Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Acta Neuropathol Commun ; 12(1): 15, 2024 01 22.
Article in English | MEDLINE | ID: mdl-38254244

ABSTRACT

Brain metastases occur in 1% of sarcoma cases and are associated with a median overall survival of 6 months. We report a rare case of a brain metastasis with unique radiologic and histopathologic features in a patient with low grade fibromyxoid sarcoma (LGFMS) previously treated with immune checkpoint inhibitor (ICI) therapy. The lone metastasis progressed in the midbrain tegmentum over 15 months as a non-enhancing, T2-hyperintense lesion with peripheral diffusion restriction, mimicking a demyelinating lesion. Histopathology of the lesion at autopsy revealed a rich infiltrate of tumor-associated macrophages (TAMs) with highest density at the leading edge of the metastasis, whereas there was a paucity of lymphocytes, suggestive of an immunologically cold environment. Given the important immunosuppressive and tumor-promoting functions of TAMs in gliomas and carcinoma/melanoma brain metastases, this unusual case provides an interesting example of a dense TAM infiltrate in a much rarer sarcoma brain metastasis.


Subject(s)
Brain Neoplasms , Glioma , Sarcoma , Humans , Tumor-Associated Macrophages , Brain , Tumor Microenvironment
2.
Blood Adv ; 8(6): 1474-1486, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38295285

ABSTRACT

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.


Subject(s)
Lymphoma, Mantle-Cell , Neurotoxicity Syndromes , Humans , Adult , Lymphoma, Mantle-Cell/therapy , Immunotherapy, Adoptive/adverse effects , Adaptor Proteins, Signal Transducing , Antigens, CD19 , Brain , Cytokine Release Syndrome
3.
J Neuropathol Exp Neurol ; 82(2): 160-168, 2023 01 20.
Article in English | MEDLINE | ID: mdl-36592076

ABSTRACT

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.


Subject(s)
Leukoencephalopathy, Progressive Multifocal , Lymphoma, Mantle-Cell , Neurotoxicity Syndromes , Receptors, Chimeric Antigen , Animals , Immunotherapy, Adoptive , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/therapy , Adaptor Proteins, Signal Transducing
4.
Mult Scler ; 28(10): 1651-1654, 2022 09.
Article in English | MEDLINE | ID: mdl-35876468

ABSTRACT

Primary central nervous system (CNS) histiocytic sarcoma is a rare hematolymphoid malignancy with features of mature histiocytes and carries a poor prognosis. We describe a unique case in which a 50-year-old woman presented with recurrent acute brainstem syndrome, area postrema syndrome, and myelitis with corresponding magnetic resonance imaging (MRI) lesions meeting diagnostic criteria for seronegative neuromyelitis optica spectrum disorder (NMOSD). Despite initial improvement with steroids and plasma exchange, she experienced recurrent symptoms over 10 months referable to new and persistently enhancing lesions. At autopsy, neuropathology revealed a diffusely infiltrative primary CNS histiocytic sarcoma. This case represents a rare clinicoradiologic mimic of NMOSD, underscoring the importance of evaluation for infiltrative diseases in cases of atypical seronegative NMOSD.


Subject(s)
Central Nervous System Neoplasms , Histiocytic Sarcoma , Area Postrema , Diagnosis, Differential , Female , Histiocytic Sarcoma/diagnosis , Humans , Magnetic Resonance Imaging , Middle Aged , Neuromyelitis Optica/diagnostic imaging
5.
Nature ; 603(7903): 934-941, 2022 03.
Article in English | MEDLINE | ID: mdl-35130560

ABSTRACT

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.


Subject(s)
Astrocytoma , Brain Stem Neoplasms , Gangliosides , Glioma , Histones , Immunotherapy, Adoptive , Mutation , Receptors, Chimeric Antigen , Astrocytoma/genetics , Astrocytoma/immunology , Astrocytoma/pathology , Astrocytoma/therapy , Brain Stem Neoplasms/genetics , Brain Stem Neoplasms/immunology , Brain Stem Neoplasms/pathology , Brain Stem Neoplasms/therapy , Child , Gangliosides/immunology , Gene Expression Profiling , Glioma/genetics , Glioma/immunology , Glioma/pathology , Glioma/therapy , Histones/genetics , Humans , Immunotherapy, Adoptive/methods , Receptors, Chimeric Antigen/immunology , Spinal Cord Neoplasms/genetics , Spinal Cord Neoplasms/immunology , Spinal Cord Neoplasms/pathology , Spinal Cord Neoplasms/therapy
6.
Exp Neurol ; 287(Pt 3): 384-394, 2017 Jan.
Article in English | MEDLINE | ID: mdl-26874223

ABSTRACT

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.


Subject(s)
Axons/physiology , Nerve Regeneration/genetics , Stroke/pathology , Stroke/physiopathology , Animals , Axons/pathology , Humans , Neuronal Plasticity/physiology , Neurons/pathology , Recovery of Function/physiology
7.
Nat Neurosci ; 18(12): 1737-45, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26502261

ABSTRACT

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.


Subject(s)
Axons/metabolism , Growth Differentiation Factor 10/biosynthesis , Growth Differentiation Factor 10/genetics , Recovery of Function/physiology , Stroke/genetics , Stroke/metabolism , Aged , Aged, 80 and over , Animals , Animals, Newborn , Axons/pathology , Humans , Macaca mulatta , Mice , Mice, Inbred C57BL , Middle Aged , Retrospective Studies , Stroke/pathology
8.
Proc Natl Acad Sci U S A ; 107(45): 19573-8, 2010 Nov 09.
Article in English | MEDLINE | ID: mdl-20974939

ABSTRACT

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.


Subject(s)
Neurons/physiology , Synapses/pathology , Synaptic Transmission/genetics , Synucleins/genetics , Synucleins/physiology , Age Factors , Animals , Gene Deletion , Mice , Mice, Knockout , Nerve Tissue Proteins/analysis , Parkinson Disease/etiology , Phenotype , Synucleins/deficiency , alpha-Synuclein/deficiency , alpha-Synuclein/genetics , beta-Synuclein/deficiency , beta-Synuclein/genetics , gamma-Synuclein/deficiency , gamma-Synuclein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...