Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 97
Filtrer
2.
Blood Cancer Discov ; 5(4): 234-248, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38904305

RÉSUMÉ

Despite advancements, acute myeloid leukemia (AML) remains unconquered by current therapies. Evidence of immune evasion during AML progression, such as HLA loss and T-cell exhaustion, suggests that antileukemic immune responses contribute to disease control and could be harnessed by immunotherapy. In this review, we discuss a spectrum of AML immunotherapy targets, encompassing cancer cell-intrinsic and surface antigens as well as targeting in the leukemic milieu, and how they can be tailored for personalized approaches. These targets are overviewed across major immunotherapy modalities applied to AML: immune checkpoint inhibitors, antibody-drug conjugates, therapeutic vaccines, bispecific/trispecific antibodies, and chimeric antigen receptor (CAR)-T and CAR-NK cells. Significance: Immune therapies in AML treatment show evolving promise. Ongoing research aims to customize approaches for varied patient profiles and clinical scenarios. This review covers immune surveillance mechanisms, therapy options like checkpoint inhibitors, antibodies, CAR-T/NK cells, and vaccines, as well as resistance mechanisms and microenvironment considerations.


Sujet(s)
Immunothérapie , Leucémie aigüe myéloïde , Humains , Leucémie aigüe myéloïde/immunologie , Leucémie aigüe myéloïde/thérapie , Immunothérapie/méthodes , Immunothérapie/tendances , Vaccins anticancéreux/usage thérapeutique , Vaccins anticancéreux/immunologie , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie
3.
Nat Immunol ; 25(6): 1020-1032, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38831106

RÉSUMÉ

The efficacy of T cell-based immunotherapies is limited by immunosuppressive pressures in the tumor microenvironment. Here we show a predominant role for the interaction between BTLA on effector T cells and HVEM (TNFRSF14) on immunosuppressive tumor microenvironment cells, namely regulatory T cells. High BTLA expression in chimeric antigen receptor (CAR) T cells correlated with poor clinical response to treatment. Therefore, we deleted BTLA in CAR T cells and show improved tumor control and persistence in models of lymphoma and solid malignancies. Mechanistically, BTLA inhibits CAR T cells via recruitment of tyrosine phosphatases SHP-1 and SHP-2, upon trans engagement with HVEM. BTLA knockout thus promotes CAR signaling and subsequently enhances effector function. Overall, these data indicate that the BTLA-HVEM axis is a crucial immune checkpoint in CAR T cell immunotherapy and warrants the use of strategies to overcome this barrier.


Sujet(s)
Immunothérapie adoptive , Récepteurs chimériques pour l'antigène , Récepteurs immunologiques , Membre-14 de la superfamille des récepteurs au TNF , Microenvironnement tumoral , Animaux , Humains , Immunothérapie adoptive/méthodes , Membre-14 de la superfamille des récepteurs au TNF/métabolisme , Membre-14 de la superfamille des récepteurs au TNF/immunologie , Membre-14 de la superfamille des récepteurs au TNF/génétique , Souris , Microenvironnement tumoral/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/métabolisme , Récepteurs chimériques pour l'antigène/génétique , Récepteurs immunologiques/métabolisme , Récepteurs immunologiques/génétique , Lymphocytes T régulateurs/immunologie , Transduction du signal , Lignée cellulaire tumorale , Tumeurs/immunologie , Tumeurs/thérapie , Souris knockout
4.
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
5.
NEJM Evid ; 3(4): EVIDoa2300213, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38776868

RÉSUMÉ

BACKGROUND: Administration of anti-CD19 chimeric antigen receptor T-cell (CART19) immunotherapy for large B-cell lymphomas (LBCLs), a subset of non-Hodgkin lymphoma (NHL), involves high costs and access to specialized tertiary care centers. We investigated whether minority health populations (MHPs) have equal access to CART19 and whether their outcomes are similar to those of non-MHPs. METHODS: We analyzed the prevalence and clinical outcomes of patients treated with commercial CART19 at two geographically and socioeconomically different institutions: the Abramson Cancer Center (ACC, Philadelphia, Pennsylvania) and the Knight Cancer Institute (KCI, Portland, Oregon). RESULTS: In the ACC catchment area, 8956 patients were diagnosed with NHL between 2015 and 2019 (latest available data from the state registry), including 17.9% MHPs. In the ACC, between 2018 and 2022 (CART became available in 2018), 1492 patients with LBCL were treated, and 194 received CART19. The proportion of MHPs was 15.7% for the entire LBCL cohort but only 6.7% for the CART19 cohort. During the same time, in the KCI catchment area, 4568 patients were diagnosed with NHL, including 4.2% MHPs. In the KCI, 396 patients with LBCL were treated, and 47 received CART19. The proportion of MHPs was 6.6% for the entire LBCL cohort and 4.2% for the CART19 cohort. The 3-month response, survival, and toxicities after CART19 infusion showed similar results, although the number of patients who were treated was limited. CONCLUSIONS: This study shows that the access of MHPs to tertiary centers for LBCL care was preserved but appeared reduced for commercial CART19 immunotherapy. Although clinical outcomes of MHPs seemed similar to those of non-MHPs, the small sample size precludes drawing firm conclusions. Further studies are needed. (Funded by the Laffey McHugh Foundation and others.).


Sujet(s)
Immunothérapie adoptive , Humains , Mâle , Femelle , Adulte d'âge moyen , Immunothérapie adoptive/effets indésirables , Sujet âgé , Adulte , Minorités/statistiques et données numériques , Récepteurs chimériques pour l'antigène/immunologie , Antigènes CD19/immunologie , Antigènes CD19/usage thérapeutique
6.
bioRxiv ; 2024 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-38659938

RÉSUMÉ

Chimeric antigen receptor (CAR) T cell therapy targeting CD19 elicits remarkable clinical efficacy in B-cell malignancies, but many patients relapse due to failed expansion and/or progressive loss of CAR-T cells. We recently reported a strategy to potently restimulate CAR-T cells in vivo, enhancing their functionality by administration of a vaccine-like stimulus comprised of surrogate peptide ligands for a CAR linked to a lymph node-targeting amphiphilic PEG-lipid (termed CAR-T-vax). Here, we demonstrate a general strategy to generate and optimize peptide mimotopes enabling CAR-T-vax generation for any CAR. Using the clinical CD19 CAR FMC63 as a test case, we employed yeast surface display to identify peptide binders to soluble IgG versions of FMC63, which were subsequently affinity matured by directed evolution. CAR-T vaccines using these optimized mimotopes triggered marked expansion of both murine CD19 CAR-T cells in a syngeneic model and human CAR-T cells in a humanized mouse model of B cell acute lymphoblastic leukemia (B-ALL), and enhanced control of leukemia progression. This approach thus enables vaccine boosting to be applied to any clinically-relevant CAR-T cell product.

7.
J Hematol Oncol ; 17(1): 19, 2024 Apr 22.
Article de Anglais | MEDLINE | ID: mdl-38644469

RÉSUMÉ

Bendamustine has been retrospectively shown to be an effective and safe lymphodepletion regimen prior to the anti-CD19 chimeric antigen receptor T cell (CART) products tisagenlecleucel and axicabtagene ciloleucel, as well as the anti-BCMA CART products idecabtagene vicleucel and ciltacabtagene autoleucel. However, bendamustine as lymphodepletion prior to lisocabtagene maraleucel (liso-cel), a 4-1BB co-stimulated, fixed CD4:CD8 ratio anti-CD19 CART product, has not been described yet. Thus, we studied a cohort of sequentially-treated patients with large B-cell lymphomas who received bendamustine lymphodepletion before liso-cel at the University of Pennsylvania between 5/2021 and 12/2023 (n = 31). Patients were evaluated for toxicities and responses. Of note, 7 patients (22.6%) would have dnot met the inclusion criteria for the registrational liso-cel clinical trials, mostly due to older age. Overall and complete response rates were 76.9% and 73.1%, respectively. At a median follow-up of 6.3 months, the 6-month progression-free and overall survival were 59.9% and 91.1%, respectively. Rates of cytokine-release syndrome (CRS) and neurotoxicity (ICANS) of any grade were 9.7% and 9.7%, respectively, with no grade ≥ 3 events. No infections were reported during the first 30 days following liso-cel infusion. Neutropenia ≥ grade 3 was observed in 29.0% of patients; thrombocytopenia ≥ grade 3 occurred in 9.7%. In conclusion, bendamustine lymphodepletion before liso-cel appears to be a strategy that can drive tumor responses while ensuring a mild toxicity profile.


Sujet(s)
Chlorhydrate de bendamustine , Immunothérapie adoptive , Humains , Chlorhydrate de bendamustine/usage thérapeutique , Adulte d'âge moyen , Mâle , Femelle , Sujet âgé , Immunothérapie adoptive/méthodes , Immunothérapie adoptive/effets indésirables , Études rétrospectives , Adulte , Lymphome B diffus à grandes cellules/traitement médicamenteux , Antinéoplasiques alcoylants/usage thérapeutique , Antinéoplasiques alcoylants/effets indésirables , Produits biologiques/usage thérapeutique , Produits biologiques/effets indésirables , Sujet âgé de 80 ans ou plus , Résultat thérapeutique
8.
Transplant Cell Ther ; 30(7): 726.e1-726.e8, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38494076

RÉSUMÉ

Brexucabtagene autoleucel (brexu-cel) is an autologous CD19-directed chimeric antigen receptor (CAR) T-cell therapy approved for treatment of relapsed/refractory mantle cell lymphoma (MCL). During a fludarabine shortage, we used bendamustine as an alternative to standard cyclophosphamide/fludarabine (cy/flu) lymphodepletion (LD) prior to brexu-cel. We assessed MCL patient outcomes as well as CAR T-cell expansion and persistence after brexu-cel following bendamustine or cy/flu LD at our center. This was a retrospective single institution study that utilized prospectively banked blood and tissue samples. Clinical efficacy was assessed by 2014 Lugano guidelines. CAR T-cell expansion and persistence in peripheral blood were assessed on day 7 and at ≥month 6 for patients with available samples. Seventeen patients received bendamustine and 5 received cy/flu. For the bendamustine cohort, 14 (82%) received bridging therapy and 4 (24%) had CNS involvement. Fifteen patients (88%) developed CRS with 4 (24%) ≥grade 3 events. Six (35%) patients developed ICANS with 4 (24%) events ≥grade 3. No patient had ≥grade 3 cytopenias at day 90. Best objective (BOR) and complete response (CRR) rates were 82% and 65%, respectively. At 24.5 months median follow-up, 12-month progression-free survival (PFS) was 45%, 24-month PFS was 25%, and median duration of response was 19 months. Median OS was not reached. BOR was 25% (1/4) for patients with CNS involvement. CAR transgene expansion after bendamustine LD was observed on day 7 in all (4/4) patients tested and persisted at ≥6 months (2/2), regardless of response. Bendamustine LD before brexu-cel for MCL is feasible and safe with a lower frequency and shorter duration of cytopenias than reported for cy/flu. Both CAR T-cell expansion and persistence were observed after bendamustine LD. Outcomes appear comparable to the real world outcomes reported with cy/flu LD.


Sujet(s)
Chlorhydrate de bendamustine , Immunothérapie adoptive , Lymphome à cellules du manteau , Humains , Chlorhydrate de bendamustine/usage thérapeutique , Chlorhydrate de bendamustine/administration et posologie , Lymphome à cellules du manteau/traitement médicamenteux , Mâle , Femelle , Adulte d'âge moyen , Sujet âgé , Études rétrospectives , Immunothérapie adoptive/méthodes , Vidarabine/analogues et dérivés , Vidarabine/usage thérapeutique , Vidarabine/administration et posologie , Adulte , Antinéoplasiques alcoylants/usage thérapeutique , Antinéoplasiques alcoylants/administration et posologie , Antigènes CD19/immunologie , Résultat thérapeutique
9.
Cytotherapy ; 26(5): 506-511, 2024 05.
Article de Anglais | MEDLINE | ID: mdl-38483365

RÉSUMÉ

BACKGROUND AIMS: The successful development of CD19-targeted chimeric antigen receptor (CAR) T-cell therapies has led to an exponential increase in the number of patients recieving treatment and the advancement of novel CAR T products. Therefore, there is a strong need to develop streamlined platforms that allow rapid, cost-effective, and accurate measurement of the key characteristics of CAR T cells during manufacturing (i.e., cell number, cell size, viability, and basic phenotype). METHODS: In this study, we compared the novel benchtop cell analyzer Moxi GO II (ORFLO Technologies), which enables simultaneous evaluation of all the aforementioned parameters, with current gold standards in the field: the Multisizer Coulter Counter (cell counter) and the BD LSRFortessa (flow cytometer). RESULTS: Our results demonstrated that the Moxi GO II can accurately measure cell number and cell size (i.e., cell volume) while simultaneously assessing simple two-color flow cytometry parameters, such as CAR T-cell viability and CD4 or CAR expression. CONCLUSIONS: These measurements are comparable with those of gold standard instruments, demonstrating that the Moxi GO II is a promising platform for quickly monitoring CAR T-cell growth and phenotype in research-grade and clinical samples.


Sujet(s)
Survie cellulaire , Cytométrie en flux , Immunothérapie adoptive , Récepteurs chimériques pour l'antigène , Lymphocytes T , Humains , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/métabolisme , Cytométrie en flux/méthodes , Immunothérapie adoptive/méthodes , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Antigènes CD19/immunologie , Antigènes CD19/métabolisme , Phénotype , Récepteurs aux antigènes des cellules T/immunologie , Récepteurs aux antigènes des cellules T/métabolisme , Immunophénotypage/méthodes , Taille de la cellule
10.
J Infect Dis ; 2024 Mar 04.
Article de Anglais | MEDLINE | ID: mdl-38437622

RÉSUMÉ

Patients with B-cell lymphomas have altered cellular components of vaccine responses due to malignancy and therapy, and the optimal timing of vaccination relative to therapy remains unknown. SARS-CoV-2 vaccines created an opportunity for new insights in vaccine timing because patients were challenged with a novel antigen across multiple phases of treatment. We studied serologic mRNA vaccine response in retrospective and prospective cohorts with lymphoma and CLL, paired with clinical and research immune parameters. Reduced serologic response was observed more frequently during active therapies, but non-response was also common within observation and post-treatment groups. Total IgA and IgM correlated with successful vaccine response. In individuals treated with CART-19, non-response was associated with reduced B and T follicular helper cells. Predictors of vaccine response varied by disease and therapeutic group, and therefore further studies of immune health during and after cancer therapies are needed to allow individualized vaccine timing.

11.
Nat Med ; 30(4): 984-989, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38266761

RÉSUMÉ

We report a T cell lymphoma (TCL) occurring 3 months after anti-CD19 chimeric antigen receptor (CAR) T cell immunotherapy for non-Hodgkin B cell lymphoma. The TCL was diagnosed from a thoracic lymph node upon surgery for lung cancer. The TCL exhibited CD8+ cytotoxic phenotype and a JAK3 variant, while the CAR transgene was very low. The T cell clone was identified at low levels in the blood before CAR T infusion and in lung cancer. To assess the overall risk of secondary primary malignancy after commercial CAR T (CD19, BCMA), we analyzed 449 patients treated at the University of Pennsylvania. At a median follow-up of 10.3 months, 16 patients (3.6%) had a secondary primary malignancy. The median onset time was 26.4 and 9.7 months for solid and hematological malignancies, respectively. The projected 5-year cumulative incidence is 15.2% for solid and 2.3% for hematological malignancies. Overall, one case of TCL was observed, suggesting a low risk of TCL after CAR T.


Sujet(s)
Tumeurs hématologiques , Tumeurs du poumon , Lymphome B , Lymphome T , Récepteurs chimériques pour l'antigène , Humains , Immunothérapie adoptive/effets indésirables , Récepteurs chimériques pour l'antigène/génétique , Récepteurs aux antigènes des cellules T/génétique , Antigènes CD19
12.
Blood ; 143(2): 139-151, 2024 Jan 11.
Article de Anglais | MEDLINE | ID: mdl-37616575

RÉSUMÉ

ABSTRACT: Patients with multiple myeloma (MM) treated with B-cell maturation antigen (BCMA)-specific chimeric antigen receptor (CAR) T cells usually relapse with BCMA+ disease, indicative of CAR T-cell suppression. CD200 is an immune checkpoint that is overexpressed on aberrant plasma cells (aPCs) in MM and is an independent negative prognostic factor for survival. However, CD200 is not present on MM cell lines, a potential limitation of current preclinical models. We engineered MM cell lines to express CD200 at levels equivalent to those found on aPCs in MM and show that these are sufficient to suppress clinical-stage CAR T-cells targeting BCMA or the Tn glycoform of mucin 1 (TnMUC1), costimulated by 4-1BB and CD2, respectively. To prevent CD200-mediated suppression of CAR T cells, we compared CRISPR-Cas9-mediated knockout of the CD200 receptor (CD200RKO), to coexpression of versions of the CD200 receptor that were nonsignaling, that is, dominant negative (CD200RDN), or that leveraged the CD200 signal to provide CD28 costimulation (CD200R-CD28 switch). We found that the CD200R-CD28 switch potently enhanced the polyfunctionality of CAR T cells, and improved cytotoxicity, proliferative capacity, CAR T-cell metabolism, and performance in a chronic antigen exposure assay. CD200RDN provided modest benefits, but surprisingly, the CD200RKO was detrimental to CAR T-cell activity, adversely affecting CAR T-cell metabolism. These patterns held up in murine xenograft models of plasmacytoma, and disseminated bone marrow predominant disease. Our findings underscore the importance of CD200-mediated immune suppression in CAR T-cell therapy of MM, and highlight a promising approach to enhance such therapies by leveraging CD200 expression on aPCs to provide costimulation via a CD200R-CD28 switch.


Sujet(s)
Immunothérapie adoptive , Myélome multiple , Humains , Souris , Animaux , Myélome multiple/métabolisme , Antigène CD28/métabolisme , Lymphocytes T , Antigène de maturation des cellules B/métabolisme , Récidive tumorale locale/métabolisme
13.
Transplant Cell Ther ; 30(2): 171-186, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-37866783

RÉSUMÉ

Chimeric antigen receptor T cell (CAR-T) immunotherapy has revolutionized the treatment of relapsed and refractory B cell-derived hematologic malignancies. Currently, there are 6 Food and Drug Administration-approved commercial CAR-T products that target antigens exclusively expressed on malignant B cells or plasma cells. However, concurrent advancement for patients with rarer and more aggressive T cell-derived hematologic malignancies have not yet been achieved. CAR-T immunotherapies are uniquely limited by challenges related to CAR-T product manufacturing and intrinsic tumor biology. In this review tailored for practicing clinician-scientists, we discuss the major barriers of CAR-T implementation against T cell-derived neoplasms and highlight specific scientific advancements poised to circumvent these obstacles. We summarize salient early-stage clinical trials implementing novel CAR-T immunotherapies specifically for patients with relapsed and/or refractory T cell neoplasms. Finally, we highlight novel manufacturing and treatment strategies that are poised to have a meaningful future clinical impact.


Sujet(s)
Tumeurs hématologiques , Tumeurs , Récepteurs chimériques pour l'antigène , États-Unis , Humains , Lymphocytes T , Récepteurs aux antigènes des cellules T/génétique , Immunothérapie/effets indésirables , Tumeurs hématologiques/thérapie
14.
Blood Adv ; 8(3): 653-666, 2024 02 13.
Article de Anglais | MEDLINE | ID: mdl-38113468

RÉSUMÉ

ABSTRACT: Lymphodepletion (LD) is an integral component of chimeric antigen receptor T-cell (CART) immunotherapies. In this study, we compared the safety and efficacy of bendamustine (Benda) to standard fludarabine/cyclophosphamide (Flu/Cy) LD before CD19-directed, CD28-costimulated CART axicabtagene ciloleucel (axi-cel) for patients with large B-cell lymphoma (LBCL) and follicular lymphoma (FL). We analyzed 59 patients diagnosed with LBCL (n = 48) and FL (n = 11) consecutively treated with axi-cel at the University of Pennsylvania. We also analyzed serum samples for cytokine levels and metabolomic changes before and after LD. Flu/Cy and Benda demonstrated similar efficacy, with complete remission rates of 51.4% and 50.0% (P = .981), respectively, and similar progression-free and overall survivals. Any-grade cytokine-release syndrome occurred in 91.9% of patients receiving Flu/Cy vs 72.7% of patients receiving Benda (P = .048); any-grade neurotoxicity after Flu/Cy occurred in 45.9% of patients and after Benda in 18.2% of patients (P = .031). In addition, Flu/Cy was associated with a higher incidence of grade ≥3 neutropenia (100% vs 54.5%; P < .001), infections (78.4% vs 27.3%; P < .001), and neutropenic fever (78.4% vs 13.6%; P < .001). These results were confirmed both in patients with LBCL and those with FL. Mechanistically, patients with Flu/Cy had a greater increase in inflammatory cytokines associated with neurotoxicity and reduced levels of metabolites critical for redox balance and biosynthesis. This study suggests that Benda LD may be a safe alternative to Flu/Cy for CD28-based CART CD19-directed immunotherapy with similar efficacy and reduced toxicities. Benda is associated with reduced levels of inflammatory cytokines and increased anabolic metabolites.


Sujet(s)
Produits biologiques , Cytokines , Lymphome folliculaire , Humains , Chlorhydrate de bendamustine/effets indésirables , Antigène CD28 , Immunothérapie adoptive/effets indésirables , Immunothérapie adoptive/méthodes , Cyclophosphamide
15.
Mol Cancer ; 22(1): 200, 2023 12 09.
Article de Anglais | MEDLINE | ID: mdl-38066564

RÉSUMÉ

BACKGROUND: Commercial anti-CD19 chimeric antigen receptor T-cell therapies (CART19) are efficacious against advanced B-cell non-Hodgkin lymphoma (NHL); however, most patients ultimately relapse. Several mechanisms contribute to this failure, including CD19-negative escape and CAR T dysfunction. All four commercial CART19 products utilize the FMC63 single-chain variable fragment (scFv) specific to a CD19 membrane-distal epitope and characterized by slow association (on) and dissociation (off) rates. We hypothesized that a novel anti-CD19 scFv that engages an alternative CD19 membrane-proximal epitope independent of FMC63 and that is characterized by faster on- and off-rates could mitigate CART19 failure and improve clinical efficacy. METHODS: We developed an autologous CART19 product with 4-1BB co-stimulation using a novel humanized chicken antibody (h1218). This antibody is specific to a membrane-proximal CD19 epitope and harbors faster on/off rates compared to FMC63. We tested h1218-CART19 in vitro and in vivo using FMC63-CART19-resistant models. We conducted a first-in-human multi-center phase I clinical trial to test AT101 (clinical-grade h1218-CART19) in patients with relapsed or refractory (r/r) NHL. RESULTS: Preclinically, h1218- but not FMC63-CART19 were able to effectively eradicate lymphomas expressing CD19 point mutations (L174V and R163L) or co-expressing FMC63-CAR19 as found in patients relapsing after FMC63-CART19. Furthermore, h1218-CART19 exhibited enhanced killing of B-cell malignancies in vitro and in vivo compared with FMC63-CART19. Mechanistically, we found that h1218-CART19 had reduced activation-induced cell death (AICD) and enhanced expansion compared to FMC63-CART19 owing to faster on- and off-rates. Based on these preclinical results, we performed a phase I dose-escalation trial, testing three dose levels (DL) of AT101 (the GMP version of h1218) using a 3 + 3 design. In 12 treated patients (7 DLBCL, 3 FL, 1 MCL, and 1 MZL), AT101 showed a promising safety profile with 8.3% grade 3 CRS (n = 1) and 8.3% grade 4 ICANS (n = 1). In the whole cohort, the overall response rate was 91.7%, with a complete response rate of 75.0%, which improved to 100% in DL-2 and -3. AT101 expansion correlates with CR and B-cell aplasia. CONCLUSIONS: We developed a novel, safe, and potent CART19 product that recognizes a membrane-proximal domain of CD19 with fast on- and off-rates and showed significant efficacy and promising safety in patients with relapsed B-cell NHL. TRIAL REGISTRATION: NCT05338931; Date: 2022-04-01.


Sujet(s)
Lymphome malin non hodgkinien , Récepteurs aux antigènes des cellules T , Récepteurs chimériques pour l'antigène , Humains , Anticorps , Antigènes CD19 , Épitopes/métabolisme , Immunothérapie adoptive/effets indésirables , Lymphome malin non hodgkinien/thérapie , Lymphome malin non hodgkinien/métabolisme , Récidive tumorale locale/métabolisme , Récepteurs chimériques pour l'antigène/métabolisme , Récepteurs aux antigènes des cellules T/antagonistes et inhibiteurs
16.
Cancer Cell ; 41(12): 2016-2018, 2023 12 11.
Article de Anglais | MEDLINE | ID: mdl-38086334

RÉSUMÉ

Chimeric antigen receptor (CAR) T cell immunotherapy in solid cancer is severely limited by the absence of ideal targets. In this issue of Cancer Cell, Bergaggio et al. find that anaplastic lymphoma kinase (ALK) inhibitors can enhance the function of ALK-specific CAR T cells against neuroblastoma by increasing target density in cancer cells.


Sujet(s)
Neuroblastome , Récepteurs chimériques pour l'antigène , Humains , Récepteurs chimériques pour l'antigène/génétique , Neuroblastome/anatomopathologie , Immunothérapie , Inhibiteurs de protéines kinases , Récepteurs à activité tyrosine kinase/génétique , Lymphocytes T , Immunothérapie adoptive , Récepteurs aux antigènes des cellules T/génétique
17.
Nat Rev Drug Discov ; 22(12): 976-995, 2023 12.
Article de Anglais | MEDLINE | ID: mdl-37907724

RÉSUMÉ

Chimeric antigen receptor (CAR)-T cells have recently emerged as a powerful therapeutic approach for the treatment of patients with chemotherapy-refractory or relapsed blood cancers, including acute lymphoblastic leukaemia, diffuse large B cell lymphoma, follicular lymphoma, mantle cell lymphoma and multiple myeloma. Nevertheless, resistance to CAR-T cell therapies occurs in most patients. In this Review, we summarize the resistance mechanisms to CAR-T cell immunotherapy by analysing CAR-T cell dysfunction, intrinsic tumour resistance and the immunosuppressive tumour microenvironment. We discuss current research strategies to overcome multiple resistance mechanisms, including optimization of the CAR design, improvement of in vivo T cell function and persistence, modulation of the immunosuppressive tumour microenvironment and synergistic combination strategies.


Sujet(s)
Tumeurs hématologiques , Récepteurs chimériques pour l'antigène , Humains , Récidive tumorale locale/étiologie , Lymphocytes T , Tumeurs hématologiques/thérapie , Immunothérapie adoptive , Récepteurs aux antigènes des cellules T , Microenvironnement tumoral
18.
Nat Commun ; 14(1): 7767, 2023 Nov 27.
Article de Anglais | MEDLINE | ID: mdl-38012187

RÉSUMÉ

Chimeric antigen receptor (CAR) T cell therapy is effective in treating B cell malignancies, but factors influencing the persistence of functional CAR+ T cells, such as product composition, patients' lymphodepletion, and immune reconstitution, are not well understood. To shed light on this issue, here we conduct a single-cell multi-omics analysis of transcriptional, clonal, and phenotypic profiles from pre- to 1-month post-infusion of CAR+ and CAR- T cells from patients from a CARTELL study (ACTRN12617001579381) who received a donor-derived 4-1BB CAR product targeting CD19. Following infusion, CAR+ T cells and CAR- T cells shows similar differentiation profiles with clonally expanded populations across heterogeneous phenotypes, demonstrating clonal lineages and phenotypic plasticity. We validate these findings in 31 patients with large B cell lymphoma treated with CD19 CAR T therapy. For these patients, we identify using longitudinal mass-cytometry data an association between NK-like subsets and clinical outcomes at 6 months with both CAR+ and CAR- T cells. These results suggest that non-CAR-derived signals can provide information about patients' immune recovery and be used as correlate of clinically relevant parameters.


Sujet(s)
Lymphome B diffus à grandes cellules , Récepteurs aux antigènes des cellules T , Humains , Lymphocytes B , Immunothérapie adoptive/méthodes , Lymphome B diffus à grandes cellules/anatomopathologie , Lymphocytes T
19.
Blood ; 142(20): 1724-1739, 2023 11 16.
Article de Anglais | MEDLINE | ID: mdl-37683180

RÉSUMÉ

Aberrant skipping of coding exons in CD19 and CD22 compromises the response to immunotherapy in B-cell malignancies. Here, we showed that the MS4A1 gene encoding human CD20 also produces several messenger RNA (mRNA) isoforms with distinct 5' untranslated regions. Four variants (V1-4) were detected using RNA sequencing (RNA-seq) at distinct stages of normal B-cell differentiation and B-lymphoid malignancies, with V1 and V3 being the most abundant. During B-cell activation and Epstein-Barr virus infection, redirection of splicing from V1 to V3 coincided with increased CD20 positivity. Similarly, in diffuse large B-cell lymphoma, only V3, but not V1, correlated with CD20 protein levels, suggesting that V1 might be translation-deficient. Indeed, the longer V1 isoform contained upstream open reading frames and a stem-loop structure, which cooperatively inhibited polysome recruitment. By modulating CD20 isoforms with splice-switching morpholino oligomers, we enhanced CD20 expression and anti-CD20 antibody rituximab-mediated cytotoxicity in a panel of B-cell lines. Furthermore, reconstitution of CD20-knockout cells with V3 mRNA led to the recovery of CD20 positivity, whereas V1-reconstituted cells had undetectable levels of CD20 protein. Surprisingly, in vitro CD20-directed chimeric antigen receptor T cells were able to kill both V3- and V1-expressing cells, but the bispecific T-cell engager mosunetuzumab was only effective against V3-expressing cells. To determine whether CD20 splicing is involved in immunotherapy resistance, we performed RNA-seq on 4 postmosunetuzumab follicular lymphoma relapses and discovered that in 2 of them, the downregulation of CD20 was accompanied by a V3-to-V1 shift. Thus, splicing-mediated mechanisms of epitope loss extend to CD20-directed immunotherapies.


Sujet(s)
Infections à virus Epstein-Barr , Tumeurs , Humains , Épissage alternatif , ARN messager/génétique , Régions 5' non traduites , Infections à virus Epstein-Barr/génétique , Herpèsvirus humain de type 4/génétique , Antigènes CD20/génétique , Isoformes de protéines/génétique , Immunothérapie , Biosynthèse des protéines , Tumeurs/génétique
20.
Immunity ; 56(10): 2388-2407.e9, 2023 Oct 10.
Article de Anglais | MEDLINE | ID: mdl-37776850

RÉSUMÉ

Chimeric antigen receptor (CAR) T cell therapy targeting CD19 has achieved tremendous success treating B cell malignancies; however, some patients fail to respond due to poor autologous T cell fitness. To improve response rates, we investigated whether disruption of the co-inhibitory receptors CTLA4 or PD-1 could restore CART function. CRISPR-Cas9-mediated deletion of CTLA4 in preclinical models of leukemia and myeloma improved CAR T cell proliferation and anti-tumor efficacy. Importantly, this effect was specific to CTLA4 and not seen upon deletion of CTLA4 and/or PDCD1 in CAR T cells. Mechanistically, CTLA4 deficiency permitted unopposed CD28 signaling and maintenance of CAR expression on the T cell surface under conditions of high antigen load. In clinical studies, deletion of CTLA4 rescued the function of T cells from patients with leukemia that previously failed CAR T cell treatment. Thus, selective deletion of CTLA4 reinvigorates dysfunctional chronic lymphocytic leukemia (CLL) patient T cells, providing a strategy for increasing patient responses to CAR T cell therapy.


Sujet(s)
Leucémie chronique lymphocytaire à cellules B , Récepteurs chimériques pour l'antigène , Humains , Récepteurs aux antigènes des cellules T/métabolisme , Antigène CTLA-4/génétique , Antigène CTLA-4/métabolisme , Lymphocytes T , Immunothérapie adoptive , Antigènes CD19
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...