Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 103
Filtrer
1.
Blood Cancer J ; 14(1): 128, 2024 Aug 07.
Article de Anglais | MEDLINE | ID: mdl-39112453

RÉSUMÉ

Follicular lymphoma (FL) exhibits considerable variability in biological features and clinical trajectories across patients. To dissect the diversity of FL, we utilized a Bernoulli mixture model to identify genetic subtypes in 713 pre-treatment tumor tissue samples. Our analysis revealed the existence of five subtypes with unique genetic profiles that correlated with clinicopathological characteristics. The clusters were enriched in specific mutations as follows: CS (CREBBP and STAT6), TT (TNFAIP3 and TP53), GM (GNA13 and MEF2B), Q (quiescent, for low mutation burden), and AR (mutations of mTOR pathway-related genes). The subtype Q was enriched for patients with stage I disease and associated with a lower proliferative history than the other subtypes. The AR subtype was unique in its enrichment for IgM-expressing FL cases and was associated with advanced-stage and more than 4 nodal sites. The existence of subtypes was validated in an independent cohort of 418 samples from the GALLIUM trial. Notably, patients assigned to the TT subtype consistently experienced inferior progression-free survival when treated with immunochemotherapy. Our findings offer insight into core pathways distinctly linked with each FL cluster and are expected to be informative in the era of targeted therapies.


Sujet(s)
Lymphome folliculaire , Humains , Lymphome folliculaire/génétique , Lymphome folliculaire/anatomopathologie , Femelle , Mâle , Mutation , Adulte d'âge moyen , Sujet âgé , Marqueurs biologiques tumoraux/génétique , Pronostic
2.
Br J Haematol ; 2024 Aug 07.
Article de Anglais | MEDLINE | ID: mdl-39112220

RÉSUMÉ

Radiotherapy is routinely used for management of limited-stage follicular lymphoma (FL), yet half of patients ultimately relapse. We hypothesized that the presence of specific gene mutations may predict outcomes. We performed targeted sequencing of a 69-gene panel in 117 limited-stage FL patients treated with radiotherapy and identified recurrently mutated genes. CREBBP was most frequently mutated, and mutated CREBBP was associated with inferior progression-free survival, though not after false discovery rate adjustment. This association failed to validate in an independent cohort. We conclude that recurrent gene mutations do not predict outcomes in this setting. Alternative biomarkers may offer better prognostic insight.

3.
Acta Haematol ; 2024 Mar 13.
Article de Anglais | MEDLINE | ID: mdl-38479365

RÉSUMÉ

INTRODUCTION: Hodgkin Lymphoma (HL) is deficient in Major Histocompatibility Complex-class I, rendering it susceptible to anti-tumoral immunity by Natural Killer (NK)-cells. Despite the functional impairment of PD-1+ NK-cells in HL, the underlying mechanisms of NK-cell dysfunction remain unclear. METHODS: This study involved 14 HL patients and SNK10/KHYG-1 cell lines to assess NK-cell activation against cancer cells. Activation was measured through transcript (PCR) and protein expression (flow cytometry). Regulatory mechanisms associated with IRE1α activation were validated through knock-down and luciferase reporter assays. RESULTS: Our findings reveal a novel role for IRE1α-endonuclease in fine-tuning NK-cell effector functions by orchestrating the XBP1s/microRNA-34a-5p/PD-1 axis. When NK-cells encounter cancer cells, IRE1α-endonuclease activates the decay of microRNA-34a-5p, resulting in increased expression of XBP1s and PD-1. IRE1α-endonuclease activation enhances NK-cells function while promoting PD-1 expression. In turn, PD-1 is directly regulated by microRNA-34a-5p, which binds to the 3'UTR of PD-1 transcript to repress PD-1 protein on the NK-cell surface. Importantly, IRE1α-pathway activation is impaired in NK-cells from HL patients. CONCLUSION: The IRE1α-endonuclease emerges as a key player, simultaneously regulating the XBP1s/microRNA-34a-5p/PD-1 axis in NK-cells, a process disrupted in HL. Targeting the IRE1α-pathway holds promise as a therapeutic strategy to optimise NK-cell functions in Hodgkin Lymphoma treatments.

6.
Haematologica ; 109(7): 2131-2143, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38268493

RÉSUMÉ

T-cell-engaging bispecific antibody (T-BsAb, also known as BiTE) therapy has emerged as a powerful therapeutic modality against multiple myeloma. Given that T-BsAb therapy redirects endogenous T cells to eliminate tumor cells, reinvigorating dysfunctional T cells may be a potential approach to improve the efficacy of T-BsAb. While various immunostimulatory cytokines can potentiate effector T-cell functions, the optimal cytokine treatment for T-BsAb therapy is yet to be established, partly due to a concern of cytokine release syndrome driven by aberrant interferon (IFN)-γ production. Here, we functionally screen immunostimulatory cytokines to determine an ideal combination partner for T-BsAb therapy. This approach reveals interleukin (IL)-21 as a potential immunostimulatory cytokine with the ability to augment T-BsAb-mediated release of granzyme B and perforin, without increasing IFN-γ release. Transcriptome profiling and functional characterization strongly support that IL-21 selectively targets the cytotoxic granule exocytosis pathway, but not pro-inflammatory responses. Notably, IL-21 modulates multiple steps of cytotoxic effector functions including upregulation of co-activating CD226 receptor, increasing cytotoxic granules, and promoting cytotoxic granule delivery at the immunological synapse. Indeed, T-BsAb-mediated myeloma killing is cytotoxic granule-dependent, and IL-21 priming significantly augments cytotoxic activities. Furthermore, in vivo IL-21 treatment induces cytotoxic effector reprogramming in bone marrow T cells, showing synergistic anti-myeloma effects in combination with T-BsAb therapy. Together, harnessing the cytotoxic granule exocytosis pathway by IL-21 may be a potential approach to achieve better responses by T-BsAb therapy.


Sujet(s)
Anticorps bispécifiques , Exocytose , Myélome multiple , Anticorps bispécifiques/pharmacologie , Anticorps bispécifiques/usage thérapeutique , Humains , Souris , Animaux , Myélome multiple/immunologie , Myélome multiple/traitement médicamenteux , Myélome multiple/thérapie , Myélome multiple/anatomopathologie , Cytotoxicité immunologique , Interleukines/métabolisme , Lignée cellulaire tumorale , Cytokines/métabolisme , Lymphocytes T cytotoxiques/immunologie , Lymphocytes T cytotoxiques/effets des médicaments et des substances chimiques , Granzymes/métabolisme , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Lymphocytes T/effets des médicaments et des substances chimiques
8.
Haematologica ; 109(3): 787-798, 2024 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-37767564

RÉSUMÉ

T-cell-engaging bispecific antibodies (T-BsAb) have produced impressive clinical responses in patients with relapsed/refractory B-cell malignancies, although treatment failure remains a major clinical challenge. Growing evidence suggests that a complex interplay between immune cells and tumor cells is implicated in the mechanism of action and therefore, understanding immune regulatory mechanisms might provide a clue for how to improve the efficacy of T-BsAb therapy. Here, we investigated the functional impact of regulatory T (Treg) cells on anti-tumor immunity elicited by T-BsAb therapy. In a preclinical model of myeloma, the activation and expansion of Treg cells in the bone marrow were observed in response to anti-B-cell maturation antigen (BCMA) T-BsAb therapy. T-BsAb triggered the generation of induced Treg cells from human conventional CD4 cells after co-culture with tumor cells. Moreover, T-BsAb directly activated freshly isolated circulating Treg cells, leading to the production of interleukin-10 and inhibition of T-BsAb-mediated CD8 T-cell responses. The activation of Treg cells was also seen in bone marrow samples from myeloma patients after ex vivo treatment with T-BsAb, further supporting that T-BsAb have an impact on Treg homeostasis. Importantly, transient ablation of Treg cells in combination with T-BsAb therapy dramatically improved effector lymphocyte activities and disease control in the preclinical myeloma model, leading to prolonged survival. Together, this information suggests that therapy-induced activation of Treg cells critically regulates anti-tumor immunity elicited by T-BsAb therapy, with important implications for improving the efficacy of such treatment.


Sujet(s)
Anticorps bispécifiques , Myélome multiple , Humains , Lymphocytes T régulateurs , Anticorps bispécifiques/pharmacologie , Anticorps bispécifiques/usage thérapeutique , Myélome multiple/traitement médicamenteux , Lymphocytes T CD4+ , Lymphocytes T CD8+
9.
Br J Haematol ; 204(2): 415-433, 2024 02.
Article de Anglais | MEDLINE | ID: mdl-38155519

RÉSUMÉ

Epstein-Barr virus (EBV)-associated lymphomas cover a range of histological B- and T-cell non-Hodgkin and Hodgkin lymphoma subtypes. The role of EBV on B-cell malignant pathogenesis and its impact on the tumour microenvironment are intriguing but incompletely understood. Both the International Consensus Classification (ICC) and 5th Edition of the World Health Organization (WHO-HAEM5) proposals give prominence to the distinct clinical, prognostic, genetic and tumour microenvironmental features of EBV in lymphoproliferative disorders. There have been major advances in our biological understanding, in how to harness features of EBV and its host immune response for targeted therapy, and in using EBV as a method to monitor disease response. In this article, we showcase the latest developments and how they may be integrated to stimulate new and innovative approaches for further lines of investigation and therapy.


Sujet(s)
Infections à virus Epstein-Barr , Maladie de Hodgkin , Lymphome malin non hodgkinien , Lymphomes , Syndromes lymphoprolifératifs , Humains , Herpèsvirus humain de type 4/génétique , Maladie de Hodgkin/anatomopathologie , Microenvironnement tumoral
10.
Blood ; 142(17): 1448-1462, 2023 10 26.
Article de Anglais | MEDLINE | ID: mdl-37595278

RÉSUMÉ

Hematopoietic stem and progenitor cells (HSPCs) rely on a complex interplay among transcription factors (TFs) to regulate differentiation into mature blood cells. A heptad of TFs (FLI1, ERG, GATA2, RUNX1, TAL1, LYL1, LMO2) bind regulatory elements in bulk CD34+ HSPCs. However, whether specific heptad-TF combinations have distinct roles in regulating hematopoietic differentiation remains unknown. We mapped genome-wide chromatin contacts (HiC, H3K27ac, HiChIP), chromatin modifications (H3K4me3, H3K27ac, H3K27me3) and 10 TF binding profiles (heptad, PU.1, CTCF, STAG2) in HSPC subsets (stem/multipotent progenitors plus common myeloid, granulocyte macrophage, and megakaryocyte erythrocyte progenitors) and found TF occupancy and enhancer-promoter interactions varied significantly across cell types and were associated with cell-type-specific gene expression. Distinct regulatory elements were enriched with specific heptad-TF combinations, including stem-cell-specific elements with ERG, and myeloid- and erythroid-specific elements with combinations of FLI1, RUNX1, GATA2, TAL1, LYL1, and LMO2. Furthermore, heptad-occupied regions in HSPCs were subsequently bound by lineage-defining TFs, including PU.1 and GATA1, suggesting that heptad factors may prime regulatory elements for use in mature cell types. We also found that enhancers with cell-type-specific heptad occupancy shared a common grammar with respect to TF binding motifs, suggesting that combinatorial binding of TF complexes was at least partially regulated by features encoded in DNA sequence motifs. Taken together, this study comprehensively characterizes the gene regulatory landscape in rare subpopulations of human HSPCs. The accompanying data sets should serve as a valuable resource for understanding adult hematopoiesis and a framework for analyzing aberrant regulatory networks in leukemic cells.


Sujet(s)
Sous-unité alpha 2 du facteur CBF , Cellules souches hématopoïétiques , Humains , Sous-unité alpha 2 du facteur CBF/génétique , Sous-unité alpha 2 du facteur CBF/métabolisme , Cellules souches hématopoïétiques/métabolisme , Régulation de l'expression des gènes , Hématopoïèse/génétique , Chromatine/métabolisme
12.
Intern Med J ; 53(7): 1105-1109, 2023 07.
Article de Anglais | MEDLINE | ID: mdl-37032307

RÉSUMÉ

Diffuse large B-cell lymphoma (DLBCL) is the most frequent aggressive lymphoma seen in clinical practice. Despite huge strides in understanding its biology, front-line therapy has remained unchanged for decades. Roughly one-third of patients have primary refractory or relapse following the end of conventional first-line therapy. The outcome of patients with primary refractory disease and those with early relapse (defined as relapse less than 1 year from the end of therapy) is markedly inferior to those with later relapse and is exemplified by dismal overall survival. In this article, the authors term patients with features that identify them as being at particularly high-risk for either primary refractory disease or early relapse, as 'ultra-high-risk'. As new treatment options become established (e.g. bispecific T-cell engagers, chimeric antigen receptor 'CAR' T-cells and antibody-drug conjugates), it is likely that there will be a push to incorporate some of these agents into the first-line setting for patients identified as ultra-high-risk. In this review, the authors outline advances in positron emission tomography, widely available laboratory assays and clinical prognosticators, which can detect a high proportion of patients with ultra-high-risk disease. Since these approaches are pragmatic and able to be adopted widely, they could be incorporated into routine clinical practice.


Sujet(s)
Lymphome B diffus à grandes cellules , Récidive tumorale locale , Humains , Lymphome B diffus à grandes cellules/diagnostic , Lymphome B diffus à grandes cellules/thérapie
13.
Leuk Lymphoma ; 64(5): 1026-1034, 2023 05.
Article de Anglais | MEDLINE | ID: mdl-36960939

RÉSUMÉ

The objective of this multicenter retrospective study was to examine the incidence, patient characteristics, pathology, and outcomes associated with Epstein-Barr virus (EBV)-related CNS lymphoma (CNSL) in older patients. Among 309 CNSL patients aged ≥60, 11.7% had EBV + tumors of which 72.2% were solid organ transplant (SOT)-related post-transplant lymphoproliferative disorders (PTLD). Younger age, SOT or autoimmune disease, and immunosuppressive treatment correlated highly with EBV-positivity. EBV + tumors were associated with absent C-MYC and BCL6 expression. EBV + PTLD was more likely to be associated with the absence of CD5 expression. EBV + non-PTLD had better median OS (not reached) compared to EBV + PTLD (10.8 months) and EBV-negative patients (43 months). Multivariable Cox regression analysis showed that age, performance status, and PTLD were negative predictors of OS. EBV status and immunosuppressive treatment were not correlated with OS. Our findings merit further investigation of EBV + PCNSL tumors and EBV-directed therapies.


Sujet(s)
Infections à virus Epstein-Barr , Lymphomes , Syndromes lymphoprolifératifs , Humains , Sujet âgé , Infections à virus Epstein-Barr/complications , Infections à virus Epstein-Barr/épidémiologie , Herpèsvirus humain de type 4 , Études rétrospectives , Incidence , Lymphomes/étiologie , Syndromes lymphoprolifératifs/étiologie , Immunosuppresseurs
16.
Leukemia ; 37(2): 379-387, 2023 02.
Article de Anglais | MEDLINE | ID: mdl-36539557

RÉSUMÉ

Redirection of tumor-associated macrophages to eliminate tumor cells holds great promise for overcoming therapeutic resistance to rituximab and other antibody drugs. Here, we determined the expression of ectonucleotidases CD39 and CD73 in diffuse large B-cell lymphoma (DLBCL), and examined the impact of extracellular ATP (eATP) metabolism on macrophage-mediated anti-lymphoma immunity. Immunostaining of tissue microarray samples showed that CD39 (the ecto-enzyme for eATP hydrolysis) was highly expressed in tumors with the non-germinal center B-cell-like (non-GCB) subtype, and to a lesser extent tumors with the GCB subtype. By contrast, the expression of CD73 (the ecto-enzyme for adenosine generation) was undetectable in tumor cells. Pharmacological blockade of CD39 prevented eATP degradation and enhanced engulfment of antibody-coated lymphoma cells by macrophages in a P2X7 receptor-dependent manner, indicating that eATP fueled antibody-dependent cellular phagocytosis (ADCP) activity. Importantly, inhibition of CD39 augmented in vivo anti-lymphoma effects by therapeutic antibodies including rituximab and daratumumab. Furthermore, the addition of a CD39 inhibitor to anti-CD20 and anti-CD47 combination therapy significantly improved survival in a disseminated model of aggressive B-cell lymphoma, supporting the benefit of dual targeting CD39-mediated eATP hydrolysis and CD47-mediated "don't eat me" signal. Together, preventing eATP degradation may be a potential approach to unleash macrophage-mediated anti-lymphoma immunity.


Sujet(s)
Lymphome B diffus à grandes cellules , Macrophages , Humains , Rituximab/pharmacologie , Rituximab/usage thérapeutique , Adénosine/métabolisme , Lymphome B diffus à grandes cellules/anatomopathologie , Phagocytose
17.
Acta Haematol ; 146(2): 166-171, 2023.
Article de Anglais | MEDLINE | ID: mdl-36273464

RÉSUMÉ

Here, we present a novel case of a patient with chronic lymphocytic leukemia (CLL) who received CTLA-4 and then PD-1 immune-checkpoint blockade (ICB) as treatment for concomitant metastatic melanoma. Whereas the metastatic melanoma was responsive to ICB, the CLL rapidly progressed (but responded to ICB cessation and ibrutinib). There were no new genetic mutational drivers to explain the altered clinical course. PD-1/PD-L1/PD-L2 and CTLA-4/CD80/CD86 expression was not increased in CLL B cells, CD8+ or CD4+ T-cell subsets, or monocytes. The patient's CLL B cells demonstrated strikingly prolonged in vitro survival during PD-1 blockade, which was not observed in samples taken before or after ICB, or with other patients. To our knowledge, a discordant clinical course to ICB coupled with these biological features has not been reported in a patient with dual malignancies.


Sujet(s)
Antinéoplasiques , Inhibiteurs de points de contrôle immunitaires , Leucémie chronique lymphocytaire à cellules B , Mélanome , Récepteur-1 de mort cellulaire programmée , Tumeurs cutanées , Humains , Antigène CTLA-4/antagonistes et inhibiteurs , Antigène CTLA-4/immunologie , Évolution de la maladie , Leucémie chronique lymphocytaire à cellules B/traitement médicamenteux , Leucémie chronique lymphocytaire à cellules B/immunologie , Leucémie chronique lymphocytaire à cellules B/anatomopathologie , Mélanome/traitement médicamenteux , Mélanome/étiologie , Mélanome/anatomopathologie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/immunologie , Tumeurs cutanées/traitement médicamenteux , Tumeurs cutanées/étiologie , Tumeurs cutanées/anatomopathologie , Antigène CD274 , Inhibiteurs de points de contrôle immunitaires/immunologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Antinéoplasiques/immunologie , Antinéoplasiques/usage thérapeutique
19.
Cancer Immunol Res ; 10(12): 1422, 2022 12 02.
Article de Anglais | MEDLINE | ID: mdl-36327248

RÉSUMÉ

Chimeric antigen receptor (CAR) T-cell therapy has yielded remarkable and durable responses for some patients with relapsed and refractory blood cancers. However, life-threatening toxicities such as immune effector cell-associated neurotoxicity syndrome (ICANS) remain a challenge for broad delivery of such therapies. In this issue, Tang and colleagues demonstrate an association between hypophosphatemia and CAR T cell-induced ICANS. Prospective studies are required to establish if phosphate monitoring is an early predictor for ICANS occurrence and if maintenance of phosphate levels has a role as a preventative strategy. See related article by Tang et al., p. 1433 (4).


Sujet(s)
Immunothérapie adoptive , Phosphates , Humains , Immunothérapie adoptive/effets indésirables
20.
Blood Rev ; 56: 100969, 2022 11.
Article de Anglais | MEDLINE | ID: mdl-35570070

RÉSUMÉ

Broad activation of host T-cell immunity by immune checkpoint blockade, has revolutionized the treatment of some but not all B-cell lymphoproliferative disorders (LPDs). The challenge for next generation immunotherapeutics, is to successfully induce anti-tumor specific T-cell immunity across a range of B-LPDs, without provoking immune-related adverse events. An emerging strategy is to target neoantigens. Neoantigens are immunogenic peptides, unique to malignant cells, that are presented to T-cells via human leukocyte antigens. Neoantigens most commonly arise from non-synonymous mutations but can also be derived from tumor specific alterations along the protein biosynthesis pathway. B-cell LPDs uniquely express a clonal B-cell receptor (BCR) idiotype, consisting of immunoglobulin genes that undergo recombination and somatic hypermutation. Notably, the BCR idiotype can also give rise to 'immunoglobulin neoantigens'. Here, we provide an overview of current strategies to identify and validate immunoglobulin and non-immunoglobulin neoantigens as well as summarizing studies investigating neoantigens within B-cell LPDs.


Sujet(s)
Syndromes lymphoprolifératifs , Tumeurs , Humains , Antigènes néoplasiques , Inhibiteurs de points de contrôle immunitaires , Immunothérapie , Tumeurs/thérapie , Peptides , Facteurs immunologiques , Antigènes HLA , Récepteurs pour l'antigène des lymphocytes B/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE