Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Arthritis Rheumatol ; 76(2): 268-278, 2024 02.
Article in English | MEDLINE | ID: mdl-37610259

ABSTRACT

OBJECTIVE: Our goal was to study the tolerance and efficacy of two B cell depletion strategies, including one with CD19-targeted chimeric antigen receptor (CAR) T cells, in a preclinical model mimicking the severe lung damages observed in systemic sclerosis. METHODS: B cell depletion strategies were evaluated in the Fra-2 transgenic (Tg) mouse model. We considered a first group of 16 untreated mice, a second group of 15 mice receiving a single dose of anti-CD20 monoclonal antibody (mAb), and a third group of 8 mice receiving CD19-targeted CAR-T cells in combination with anti-CD20 monoclonal antibody. After six weeks of clinical evaluation, different validated markers of inflammation, lung fibrosis, and pulmonary vascular remodeling were assessed. RESULTS: CD19-targeted CAR-T cells infusion in combination with anti-CD20 mAb resulted in a deeper B cell depletion than anti-CD20 mAb alone in the peripheral blood and lesional lungs of Fra-2 Tg mice. CAR-T cell infusion worsened the clinical score and increased mortality in Fra-2 Tg mice. In line with the above findings, CAR-T cell infusion significantly increased lung collagen content, the histological fibrosis score, and right ventricular systolic pressure. CAR-T cells accumulated in lesional lungs and promoted T activation and inflammatory cytokine production. Treatment with anti-CD20 mAb in monotherapy had no impact on lung inflammation-driven fibrosis and pulmonary hypertension. CONCLUSION: B cell therapies failed to show efficacy in the Fra2 Tg mice. The exacerbated Fra-2 lung inflammatory burden stimulated accumulation and expansion of activated CD19-targeted CAR-T cells, secondarily inducing T cell activation and systemic inflammation, finally leading to disease worsening.


Subject(s)
Receptors, Chimeric Antigen , Scleroderma, Systemic , Mice , Animals , T-Lymphocytes , Disease Models, Animal , Antibodies, Monoclonal/pharmacology , Antigens, CD19/metabolism , Mice, Transgenic , Scleroderma, Systemic/metabolism , Fibrosis
2.
Cell Rep Med ; 4(9): 101161, 2023 09 19.
Article in English | MEDLINE | ID: mdl-37595589

ABSTRACT

Anti-CD19 chimeric antigen receptor (CAR) T cell therapy represents a breakthrough for the treatment of B cell malignancies. Yet, it can lead to severe adverse events, including cytokine release syndrome (CRS), which may require urgent clinical management. Whether interpatient variability in CAR T cell subsets contributes to CRS is unclear. Here, we show that CD4+ CAR T cells are the main drivers of CRS. Using an immunocompetent model of anti-CD19 CAR T cell therapy, we report that CD4+, but not CD8+, CAR T cells elicit physiological CRS-like manifestations associated with the release of inflammatory cytokines. In CAR T cell-treated patients, CRS occurrence and severity are significantly associated with high absolute values of CD4+ CAR T cells in the blood. CRS in mice occurs independently of CAR T cell-derived interferon γ (IFN-γ) but requires elevated tumor burden. Thus, adjusting the CD4:CD8 CAR T cell ratio to patient tumor load may help mitigate CAR T cell-associated toxicities.


Subject(s)
Cytokine Release Syndrome , Immunotherapy, Adoptive , Humans , Animals , Mice , Cytokine Release Syndrome/etiology , Immunotherapy, Adoptive/adverse effects , CD8-Positive T-Lymphocytes , Antigens, CD19 , CD4-Positive T-Lymphocytes
3.
Nat Cancer ; 4(7): 968-983, 2023 07.
Article in English | MEDLINE | ID: mdl-37248395

ABSTRACT

CD4+ T cells and CD4+ chimeric antigen receptor (CAR) T cells display highly variable antitumor activity in preclinical models and in patients; however, the mechanisms dictating how and when CD4+ T cells promote tumor regression are incompletely understood. With the help of functional intravital imaging, we report that interferon (IFN)-γ production but not perforin-mediated cytotoxicity was the dominant mechanism for tumor elimination by anti-CD19 CD4+ CAR T cells. Mechanistically, mouse or human CD4+ CAR T-cell-derived IFN-γ diffused extensively to act on tumor cells at distance selectively killing tumors sensitive to cytokine-induced apoptosis, including antigen-negative variants. In anti-CD19 CAR T-cell-treated patients exhibiting elevated CAR CD4:CD8 ratios, strong induction of serum IFN-γ was associated with increased survival. We propose that the sensitivity of tumor cells to the pro-apoptotic activity of IFN-γ is a major determinant of CD4+ CAR T-cell efficacy and may be considered to guide the use of CD4+ T cells during immunotherapy.


Subject(s)
Neoplasms , T-Lymphocytes , Humans , Animals , Mice , Receptors, Antigen, T-Cell , Cytokines , Interferon-gamma , CD4-Positive T-Lymphocytes
4.
Arthritis Rheumatol ; 73(11): 1954-1965, 2021 11.
Article in English | MEDLINE | ID: mdl-34042325

ABSTRACT

Chimeric antigen receptor-T (CAR-T) cell therapy is based on specific targeting of tumor antigens, leading to lysis and destruction of tumor cells. The high potency of CAR-T cells in the management of B cell malignancies has been demonstrated. Following the success of this therapeutic strategy, new CAR-T cell-derived constructs that have the ability to eradicate pathogenic B cells or restore tolerance have been developed. The present review discusses how the knowledge and technology generated by the use of CAR-T cells may be translated and integrated into ongoing therapeutic strategies for autoimmune rheumatic diseases. To this end, we describe the details of CAR-T cell technology, as well as the meaningful achievements attained with the use of CAR-T cells in onco-hematology. In addition, we review the preliminary data obtained with CAR-T cells and their derivative constructs in experimental models of autoimmune diseases. Finally, we focus on how CAR-T cell engineering interferes with the pathogenesis of 3 chronic autoimmune rheumatic diseases-rheumatoid arthritis, systemic lupus erythematosus, and systemic sclerosis-and discuss whether these constructs might yield greater efficacy and be associated with fewer adverse events compared to current treatment strategies.


Subject(s)
Autoimmune Diseases/therapy , Immunotherapy, Adoptive/methods , Receptors, Chimeric Antigen/immunology , Rheumatic Diseases/therapy , T-Lymphocytes/immunology , Autoimmune Diseases/immunology , Humans , Rheumatic Diseases/immunology
5.
Sci Immunol ; 6(57)2021 03 26.
Article in English | MEDLINE | ID: mdl-33771887

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapy relies on the activity of a large pool of tumor-targeting cytotoxic effectors. Whether CAR T cells act autonomously or require interactions with the tumor microenvironment (TME) remains incompletely understood. Here, we report an essential cross-talk between CAR T cell subsets and the TME for tumor control in an immunocompetent mouse B cell lymphoma model of anti-CD19 CAR T cell therapy. Using single-cell RNA sequencing, we revealed substantial modification of the TME during CAR T cell therapy. Interferon-γ (IFN-γ) produced by CAR T cells not only enhanced endogenous T and natural killer cell activity but was also essential for sustaining CAR T cell cytotoxicity, as revealed by intravital imaging. CAR T cell-derived IFN-γ facilitated host interleukin-12 production that supported host immune and CAR T cell responses. Compared with CD8+ CAR T cells, CD4+ CAR T cells were more efficient at host immune activation but less capable of direct tumor killing. In summary, CAR T cells do not act independently in vivo but rely instead on cytokine-mediated cross-talk with the TME for optimal activity. Invigorating CAR T cell interplay with the host represents an attractive strategy to prevent relapses after therapy.


Subject(s)
Cell Communication/immunology , Cytotoxicity Tests, Immunologic , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tumor Microenvironment/immunology , Animals , Antigens, CD19/immunology , Antigens, Neoplasm/immunology , Cell Communication/genetics , Cell Line, Tumor , Computational Biology/methods , Cytokines/metabolism , Disease Models, Animal , Gene Expression Profiling , Humans , Immunotherapy, Adoptive , Interferon-gamma/biosynthesis , Lymphocyte Activation/immunology , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Lymphoma, B-Cell/therapy , Mice , Neoplasms/immunology , Neoplasms/therapy , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics
6.
Curr Biol ; 30(14): 2761-2776.e7, 2020 07 20.
Article in English | MEDLINE | ID: mdl-32502410

ABSTRACT

Neutrophils are major inflammatory cells that rapidly infiltrate wounds to provide antimicrobial functions. Within the damaged tissue, neutrophil migration behavior often switches from exploratory patrolling to coordinated swarming, giving rise to dense clusters that further disrupt tissue architecture. This aggregation response is self-organized by neutrophil paracrine chemoattractant signaling (most notably of the inflammatory mediator leukotriene B4 [LTB4]). The coordination mechanism and possible evolutionary benefits of neutrophil swarms are elusive. Here, we show that neutrophil swarms require mutual reinforcement of damage signaling at the wound core. New biosensors and live imaging in zebrafish revealed that neutrophil chemoattractant synthesis is triggered by a sustained calcium flux upon contact with necrotic tissue that requires sensing of the damage signal ATP. This "calcium alarm" signal rapidly propagates in the nascent neutrophil cluster in a contact-dependent manner via connexin-43 (Cx43) hemichannels, which are mediators of active ATP release. This enhances chemoattractant biosynthesis in the growing cluster, which is instrumental for coordinated motion and swarming. Inhibition of neutrophil Cx43 compromises clearance of wound-colonizing P. aeruginosa bacteria and exacerbates infection-induced morbidity. Thus, cooperative production of alarm signals among pioneer clustering neutrophils fuels the growth of dense antimicrobial cell masses that effectively seal off breached tissue barriers from opportunistic pathogens.


Subject(s)
Calcium/physiology , Connexins/physiology , Neutrophil Infiltration/genetics , Neutrophil Infiltration/physiology , Neutrophils/immunology , Neutrophils/pathology , Signal Transduction/genetics , Signal Transduction/physiology , Wounds and Injuries/pathology , Adenosine Triphosphate/metabolism , Animals , Cell Aggregation/genetics , Cell Aggregation/physiology , Connexin 43 , Leukotriene B4/physiology , Neutrophil Infiltration/immunology , Pseudomonas aeruginosa , Wounds and Injuries/immunology , Zebrafish
7.
Trends Immunol ; 40(11): 1022-1034, 2019 11.
Article in English | MEDLINE | ID: mdl-31668676

ABSTRACT

Understanding complex interactions between the immune system and the tumor microenvironment is an essential step towards the rational development and optimization of immunotherapies. Several experimental approaches are available to tackle this complexity but most are not designed to address the dynamic features of immune reactions, including cell migration, cellular interactions, and transient signaling events. By providing a unique means to access these precious parameters, intravital imaging offers a fresh look at intratumoral immune responses at the single-cell level. Here, we discuss how in vivo imaging sheds light on fundamental aspects of tumor immunity and helps elucidate modes of action of immunotherapies. We conclude by discussing future developments that may consolidate the unique contribution of intravital imaging for our understanding of tumor immunity.


Subject(s)
Immunotherapy/methods , Intravital Microscopy/methods , Neoplasms/immunology , Animals , Cell Movement , Disease Models, Animal , Humans , Immune Tolerance , Immunologic Surveillance , Monitoring, Immunologic , Neoplasms/therapy , Tumor Escape , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...