Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 108
Filtrer
1.
Nat Cancer ; 5(7): 1010-1023, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38750245

RÉSUMÉ

Chimeric antigen receptor (CAR) T cells used for the treatment of B cell malignancies can identify T cell subsets with superior clinical activity. Here, using infusion products of individuals with large B cell lymphoma, we integrated functional profiling using timelapse imaging microscopy in nanowell grids with subcellular profiling and single-cell RNA sequencing to identify a signature of multifunctional CD8+ T cells (CD8-fit T cells). CD8-fit T cells are capable of migration and serial killing and harbor balanced mitochondrial and lysosomal volumes. Using independent datasets, we validate that CD8-fit T cells (1) are present premanufacture and are associated with clinical responses in individuals treated with axicabtagene ciloleucel, (2) longitudinally persist in individuals after treatment with CAR T cells and (3) are tumor migrating cytolytic cells capable of intratumoral expansion in solid tumors. Our results demonstrate the power of multimodal integration of single-cell functional assessments for the discovery and application of CD8-fit T cells as a T cell subset with optimal fitness in cell therapy.


Sujet(s)
Lymphocytes T CD8+ , Immunothérapie adoptive , Lymphome B diffus à grandes cellules , Récepteurs chimériques pour l'antigène , Analyse sur cellule unique , Humains , Lymphome B diffus à grandes cellules/immunologie , Lymphome B diffus à grandes cellules/thérapie , Analyse sur cellule unique/méthodes , Immunothérapie adoptive/méthodes , Lymphocytes T CD8+/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Sous-populations de lymphocytes T/immunologie , Produits biologiques
3.
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
4.
J Immunother Cancer ; 12(2)2024 Feb 12.
Article de Anglais | MEDLINE | ID: mdl-38350684

RÉSUMÉ

BACKGROUND: Chimeric antigen receptor (CAR)-T cells are approved for use in the treatment of hematological malignancies. Axicabtagene ciloleucel (YESCARTA) and brexucabtagene autoleucel (TECARTUS) genetically modified autologous T cells expressing an anti-CD19 scFv based on the FMC63 clone have shown impressive response rates for the treatment of CD19+B cell malignancies, but there remain challenges in monitoring long-term persistence as well as the functional characterization of low-level persisting CAR-T cells in patients. Furthermore, due to CD19-negative driven relapse, having the capability to monitor patients with simultaneous detection of the B cell malignancy and persisting CAR-T cells in patient peripheral blood is important for ensuring timely treatment optionality and understanding relapse. METHODS: This study demonstrates the development and technical validation of a comprehensive liquid biopsy, high-definition single cell assay (HDSCA)-HemeCAR for (1) KTE-X19 CAR-T cell identification and analysis and (2) simultaneously monitoring the CD19-epitope landscape on neoplastic B cells in cryopreserved or fresh peripheral blood. Proprietary anti-CD19 CAR reagents, healthy donor transduced CAR-T cells, and patient samples consisting of malignant B cell fractions from manufacturing were used for assay development. RESULTS: The CAR-T assay showed an approximate limit of detection at 1 cell in 3 million with a sensitivity of 91%. Genomic analysis was additionally used to confirm the presence of the CAR transgene. This study additionally reports the successful completion of two B cell assays with multiple CD19 variants (FMC63 and LE-CD19) and a unique fourth channel biomarker (CD20 or CD22). In patient samples, we observed that CD19 isoforms were highly heterogeneous both intrapatient and interpatient. CONCLUSIONS: With the simultaneous detection of the CAR-T cells and the B cell malignancy in patient peripheral blood, the HDSCA-HemeCAR workflow may be considered for risk monitoring and patient management.


Sujet(s)
Tumeurs , Récepteurs chimériques pour l'antigène , Humains , Récepteurs chimériques pour l'antigène/génétique , Récepteurs aux antigènes des cellules T/génétique , Récidive , Antigènes CD19 , Thérapie cellulaire et tissulaire
5.
Nat Med ; 30(2): 507-518, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38233586

RÉSUMÉ

The phase 3 ZUMA-7 trial in second-line large B cell lymphoma demonstrated superiority of anti-CD19 CAR T cell therapy (axicabtagene ciloleucel (axi-cel)) over standard of care (SOC; salvage chemotherapy followed by hematopoietic transplantation) ( NCT03391466 ). Here, we present a prespecified exploratory analysis examining the association between pretreatment tumor characteristics and the efficacy of axi-cel versus SOC. B cell gene expression signature (GES) and CD19 expression associated significantly with improved event-free survival for axi-cel (P = 0.0002 for B cell GES; P = 0.0165 for CD19 expression) but not SOC (P = 0.9374 for B cell GES; P = 0.5526 for CD19 expression). Axi-cel showed superior event-free survival over SOC irrespective of B cell GES and CD19 expression (P = 8.56 × 10-9 for B cell GES high; P = 0.0019 for B cell GES low; P = 3.85 × 10-9 for CD19 gene high; P = 0.0017 for CD19 gene low). Low CD19 expression in malignant cells correlated with a tumor GES consisting of immune-suppressive stromal and myeloid genes, highlighting the inter-relation between malignant cell features and immune contexture substantially impacting axi-cel outcomes. Tumor burden, lactate dehydrogenase and cell-of-origin impacted SOC more than axi-cel outcomes. T cell activation and B cell GES, which are associated with improved axi-cel outcome, decreased with increasing lines of therapy. These data highlight differences in resistance mechanisms to axi-cel and SOC and support earlier intervention with axi-cel.


Sujet(s)
Produits biologiques , Lymphome B diffus à grandes cellules , Humains , Immunothérapie adoptive , Microenvironnement tumoral , Lymphome B diffus à grandes cellules/génétique , Lymphome B diffus à grandes cellules/thérapie , Lymphocytes B , Protéines adaptatrices de la transduction du signal , Antigènes CD19
6.
Blood Cancer Discov ; 5(1): 21-33, 2024 01 08.
Article de Anglais | MEDLINE | ID: mdl-37983485

RÉSUMÉ

Treatment resistance and toxicities remain a risk following chimeric antigen receptor (CAR) T-cell therapy. Herein, we report pharmacokinetics, pharmacodynamics, and product and apheresis attributes associated with outcomes among patients with relapsed/refractory large B-cell lymphoma (LBCL) treated with axicabtagene ciloleucel (axi-cel) in ZUMA-7. Axi-cel peak expansion associated with clinical response and toxicity, but not response durability. In apheresis material and final product, a naive T-cell phenotype (CCR7+CD45RA+) expressing CD27 and CD28 associated with improved response durability, event-free survival, progression-free survival, and a lower number of prior therapies. This phenotype was not associated with high-grade cytokine release syndrome (CRS) or neurologic events. Higher baseline and postinfusion levels of serum inflammatory markers associated with differentiated/effector products, reduced efficacy, and increased CRS and neurologic events, thus suggesting targets for intervention. These data support better outcomes with earlier CAR T-cell intervention and may improve patient care by informing on predictive biomarkers and development of next-generation products. SIGNIFICANCE: In ZUMA-7, the largest randomized CAR T-cell trial in LBCL, a naive T-cell product phenotype (CCR7+CD45RA+) expressing CD27 and CD28 associated with improved efficacy, decreased toxicity, and a lower number of prior therapies, supporting earlier intervention with CAR T-cell therapy. In addition, targets for improvement of therapeutic index are proposed. This article is featured in Selected Articles from This Issue, p. 4.


Sujet(s)
Immunothérapie adoptive , Lymphome B diffus à grandes cellules , Humains , Immunothérapie adoptive/effets indésirables , Antigène CD28 , Récepteurs CCR7 , Lymphome B diffus à grandes cellules/thérapie , Personnel de recherche , Syndrome de libération de cytokines , Antigènes CD45
7.
Blood ; 141(19): 2307-2315, 2023 05 11.
Article de Anglais | MEDLINE | ID: mdl-36821768

RÉSUMÉ

In phase 2 of ZUMA-1, a single-arm, multicenter, registrational trial, axicabtagene ciloleucel (axi-cel) autologous anti-CD19 chimeric antigen receptor (CAR) T-cell therapy demonstrated durable responses at 2 years in patients with refractory large B-cell lymphoma (LBCL). Here, we assessed outcomes in ZUMA-1 after 5 years of follow-up. Eligible adults received lymphodepleting chemotherapy followed by axi-cel (2 × 106 cells per kg). Investigator-assessed response, survival, safety, and pharmacokinetics were assessed in patients who had received treatment. The objective response rate in these 101 patients was 83% (58% complete response rate); with a median follow-up of 63.1 months, responses were ongoing in 31% of patients at data cutoff. Median overall survival (OS) was 25.8 months, and the estimated 5-year OS rate was 42.6%. Disease-specific survival (excluding deaths unrelated to disease progression) estimated at 5 years was 51.0%. No new serious adverse events or deaths related to axi-cel were observed after additional follow-up. Peripheral blood B cells were detectable in all evaluable patients at 3 years with polyclonal B-cell recovery in 91% of patients. Ongoing responses at 60 months were associated with early CAR T-cell expansion. In conclusion, this 5-year follow-up analysis of ZUMA-1 demonstrates sustained overall and disease-specific survival, with no new safety signals in patients with refractory LBCL. Protracted B-cell aplasia was not required for durable responses. These findings support the curative potential of axi-cel in a subset of patients with aggressive B-cell lymphomas. This trial was registered at ClinicalTrials.gov, as #NCT02348216.


Sujet(s)
Produits biologiques , Lymphome B diffus à grandes cellules , Récepteurs chimériques pour l'antigène , Adulte , Humains , Études de suivi , Immunothérapie adoptive/effets indésirables , Lymphome B diffus à grandes cellules/anatomopathologie , Antigènes CD19/usage thérapeutique
8.
Nat Med ; 28(9): 1872-1882, 2022 09.
Article de Anglais | MEDLINE | ID: mdl-36038629

RÉSUMÉ

Axicabtagene ciloleucel (axi-cel) is an anti-CD19 chimeric antigen receptor (CAR) T cell therapy approved for relapsed/refractory large B cell lymphoma (LBCL) and has treatment with similar efficacy across conventional LBCL subtypes. Toward patient stratification, we assessed whether tumor immune contexture influenced clinical outcomes after axi-cel. We evaluated the tumor microenvironment (TME) of 135 pre-treatment and post-treatment tumor biopsies taken from 51 patients in the ZUMA-1 phase 2 trial. We uncovered dynamic patterns that occurred within 2 weeks after axi-cel. The biological associations among Immunoscore (quantification of tumor-infiltrating T cell density), Immunosign 21 (expression of pre-defined immune gene panel) and cell subsets were validated in three independent LBCL datasets. In the ZUMA-1 trial samples, clinical response and overall survival were associated with pre-treatment immune contexture as characterized by Immunoscore and Immunosign 21. Circulating CAR T cell levels were associated with post-treatment TME T cell exhaustion. TME enriched for chemokines (CCL5 and CCL22), γ-chain receptor cytokines (IL-15, IL-7 and IL-21) and interferon-regulated molecules were associated with T cell infiltration and markers of activity. Finally, high density of regulatory T cells in pre-treatment TME associated with reduced axi-cel-related neurologic toxicity. These findings advance the understanding of LBCL TME characteristics associated with clinical responses to anti-CD19 CAR T cell therapy and could foster biomarker development and treatment optimization for patients with LBCL.


Sujet(s)
Produits biologiques , Lymphome B diffus à grandes cellules , Récepteurs chimériques pour l'antigène , Antigènes CD19 , Numération cellulaire , Humains , Immunothérapie adoptive/effets indésirables , Interférons/usage thérapeutique , Interleukine-15 , Interleukine-7/usage thérapeutique , Lymphome B diffus à grandes cellules/thérapie , Récepteurs chimériques pour l'antigène/génétique , Récepteurs chimériques pour l'antigène/usage thérapeutique , Microenvironnement tumoral
9.
J Clin Invest ; 132(17)2022 09 01.
Article de Anglais | MEDLINE | ID: mdl-35881486

RÉSUMÉ

The in vivo persistence of adoptively transferred T cells is predictive of antitumor response. Identifying functional properties of infused T cells that lead to in vivo persistence and tumor eradication has remained elusive. We profiled CD19-specific chimeric antigen receptor (CAR) T cells as the infusion products used to treat large B cell lymphomas using high-throughput single-cell technologies based on time-lapse imaging microscopy in nanowell grids (TIMING), which integrates killing, cytokine secretion, and transcriptional profiling. Our results show that the directional migration of CD19-specific CAR T cells is correlated with multifunctionality. We showed that CD2 on T cells is associated with directional migration and that the interaction between CD2 on T cells and CD58 on lymphoma cells accelerates killing and serial killing. Consistent with this, we observed that elevated CD58 expression on pretreatment tumor samples in patients with relapsed or refractory large B cell lymphomas treated with CD19-specific CAR T cell therapy was associated with complete clinical response and survival. These results highlight the importance of studying dynamic T cell-tumor cell interactions in identifying optimal antitumor responses.


Sujet(s)
Antigènes CD2/métabolisme , Antigènes CD58/métabolisme , Lymphome B diffus à grandes cellules , Lymphocytes T , Antigènes CD19 , Humains , Immunothérapie adoptive/méthodes , Lymphome B diffus à grandes cellules/génétique , Lymphome B diffus à grandes cellules/thérapie , Récepteurs aux antigènes des cellules T , Analyse sur cellule unique
11.
Br J Haematol ; 195(3): 388-398, 2021 11.
Article de Anglais | MEDLINE | ID: mdl-34590303

RÉSUMÉ

Axicabtagene ciloleucel (axi-cel) is an autologous anti-CD19 chimeric antigen receptor (CAR) T-cell therapy approved for relapsed or refractory large B-cell lymphoma (R/R LBCL). To reduce axi-cel-related toxicity, several exploratory safety management cohorts were added to ZUMA-1 (NCT02348216), the pivotal phase 1/2 study of axi-cel in refractory LBCL. Cohort 4 evaluated the rates and severity of cytokine release syndrome (CRS) and neurologic events (NEs) with earlier corticosteroid and tocilizumab use. Primary endpoints were incidence and severity of CRS and NEs. Patients received 2 × 106 anti-CD19 CAR T cells/kg after conditioning chemotherapy. Forty-one patients received axi-cel. Incidences of any-grade CRS and NEs were 93% and 61%, respectively (grade ≥ 3, 2% and 17%). There was no grade 4 or 5 CRS or NE. Despite earlier dosing, the cumulative cortisone-equivalent corticosteroid dose in patients requiring corticosteroid therapy was lower than that reported in the pivotal ZUMA-1 cohorts. With a median follow-up of 14·8 months, objective and complete response rates were 73% and 51%, respectively, and 51% of treated patients were in ongoing response. Earlier and measured use of corticosteroids and/or tocilizumab has the potential to reduce the incidence of grade ≥ 3 CRS and NEs in patients with R/R LBCL receiving axi-cel.


Sujet(s)
Hormones corticosurrénaliennes/usage thérapeutique , Produits biologiques/effets indésirables , Syndrome de libération de cytokines/prévention et contrôle , Immunothérapie adoptive/effets indésirables , Lymphome B diffus à grandes cellules/traitement médicamenteux , Maladies du système nerveux/prévention et contrôle , Adulte , Sujet âgé , Anticorps monoclonaux humanisés/usage thérapeutique , Produits biologiques/usage thérapeutique , Marqueurs biologiques , Cyclophosphamide/usage thérapeutique , Syndrome de libération de cytokines/induit chimiquement , Association de médicaments , Femelle , Humains , Leucaphérèse , Lévétiracétam/usage thérapeutique , Mâle , Adulte d'âge moyen , Maladies du système nerveux/induit chimiquement , Neutropénie/induit chimiquement , Score de propension , Vidarabine/analogues et dérivés , Vidarabine/usage thérapeutique , Jeune adulte
13.
Blood ; 138(1): 11-22, 2021 07 08.
Article de Anglais | MEDLINE | ID: mdl-33827116

RÉSUMÉ

ZUMA-3 is a phase 1/2 study evaluating KTE-X19, an autologous anti-CD19 chimeric antigen receptor (CAR) T-cell therapy, in adult relapsed/refractory (R/R) B-cell acute lymphoblastic leukemia (B-ALL). We report the phase 1 results. After fludarabine-cyclophosphamide lymphodepletion, patients received a single infusion of KTE-X19 at 2 × 106, 1 × 106, or 0.5 × 106 cells per kg. The rate of dose-limiting toxicities (DLTs) within 28 days after KTE-X19 infusion was the primary end point. KTE-X19 was manufactured for 54 enrolled patients and administered to 45 (median age, 46 years; range, 18-77 years). No DLTs occurred in the DLT-evaluable cohort. Grade ≥3 cytokine release syndrome (CRS) and neurologic events (NEs) occurred in 31% and 38% of patients, respectively. To optimize the risk-benefit ratio, revised adverse event (AE) management for CRS and NEs (earlier steroid use for NEs and tocilizumab only for CRS) was evaluated at 1 × 106 cells per kg KTE-X19. In the 9 patients treated under revised AE management, 33% had grade 3 CRS and 11% had grade 3 NEs, with no grade 4 or 5 NEs. The overall complete remission rate correlated with CAR T-cell expansion and was 83% in patients treated with 1 × 106 cells per kg and 69% in all patients. Minimal residual disease was undetectable in all responding patients. At a median follow-up of 22.1 months (range, 7.1-36.1 months), the median duration of remission was 17.6 months (95% confidence interval [CI], 5.8-17.6 months) in patients treated with 1 × 106 cells per kg and 14.5 months (95% CI, 5.8-18.1 months) in all patients. KTE-X19 treatment provided a high response rate and tolerable safety in adults with R/R B-ALL. Phase 2 is ongoing at 1 × 106 cells per kg with revised AE management. This trial is registered at www.clinicaltrials.gov as #NCT02614066.


Sujet(s)
Antigènes CD19/métabolisme , Immunothérapie adoptive , Leucémie-lymphome lymphoblastique à précurseurs B et T/traitement médicamenteux , Récepteurs chimériques pour l'antigène/usage thérapeutique , Adolescent , Adulte , Sujet âgé , Marqueurs biologiques tumoraux/métabolisme , Prolifération cellulaire , Syndrome de libération de cytokines/induit chimiquement , Survie sans rechute , Relation dose-effet des médicaments , Femelle , Humains , Immunothérapie adoptive/effets indésirables , Médiateurs de l'inflammation/métabolisme , Mâle , Adulte d'âge moyen , Maladie résiduelle/anatomopathologie , Résultat thérapeutique , Jeune adulte
14.
Nat Med ; 27(3): 419-425, 2021 03.
Article de Anglais | MEDLINE | ID: mdl-33558725

RÉSUMÉ

Genetically engineered T cell therapy can induce remarkable tumor responses in hematologic malignancies. However, it is not known if this type of therapy can be applied effectively to epithelial cancers, which account for 80-90% of human malignancies. We have conducted a first-in-human, phase 1 clinical trial of T cells engineered with a T cell receptor targeting HPV-16 E7 for the treatment of metastatic human papilloma virus-associated epithelial cancers (NCT02858310). The primary endpoint was maximum tolerated dose. Cell dose was not limited by toxicity with a maximum dose of 1 × 1011 engineered T cells administered. Tumor responses following treatment were evaluated using RECIST (Response Evaluation Criteria in Solid Tumors) guidelines. Robust tumor regression was observed with objective clinical responses in 6 of 12 patients, including 4 of 8 patients with anti-PD-1 refractory disease. Responses included extensive regression of bulky tumors and complete regression of most tumors in some patients. Genomic studies, which included intra-patient tumors with dichotomous treatment responses, revealed resistance mechanisms from defects in critical components of the antigen presentation and interferon response pathways. These findings demonstrate that engineered T cells can mediate regression of common carcinomas, and they reveal immune editing as a constraint on the curative potential of cellular therapy and possibly other immunotherapies in advanced epithelial cancer.


Sujet(s)
Tumeurs épithéliales épidermoïdes et glandulaires/anatomopathologie , Papillomaviridae/métabolisme , Protéines E7 de papillomavirus/métabolisme , Infections à papillomavirus/métabolisme , Récepteurs aux antigènes des cellules T/métabolisme , Lymphocytes T/métabolisme , Lignée cellulaire tumorale , Humains , Métastase tumorale , Tumeurs épithéliales épidermoïdes et glandulaires/métabolisme , Tumeurs épithéliales épidermoïdes et glandulaires/virologie
15.
Cancer Discov ; 11(3): 599-613, 2021 03.
Article de Anglais | MEDLINE | ID: mdl-33334730

RÉSUMÉ

T cell-based therapies have induced cancer remissions, though most tumors ultimately progress, reflecting inherent or acquired resistance including antigen escape. Better understanding of how T cells eliminate tumors will help decipher resistance mechanisms. We used a CRISPR/Cas9 screen and identified a necessary role for Fas-FasL in antigen-specific T-cell killing. We also found that Fas-FasL mediated off-target "bystander" killing of antigen-negative tumor cells. This localized bystander cytotoxicity enhanced clearance of antigen-heterogeneous tumors in vivo, a finding that has not been shown previously. Fas-mediated on-target and bystander killing was reproduced in chimeric antigen receptor (CAR-T) and bispecific antibody T-cell models and was augmented by inhibiting regulators of Fas signaling. Tumoral FAS expression alone predicted survival of CAR-T-treated patients in a large clinical trial (NCT02348216). These data suggest strategies to prevent immune escape by targeting both the antigen expression of most tumor cells and the geography of antigen-loss variants. SIGNIFICANCE: This study demonstrates the first report of in vivo Fas-dependent bystander killing of antigen-negative tumors by T cells, a phenomenon that may be contributing to the high response rates of antigen-directed immunotherapies despite tumoral heterogeneity. Small molecules that target the Fas pathway may potentiate this mechanism to prevent cancer relapse.This article is highlighted in the In This Issue feature, p. 521.


Sujet(s)
Cytotoxicité immunologique , Immunothérapie , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Antigènes CD95/métabolisme , Animaux , Antigènes néoplasiques/immunologie , Effet bystander/immunologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Systèmes CRISPR-Cas , Modèles animaux de maladie humaine , Édition de gène , Génie génétique , Humains , Immunothérapie/effets indésirables , Immunothérapie/méthodes , Immunothérapie adoptive/effets indésirables , Immunothérapie adoptive/méthodes , Souris , Souris knockout , Tumeurs/étiologie , Tumeurs/thérapie , Récepteurs chimériques pour l'antigène , Spécificité antigénique des récepteurs des lymphocytes T , Résultat thérapeutique , Tests d'activité antitumorale sur modèle de xénogreffe
16.
Blood Adv ; 4(19): 4898-4911, 2020 10 13.
Article de Anglais | MEDLINE | ID: mdl-33035333

RÉSUMÉ

ZUMA-1 demonstrated a high rate of durable response and a manageable safety profile with axicabtagene ciloleucel (axi-cel), an anti-CD19 chimeric antigen receptor (CAR) T-cell therapy, in patients with refractory large B-cell lymphoma. As previously reported, prespecified clinical covariates for secondary end point analysis were not clearly predictive of efficacy; these included Eastern Cooperative Oncology Group performance status (0 vs 1), age, disease subtype, disease stage, and International Prognostic Index score. We interrogated covariates included in the statistical analysis plan and an extensive panel of biomarkers according to an expanded translational biomarker plan. Univariable and multivariable analyses indicated that rapid CAR T-cell expansion commensurate with pretreatment tumor burden (influenced by product T-cell fitness), the number of CD8 and CCR7+CD45RA+ T cells infused, and host systemic inflammation, were the most significant determining factors for durable response. Key parameters differentially associated with clinical efficacy and toxicities, with both theoretical and practical implications for optimizing CAR T-cell therapy. This trial was registered at www.clinicaltrials.gov as #NCT02348216.


Sujet(s)
Antigènes CD19 , Immunothérapie adoptive , Antigènes CD19/usage thérapeutique , Produits biologiques , Humains , Inflammation , Charge tumorale
17.
J Immunother ; 43(9): 273-282, 2020.
Article de Anglais | MEDLINE | ID: mdl-32925563

RÉSUMÉ

Expression of carbonic-anhydrase IX (CAIX) in clear cell renal cell carcinoma (RCC) makes it an attractive vaccine target. We developed a fusion-gene construct, granulocyte-macrophage (GM) colony-stimulating factor+CAIX, delivered by an adenoviral vector (Ad) into autologous dendritic cells (DCs) in this phase 1 study. The injected immature DCs were expected to stimulate an antigen-specific immune response against CAIX expressing RCC. Three dose-escalation cohorts (5, 15, and 50×10 cells/administration) were injected intradermally q2wk×3 doses based on a 3+3 design. The primary objective was the safety of the injections. Secondary objectives were immune responses using enzyme-linked immunosorbent spot, a serum biomarker panel, and clinical response. Fifteen patients with metastatic RCC were enrolled, and 9 patients received all 3 doses. No serious adverse events were seen. There were 3 (33%) patients with grade 1 fatigue, 1 of whom subsequently experienced grade 2 fatigue. One patient (11%) experienced grade 1-2 leukopenia. Only 1 patient (11%) experienced grade 2 flu-like symptoms. Of the 9 patients who received treatment, 1 expired of progressive disease, 2 patients were lost to follow-up and 6 patients are alive. Of the 6 patients, 5 have progressive disease, and 1 has completed treatment with stable disease at 27 months follow-up. Immune response measurements appeared more robust in higher dose cohorts, which appeared to be related to patients with stable disease at 3 months. These early data show that autologous immature DC-AdGMCAIX can be safely given to metastatic RCC patients without any serious adverse events with CAIX-specific immune response elicited by the treatment. These preliminary data support further study of Ad-GMCAIX, particularly with combination therapies that may enhance clinical activity.


Sujet(s)
Antigènes néoplasiques/génétique , Vaccins anticancéreux/administration et posologie , Carbonic anhydrase IX/génétique , Néphrocarcinome/thérapie , Cellules dendritiques/immunologie , Facteur de stimulation des colonies de granulocytes et de macrophages/génétique , Tumeurs du rein/thérapie , Antigènes néoplasiques/immunologie , Vaccins anticancéreux/effets indésirables , Vaccins anticancéreux/génétique , Vaccins anticancéreux/métabolisme , Carbonic anhydrase IX/immunologie , Néphrocarcinome/immunologie , Néphrocarcinome/anatomopathologie , Cellules dendritiques/métabolisme , Prise en charge de la maladie , Facteur de stimulation des colonies de granulocytes et de macrophages/métabolisme , Humains , Immunothérapie/effets indésirables , Immunothérapie/méthodes , Tumeurs du rein/immunologie , Tumeurs du rein/anatomopathologie , Résultat thérapeutique
18.
JCI Insight ; 5(12)2020 06 18.
Article de Anglais | MEDLINE | ID: mdl-32484797

RÉSUMÉ

Mechanisms of chimeric antigen receptor (CAR) T cell-mediated antitumor immunity and toxicity remain poorly characterized because few studies examine the intact tumor microenvironment (TME) following CAR T cell infusion. Axicabtagene ciloleucel is an autologous anti-CD19 CAR T cell therapy approved for patients with large B cell lymphoma. We devised multiplex immunostaining and ISH assays to interrogate CAR T cells and other immune cell infiltrates in biopsies of diffuse large B cell lymphoma following axicabtagene ciloleucel infusion. We found that a majority of intratumoral CAR T cells expressed markers of T cell activation but, unexpectedly, constituted ≤5% of all T cells within the TME 5 days or more after therapy. Large numbers of T cells without CAR were also activated within the TME after axicabtagene ciloleucel infusion; these cells were positive for Ki-67, IFN-γ, granzyme B (GzmB), and/or PD-1 and were found at the highest levels in biopsies with CAR T cells. Additionally, non-CAR immune cells were the exclusive source of IL-6, a cytokine associated with cytokine release syndrome, and were found at their highest numbers in biopsies with CAR T cells. These data suggest that intratumoral CAR T cells are associated with non-CAR immune cell activation within the TME with both beneficial and pathological effects.


Sujet(s)
Antigènes CD19/usage thérapeutique , Lymphome B diffus à grandes cellules/thérapie , Récepteurs aux antigènes des cellules T/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Microenvironnement tumoral/immunologie , Antigènes CD19/immunologie , Produits biologiques , Marqueurs biologiques/analyse , Humains , Immunothérapie adoptive/méthodes , Lymphome B diffus à grandes cellules/anatomopathologie , Lymphocytes T/immunologie
20.
Proteomics ; 20(13): e1900270, 2020 07.
Article de Anglais | MEDLINE | ID: mdl-32108428

RÉSUMÉ

Highly multiplexed single-cell functional proteomics has emerged as one of the next-generation toolkits for a deeper understanding of functional heterogeneity in cell. Different from the conventional population-based bulk and single-cell RNA-Seq assays, the microchip-based proteomics at the single-cell resolution enables a unique identification of highly polyfunctional cell subsets that co-secrete many proteins from live single cells and most importantly correlate with patient response to a therapy. The 32-plex IsoCode chip technology has defined a polyfunctional strength index (PSI) of pre-infusion anti-CD19 chimeric antigen receptor (CAR)-T products, that is significantly associated with patient response to the CAR-T cell therapy. To complement the clinical relevance of the PSI, a comprehensive visualization toolkit of 3D uniform manifold approximation and projection (UMAP) and t-distributed stochastic neighbor embedding (t-SNE) in a proteomic analysis pipeline is developed, providing more advanced analytical algorithms for more intuitive data visualizations. The UMAP and t-SNE of anti-CD19 CAR-T products reveal distinct cytokine profiles between nonresponders and responders and demonstrate a marked upregulation of antitumor-associated cytokine signatures in CAR-T cells from responding patients. Using this powerful while user-friendly analytical tool, the multi-dimensional single-cell data can be dissected from complex immune responses and uncover underlying mechanisms, which can promote correlative biomarker discovery, improved bioprocessing, and personalized treatment development.


Sujet(s)
Algorithmes , Protéomique , Cytokines , Humains
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE