Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Neurology ; 102(12): e209527, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38830184

ABSTRACT

OBJECTIVES: Despite a high response rate at the first evaluation during induction chemotherapy, the risk of early relapse remains high and unpredictable in primary CNS lymphomas (PCSNLs). We aimed to assess the prognostic value of early IL-10 levels in CSF (e-IL-10) after 2 months of induction chemotherapy. METHODS: We retrospectively selected from the LOC (Lymphomes Oculo-Cérébraux) network database patients with PCSNLs who had complete or partial response at the 2-month evaluation of a high-dose methotrexate-based first-line chemotherapy for whom e-IL-10 was available. RESULTS: Thirty patients (median age: 62 years, brain involvement in 30/30, CSF involvement in 10/30, median baseline CSF IL-10: 27.5 pg/mL) met the selection criteria. e-IL-10 was undetectable in 22 patients and detectable in 8 patients. At the end of induction treatment, 7 of 8 and 4 of 22 of the patients with detectable and undetectable e-IL-10 had experienced progressive disease, respectively (p = 0.001, OR: 26.8, 95% CI 2-1,478). The median progression-free survival times were 5.8 months (95% CI 2.8-8.8) and 28.7 months (95% CI 13.4-43.9) in the groups with detectable and undetectable e-IL-10, respectively (p < 0.001). DISCUSSION: Our results suggest that despite an objective response, the persistence of detectable e-IL-10 is associated with a high risk of early relapse in PCNSL. A closer follow-up of such patients is warranted.


Subject(s)
Central Nervous System Neoplasms , Induction Chemotherapy , Interleukin-10 , Humans , Middle Aged , Female , Male , Interleukin-10/cerebrospinal fluid , Central Nervous System Neoplasms/cerebrospinal fluid , Central Nervous System Neoplasms/drug therapy , Aged , Retrospective Studies , Prognosis , Adult , Lymphoma/cerebrospinal fluid , Lymphoma/drug therapy , Methotrexate/therapeutic use , Methotrexate/administration & dosage
2.
Cancer Cell ; 42(3): 358-377.e8, 2024 Mar 11.
Article in English | MEDLINE | ID: mdl-38215747

ABSTRACT

The evolutionary trajectory of glioblastoma (GBM) is a multifaceted biological process that extends beyond genetic alterations alone. Here, we perform an integrative proteogenomic analysis of 123 longitudinal glioblastoma pairs and identify a highly proliferative cellular state at diagnosis and replacement by activation of neuronal transition and synaptogenic pathways in recurrent tumors. Proteomic and phosphoproteomic analyses reveal that the molecular transition to neuronal state at recurrence is marked by post-translational activation of the wingless-related integration site (WNT)/ planar cell polarity (PCP) signaling pathway and BRAF protein kinase. Consistently, multi-omic analysis of patient-derived xenograft (PDX) models mirror similar patterns of evolutionary trajectory. Inhibition of B-raf proto-oncogene (BRAF) kinase impairs both neuronal transition and migration capability of recurrent tumor cells, phenotypic hallmarks of post-therapy progression. Combinatorial treatment of temozolomide (TMZ) with BRAF inhibitor, vemurafenib, significantly extends the survival of PDX models. This study provides comprehensive insights into the biological mechanisms of glioblastoma evolution and treatment resistance, highlighting promising therapeutic strategies for clinical intervention.


Subject(s)
Brain Neoplasms , Glioblastoma , Proteogenomics , Animals , Humans , Glioblastoma/genetics , Proto-Oncogene Proteins B-raf , Proteomics , Cell Line, Tumor , Neoplasm Recurrence, Local , Disease Models, Animal , Brain Neoplasms/genetics , Drug Resistance, Neoplasm , Xenograft Model Antitumor Assays
4.
Biomedicines ; 11(2)2023 Feb 09.
Article in English | MEDLINE | ID: mdl-36831042

ABSTRACT

INTRODUCTION: Autoimmune encephalitis (AE) diagnosis and follow-up remain challenging. Brain 18F-fluoro-deoxy-glucose positron emission tomography (FDG PET) has shown promising results in AE. Our aim was to investigate FDG PET alterations in AE, according to antibody subtype. METHODS: We retrospectively included patients with available FDG PET and seropositive AE diagnosed in our center between 2015 and 2020. Brain PET Z-score maps (relative to age matched controls) were analyzed, considering metabolic changes significant if |Z-score| ≥ 2. RESULTS: Forty-six patients were included (49.4 yrs [18; 81]): 13 with GAD autoantibodies, 11 with anti-LGI1, 9 with NMDAR, 5 with CASPR2, and 8 with other antibodies. Brain PET was abnormal in 98% of patients versus 53% for MRI. The most frequent abnormalities were medial temporal lobe (MTL) and/or striatum hypermetabolism (52% and 43% respectively), cortical hypometabolism (78%), and cerebellum abnormalities (70%). LGI1 AE tended to have more frequent MTL hypermetabolism. NMDAR AE was prone to widespread cortical hypometabolism. Fewer abnormalities were observed in GAD AE. Striatum hypermetabolism was more frequent in patients treated for less than 1 month (p = 0.014), suggesting a relation to disease activity. CONCLUSION: FDG PET could serve as an imaging biomarker for early diagnosis and follow-up in AE.

5.
Adv Exp Med Biol ; 1394: 85-101, 2023.
Article in English | MEDLINE | ID: mdl-36587383

ABSTRACT

A better understanding of cellular and molecular biology of primary central nervous system (CNS) tumors is a critical step toward the design of innovative treatments. In addition to improving knowledge, identification of the cell of origin in tumors allows for sharp and efficient targeting of specific tumor cells promoting and driving oncogenic processes. The World Health Organization identifies approximately 150 primary brain tumor subtypes with various ontogeny and clinical outcomes. Identification of the cell of origin of each tumor type with its lineage and differentiation level is challenging. In the current chapter, we report the suspected cell of origin of various CNS primary tumors including gliomas, glioneuronal tumors, medulloblastoma, meningioma, atypical teratoid rhabdoid tumor, germinomas, and lymphoma. Most of them have been pinpointed through transgenic mouse models and analysis of molecular signatures of tumors. Identification of the cell or cells of origin in primary brain tumors will undoubtedly open new therapeutic avenues, including the reactivation of differentiation programs for therapeutic perspectives.


Subject(s)
Brain Neoplasms , Central Nervous System Neoplasms , Cerebellar Neoplasms , Glioma , Medulloblastoma , Rhabdoid Tumor , Spinal Cord Neoplasms , Animals , Mice , Medulloblastoma/genetics , Medulloblastoma/pathology , Glioma/pathology , Brain/pathology , Central Nervous System Neoplasms/pathology , Spinal Cord Neoplasms/pathology , Rhabdoid Tumor/genetics , Rhabdoid Tumor/pathology , Cerebellar Neoplasms/pathology , Brain Neoplasms/genetics , Brain Neoplasms/pathology
7.
Curr Opin Neurol ; 35(6): 794-802, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36226710

ABSTRACT

PURPOSE OF REVIEW: Adult-type diffuse gliomas are highly heterogeneous tumors. Bulk transcriptome analyses suggested that the composition of the tumor microenvironment (TME) corresponds to genetic and clinical features. In this review, we highlight novel findings on the intratumoral heterogeneity of IDH-wildtype and IDH-mutant gliomas characterized at single-cell resolution, and emphasize the mechanisms shaping the immune TME and therapeutic implications. RECENT FINDINGS: Emergent evidence indicates that in addition to genetic drivers, epigenetic mechanisms and microenvironmental factors influence the glioma subtypes. Interactions between glioma and immune cells contribute to immune evasion, particularly in aggressive tumors. Spatial and temporal heterogeneity of malignant and immune cell subpopulations is high in recurrent gliomas. IDH-wildtype and IDH-mutant tumors display distinctive changes in their myeloid and lymphoid compartments, and D-2HG produced by IDH-mutant cells impacts the immune TME. SUMMARY: The comprehensive dissection of the intratumoral ecosystem of human gliomas using single-cell and spatial transcriptomic approaches advances our understanding of the mechanisms underlying the immunosuppressed state of the TME, supports the prognostic value of tumor-associated macrophages and microglial cells, and sheds light on novel therapeutic options.


Subject(s)
Brain Neoplasms , Glioma , Adult , Humans , Isocitrate Dehydrogenase/genetics , Tumor Microenvironment/genetics , Ecosystem , Mutation , Neoplasm Recurrence, Local , Glioma/genetics , Glioma/pathology
8.
Br J Haematol ; 199(4): 507-519, 2022 11.
Article in English | MEDLINE | ID: mdl-35945164

ABSTRACT

To optimise management of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection identifying high-risk patients and maintaining treatment dose intensity is an important issue in patients with aggressive lymphomas. In the present study, we report on the presentation, management, and outcome of an international series of 91 patients with primary central nervous system lymphoma and SARS-CoV-2 infection. SARS-CoV-2 was diagnosed before/during first-line treatment in 64 patients, during follow-up in 21, and during salvage therapy in six. Among the 64 patients infected before/during first-line chemotherapy, 38 (59%) developed pneumonia and 26 (41%) did not clear the virus. Prolonged exposure to steroids before viral infection and/or treatment with high-dose cytarabine favoured pneumonia development and virus persistence and were associated with poorer survival; 81% of patients who did not clear virus died early from coronavirus disease 2019 (COVID-19). Vaccination was associated with lower pneumonia incidence and in-hospital mortality. Chemotherapy was initiated/resumed in 43 (67%) patients, more commonly among patients who did not develop pneumonia, cleared the virus, or did not receive steroids during infection. Chemotherapy resumption in patients with viral persistence should be indicated cautiously as it was associated with a poorer survival (6-month, 70% and 87%, p = 0.07). None of the 21 patients infected during follow-up died from COVID-19, requiring similar measures as infected subjects in the general population.


Subject(s)
COVID-19 , Lymphoma , Humans , SARS-CoV-2 , Central Nervous System , Lymphoma/drug therapy
10.
J Neurol ; 268(9): 3072-3080, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33387015

ABSTRACT

BACKGROUND: Cancer patients may be at higher risk for severe coronavirus infectious disease-19 (COVID-19); however, the outcome of Primary Central Nervous System Lymphoma (PCNSL) patients with SARS-CoV-2 infection has not been described yet. METHODS: We conducted a retrospective study within the Lymphomes Oculo-Cérébraux national network (LOC) to assess the clinical characteristics and outcome of SARS-CoV-2 infection in PCNSL patients (positive real-time polymerase chain reaction of nasopharyngeal swab or evocative lung computed tomography scan). We compared clinical characteristics between patients with severe (death and/or intensive care unit admission) and mild disease. RESULTS: Between March and May 2020, 13 PCNSL patients were diagnosed with SARS-CoV-2 infection, 11 (85%) of whom were undergoing chemotherapy at the time of infection. The mortality rate was 23% (3/13), and two additional patients (15%) required mechanical ventilation. Two patients (15%) had no COVID-19 symptoms. History of diabetes mellitus was more common in severe patients (3/5 vs 0/8, p = 0.03). Two patients recovered from COVID-19 after mechanical ventilation during more than two weeks and resumed chemotherapy. In all, chemotherapy was resumed after COVID-19 recovery in nine patients (69%) after a median delay of 16 days (range 3-32), none of whom developed unusual chemotherapy complication nor SARS-Cov2 reactivation. CONCLUSION: This preliminary analysis suggests that, while being at higher risk be for severe illness, PCNSL patients with COVID-19 might be treated maximally especially if they achieved oncological response at the time of SARS-CoV-2 infection. Chemotherapy might be resumed without prolonged delay in PCNSL patients with COVID-19.


Subject(s)
COVID-19 , Lymphoma , Central Nervous System , Humans , Lymphoma/complications , Lymphoma/epidemiology , Lymphoma/therapy , RNA, Viral , Retrospective Studies , SARS-CoV-2
11.
Neuro Oncol ; 23(6): 955-966, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33173941

ABSTRACT

BACKGROUND: Isocitrate dehydrogenase (IDH) wildtype (wt) grade II gliomas are a rare and heterogeneous entity. Survival and prognostic factors are poorly defined. METHODS: We searched retrospectively all patients diagnosed with diffuse World Health Organization (WHO) grades II and III gliomas at our center (1989-2020). RESULTS: Out of 517 grade II gliomas, 47 were "diffuse astrocytomas, IDHwt." Tumors frequently had fronto-temporo-insular location (28/47, 60%) and infiltrative behavior. We found telomerase reverse transcriptase (TERT) promoter mutations (23/45, 51%), whole chromosome 7 gains (10/37, 27%), whole chromosome 10 losses (10/41, 24%), and EGFR amplifications (4/43, 9%), but no TP53 mutations (0/22, 0%). Median overall survival (OS) was 59 months (vs 19 mo for IDHwt grade III gliomas) (P < 0.0001). Twenty-nine patients (29/43, 67%) met the definition of molecular glioblastoma according to cIMPACT-NOW update 3. Median OS in this subset was 42 months, which was shorter compared with patients with IDHwt grade II gliomas not meeting this definition (median OS: 57 mo), but substantially longer compared with IDHwt grade III gliomas meeting the definition for molecular glioblastoma (median OS: 17 mo, P < 0.0001). Most patients with IDHwt grade II gliomas met cIMPACT criteria because of isolated TERT promoter mutations (16/26, 62%), which were not predictive of poor outcome (median OS: 88 mo). Actionable targets, including 5 gene fusions involving FGFR3, were found in 7 patients (24%). CONCLUSIONS: Our findings highlight the importance of histological grading and molecular profiling for the prognostic stratification of IDHwt gliomas and suggest some caution when assimilating IDHwt grade II gliomas to molecular glioblastomas, especially those with isolated TERT promoter mutation.


Subject(s)
Brain Neoplasms , Glioma , Brain Neoplasms/genetics , Glioma/genetics , Humans , Isocitrate Dehydrogenase/genetics , Mutation , Neoplasm Grading , Prognosis , Retrospective Studies
12.
Clin Cancer Res ; 18(19): 5203-11, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22837180

ABSTRACT

PURPOSE: Our objective was to identify the genetic changes involved in primary central nervous system lymphoma (PCNSL) oncogenesis and evaluate their clinical relevance. EXPERIMENTAL DESIGN: We investigated a series of 29 newly diagnosed, HIV-negative, PCNSL patients using high-resolution single-nucleotide polymorphism (SNP) arrays (n = 29) and whole-exome sequencing (n = 4) approaches. Recurrent homozygous deletions and somatic gene mutations found were validated by quantitative real-time PCR and Sanger sequencing, respectively. Molecular results were correlated with prognosis. RESULTS: All PCNSLs were diffuse large B-cell lymphomas, and the patients received chemotherapy without radiotherapy as initial treatment. The SNP analysis revealed recurrent large and focal chromosome imbalances that target candidate genes in PCNSL oncogenesis. The most frequent genomic abnormalities were (i) 6p21.32 loss (HLA locus), (ii) 6q loss, (iii) CDKN2A homozygous deletions, (iv) 12q12-q22, and (v) chromosome 7q21 and 7q31 gains. Homozygous deletions of PRMD1, TOX, and DOCK5 and the amplification of HDAC9 were also detected. Sequencing of matched tumor and blood DNA samples identified novel somatic mutations in MYD88 and TBL1XR1 in 38% and 14% of the cases, respectively. The correlation of genetic abnormalities with clinical outcomes using multivariate analysis showed that 6q22 loss (P = 0.006 and P = 0.01) and CDKN2A homozygous deletion (P = 0.02 and P = 0.01) were significantly associated with shorter progression-free survival and overall survival. CONCLUSIONS: Our study provides new insights into the molecular tumorigenesis of PCNSL and identifies novel genetic alterations in this disease, especially MYD88 and TBL1XR1 mutations activating the NF-κB signaling pathway, which may be promising targets for future therapeutic strategies.


Subject(s)
Central Nervous System Neoplasms , Chromosomal Instability , Lymphoma, Large B-Cell, Diffuse , Myeloid Differentiation Factor 88 , Nuclear Proteins , Receptors, Cytoplasmic and Nuclear , Repressor Proteins , Adult , Aged , Aged, 80 and over , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/metabolism , Disease-Free Survival , Female , Humans , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Male , Middle Aged , Mutation , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Polymorphism, Single Nucleotide , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...