Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.681
Filtrer
1.
Mol Imaging ; 23: 15353508241257924, 2024.
Article de Anglais | MEDLINE | ID: mdl-38952399

RÉSUMÉ

Chimeric antigen receptor (CAR)-T cell-based immunotherapy has emerged as a path-breaking strategy for certain hematological malignancies. Assessment of the response to CAR-T therapy using quantitative imaging techniques such as positron emission tomography/computed tomography (PET/CT) has been broadly investigated. However, the definitive role of PET/CT in CAR-T therapy remains to be established. [18F]FDG PET/CT has demonstrated high sensitivity and specificity for differentiating patients with a partial and complete response after CAR-T therapy in lymphoma. The early therapeutic response and immune-related adverse effects such as cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome can also be detected on [18F]FDG PET images. In otherwise asymptomatic lymphoma patients with partial response following CAR-T therapy, the only positive findings could be abnormal PET/CT results. In multiple myeloma, a negative [18F]FDG PET/CT after receiving B-cell maturation antigen-directed CAR-T therapy has been associated with a favorable prognosis. In leukemia, [18F]FDG PET/CT can detect extramedullary metastases and treatment responses after therapy. Hence, PET/CT is a valuable imaging tool for patients undergoing CAR-T therapy for pretreatment evaluation, monitoring treatment response, assessing safety, and guiding therapeutic strategies. Developing guidelines with standardized cutoff values for various PET parameters and tumor cell-specific tracers may improve the efficacy and safety of CAR-T therapy.


Sujet(s)
Tumeurs hématologiques , Tomographie par émission de positons couplée à la tomodensitométrie , Humains , Tomographie par émission de positons couplée à la tomodensitométrie/méthodes , Tumeurs hématologiques/thérapie , Tumeurs hématologiques/imagerie diagnostique , Tumeurs hématologiques/immunologie , Immunothérapie adoptive/méthodes , Immunothérapie/méthodes , Récepteurs chimériques pour l'antigène/usage thérapeutique , Fluorodésoxyglucose F18
3.
Front Immunol ; 15: 1383894, 2024.
Article de Anglais | MEDLINE | ID: mdl-38962014

RÉSUMÉ

Chimeric antigen receptor (CAR) T cell therapy has effectively complemented the treatment of advanced relapsed and refractory hematological cancers. The remarkable achievements of CD19- and BCMA-CAR T therapies have raised high expectations within the fields of hematology and oncology. These groundbreaking successes are propelling a collective aspiration to extend the reach of CAR therapies beyond B-lineage malignancies. Advanced CAR technologies have created a momentum to surmount the limitations of conventional CAR concepts. Most importantly, innovations that enable combinatorial targeting to address target antigen heterogeneity, using versatile adapter CAR concepts in conjunction with recent transformative next-generation CAR design, offer the promise to overcome both the bottleneck associated with CAR manufacturing and patient-individualized treatment regimens. In this comprehensive review, we delineate the fundamental prerequisites, navigate through pivotal challenges, and elucidate strategic approaches, all aimed at paving the way for the future establishment of multitargeted immunotherapies using universal CAR technologies.


Sujet(s)
Immunothérapie adoptive , Récepteurs chimériques pour l'antigène , Humains , Immunothérapie adoptive/méthodes , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/génétique , Animaux , Lymphocytes T/immunologie , Antigènes CD19/immunologie , Tumeurs hématologiques/thérapie , Tumeurs hématologiques/immunologie , Récepteurs aux antigènes des cellules T/immunologie , Récepteurs aux antigènes des cellules T/génétique , Tumeurs/thérapie , Tumeurs/immunologie
4.
Expert Opin Biol Ther ; 24(6): 425-432, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38943466

RÉSUMÉ

INTRODUCTION: CAR T cells have generated great excitement due to their remarkable clinical response rates in selected hematologic malignancies. However, these engineered immune cells are living drugs which are hard to control after administration. AREAS COVERED: We discuss small molecule-regulated switch systems which can potentially be used to control CAR T cell function within the patient, as well as the most important obstacles in the CAR T cell field, which might be overcome with those switch systems. EXPERT OPINION: There is an urgent need to develop advanced switch systems. Once available, we expect that they will open up new avenues for future CAR T cell generations.


Sujet(s)
Immunothérapie adoptive , Récepteurs chimériques pour l'antigène , Humains , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/métabolisme , Lymphocytes T/immunologie , Lymphocytes T/effets des médicaments et des substances chimiques , Animaux , Tumeurs hématologiques/immunologie , Tumeurs hématologiques/thérapie
5.
J Hematol Oncol ; 17(1): 49, 2024 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-38915099

RÉSUMÉ

Significant advances have been made in chimeric antigen receptor T (CAR-T)-cell therapy for the treatment of recurrent or refractory B-cell hematologic malignancies. However, CAR-T-cell therapy has not yet achieved comparable success in the management of aggressive T-cell malignancies. This article reviews the challenges of CAR-T-cell therapy in treating T-cell malignancies and summarizes the progress of preclinical and clinical studies in this area. We present an analysis of clinical trials of CAR-T-cell therapies for the treatment of T-cell malignancies grouped by target antigen classification. Moreover, this review focuses on the major challenges encountered by CAR-T-cell therapies, including the nonspecific killing due to T-cell target antigen sharing and contamination with cell products during preparation. This review discusses strategies to overcome these challenges, presenting novel therapeutic approaches that could enhance the efficacy and applicability of CAR-T-cell therapy in the treatment of T-cell malignancies. These ideas and strategies provide important information for future studies to promote the further development and application of CAR-T-cell therapy in this field.


Sujet(s)
Immunothérapie adoptive , Récepteurs chimériques pour l'antigène , Lymphocytes T , Humains , Immunothérapie adoptive/méthodes , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/usage thérapeutique , Lymphocytes T/immunologie , Lymphocytes T/transplantation , Tumeurs hématologiques/thérapie , Tumeurs hématologiques/immunologie , Animaux , Récepteurs aux antigènes des cellules T/immunologie , Récepteurs aux antigènes des cellules T/usage thérapeutique
6.
Int J Hematol ; 120(1): 3-5, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38861242

RÉSUMÉ

The introduction of immunotherapies has led to remarkable progress in the treatment of hematological malignancies, including B-cell malignancies such as B-cell lymphoma and multiple myeloma (MM). Although conventional therapeutic antibodies are effective as immunotherapy for newly diagnosed and relapsed/refractory B-cell lymphoma and MM, some cases are resistant. Chimeric antigen receptor (CAR) T-cell therapies targeting B-cell lymphoma and MM have progressed through several generations, and have improved treatment strategies for relapsed/refractory disease. In addition to conventional therapeutic antibodies, bispecific antibodies targeting both tumor cells and T cells have been developed for MM. Both CAR T-cell therapies and bispecific antibodies are effective for heavily treated patients with relapsed/refractory disease. However, most patients treated with these therapies relapse, and serious adverse events like cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) are problematic. This Progress in Hematology, "Novel treatment strategies for hematological malignancies in the immunotherapy era," focuses on such limitations and the future outlook for CAR T-cell therapies and bispecific antibodies for B-cell malignancies. The role of NK cells in anti-tumor immunity for AML and various therapeutic strategies for NK-cell therapy in AML is also discussed.


Sujet(s)
Anticorps bispécifiques , Tumeurs hématologiques , Immunothérapie adoptive , Humains , Anticorps bispécifiques/usage thérapeutique , Tumeurs hématologiques/thérapie , Tumeurs hématologiques/immunologie , Immunothérapie adoptive/effets indésirables , Immunothérapie adoptive/méthodes , Immunothérapie/méthodes , Cellules tueuses naturelles/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Lymphome B/thérapie , Lymphome B/immunologie , Myélome multiple/thérapie , Myélome multiple/immunologie , Syndrome de libération de cytokines/étiologie , Syndrome de libération de cytokines/thérapie
7.
Front Immunol ; 15: 1374390, 2024.
Article de Anglais | MEDLINE | ID: mdl-38868768

RÉSUMÉ

Immunotherapy for hematological malignancies is a rapidly advancing field that has gained momentum in recent years, primarily encompassing chimeric antigen receptor T-cell (CAR-T) therapies, immune checkpoint inhibitors, and other modalities. However, its clinical efficacy remains limited, and drug resistance poses a significant challenge. Therefore, novel immunotherapeutic targets and agents need to be identified. Recently, N6-methyladenosine (m6A), the most prevalent RNA epitope modification, has emerged as a pivotal factor in various malignancies. Reportedly, m6A mutations influence the immunological microenvironment of hematological malignancies, leading to immune evasion and compromising the anti-tumor immune response in hematological malignancies. In this review, we comprehensively summarize the roles of the currently identified m6A modifications in various hematological malignancies, with a particular focus on their impact on the immune microenvironment. Additionally, we provide an overview of the research progress made in developing m6A-targeted drugs for hematological tumor therapy, to offer novel clinical insights.


Sujet(s)
Adénosine , Tumeurs hématologiques , Microenvironnement tumoral , Humains , Microenvironnement tumoral/immunologie , Tumeurs hématologiques/immunologie , Tumeurs hématologiques/thérapie , Adénosine/analogues et dérivés , Adénosine/immunologie , Adénosine/métabolisme , Animaux , Immunothérapie/méthodes
9.
Cancer Control ; 31: 10732748241263713, 2024.
Article de Anglais | MEDLINE | ID: mdl-38910268

RÉSUMÉ

Chimeric antigen receptor T cell therapy is used to treat hematological malignancies which are refractory to standard therapy. It is a form of immunotherapy in which a patient's T cells are programmed to act against tumor cells. We discuss the process of manufacturing CAR-T cells, the common side effects of therapy, and the recent emerging risk of T-cell malignancies after treatment.


Sujet(s)
Immunothérapie adoptive , Récepteurs chimériques pour l'antigène , Humains , Immunothérapie adoptive/méthodes , Immunothérapie adoptive/effets indésirables , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/usage thérapeutique , Lymphocytes T/immunologie , Tumeurs hématologiques/thérapie , Tumeurs hématologiques/immunologie , Récepteurs aux antigènes des cellules T/immunologie , Récepteurs aux antigènes des cellules T/usage thérapeutique
10.
Cells ; 13(11)2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38891125

RÉSUMÉ

Chimerism analysis after allogeneic hematopoietic stem cell transplantation serves to confirm engraftment, indicate relapse of hematologic malignancy, and attribute graft failure to either immune rejection or poor graft function. Short tandem repeat PCR (STR-PCR) is the prevailing method, followed by quantitative real-time PCR (qPCR), with detection limits of 1-5% and 0.1%, respectively. Chimerism assays using digital PCR or next-generation sequencing, both of which are more sensitive than STR-PCR, are increasingly used. Stable mixed chimerism is usually not associated with poor outcomes in non-malignant diseases, but recipient chimerism may foretell relapse of hematologic malignancies, so higher detection sensitivity may be beneficial in such cases. Thus, the need for and the type of intervention, e.g., immunosuppression regimen, donor lymphocyte infusion, and/or salvage second transplantation, should be guided by donor chimerism in the context of the feature and/or residual malignant cells of the disease to be treated.


Sujet(s)
Chimérisme , Transplantation de cellules souches hématopoïétiques , Transplantation homologue , Humains , Transplantation de cellules souches hématopoïétiques/méthodes , Chimère obtenue par transplantation , Tumeurs hématologiques/thérapie , Tumeurs hématologiques/génétique , Tumeurs hématologiques/immunologie
11.
Cancer Med ; 13(11): e7375, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38864474

RÉSUMÉ

Chimeric antigen receptor T-cell (CAR-T) therapy is becoming an effective technique for the treatment of patients with relapsed/refractory hematologic malignancies. After analyzing patients with tumor progression and sustained remission after CAR-T cell therapy, many factors were found to be associated with the efficacy of CAR-T therapy. This paper reviews the factors affecting the effect of CAR-T such as tumor characteristics, tumor microenvironment and immune function of patients, CAR-T cell structure, construction method and in vivo expansion values, lymphodepletion chemotherapy, and previous treatment, and provides a preliminary outlook on the corresponding therapeutic strategies.


Sujet(s)
Immunothérapie adoptive , Récepteurs chimériques pour l'antigène , Microenvironnement tumoral , Humains , Récepteurs chimériques pour l'antigène/immunologie , Immunothérapie adoptive/méthodes , Microenvironnement tumoral/immunologie , Lymphocytes T/immunologie , Tumeurs hématologiques/thérapie , Tumeurs hématologiques/immunologie , Résultat thérapeutique , Récepteurs aux antigènes des cellules T/génétique , Récepteurs aux antigènes des cellules T/immunologie , Animaux
13.
Nat Rev Clin Oncol ; 21(7): 501-521, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38769449

RÉSUMÉ

Chimeric antigen receptor (CAR) T cell therapy has revolutionized the treatment of several haematological malignancies and is being investigated in patients with various solid tumours. Characteristic CAR T cell-associated toxicities such as cytokine-release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) are now well-recognized, and improved supportive care and management with immunosuppressive agents has made CAR T cell therapy safer and more feasible than it was when the first regulatory approvals of such treatments were granted in 2017. The increasing clinical experience with these therapies has also improved recognition of previously less well-defined toxicities, including movement disorders, immune effector cell-associated haematotoxicity (ICAHT) and immune effector cell-associated haemophagocytic lymphohistiocytosis-like syndrome (IEC-HS), as well as the substantial risk of infection in patients with persistent CAR T cell-induced B cell aplasia and hypogammaglobulinaemia. A more diverse selection of immunosuppressive and supportive-care pharmacotherapies is now being utilized for toxicity management, yet no universal algorithm for their application exists. As CAR T cell products targeting new antigens are developed, additional toxicities involving damage to non-malignant tissues expressing the target antigen are a potential hurdle. Continued prospective evaluation of toxicity management strategies and the design of less-toxic CAR T cell products are both crucial for ongoing success in this field. In this Review, we discuss the evolving understanding and clinical management of CAR T cell-associated toxicities.


Sujet(s)
Immunothérapie adoptive , Récepteurs chimériques pour l'antigène , Humains , Immunothérapie adoptive/effets indésirables , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/usage thérapeutique , Syndrome de libération de cytokines/étiologie , Syndrome de libération de cytokines/immunologie , Syndromes neurotoxiques/étiologie , Syndromes neurotoxiques/immunologie , Tumeurs hématologiques/thérapie , Tumeurs hématologiques/immunologie , Tumeurs/immunologie , Tumeurs/thérapie , Lymphocytes T/immunologie
14.
Nature ; 630(8017): 728-735, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38778101

RÉSUMÉ

Haematopoietic stem cell (HSC) transplantation (HSCT) is the only curative treatment for a broad range of haematological malignancies, but the standard of care relies on untargeted chemotherapies and limited possibilities to treat malignant cells after HSCT without affecting the transplanted healthy cells1. Antigen-specific cell-depleting therapies hold the promise of much more targeted elimination of diseased cells, as witnessed in the past decade by the revolution of clinical practice for B cell malignancies2. However, target selection is complex and limited to antigens expressed on subsets of haematopoietic cells, resulting in a fragmented therapy landscape with high development costs2-5. Here we demonstrate that an antibody-drug conjugate (ADC) targeting the pan-haematopoietic marker CD45 enables the antigen-specific depletion of the entire haematopoietic system, including HSCs. Pairing this ADC with the transplantation of human HSCs engineered to be shielded from the CD45-targeting ADC enables the selective eradication of leukaemic cells with preserved haematopoiesis. The combination of CD45-targeting ADCs and engineered HSCs creates an almost universal strategy to replace a diseased haematopoietic system, irrespective of disease aetiology or originating cell type. We propose that this approach could have broad implications beyond haematological malignancies.


Sujet(s)
Tumeurs hématologiques , Hématopoïèse , Immunoconjugués , Antigènes CD45 , Animaux , Femelle , Humains , Mâle , Souris , Tumeurs hématologiques/traitement médicamenteux , Tumeurs hématologiques/thérapie , Tumeurs hématologiques/immunologie , Hématopoïèse/effets des médicaments et des substances chimiques , Transplantation de cellules souches hématopoïétiques , Cellules souches hématopoïétiques/cytologie , Cellules souches hématopoïétiques/effets des médicaments et des substances chimiques , Cellules souches hématopoïétiques/métabolisme , Immunoconjugués/pharmacologie , Immunoconjugués/usage thérapeutique , Antigènes CD45/immunologie , Antigènes CD45/métabolisme , Lignée cellulaire tumorale , Spécificité des anticorps
15.
Front Immunol ; 15: 1409021, 2024.
Article de Anglais | MEDLINE | ID: mdl-38751430

RÉSUMÉ

Chimeric antigen receptor-T (CAR-T) cell therapy has made remarkable strides in treating hematological malignancies. However, the widespread adoption of CAR-T cell therapy is hindered by several challenges. These include concerns about the long-term and complex manufacturing process, as well as efficacy factors such as tumor antigen escape, CAR-T cell exhaustion, and the immunosuppressive tumor microenvironment. Additionally, safety issues like the risk of secondary cancers post-treatment, on-target off-tumor toxicity, and immune effector responses triggered by CAR-T cells are significant considerations. To address these obstacles, researchers have explored various strategies, including allogeneic universal CAR-T cell development, infusion of non-activated quiescent T cells within a 24-hour period, and in vivo induction of CAR-T cells. This review comprehensively examines the clinical challenges of CAR-T cell therapy and outlines strategies to overcome them, aiming to chart pathways beyond its current Achilles heels.


Sujet(s)
Thérapie cellulaire et tissulaire , Immunothérapie adoptive , Récepteurs chimériques pour l'antigène , Lymphocytes T , Animaux , Humains , Antigènes néoplasiques/immunologie , Tumeurs hématologiques/thérapie , Tumeurs hématologiques/immunologie , Immunothérapie adoptive/méthodes , Immunothérapie adoptive/effets indésirables , Récepteurs aux antigènes des cellules T/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/génétique , Lymphocytes T/immunologie , Lymphocytes T/transplantation , Microenvironnement tumoral/immunologie
16.
Expert Rev Clin Immunol ; 20(7): 745-763, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38739466

RÉSUMÉ

INTRODUCTION: While CAR T-cell therapy has led to remarkable responses in relapsed B-cell hematologic malignancies, only 50% of patients ultimately have a complete, sustained response. Understanding the mechanisms of resistance and relapse after CAR T-cell therapy is crucial to future development and improving outcomes. AREAS COVERED: We review reasons for both primary resistance and relapse after CAR T-cell therapies. Reasons for primary failure include CAR T-cell manufacturing problems, suboptimal fitness of autologous T-cells themselves, and intrinsic features of the underlying cancer and tumor microenvironment. Relapse after initial response to CAR T-cell therapy may be antigen-positive, due to CAR T-cell exhaustion or limited persistence, or antigen-negative, due to antigen-modulation on the target cells. Finally, we discuss ongoing efforts to overcome resistance to CAR T-cell therapy with enhanced CAR constructs, manufacturing methods, alternate cell types, combinatorial strategies, and optimization of both pre-infusion conditioning regimens and post-infusion consolidative strategies. EXPERT OPINION: There is a continued need for novel approaches to CAR T-cell therapy for both hematologic and solid malignancies to obtain sustained remissions. Opportunities for improvement include development of new targets, optimally combining existing CAR T-cell therapies, and defining the role for adjunctive immune modulators and stem cell transplant in enhancing long-term survival.


Sujet(s)
Tumeurs hématologiques , Immunothérapie adoptive , Récepteurs chimériques pour l'antigène , Lymphocytes T , Humains , Immunothérapie adoptive/méthodes , Récepteurs chimériques pour l'antigène/immunologie , Tumeurs hématologiques/thérapie , Tumeurs hématologiques/immunologie , Lymphocytes T/immunologie , Récidive , Animaux , Microenvironnement tumoral/immunologie
17.
Int J Cancer ; 155(4): 618-626, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38721724

RÉSUMÉ

Immunocompromised patients are at high risk to fail clearance of SARS-CoV-2. Prolonged COVID-19 constitutes a health risk and a management problem as cancer treatments often have to be disrupted. As SARS-CoV-2 evolves, new variants of concern have emerged that evade available monoclonal antibodies. Moreover, antiviral therapy promotes SARS-CoV-2 escape mutations, particularly in immunocompromised patients. These patients frequently suffer from prolonged infection. No successful treatment has been established for persistent COVID-19 infection. Here, we report on a series of 21 immunocompromised patients with COVID-19-most of them hematologic malignancies-treated with plasma obtained from recently convalescent or vaccinated donors or a combination thereof. Repeated dosing of SARS-CoV-2-antibody-containing plasma could clear SARS-CoV-2 infection in 16 out of 21 immunocompromised patients even if COVID-19-specific treatments failed to induce sustained viral clearance or to improve clinical course of SARS-CoV-2 infection. Ten patients were major responders defined as an increase delta(d)Ct of > = 5 after the first administration of convalescent and/or vaccinated plasma (C/VP). On average, SARS-CoV-2 PCR Ct values increased from a median value of 22.55 (IQR = 19.10-24.25) to a median value of 29.57 (IQR = 27.55-34.63; p = <.0001) in the major response subgroup. Furthermore, when treated a second time with C/VP, even 4 out of 5 of the initial nonresponders showed an increase in Ct-values from a median value of 23.13 (IQR = 17.75-28.05) to a median value of 32.79 (IQR = 31.75-33.75; p = .013). Our results suggest that C/VP could be a feasible treatment of COVID-19 infection in patients with hematologic malignancies who did not respond to antiviral treatment.


Sujet(s)
Sérothérapie COVID-19 , COVID-19 , Tumeurs hématologiques , Immunisation passive , Sujet immunodéprimé , SARS-CoV-2 , Humains , COVID-19/immunologie , COVID-19/virologie , COVID-19/prévention et contrôle , COVID-19/thérapie , Tumeurs hématologiques/thérapie , Tumeurs hématologiques/immunologie , Tumeurs hématologiques/virologie , Femelle , Adulte d'âge moyen , Mâle , Sujet âgé , SARS-CoV-2/immunologie , Immunisation passive/méthodes , Sujet immunodéprimé/immunologie , Adulte , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Vaccins contre la COVID-19/immunologie , Vaccins contre la COVID-19/administration et posologie , Maladie chronique , Résultat thérapeutique
18.
mSystems ; 9(6): e0138523, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38752789

RÉSUMÉ

A dysfunction of human host genes and proteins in coronavirus infectious disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a key factor impacting clinical symptoms and outcomes. Yet, a detailed understanding of human host immune responses is still incomplete. Here, we applied RNA sequencing to 94 samples of COVID-19 patients with and without hematological tumors as well as COVID-19 uninfected non-tumor individuals to obtain a comprehensive transcriptome landscape of both hematological tumor patients and non-tumor individuals. In our analysis, we further accounted for the human-SARS-CoV-2 protein interactome, human protein interactome, and human protein complex subnetworks to understand the mechanisms of SARS-CoV-2 infection and host immune responses. Our data sets enabled us to identify important SARS-CoV-2 (non-)targeted differentially expressed genes and complexes post-SARS-CoV-2 infection in both hematological tumor and non-tumor individuals. We found several unique differentially expressed genes, complexes, and functions/pathways such as blood coagulation (APOE, SERPINE1, SERPINE2, and TFPI), lipoprotein particle remodeling (APOC2, APOE, and CETP), and pro-B cell differentiation (IGHM, VPREB1, and IGLL1) during COVID-19 infection in patients with hematological tumors. In particular, APOE, a gene that is associated with both blood coagulation and lipoprotein particle remodeling, is not only upregulated in hematological tumor patients post-SARS-CoV-2 infection but also significantly expressed in acute dead patients with hematological tumors, providing clues for the design of future therapeutic strategies specifically targeting COVID-19 in patients with hematological tumors. Our data provide a rich resource for understanding the specific pathogenesis of COVID-19 in immunocompromised patients, such as those with hematological malignancies, and developing effective therapeutics for COVID-19. IMPORTANCE: A majority of previous studies focused on the characterization of coronavirus infectious disease 2019 (COVID-19) disease severity in people with normal immunity, while the characterization of COVID-19 in immunocompromised populations is still limited. Our study profiles changes in the transcriptome landscape post-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in hematological tumor patients and non-tumor individuals. Furthermore, our integrative and comparative systems biology analysis of the interactome, complexome, and transcriptome provides new insights into the tumor-specific pathogenesis of COVID-19. Our findings confirm that SARS-CoV-2 potentially tends to target more non-functional host proteins to indirectly affect host immune responses in hematological tumor patients. The identified unique genes, complexes, functions/pathways, and expression patterns post-SARS-CoV-2 infection in patients with hematological tumors increase our understanding of how SARS-CoV-2 manipulates the host molecular mechanism. Our observed differential genes/complexes and clinical indicators of normal/long infection and deceased COVID-19 patients provide clues for understanding the mechanism of COVID-19 progression in hematological tumors. Finally, our study provides an important data resource that supports the increasing value of the application of publicly accessible data sets to public health.


Sujet(s)
COVID-19 , Sujet immunodéprimé , SARS-CoV-2 , Transcriptome , Humains , COVID-19/génétique , COVID-19/immunologie , COVID-19/virologie , Transcriptome/génétique , SARS-CoV-2/génétique , Tumeurs hématologiques/génétique , Tumeurs hématologiques/immunologie , Mâle , Femelle , Cartes d'interactions protéiques/génétique , Adulte d'âge moyen , Analyse de profil d'expression de gènes/méthodes
19.
Int J Rheum Dis ; 27(5): e15182, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38742463

RÉSUMÉ

Chimeric antigen receptor (CAR) T-cell therapy is a form of immunotherapy where the lymphocytes, mostly T-cells, are redirected to specifically recognize and eliminate a target antigen by coupling them with CARs. The binding of CAR and target cell surface antigens leads to vigorous T cell activation and robust anti-tumor immune responses. Areas of implication of CAR T-cell therapies include mainly hematological malignancies (i.e., advanced B-cell cancers); however, recent studies have proven the unprecedented success of the new immunotherapy also in autoimmune rheumatic diseases. We aim to review the recent advances in CAR T-cell therapies in rheumatology but also to address the limitations of their use in the real clinical practice based on the data on their efficacy and safety.


Sujet(s)
Maladies auto-immunes , Tumeurs hématologiques , Immunothérapie adoptive , Récepteurs chimériques pour l'antigène , Rhumatismes , Humains , Immunothérapie adoptive/effets indésirables , Immunothérapie adoptive/méthodes , Rhumatismes/immunologie , Rhumatismes/thérapie , Récepteurs chimériques pour l'antigène/immunologie , Maladies auto-immunes/immunologie , Maladies auto-immunes/thérapie , Tumeurs hématologiques/immunologie , Tumeurs hématologiques/thérapie , Résultat thérapeutique , Lymphocytes T/immunologie , Animaux
20.
Scand J Immunol ; 99(6): e13364, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38720521

RÉSUMÉ

Mucosal-associated invariant T-cells (MAIT) are unconventional T-cells with cytotoxic and pro-inflammatory properties. Previous research has reported contradictory findings on their role in cancerogenesis with data being even scarcer in haematological malignancies. Here, we report the results of a systematic analysis of MAIT cells in treatment-naïve patients with a broad range of haematological malignancies. We analysed peripheral blood of 204 patients and 50 healthy subjects. The pool of haematological patients had a statistically significant lower both the absolute value (median values, 0.01 × 109/L vs. 0.05 × 109/L) of MAIT cells and their percentage (median values 0.94% vs. 2.56%) among T-cells compared to the control group. Separate analysis showed that the decrease in the absolute number of MAIT cells is significant in patients with acute myeloid leukaemia, myeloproliferative neoplasms, plasma cell myeloma, B-cell non-Hodgkin lymphomas, otherwise not specified, diffuse large B-cell lymphoma, follicular lymphoma, mantle cell lymphoma, marginal zone lymphoma compared to the control population. Furthermore, in haematological malignancies, MAIT cells overexpress PD-1 (average values, 51.7% vs. 6.7%), HLA-DR (average values, 40.2% vs. 7%), CD38 (average values, 25.9% vs. 4.9%) and CD69 (average values, 40.2% vs. 9.2%). Similar results were obtained when comparing patients with individual malignancies to the control population. Our data show that the depletion of circulating MAIT cells is a common observation in a broad spectrum of haematological malignancies. In addition to their reduced numbers, MAIT cells acquire an activated/exhausted phenotype.


Sujet(s)
Tumeurs hématologiques , Cellules T invariantes associées aux muqueuses , Récepteur-1 de mort cellulaire programmée , Humains , Cellules T invariantes associées aux muqueuses/immunologie , Tumeurs hématologiques/immunologie , Mâle , Femelle , Adulte d'âge moyen , Sujet âgé , Adulte , Récepteur-1 de mort cellulaire programmée/immunologie , Récepteur-1 de mort cellulaire programmée/métabolisme , Antigènes CD/métabolisme , Sujet âgé de 80 ans ou plus , Antigènes de différenciation des lymphocytes T/métabolisme , Numération des lymphocytes , Antigènes CD38/métabolisme , Antigènes CD38/immunologie , Immunophénotypage , Jeune adulte , Glycoprotéines membranaires/immunologie , Lectines de type C
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...