nervous system1. We have previously shown that the disialoganglioside GD2 is highly expressed on H3K27M-mutated glioma cells and have demonstrated promising preclinical efficacy of GD2-directed chimeric antigen receptor (CAR) T cells2, providing the rationale for a first-in-human phase I clinical trial (NCT04196413). Because CAR T cell-induced brainstem inflammation can result in obstructive hydrocephalus, increased intracranial pressure and dangerous tissue shifts, neurocritical care precautions were incorporated. Here we present the clinical experience from the first four patients with H3K27M-mutated DIPG or spinal cord DMG treated with GD2-CAR T cells at dose level 1 (1 × 106 GD2-CAR T cells per kg administered intravenously). Patients who exhibited clinical benefit were eligible for subsequent GD2-CAR T cell infusions administered intracerebroventricularly3. Toxicity was largely related to the location of the tumour and was reversible with intensive supportive care. On-target, off-tumour toxicity was not observed. Three of four patients exhibited clinical and radiographic improvement. Pro-inflammatory cytokine levels were increased in the plasma and cerebrospinal fluid. Transcriptomic analyses of 65,598 single cells from CAR T cell products and cerebrospinal fluid elucidate heterogeneity in response between participants and administration routes. These early results underscore the promise of this therapeutic approach for patients with H3K27M-mutated DIPG or spinal cord DMG.


Asunto(s)
Astrocitoma , Neoplasias del Tronco Encefálico , Gangliósidos , Glioma , Histonas , Inmunoterapia Adoptiva , Mutación , Receptores Quiméricos de Antígenos , Astrocitoma/genética , Astrocitoma/inmunología , Astrocitoma/patología , Astrocitoma/terapia , Neoplasias del Tronco Encefálico/genética , Neoplasias del Tronco Encefálico/inmunología , Neoplasias del Tronco Encefálico/patología , Neoplasias del Tronco Encefálico/terapia , Niño , Gangliósidos/inmunología , Perfilación de la Expresión Génica , Glioma/genética , Glioma/inmunología , Glioma/patología , Glioma/terapia , Histonas/genética , Humanos , Inmunoterapia Adoptiva/métodos , Receptores Quiméricos de Antígenos/inmunología , Neoplasias de la Médula Espinal/genética , Neoplasias de la Médula Espinal/inmunología , Neoplasias de la Médula Espinal/patología , Neoplasias de la Médula Espinal/terapia
2.
Cancer Res Commun ; 2024 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-39269033

RESUMEN

A common complication of chimeric antigen receptor (CAR)-T cell therapy is immune effector cell associated neurotoxicity syndrome (ICANS), which presents with encephalopathy, aphasia, inattention, somnolence, seizures, weakness, or cerebral edema. Despite its significant morbidity, there are currently no effective targeted treatments. Given the clinical similarities between ICANS and the neurological manifestations of acute hypophosphatemia, we retrospectively reviewed 499 patients treated with CD19-targeted CAR-T cell therapy across multiple clinical trials between 2015 and 2020. In addition to clinical toxicities experienced by the patients, we also interrogated the impact of serum electrolyte data and repletion of corresponding electrolyte deficiencies with ICANS incidence, severity, and duration. Hypophosphatemia was a common occurrence in CAR T-cell recipients, and the only electrolyte derangement associated with a significantly higher cumulative incidence of ICANS. Moreover, phosphorus repletion in hypophosphatemic patients was associated with significantly decreased ICANS incidence and duration. Hypophosphatemia was uniquely associated with encephalopathy neurological adverse events, which also showed the strongest positive correlation with both ICANS and CRS severity. These findings suggest that serum phosphorus could be a reliable biomarker for ICANS, and expeditious, goal-directed phosphorus repletion in response to serum hypophosphatemia could be a safe, inexpensive, and widely available intervention for such patients.

3.
Blood Adv ; 8(6): 1474-1486, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38295285

RESUMEN

ABSTRACT: CD19 chimeric antigen receptor (CAR) T-cell therapy has proven highly effective for treating relapsed/refractory mantle cell lymphoma (MCL). However, immune effector cell-associated neurotoxicity syndrome (ICANS) remains a significant concern. This study aimed to evaluate the clinical, radiological, and laboratory correlatives associated with ICANS development after CD19 CAR T-cell therapy in patients with MCL. All patients (N = 26) who received standard-of-care brexucabtagene autoleucel until July 2022 at our institution were evaluated. Laboratory and radiographic correlatives including brain magnetic resonance imaging (MRI) and electroencephalogram (EEG) were evaluated to determine the clinical impact of ICANS. Seventeen (65%) patients experienced ICANS after treatment, with a median onset on day 6. Ten (38%) patients experienced severe (grade ≥3) ICANS. All patients with ICANS had antecedent cytokine release syndrome (CRS), but no correlation was observed between ICANS severity and CRS grade. Overall, 92% of EEGs revealed interictal changes; no patients experienced frank seizures because of ICANS. In total, 86% of patients with severe ICANS with postinfusion brain MRIs demonstrated acute neuroimaging findings not seen on pretreatment MRI. Severe ICANS was also associated with higher rates of cytopenia, coagulopathy, increased cumulative steroid exposure, and prolonged hospitalization. However, severe ICANS did not affect treatment outcomes of patients with MCL. Severe ICANS is frequently associated with a range of postinfusion brain MRI changes and abnormal EEG findings. Longer hospitalization was observed in patients with severe ICANS, especially those with abnormal acute MRI or EEG findings, but there was no discernible impact on overall treatment response and survival.


Asunto(s)
Linfoma de Células del Manto , Síndromes de Neurotoxicidad , Humanos , Adulto , Linfoma de Células del Manto/terapia , Inmunoterapia Adoptiva/efectos adversos , Proteínas Adaptadoras Transductoras de Señales , Antígenos CD19 , Encéfalo , Síndrome de Liberación de Citoquinas
4.
Proc Natl Acad Sci U S A ; 107(45): 19573-8, 2010 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-20974939

RESUMEN

Synucleins are a vertebrate-specific family of abundant neuronal proteins. They comprise three closely related members, α-, ß-, and γ-synuclein. α-Synuclein has been the focus of intense attention since mutations in it were identified as a cause for familial Parkinson's disease. Despite their disease relevance, the normal physiological function of synucleins has remained elusive. To address this, we generated and characterized αßγ-synuclein knockout mice, which lack all members of this protein family. Deletion of synucleins causes alterations in synaptic structure and transmission, age-dependent neuronal dysfunction, as well as diminished survival. Abrogation of synuclein expression decreased excitatory synapse size by ∼30% both in vivo and in vitro, revealing that synucleins are important determinants of presynaptic terminal size. Young synuclein null mice show improved basic transmission, whereas older mice show a pronounced decrement. The late onset phenotypes in synuclein null mice were not due to a loss of synapses or neurons but rather reflect specific changes in synaptic protein composition and axonal structure. Our results demonstrate that synucleins contribute importantly to the long-term operation of the nervous system and that alterations in their physiological function could contribute to the development of Parkinson's disease.


Asunto(s)
Neuronas/fisiología , Sinapsis/patología , Transmisión Sináptica/genética , Sinucleínas/genética , Sinucleínas/fisiología , Factores de Edad , Animales , Eliminación de Gen , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/análisis , Enfermedad de Parkinson/etiología , Fenotipo , Sinucleínas/deficiencia , alfa-Sinucleína/deficiencia , alfa-Sinucleína/genética , Sinucleína beta/deficiencia , Sinucleína beta/genética , gamma-Sinucleína/deficiencia , gamma-Sinucleína/genética
5.
J Neuropathol Exp Neurol ; 82(2): 160-168, 2023 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-36592076

RESUMEN

Immune effector cell-associated neurotoxicity syndrome (ICANS) is a prevalent condition seen after treatment with chimeric antigen receptor T-cell (CAR T) therapy and other cancer cell therapies. The underlying pathophysiology and neuropathology of the clinical syndrome are incompletely understood due to the limited availability of brain tissue evaluation from patient cases, and a lack of high-fidelity preclinical animal models for translational research. Here, we present the cellular and tissue neuropathologic analysis of a patient who experienced grade 4 ICANS after treatment with anti-CD19 CAR T therapy for mantle cell lymphoma. Our pathologic evaluation reveals a pattern of multifocal demyelinating leukoencephalopathy associated with a clinical course of severe ICANS. A focused analysis of glial subtypes further suggests region-specific oligodendrocyte lineage cell loss as a potential cellular and pathophysiologic correlate in severe ICANS. We propose a framework for the continuum of neuropathologic changes thus far reported across ICANS cases. Future elucidation of the mechanistic processes underlying ICANS will be critical in minimizing neurotoxicity following CAR T-cell and related immunotherapy treatments across oncologic and autoimmune diseases.


Asunto(s)
Leucoencefalopatía Multifocal Progresiva , Linfoma de Células del Manto , Síndromes de Neurotoxicidad , Receptores Quiméricos de Antígenos , Animales , Inmunoterapia Adoptiva , Síndromes de Neurotoxicidad/etiología , Síndromes de Neurotoxicidad/terapia , Proteínas Adaptadoras Transductoras de Señales
6.
Exp Neurol ; 287(Pt 3): 384-394, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26874223

RESUMEN

Stroke is the leading cause of adult disability. Yet there is a limited degree of recovery in this disease. One of the mechanisms of recovery is the formation of new connections in the brain and spinal cord after stroke: post-stroke axonal sprouting. Studies indicate that post-stroke axonal sprouting occurs in mice, rats, primates and humans. Inducing post-stroke axonal sprouting in specific connections enhances recovery; blocking axonal sprouting impairs recovery. Behavioral activity patterns after stroke modify the axonal sprouting response. A unique regenerative molecular program mediates this aspect of tissue repair in the CNS. The types of connections that are formed after stroke indicate three patterns of axonal sprouting after stroke: reactive, reparative and unbounded axonal sprouting. These differ in mechanism, location, relationship to behavioral recovery and, importantly, in their prospect for therapeutic manipulation to enhance tissue repair.


Asunto(s)
Axones/fisiología , Regeneración Nerviosa/genética , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/fisiopatología , Animales , Axones/patología , Humanos , Plasticidad Neuronal/fisiología , Neuronas/patología , Recuperación de la Función/fisiología
7.
Nat Neurosci ; 18(12): 1737-45, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26502261

RESUMEN

Stroke produces a limited process of neural repair. Axonal sprouting in cortex adjacent to the infarct is part of this recovery process, but the signal that initiates axonal sprouting is not known. Growth and differentiation factor 10 (GDF10) is induced in peri-infarct neurons in mice, non-human primates and humans. GDF10 promotes axonal outgrowth in vitro in mouse, rat and human neurons through TGFßRI and TGFßRII signaling. Using pharmacogenetic gain- and loss-of-function studies, we found that GDF10 produced axonal sprouting and enhanced functional recovery after stroke; knocking down GDF10 blocked axonal sprouting and reduced recovery. RNA sequencing from peri-infarct cortical neurons revealed that GDF10 downregulated PTEN, upregulated PI3 kinase signaling and induced specific axonal guidance molecules. Using unsupervised genome-wide association analysis of the GDF10 transcriptome, we found that it was not related to neurodevelopment, but may partially overlap with other CNS injury patterns. Thus, GDF10 is a stroke-induced signal for axonal sprouting and functional recovery.


Asunto(s)
Axones/metabolismo , Factor 10 de Diferenciación de Crecimiento/biosíntesis , Factor 10 de Diferenciación de Crecimiento/genética , Recuperación de la Función/fisiología , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Animales Recién Nacidos , Axones/patología , Humanos , Macaca mulatta , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Estudios Retrospectivos , Accidente Cerebrovascular/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA