Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 408
Filtrer
1.
Nat Rev Rheumatol ; 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39107407

RÉSUMÉ

Chimeric antigen receptor (CAR) T cells are highly effective at targeting and eliminating cells of the B cell lineage. CAR T cell therapy has become a standard-of-care treatment for patients with relapsed or refractory B cell malignancies. In addition, the administration of genetically modified T cells with the capacity to deplete B cells and/or plasma cells has tremendous therapeutic potential in autoimmune diseases. In the past few years, CD19-based and B cell maturation antigen (BCMA)-based CAR T cell therapies have been applied to various B cell-mediated autoimmune diseases including systemic lupus erythematosus, idiopathic inflammatory myopathy, systemic sclerosis, neuromyelitis optica spectrum disorder, myasthenia gravis and multiple sclerosis. The scientific rationale behind this approach is that deep depletion of B cells, including autoreactive B cell clones, could restore normal immune function, referred to as an immune reset. In this Review, we discuss important aspects of CAR T cell therapy in autoimmune disease, including considerations relating to patient selection, safety, efficacy and medical management. These considerations are based on the early experiences of CAR T cell therapy in autoimmune diseases, and as the field of CAR T cell therapy in autoimmune diseases continues to rapidly evolve, these issues will remain subject to ongoing refinement and adaptation.

2.
Nat Rev Cancer ; 2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-39048767

RÉSUMÉ

Adoptive cell therapies engineered to express chimeric antigen receptors (CARs) or transgenic T cell receptors (TCRs) to recognize and eliminate cancer cells have emerged as a promising approach for achieving long-term remissions in patients with cancer. To be effective, the engineered cells must persist at therapeutically relevant levels while avoiding off-tumour toxicities, which has been challenging to realize outside of B cell and plasma cell malignancies. This Review discusses concepts to enhance the efficacy, safety and accessibility of cellular immunotherapies by endowing cells with selective resistance to small-molecule drugs or antibody-based therapies to facilitate combination therapies with substances that would otherwise interfere with the functionality of the effector cells. We further explore the utility of engineering healthy haematopoietic stem cells to confer resistance to antigen-directed immunotherapies and small-molecule targeted therapies to expand the therapeutic index of said targeted anticancer agents as well as to facilitate in vivo selection of gene-edited haematopoietic stem cells for non-malignant applications. Lastly, we discuss approaches to evade immune rejection, which may be required in the setting of allogeneic cell therapies. Increasing confidence in the tools and outcomes of genetically modified cell therapy now paves the way for rational combinations that will open new therapeutic horizons.

3.
Proc Natl Acad Sci U S A ; 121(31): e2403002121, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-39047033

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) represents a challenge in oncology, with limited treatment options for advanced-stage patients. Chimeric antigen receptor T cell (CAR T) therapy targeting mesothelin (MSLN) shows promise, but challenges such as the hostile immunosuppressive tumor microenvironment (TME) hinder its efficacy. This study explores the synergistic potential of combining proton radiation therapy (RT) with MSLN-targeting CAR T therapy in a syngeneic PDAC model. Proton RT significantly increased MSLN expression in tumor cells and caused a significant increase in CAR T cell infiltration into tumors. The combination therapy reshaped the immunosuppressive TME, promoting antitumorigenic M1 polarized macrophages and reducing myeloid-derived suppressor cells (MDSC). In a flank PDAC model, the combination therapy demonstrated superior attenuation of tumor growth and improved survival compared to individual treatments alone. In an orthotopic PDAC model treated with image-guided proton RT, tumor growth was significantly reduced in the combination group compared to the RT treatment alone. Further, the combination therapy induced an abscopal effect in a dual-flank tumor model, with increased serum interferon-γ levels and enhanced proliferation of extratumoral CAR T cells. In conclusion, combining proton RT with MSLN-targeting CAR T therapy proves effective in modulating the TME, enhancing CAR T cell trafficking, and exerting systemic antitumor effects. Thus, this combinatorial approach could present a promising strategy for improving outcomes in unresectable PDAC.


Sujet(s)
Carcinome du canal pancréatique , Protéines liées au GPI , Immunothérapie adoptive , Mésothéline , Tumeurs du pancréas , Récepteurs chimériques pour l'antigène , Microenvironnement tumoral , Animaux , Tumeurs du pancréas/thérapie , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/radiothérapie , Tumeurs du pancréas/anatomopathologie , Souris , Protéines liées au GPI/métabolisme , Protéines liées au GPI/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/métabolisme , Immunothérapie adoptive/méthodes , Microenvironnement tumoral/immunologie , Humains , Carcinome du canal pancréatique/thérapie , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/radiothérapie , Carcinome du canal pancréatique/anatomopathologie , Lignée cellulaire tumorale , Protonthérapie/méthodes , Association thérapeutique , Lymphocytes T/immunologie , Femelle
4.
Cancer Immunol Res ; : OF1-OF4, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-39018097

RÉSUMÉ

Methods to engineer the genomes of human cells for therapeutic intervention continue to advance at a remarkable pace. Chimeric antigen receptor-engineered T lymphocytes have pioneered the way for these therapies, initially beginning with insertions of chimeric antigen receptor transgenes into T-cell genomes using classical gene therapy vectors. The broad use of clustered regularly interspaced short palindromic repeats (CRISPR)-based technologies to edit endogenous genes has now opened the door to a new era of precision medicine. To add complexity, many engineered cellular therapies under development integrate gene therapy with genome editing to introduce novel biological functions and enhance therapeutic efficacy. Here, we review the current state of scientific, translational, and regulatory oversight of gene-edited cell products.

5.
Cancer Cell ; 42(8): 1319-1325, 2024 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-39059390

RÉSUMÉ

Chimeric antigen receptor (CAR) T cells are effectively used in certain hematological malignancies, though tumor relapse and limited success in solid tumors persist. Recent efforts focus on developing combination treatments to enhance outcomes and safety. Here, we provide a comprehensive overview of such combinatorial approaches and a consideration of ongoing clinical trials.


Sujet(s)
Immunothérapie adoptive , Tumeurs , Récepteurs chimériques pour l'antigène , Humains , Immunothérapie adoptive/méthodes , Tumeurs/thérapie , Tumeurs/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Lymphocytes T/immunologie , Association thérapeutique , Essais cliniques comme sujet , Récepteurs aux antigènes des cellules T/immunologie , Animaux
6.
Cancer Immunol Res ; 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38869428

RÉSUMÉ

Genome editing technologies have seen remarkable progress in recent years, enabling precise regulation of exogenous and endogenous genes. These advances have been extensively applied to the engineering of human T lymphocytes, leading to the development of practice changing therapies for patients with cancer and the promise of synthetic immune cell therapies for a variety of non-malignant diseases. Many distinct conceptual and technical approaches have been used to edit T-cell genomes, however targeted assessments of which techniques are most effective for manufacturing, gene editing and transgene expression are rarely reported. Through extensive comparative evaluation, we identified methods that most effectively enhance engineering of research-scale and pre-clinical T-cell products at critical stages of manufacturing.

7.
J Immunother Cancer ; 12(6)2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38834201

RÉSUMÉ

BACKGROUND: Advanced clear cell renal cell carcinoma (ccRCC) is a prevalent kidney cancer for which long-term survival rates are abysmal, though immunotherapies are showing potential. Not yet clinically vetted are bispecific T cell engagers (BTEs) that activate T cell-mediated cancer killing through intercellular synapsing. Multiple BTE formats exist, however, with limited cross-characterizations to help optimize new drug design. Here, we developed BTEs to treat ccRCC by targeting carbonic anhydrase 9 (CA9) while characterizing the persistent BTE (PBTE) format and comparing it to a new format, the persistent multivalent T cell engager (PMTE). These antibody therapies against ccRCC are developed as both recombinant and synthetic DNA (synDNA) medicines. METHODS: Antibody formatting effects on binding kinetics were assessed by flow cytometry and intercellular synaptic strength assays while potency was tested using T-cell activation and cytotoxicity assays. Mouse models were used to study antibody plasma and tumor pharmacokinetics, as well as antitumor efficacy as both recombinant and synDNA medicines. Specifically, three models using ccRCC cell line xenografts and human donor T cells in immunodeficient mice were used to support this study. RESULTS: Compared with a first-generation BTE, we show that the PBTE reduced avidity, intercellular synaptic strength, cytotoxic potency by as much as 33-fold, and ultimately efficacy against ccRCC tumors in vivo. However, compared with the PBTE, we demonstrate that the PMTE improved cell avidity, restored intercellular synapses, augmented cytotoxic potency by 40-fold, improved tumor distribution pharmacokinetics by 2-fold, and recovered synDNA efficacy in mouse tumor models by 20-fold. All the while, the PMTE displayed a desirable half-life of 4 days in mice compared with the conventional BTE's 2 hours. CONCLUSIONS: With impressive efficacy, the CA9-targeted PMTE is a promising new therapy for advanced ccRCC, which can be effectively delivered through synDNA. The highly potent PMTE format itself is a promising new tool for future applications in the multispecific antibody space.


Sujet(s)
Anticorps bispécifiques , Néphrocarcinome , Tumeurs du rein , Lymphocytes T , Néphrocarcinome/traitement médicamenteux , Néphrocarcinome/immunologie , Humains , Animaux , Souris , Tumeurs du rein/traitement médicamenteux , Tumeurs du rein/immunologie , Lymphocytes T/immunologie , Anticorps bispécifiques/pharmacologie , Anticorps bispécifiques/usage thérapeutique , Lignée cellulaire tumorale , Immunothérapie/méthodes , Carbonic anhydrase IX/métabolisme , Femelle , Tests d'activité antitumorale sur modèle de xénogreffe
8.
Trends Cancer ; 10(8): 687-695, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38853073

RÉSUMÉ

Chimeric antigen receptor (CAR) T cell therapy has emerged as a revolutionary treatment for hematological malignancies, but its adaptation to solid tumors is impeded by multiple challenges, particularly T cell dysfunction and exhaustion. The heterogeneity and inhospitableness of the solid tumor microenvironment (TME) contribute to diminished CAR T cell efficacy exhibited by reduced cytotoxicity, proliferation, cytokine secretion, and the upregulation of inhibitory receptors, similar to the phenotype of tumor-infiltrating lymphocytes (TILs). In this review, we highlight recent advances in T cell therapy for solid tumors, particularly brain cancer. Innovative strategies, including locoregional delivery and 'armoring' CAR T cells with cytokines such as interleukin (IL)-18, are under investigation to improve efficacy and safety. We also highlight emerging issues with toxicity management of CAR T cell adverse events. This review discusses the obstacles associated with CAR T cell therapy in the context of solid tumors and outlines current and future strategies to overcome these challenges.


Sujet(s)
Immunothérapie adoptive , Tumeurs , Récepteurs chimériques pour l'antigène , Lymphocytes T , Microenvironnement tumoral , Humains , Immunothérapie adoptive/méthodes , Immunothérapie adoptive/effets indésirables , Tumeurs/immunologie , Tumeurs/thérapie , Tumeurs/génétique , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Microenvironnement tumoral/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/génétique , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Animaux , Récepteurs aux antigènes des cellules T/immunologie , Récepteurs aux antigènes des cellules T/métabolisme , Récepteurs aux antigènes des cellules T/génétique
10.
Nat Commun ; 15(1): 3937, 2024 May 10.
Article de Anglais | MEDLINE | ID: mdl-38729924

RÉSUMÉ

Human natural killer (NK) cell-based therapies are under assessment for treating various cancers, but cryopreservation reduces both the recovery and function of NK cells, thereby limiting their therapeutic feasibility. Using cryopreservation protocols optimized for T cells, here we find that ~75% of NK cells die within 24 h post-thaw, with the remaining cells displaying reduced cytotoxicity. Using CRISPR-Cas9 gene editing and confocal microscopy, we find that cryopreserved NK cells largely die via apoptosis initiated by leakage of granzyme B from cytotoxic vesicles. Pretreatment of NK cells with a combination of Interleukins-15 (IL-15) and IL-18 prior to cryopreservation improves NK cell recovery to ~90-100% and enables equal tumour control in a xenograft model of disseminated Raji cell lymphoma compared to non-cryopreserved NK cells. The mechanism of IL-15 and IL-18-induced protection incorporates two mechanisms: a transient reduction in intracellular granzyme B levels via degranulation, and the induction of antiapoptotic genes.


Sujet(s)
Apoptose , Cryoconservation , Granzymes , Interleukine-15 , Interleukine-18 , Cellules tueuses naturelles , Granzymes/métabolisme , Interleukine-15/métabolisme , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Humains , Interleukine-18/métabolisme , Animaux , Cryoconservation/méthodes , Souris , Lignée cellulaire tumorale , Systèmes CRISPR-Cas
11.
Nat Commun ; 15(1): 3933, 2024 May 10.
Article de Anglais | MEDLINE | ID: mdl-38730243

RÉSUMÉ

As a strategy to improve the therapeutic success of chimeric antigen receptor T cells (CART) directed against solid tumors, we here test the combinatorial use of CART and IMSA101, a newly developed stimulator of interferon genes (STING) agonist. In two syngeneic tumor models, improved overall survival is observed when mice are treated with intratumorally administered IMSA101 in addition to intravenous CART infusion. Transcriptomic analyses of CART isolated from tumors show elevated T cell activation, as well as upregulated cytokine pathway signatures, in particular IL-18, in the combination treatment group. Also, higher levels of IL-18 in serum and tumor are detected with IMSA101 treatment. Consistent with this, the use of IL-18 receptor negative CART impair anti-tumor responses in mice receiving combination treatment. In summary, we find that IMSA101 enhances CART function which is facilitated through STING agonist-induced IL-18 secretion.


Sujet(s)
Interleukine-18 , Protéines membranaires , Récepteurs chimériques pour l'antigène , Animaux , Interleukine-18/métabolisme , Protéines membranaires/agonistes , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Souris , Récepteurs chimériques pour l'antigène/métabolisme , Récepteurs chimériques pour l'antigène/immunologie , Humains , Lignée cellulaire tumorale , Souris de lignée C57BL , Lymphocytes T/immunologie , Lymphocytes T/effets des médicaments et des substances chimiques , Lymphocytes T/métabolisme , Activation des lymphocytes/effets des médicaments et des substances chimiques , Immunothérapie adoptive/méthodes , Femelle , Tumeurs/immunologie , Tumeurs/thérapie , Tumeurs/traitement médicamenteux
12.
Blood Cancer Discov ; 5(4): 229-233, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38713827

RÉSUMÉ

In this commentary, we discuss the investigation into reports of T-cell malignancies following chimeric antigen receptor T-cell therapy. We argue that although these cases should be thoroughly examined, current data suggest that such risks with autologous chimeric antigen receptor T cells are remarkably low compared with other cancer treatments. We also emphasize the importance of continued research, transparent reporting, and participation in postauthorization safety studies.


Sujet(s)
Transformation cellulaire néoplasique , Immunothérapie adoptive , Récepteurs chimériques pour l'antigène , Lymphocytes T , Humains , Immunothérapie adoptive/méthodes , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/génétique , Transformation cellulaire néoplasique/immunologie , Transformation cellulaire néoplasique/génétique , Lymphocytes T/immunologie , Récepteurs aux antigènes des cellules T/immunologie , Récepteurs aux antigènes des cellules T/génétique , Récepteurs aux antigènes des cellules T/métabolisme , Tumeurs/immunologie , Tumeurs/thérapie
14.
Nat Med ; 30(5): 1320-1329, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38480922

RÉSUMÉ

Recurrent glioblastoma (rGBM) remains a major unmet medical need, with a median overall survival of less than 1 year. Here we report the first six patients with rGBM treated in a phase 1 trial of intrathecally delivered bivalent chimeric antigen receptor (CAR) T cells targeting epidermal growth factor receptor (EGFR) and interleukin-13 receptor alpha 2 (IL13Rα2). The study's primary endpoints were safety and determination of the maximum tolerated dose. Secondary endpoints reported in this interim analysis include the frequency of manufacturing failures and objective radiographic response (ORR) according to modified Response Assessment in Neuro-Oncology criteria. All six patients had progressive, multifocal disease at the time of treatment. In both dose level 1 (1 ×107 cells; n = 3) and dose level 2 (2.5 × 107 cells; n = 3), administration of CART-EGFR-IL13Rα2 cells was associated with early-onset neurotoxicity, most consistent with immune effector cell-associated neurotoxicity syndrome (ICANS), and managed with high-dose dexamethasone and anakinra (anti-IL1R). One patient in dose level 2 experienced a dose-limiting toxicity (grade 3 anorexia, generalized muscle weakness and fatigue). Reductions in enhancement and tumor size at early magnetic resonance imaging timepoints were observed in all six patients; however, none met criteria for ORR. In exploratory endpoint analyses, substantial CAR T cell abundance and cytokine release in the cerebrospinal fluid were detected in all six patients. Taken together, these first-in-human data demonstrate the preliminary safety and bioactivity of CART-EGFR-IL13Rα2 cells in rGBM. An encouraging early efficacy signal was also detected and requires confirmation with additional patients and longer follow-up time. ClinicalTrials.gov identifier: NCT05168423 .


Sujet(s)
Récepteurs ErbB , Glioblastome , Immunothérapie adoptive , Sous-unité alpha2 du récepteur à l'interleukine-13 , Récepteurs chimériques pour l'antigène , Humains , Glioblastome/thérapie , Glioblastome/immunologie , Glioblastome/imagerie diagnostique , Glioblastome/anatomopathologie , Sous-unité alpha2 du récepteur à l'interleukine-13/immunologie , Adulte d'âge moyen , Mâle , Récepteurs chimériques pour l'antigène/immunologie , Femelle , Immunothérapie adoptive/effets indésirables , Immunothérapie adoptive/méthodes , Récidive tumorale locale/immunologie , Récidive tumorale locale/anatomopathologie , Adulte , Sujet âgé , Tumeurs du cerveau/immunologie , Tumeurs du cerveau/thérapie , Tumeurs du cerveau/anatomopathologie , Injections rachidiennes , Dose maximale tolérée
16.
Proc Natl Acad Sci U S A ; 121(13): e2319856121, 2024 Mar 26.
Article de Anglais | MEDLINE | ID: mdl-38513098

RÉSUMÉ

The use of lipid nanoparticles (LNP) to encapsulate and deliver mRNA has become an important therapeutic advance. In addition to vaccines, LNP-mRNA can be used in many other applications. For example, targeting the LNP with anti-CD5 antibodies (CD5/tLNP) can allow for efficient delivery of mRNA payloads to T cells to express protein. As the percentage of protein expressing T cells induced by an intravenous injection of CD5/tLNP is relatively low (4-20%), our goal was to find ways to increase mRNA-induced translation efficiency. We showed that T cell activation using an anti-CD3 antibody improved protein expression after CD5/tLNP transfection in vitro but not in vivo. T cell health and activation can be increased with cytokines, therefore, using mCherry mRNA as a reporter, we found that culturing either mouse or human T cells with the cytokine IL7 significantly improved protein expression of delivered mRNA in both CD4+ and CD8+ T cells in vitro. By pre-treating mice with systemic IL7 followed by tLNP administration, we observed significantly increased mCherry protein expression by T cells in vivo. Transcriptomic analysis of mouse T cells treated with IL7 in vitro revealed enhanced genomic pathways associated with protein translation. Improved translational ability was demonstrated by showing increased levels of protein expression after electroporation with mCherry mRNA in T cells cultured in the presence of IL7, but not with IL2 or IL15. These data show that IL7 selectively increases protein translation in T cells, and this property can be used to improve expression of tLNP-delivered mRNA in vivo.


Sujet(s)
Lymphocytes T CD4+ , Lymphocytes T CD8+ , Interleukine-7 , Liposomes , Nanoparticules , Biosynthèse des protéines , ARN messager , Animaux , Humains , Souris , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Lymphocytes T CD8+/métabolisme , Interleukine-7/pharmacologie , Biosynthèse des protéines/effets des médicaments et des substances chimiques , ARN messager/métabolisme , Souris de lignée C57BL , Cellules cultivées , Lymphocytes T CD4+/effets des médicaments et des substances chimiques , Lymphocytes T CD4+/immunologie
17.
Adv Mater ; 36(26): e2313226, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38419362

RÉSUMÉ

Chimeric antigen receptor (CAR) T cell therapy has achieved remarkable clinical success in the treatment of hematological malignancies. However, producing these bespoke cancer-killing cells is a complicated ex vivo process involving leukapheresis, artificial T cell activation, and CAR construct introduction. The activation step requires the engagement of CD3/TCR and CD28 and is vital for T cell transfection and differentiation. Though antigen-presenting cells (APCs) facilitate activation in vivo, ex vivo activation relies on antibodies against CD3 and CD28 conjugated to magnetic beads. While effective, this artificial activation adds to the complexity of CAR T cell production as the beads must be removed prior to clinical implementation. To overcome this challenge, this work develops activating lipid nanoparticles (aLNPs) that mimic APCs to combine the activation of magnetic beads and the transfection capabilities of LNPs. It is shown that aLNPs enable one-step activation and transfection of primary human T cells with the resulting mRNA CAR T cells reducing tumor burden in a murine xenograft model, validating aLNPs as a promising platform for the rapid production of mRNA CAR T cells.


Sujet(s)
Cellules présentatrices d'antigène , Immunothérapie adoptive , Nanoparticules , ARN messager , Récepteurs chimériques pour l'antigène , Lymphocytes T , Humains , Nanoparticules/composition chimique , Animaux , Souris , Cellules présentatrices d'antigène/immunologie , Immunothérapie adoptive/méthodes , Lymphocytes T/immunologie , ARN messager/génétique , ARN messager/métabolisme , Tumeurs/thérapie , Tumeurs/immunologie , Immunothérapie/méthodes , Lignée cellulaire tumorale , Lipides/composition chimique , Transfection/méthodes , Liposomes
18.
Blood Adv ; 8(9): 2182-2192, 2024 May 14.
Article de Anglais | MEDLINE | ID: mdl-38386999

RÉSUMÉ

ABSTRACT: Relapse after CD19-directed chimeric antigen receptor (CAR)-modified T cells remains a substantial challenge. Short CAR T-cell persistence contributes to relapse risk, necessitating novel approaches to prolong durability. CAR T-cell reinfusion (CARTr) represents a potential strategy to reduce the risk of or treat relapsed disease after initial CAR T-cell infusion (CARTi). We conducted a retrospective review of reinfusion of murine (CTL019) or humanized (huCART19) anti-CD19/4-1BB CAR T cells across 3 clinical trials or commercial tisagenlecleucel for relapse prevention (peripheral B-cell recovery [BCR] or marrow hematogones ≤6 months after CARTi), minimal residual disease (MRD) or relapse, or nonresponse to CARTi. The primary endpoint was complete response (CR) at day 28 after CARTr, defined as complete remission with B-cell aplasia. Of 262 primary treatments, 81 were followed by ≥1 reinfusion (investigational CTL019, n = 44; huCART19, n = 26; tisagenlecleucel, n = 11), representing 79 patients. Of 63 reinfusions for relapse prevention, 52% achieved CR (BCR, 15/40 [38%]; hematogones, 18/23 [78%]). Lymphodepletion was associated with response to CARTr for BCR (odds ratio [OR], 33.57; P = .015) but not hematogones (OR, 0.30; P = .291). The cumulative incidence of relapse was 29% at 24 months for CR vs 61% for nonresponse to CARTr (P = .259). For MRD/relapse, CR rate to CARTr was 50% (5/10), but 0/8 for nonresponse to CARTi. Toxicity was generally mild, with the only grade ≥3 cytokine release syndrome (n = 6) or neurotoxicity (n = 1) observed in MRD/relapse treatment. Reinfusion of CTL019/tisagenlecleucel or huCART19 is safe, may reduce relapse risk in a subset of patients, and can reinduce remission in CD19+ relapse.


Sujet(s)
Antigènes CD19 , Immunothérapie adoptive , Leucémie-lymphome lymphoblastique à précurseurs B et T , Humains , Enfant , Antigènes CD19/immunologie , Antigènes CD19/usage thérapeutique , Leucémie-lymphome lymphoblastique à précurseurs B et T/thérapie , Immunothérapie adoptive/méthodes , Immunothérapie adoptive/effets indésirables , Enfant d'âge préscolaire , Femelle , Mâle , Récepteurs chimériques pour l'antigène/usage thérapeutique , Adolescent , Récidive , Études rétrospectives , Nourrisson , Récepteurs aux antigènes des cellules T/usage thérapeutique , Résultat thérapeutique , Lymphocytes T/immunologie
19.
Nat Biomed Eng ; 8(5): 513-528, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38378820

RÉSUMÉ

The broader clinical use of bispecific T cell engagers for inducing anti-tumour toxicity is hindered by their on-target off-tumour toxicity and the associated neurotoxicity and cytokine-release syndrome. Here we show that the off-tumour toxicity of a supramolecular bispecific T cell engager binding to the T cell co-receptor CD3 and to the human epidermal growth factor receptor 2 on breast tumour cells can be halted by disengaging the T cells from the tumour cells via the infusion of the small-molecule drug amantadine, which disassembles the supramolecular aggregate. In mice bearing human epidermal growth factor receptor 2-expressing tumours and with a human immune system, high intravenous doses of such a 'switchable T cell nanoengager' elicited strong tumour-specific adaptive immune responses that prevented tumour relapse, while the infusion of amantadine restricted off-tumour toxicity, cytokine-release syndrome and neurotoxicity. Supramolecular chemistry may be further leveraged to control the anti-tumour activity and off-tumour toxicity of bispecific antibodies.


Sujet(s)
Amantadine , Anticorps bispécifiques , Antigènes CD3 , Lymphocytes T , Animaux , Humains , Lymphocytes T/immunologie , Lymphocytes T/effets des médicaments et des substances chimiques , Anticorps bispécifiques/pharmacologie , Anticorps bispécifiques/immunologie , Souris , Antigènes CD3/immunologie , Amantadine/pharmacologie , Lignée cellulaire tumorale , Femelle , Récepteur ErbB-2/immunologie , Récepteur ErbB-2/métabolisme , Tumeurs du sein/immunologie , Tumeurs du sein/traitement médicamenteux
20.
Proc Natl Acad Sci U S A ; 121(10): e2317735121, 2024 Mar 05.
Article de Anglais | MEDLINE | ID: mdl-38408246

RÉSUMÉ

Chimeric antigen receptor (CAR) T cell dysfunction is a major barrier to achieving lasting remission in hematologic cancers, especially in chronic lymphocytic leukemia (CLL). We have shown previously that Δ133p53α, an endogenous isoform of the human TP53 gene, decreases in expression with age in human T cells, and that reconstitution of Δ133p53α in poorly functional T cells can rescue proliferation [A. M. Mondal et al., J. Clin. Invest. 123, 5247-5257 (2013)]. Although Δ133p53α lacks a transactivation domain, it can form heterooligomers with full-length p53 and modulate the p53-mediated stress response [I. Horikawa et al., Cell Death Differ. 24, 1017-1028 (2017)]. Here, we show that constitutive expression of Δ133p53α potentiates the anti-tumor activity of CD19-directed CAR T cells and limits dysfunction under conditions of high tumor burden and metabolic stress. We demonstrate that Δ133p53α-expressing CAR T cells exhibit a robust metabolic phenotype, maintaining the ability to execute effector functions and continue proliferating under nutrient-limiting conditions, in part due to upregulation of critical biosynthetic processes and improved mitochondrial function. Importantly, we show that our strategy to constitutively express Δ133p53α improves the anti-tumor efficacy of CAR T cells generated from CLL patients that previously failed CAR T cell therapy. More broadly, our results point to the potential role of the p53-mediated stress response in limiting the prolonged antitumor functions required for complete tumor clearance in patients with high disease burden, suggesting that modulation of the p53 signaling network with Δ133p53α may represent a translationally viable strategy for improving CAR T cell therapy.


Sujet(s)
Leucémie chronique lymphocytaire à cellules B , Récepteurs chimériques pour l'antigène , Humains , Immunothérapie adoptive/méthodes , Leucémie chronique lymphocytaire à cellules B/génétique , Leucémie chronique lymphocytaire à cellules B/thérapie , Récepteurs chimériques pour l'antigène/génétique , Récepteurs chimériques pour l'antigène/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Antigènes CD19 , Thérapie cellulaire et tissulaire , Récepteurs aux antigènes des cellules T/génétique , Récepteurs aux antigènes des cellules T/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE