Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 390
Filtrer
2.
J Hematol Oncol ; 17(1): 29, 2024 May 06.
Article de Anglais | MEDLINE | ID: mdl-38711046

RÉSUMÉ

Currently, many off-the-shelf chimeric antigen receptor (CAR)-T cell products are under investigation for the treatment of relapsed or refractory (R/R) B-cell neoplasms. Compared with autologous CAR-T cell therapy, off-the-shelf universal CAR-T cell therapies have many potential benefits, such as immediate accessibility for patients, stable quality due to industrialized manufacturing and additional infusions of CAR-T cells with different targets. However, critical challenges, including graft-versus-host disease and CAR-T cell elimination by the host immune system, still require extensive research. The most common technological approaches involve modifying healthy donor T cells via gene editing technology and altering different types of T cells. This article summarizes some of the latest data from preclinical and clinical studies of off-the-shelf CAR-T cell therapies in the treatment of R/R B-cell malignancies from the 2023 ASH Annual Meeting (ASH 2023).


Sujet(s)
Immunothérapie adoptive , Récepteurs chimériques pour l'antigène , Humains , Immunothérapie adoptive/méthodes , Leucémie B/thérapie , Leucémie B/immunologie , Lymphome B/thérapie , Lymphome B/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/usage thérapeutique , Lymphocytes T/immunologie , Lymphocytes T/transplantation
3.
Bull Cancer ; 108(10S): S143-S154, 2021 Oct.
Article de Français | MEDLINE | ID: mdl-34920797

RÉSUMÉ

CAR-T Cells have opened new doors for cellular immunotherapies and provides new therapeutic options for patients with refractory B-cell malignancies, B-cell acute lymphoblastic leukemia and diffuse large B-cel lymphoma. CAR-T Cells have benefited from an accelerated approval procedure in many countries. Indeed, The French health authorities have approved the specialties Tisacel ® and Axicel ®, but additional data including the use of CAR-T Cells in real life were also mandatory. In regard to the scientific interest of the project, LYSA-LYSARC committed itself to prospectively and retrospectively collect information on patients eligible for CAR-T Cells as required by French health authorities. Other academic cooperating groups (GRAALL, IFM, SFCE, FILO and the scientific society SFGM-TC) were associated to this initiative which aims to build a nationwide CAR-T Cells devoted registry, so-called DESCART (Dispositif d'Enregistrement et Suivi des patients traités par CAR-T cells). DESCAR-T is a real-life multicentric registry set up in French sites qualified for CAR-T Cells treatment. DESCAR-T objective is to describe the use of CAR-T Cells in real life. All paediatric and adult patients with hematological malignancy fulfilling CAR-T Cells approval criteria and for whom a CAR-T Cells therapy has been discussed are included from 1 July 2018. Clinical data are directly collected from medical records and patients are treated according to the centers' routine practices. One of the distinctive features of DESCAR-T is its link with HTA for CAR-T Cells s reimbursement by the French public health system. DESCAR-T is the first national registry promoted by an academic group allowing centralized data collection for both academic and HTA/health authorities' purposes.


Sujet(s)
Tumeurs hématologiques/thérapie , Immunothérapie adoptive/statistiques et données numériques , Récepteurs chimériques pour l'antigène/immunologie , Enregistrements/statistiques et données numériques , Lymphocytes T/transplantation , Adolescent , Enfant , Collecte de données/méthodes , France , Tumeurs hématologiques/immunologie , Humains , Immunothérapie adoptive/législation et jurisprudence , Leucémie B/immunologie , Leucémie B/thérapie , Lymphome B diffus à grandes cellules/immunologie , Lymphome B diffus à grandes cellules/thérapie , Dossiers médicaux/statistiques et données numériques , Leucémie-lymphome lymphoblastique à précurseurs B/immunologie , Leucémie-lymphome lymphoblastique à précurseurs B/thérapie , Leucémie-lymphome lymphoblastique à précurseurs B et T , Enregistrements/éthique , Lymphocytes T/immunologie , Jeune adulte
4.
Bull Cancer ; 108(10S): S40-S54, 2021 Oct.
Article de Anglais | MEDLINE | ID: mdl-34920807

RÉSUMÉ

The marketing authorization of tisagenlecleucel, a 2nd generation of CD19-directed CAR T-cells, containing the 4-1 BB co-stimulatory domain, in 2017 in USA and in 2018 in EU, has revolutionized the therapeutic strategy in advanced B-cell acute lymphoblastic leukemia (B-ALL) in children, adolescents and young adults (AYAs) with relapsed or refractory disease. This innovative treatment, based on a "living drug", has shown very impressive short-term responses. However, safety profile and complex logistics require high expertise centers and tight collaborations between addressing and treating centers. Current research is exploring the possibility to move to first line ALL with high-risk features and/or first high-risk relapse. More efficient CAR T-cells products, are still lacking to counteract the escape mechanisms already described. Moreover, to define the bridge-to-CAR time for each patient remains a challenge to obtain optimal disease burden allowing expansion and persistence of CAR T-cells. Also difficult is to identify patients who will benefit from further therapy after infusion, such as allogeneic HSCT or may be immuno-modulatory treatment. Finally, CAR T-cells directed against T-ALL are only in their beginning but require more complex engineering process to avoid T- cell immune-deficiency or fratricide.


Sujet(s)
Antinéoplasiques immunologiques/usage thérapeutique , Immunothérapie adoptive/méthodes , Leucémie-lymphome lymphoblastique à précurseurs B et T/thérapie , Récepteurs aux antigènes des cellules T/usage thérapeutique , Récepteurs chimériques pour l'antigène/immunologie , Adolescent , Antigènes CD19/immunologie , Antigène CD28/immunologie , Ingénierie cellulaire , Enfant , Essais cliniques comme sujet , Coûts indirects de la maladie , Humains , Immunomodulation , Immunothérapie adoptive/effets indésirables , Leucémie B/immunologie , Leucémie B/anatomopathologie , Leucémie B/thérapie , Déplétion lymphocytaire , Leucémie-lymphome lymphoblastique à précurseurs B et T/immunologie , Leucémie-lymphome lymphoblastique à précurseurs B et T/anatomopathologie , Récidive , Échappement de la tumeur à la surveillance immunitaire/immunologie , Jeune adulte
5.
BMC Cancer ; 21(1): 1331, 2021 Dec 14.
Article de Anglais | MEDLINE | ID: mdl-34906116

RÉSUMÉ

BACKGROUND: The clinical outcome of Philadelphia chromosome-negative B cell acute lymphoblastic leukemia (Ph-neg B-ALL) varies considerably from one person to another after clinical treatment due to lack of targeted therapies and leukemia's heterogeneity. Ferroptosis is a recently discovered programmed cell death strongly correlated with cancers. Nevertheless, few related studies have reported its significance in acute lymphoblastic leukemia. METHODS: Herein, we collected clinical data of 80 Ph-neg B-ALL patients diagnosed in our center and performed RNA-seq with their initial bone marrow fluid samples. Throughout unsupervised machine learning K-means clustering with 24 ferroptosis related genes (FRGs), the clustered patients were parted into three variant risk groups and were performed with bioinformatics analysis. RESULTS: As a result, we discovered significant heterogeneity of both immune microenvironment and genomic variance. Furthermore, the immune check point inhibitors response and potential implementation of Sorafenib in Ph-neg B-ALL was also analyzed in our cohort. Lastly, one prognostic model based on 8 FRGs was developed to evaluate the risk of Ph-neg B-ALL patients. CONCLUSION: Jointly, our study proved the crucial role of ferroptosis in Ph-neg B-ALL and Sorafenib is likely to improve the survival of high-risk Ph-neg B-ALL patients.


Sujet(s)
Ferroptose/génétique , Leucémie B/génétique , Leucémie B/immunologie , Leucémie-lymphome lymphoblastique à précurseurs B et T/génétique , Leucémie-lymphome lymphoblastique à précurseurs B et T/immunologie , Adolescent , Adulte , Antinéoplasiques/usage thérapeutique , Apoptose , Enfant , Analyse de regroupements , Femelle , Ferroptose/immunologie , Humains , Leucémie B/traitement médicamenteux , Mâle , Adulte d'âge moyen , Chromosome Philadelphie , Leucémie-lymphome lymphoblastique à précurseurs B et T/traitement médicamenteux , Pronostic , RNA-Seq , Facteurs de risque , Sorafénib/usage thérapeutique , Résultat thérapeutique , Microenvironnement tumoral/immunologie , Apprentissage machine non supervisé , Jeune adulte
7.
JCI Insight ; 6(16)2021 08 23.
Article de Anglais | MEDLINE | ID: mdl-34423790

RÉSUMÉ

Targeting T cell malignancies with universal CD7-targeting chimeric antigen receptor T cells (UCART7) can lead to profound immune deficiency due to loss of normal T and NK cells. While a small population of endogenous CD7- T cells exists, these cells are unlikely to be able to repopulate the entire immune repertoire after UCART7 treatment, as they are limited in number and proliferative capacity. To rescue T and NK cells after UCART7, we created hematopoietic stem cells genetically deleted for CD7 (CD7-KO HSCs). CD7-KO HSCs were able to engraft immunodeficient mice and differentiate into T and NK cells lacking CD7 expression. CD7-KO T and NK cells could perform effector functions as robustly as control T and NK cells. Furthermore, CD7-KO T cells were phenotypically and functionally distinct from endogenous CD7- T cells, indicating that CD7-KO T cells can supplement immune functions lacking in CD7- T cells. Mice engrafted with CD7-KO HSCs maintained T and NK cell numbers after UCART7 treatment, while these were significantly decreased in control mice. These studies support the development of CD7-KO HSCs to augment host immunity in patients with T cell malignancies after UCART7 treatment.


Sujet(s)
Antigènes CD7/génétique , Cytotoxicité immunologique , Transplantation de cellules souches hématopoïétiques/méthodes , Immunothérapie adoptive/effets indésirables , Animaux , Ingénierie cellulaire/méthodes , Édition de gène , Techniques de knock-out de gènes , Cellules souches hématopoïétiques/métabolisme , Humains , Immunothérapie adoptive/méthodes , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Leucémie B/immunologie , Leucémie B/thérapie , Souris , RNA-Seq , Récepteurs chimériques pour l'antigène/génétique , Récepteurs chimériques pour l'antigène/immunologie , Analyse sur cellule unique , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Lymphocytes T/transplantation , Chimère obtenue par transplantation
8.
Blood ; 138(23): 2360-2371, 2021 12 09.
Article de Anglais | MEDLINE | ID: mdl-34255829

RÉSUMÉ

B-cell-activating factor (BAFF) mediates B-cell survival and, when deregulated, contributes to autoimmune diseases and B-cell malignancies. The mechanism connecting BAFF receptor (BAFFR) signal to downstream pathways and pathophysiological functions is not well understood. Here we identified DYRK1a as a kinase that responds to BAFF stimulation and mediates BAFF-induced B-cell survival. B-cell-specific DYRK1a deficiency causes peripheral B-cell reduction and ameliorates autoimmunity in a mouse model of lupus. An unbiased screen identified DYRK1a as a protein that interacts with TRAF3, a ubiquitin ligase component mediating degradation of the noncanonical nuclear factor (NF)-κB-inducing kinase (NIK). DYRK1a phosphorylates TRAF3 at serine-29 to interfere with its function in mediating NIK degradation, thereby facilitating BAFF-induced NIK accumulation and noncanonical NF-κB activation. Interestingly, B-cell acute lymphoblastic leukemia (B-ALL) cells express high levels of BAFFR and respond to BAFF for noncanonical NF-κB activation and survival in a DYRK1a-dependent manner. Furthermore, DYRK1a promotes a mouse model of B-ALL through activation of the noncanonical NF-κB pathway. These results establish DYRK1a as a critical BAFFR signaling mediator and provide novel insight into B-ALL pathogenesis.


Sujet(s)
Auto-immunité , Facteur d'activation des lymphocytes B/immunologie , Leucémie B/immunologie , Facteur de transcription NF-kappa B/immunologie , Protein-Serine-Threonine Kinases/immunologie , Protein-tyrosine kinases/immunologie , Animaux , Maladies auto-immunes/immunologie , Maladies auto-immunes/anatomopathologie , Lymphocytes B/immunologie , Lymphocytes B/anatomopathologie , Carcinogenèse/immunologie , Carcinogenèse/anatomopathologie , Lignée cellulaire tumorale , Humains , Leucémie B/anatomopathologie , Souris , Souris de lignée C57BL , Leucémie-lymphome lymphoblastique à précurseurs B/immunologie , Leucémie-lymphome lymphoblastique à précurseurs B/anatomopathologie ,
9.
Biochem Soc Trans ; 49(3): 1467-1478, 2021 06 30.
Article de Anglais | MEDLINE | ID: mdl-34196360

RÉSUMÉ

B-cells are antibody-producing cells of the adaptive immune system. Approximately 75% of all newly generated B-cells in the bone marrow are autoreactive and express potentially harmful autoantibodies. To prevent autoimmune disease, the immune system has evolved a powerful mechanism to eliminate autoreactive B-cells, termed negative B-cell selection. While designed to remove autoreactive clones during early B-cell development, our laboratory recently discovered that transformed B-cells in leukemia and lymphoma are also subject to negative selection. Indeed, besides the risk of developing autoimmune disease, B-cells are inherently prone to malignant transformation: to produce high-affinity antibodies, B-cells undergo multiple rounds of somatic immunoglobulin gene recombination and hypermutation. Reflecting high frequencies of DNA-breaks, adaptive immune protection by B-cells comes with a dramatically increased risk of development of leukemia and lymphoma. Of note, B-cells exist under conditions of chronic restriction of energy metabolism. Here we discuss how these metabolic gatekeeper functions during B-cell development provide a common mechanism for the removal of autoreactive and premalignant B-cells to safeguard against both autoimmune diseases and B-cell malignancies.


Sujet(s)
Immunité acquise/immunologie , Autoanticorps/immunologie , Maladies auto-immunes/immunologie , Auto-immunité/immunologie , Lymphocytes B/immunologie , Animaux , Autoanticorps/métabolisme , Maladies auto-immunes/métabolisme , Lymphocytes B/métabolisme , Transformation cellulaire néoplasique/immunologie , Transformation cellulaire néoplasique/métabolisme , Humains , Leucémie B/immunologie , Leucémie B/métabolisme , Activation des lymphocytes/immunologie
10.
Medicine (Baltimore) ; 100(25): e26446, 2021 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-34160438

RÉSUMÉ

RATIONALE: Viruses are the most common pathogens that can cause infection-related non-recurrent death after transplantation, occurring mostly from the early stages of hematopoietic stem cell transplantation (HSCT) to within 1 year after transplantation. Human coronavirus (HCoV)-NL63 is a coronavirus that could cause mortality among patients with underlying disease complications. Serological tests are of limited diagnostic value in immunocompromised hosts and cases of latent infection reactivation. In contrast, macro-genomic high-throughput (DNA and RNA) sequencing allows for rapid and accurate diagnosis of infecting pathogens for targeted treatment. PATIENT CONCERNS: In this report, we describe a patient who exhibited acute B-lymphocytic leukemia and developed complicated pulmonary HCoV-NL63 infection after a second allogeneic HSCT (allo-HSCT). Six months after the second allo-HSCT, he developed sudden-onset hyperthermia and cough with decreased oxygen saturation. Chest computed tomography (CT) suggested bilateral multiple rounded ground-glass opacities with the pulmonary lobules as units. DIAGNOSES: HCoV-NL63 was detected by metagenomic next-generation sequencing (NGS), and HCoV-NL63 viral pneumonia was diagnosed. INTERVENTIONS: The treatment was mainly based on the use of antiviral therapy, hormone administration, and gamma-globulin. OUTCOMES: After the therapy, the body temperature returned to normal, the chest CT findings had improved on review, and the viral copy number eventually became negative. LESSONS: The latest NGS is an effective method for early infection diagnosis. The HCoV-NL63 virus can cause inflammatory factor storm and alter the neutrophil-to-lymphocyte ratio (NLR). This case suggests that the patient's NLR and cytokine levels could be monitored during the clinical treatment to assess the disease and its treatment outcome in a timely manner.


Sujet(s)
Infections à coronavirus/diagnostic , Coronavirus humain NL63/isolement et purification , Transplantation de cellules souches hématopoïétiques/effets indésirables , Leucémie B/thérapie , Pneumopathie virale/diagnostic , Antiviraux/administration et posologie , Infections à coronavirus/traitement médicamenteux , Infections à coronavirus/immunologie , Infections à coronavirus/virologie , Coronavirus humain NL63/génétique , Coronavirus humain NL63/immunologie , Association de médicaments/méthodes , Séquençage nucléotidique à haut débit , Humains , Sujet immunodéprimé , Leucémie B/immunologie , Poumon/imagerie diagnostique , Mâle , Métagénomique , Pneumopathie virale/traitement médicamenteux , Pneumopathie virale/immunologie , Pneumopathie virale/virologie , Tomodensitométrie , Transplantation homologue/effets indésirables , Jeune adulte , Gammaglobulines/administration et posologie
11.
J Immunother Cancer ; 9(6)2021 06.
Article de Anglais | MEDLINE | ID: mdl-34135100

RÉSUMÉ

Chimeric antigen receptor T-cells (CAR T-cells) for the treatment of relapsing/refractory B-cell precursor acute lymphoblastic leukemia have led to exciting clinical results. However, CAR T-cell approaches revealed a potential risk of CD19-/CAR+ leukemic relapse due to inadvertent transduction of leukemia cells. BACKGROUND: METHODS: We evaluated the impact of a high percentage of leukemia blast contamination in patient-derived starting material (SM) on CAR T-cell drug product (DP) manufacturing. In vitro as well as in vivo models were employed to identify characteristics of the construct associated with better profile of safety in case of inadvertent B-cell leukemia transduction during CAR T-cell manufacturing. RESULTS: The presence of large amounts of CD19+ cells in SM did not affect the transduction level of DPs, as well as the CAR T-cell rate of expansion at the end of standard production of 14 days. DPs were deeply characterized by flow cytometry and molecular biology for Ig-rearrangements, showing that the level of B-cell contamination in DPs did not correlate with the percentage of CD19+ cells in SM, in the studied patient cohort. Moreover, we investigated whether CAR design may affect the control of CAR+ leukemia cells. We provided evidences that CAR.CD19 short linker (SL) prevents complete epitope masking in CD19+CAR+ leukemia cells and we demonstrated in vitro and in vivo that CD19 +CAR(SL)+leukemic cells are killed by CAR.CD19 T-cells. CONCLUSIONS: Taken together, these data suggest that a VL-VH SL may result in a safe CAR-T product, even when manufacturing starts from biological materials characterized by heavy contamination of leukemia blasts.


Sujet(s)
Épitopes/immunologie , Leucémie B/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Animaux , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Humains , Souris
12.
Curr Hematol Malig Rep ; 16(1): 32-39, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-33630232

RÉSUMÉ

PURPOSE OF REVIEW: Chimeric antigen receptor T-cell (CAR-T) therapy is a form of adoptive cellular therapy that has revolutionized the treatment landscape in hematologic malignancies, especially B-cell lymphomas. In this review, we will discuss some of the landmark data behind these therapies and then lay out our approach to utilizing this new therapy. RECENT FINDINGS: CD19-directed CAR-Ts are the most common type currently used in treatment of relapsed B-cell lymphoid neoplasms. There are currently three FDA-approved products: axicabtagene ciluecel and tisagenlecleucel for the treatment of relapsed/refractory large B-cell lymphoma and pediatric B-cell acute lymphocytic leukemia (tisagenlecleucel only) and brexucabtagene autoleucel for the treatment of relapsed/refractory mantle cell lymphoma. These therapies are associated with distinctive acute toxicities such as cytokine release syndrome and neurotoxicity and chronic toxicities such as cytopenias and hypogammaglobulinemia. CAR-T therapy provides significant potential in the treatment of relapsed B-cell lymphomas despite current limitations. Several novel CAR cell designs are currently being studied in clinical trials which include tandem CAR-Ts, allogeneic CAR-Ts, and CAR-NK cells.


Sujet(s)
Immunothérapie adoptive/méthodes , Leucémie B/thérapie , Lymphome B/thérapie , Animaux , Antigènes CD19/usage thérapeutique , Lymphocytes B/immunologie , Lymphocytes B/anatomopathologie , Produits biologiques , Humains , Leucémie B/immunologie , Leucémie B/anatomopathologie , Lymphome B/immunologie , Lymphome B/anatomopathologie , Récepteurs aux antigènes des cellules T/usage thérapeutique
13.
Leukemia ; 35(1): 75-89, 2021 01.
Article de Anglais | MEDLINE | ID: mdl-32205861

RÉSUMÉ

Chimeric antigen receptor (CAR) T-cells targeting CD19 demonstrate remarkable efficacy in treating B-lineage acute lymphoblastic leukemia (BL-ALL), yet up to 39% of treated patients relapse with CD19(-) disease. We report that CD19(-) escape is associated with downregulation, but preservation, of targetable expression of CD20 and CD22. Accordingly, we reasoned that broadening the spectrum of CD19CAR T-cells to include both CD20 and CD22 would enable them to target CD19(-) escape BL-ALL while preserving their upfront efficacy. We created a CD19/20/22-targeting CAR T-cell by coexpressing individual CAR molecules on a single T-cell using one tricistronic transgene. CD19/20/22CAR T-cells killed CD19(-) blasts from patients who relapsed after CD19CAR T-cell therapy and CRISPR/Cas9 CD19 knockout primary BL-ALL both in vitro and in an animal model, while CD19CAR T-cells were ineffective. At the subcellular level, CD19/20/22CAR T-cells formed dense immune synapses with target cells that mediated effective cytolytic complex formation, were efficient serial killers in single-cell tracking studies, and were as efficacious as CD19CAR T-cells against primary CD19(+) disease. In conclusion, independent of CD19 expression, CD19/20/22CAR T-cells could be used as salvage or front-line CAR therapy for patients with recalcitrant disease.


Sujet(s)
Antigènes CD19/immunologie , Immunothérapie adoptive , Leucémie B/immunologie , Leucémie B/métabolisme , Récepteurs aux antigènes des cellules T/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Animaux , Antigènes CD19/composition chimique , Antigènes néoplasiques , Marqueurs biologiques , Lignée cellulaire tumorale , Cytokines/métabolisme , Cytotoxicité immunologique , Modèles animaux de maladie humaine , Expression des gènes , Humains , Immunothérapie adoptive/méthodes , Leucémie B/génétique , Leucémie B/thérapie , Souris transgéniques , Liaison aux protéines , Récepteurs aux antigènes des cellules T/métabolisme , Récepteurs chimériques pour l'antigène/génétique , Récepteurs chimériques pour l'antigène/métabolisme , Relation structure-activité , Transduction génétique , Transgènes , Résultat thérapeutique , Tests d'activité antitumorale sur modèle de xénogreffe
14.
Blood ; 137(3): 323-335, 2021 01 21.
Article de Anglais | MEDLINE | ID: mdl-32967009

RÉSUMÉ

CD19-targeted chimeric antigen receptor-engineered (CD19 CAR) T-cell therapy has shown significant efficacy for relapsed or refractory (R/R) B-cell malignancies. Yet, CD19 CAR T cells fail to induce durable responses in most patients. Second infusions of CD19 CAR T cells (CART2) have been considered as a possible approach to improve outcomes. We analyzed data from 44 patients with R/R B-cell malignancies (acute lymphoblastic leukemia [ALL], n = 14; chronic lymphocytic leukemia [CLL], n = 9; non-Hodgkin lymphoma [NHL], n = 21) who received CART2 on a phase 1/2 trial (NCT01865617) at our institution. Despite a CART2 dose increase in 82% of patients, we observed a low incidence of severe toxicity after CART2 (grade ≥3 cytokine release syndrome, 9%; grade ≥3 neurotoxicity, 11%). After CART2, complete response (CR) was achieved in 22% of CLL, 19% of NHL, and 21% of ALL patients. The median durations of response after CART2 in CLL, NHL, and ALL patients were 33, 6, and 4 months, respectively. Addition of fludarabine to cyclophosphamide-based lymphodepletion before the first CAR T-cell infusion (CART1) and an increase in the CART2 dose compared with CART1 were independently associated with higher overall response rates and longer progression-free survival after CART2. We observed durable CAR T-cell persistence after CART2 in patients who received cyclophosphamide and fludarabine (Cy-Flu) lymphodepletion before CART1 and a higher CART2 compared with CART1 cell dose. The identification of 2 modifiable pretreatment factors independently associated with better outcomes after CART2 suggests strategies to improve in vivo CAR T-cell kinetics and responses after repeat CAR T-cell infusions, and has implications for the design of trials of novel CAR T-cell products after failure of prior CAR T-cell immunotherapies.


Sujet(s)
Antigènes CD19/métabolisme , Immunothérapie adoptive , Leucémie B/thérapie , Leucémie chronique lymphocytaire à cellules B/thérapie , Lymphome malin non hodgkinien/thérapie , Leucémie-lymphome lymphoblastique à précurseurs B et T/thérapie , Adulte , Sujet âgé , Prolifération cellulaire , Cyclophosphamide/usage thérapeutique , Syndrome de libération de cytokines/complications , Femelle , Humains , Leucémie B/immunologie , Leucémie chronique lymphocytaire à cellules B/immunologie , Lymphome malin non hodgkinien/immunologie , Mâle , Adulte d'âge moyen , Analyse multifactorielle , Leucémie-lymphome lymphoblastique à précurseurs B et T/immunologie , Survie sans progression , Lymphocytes T/immunologie , Résultat thérapeutique , Vidarabine/analogues et dérivés , Vidarabine/usage thérapeutique
15.
Front Immunol ; 11: 581119, 2020.
Article de Anglais | MEDLINE | ID: mdl-33240268

RÉSUMÉ

Congenital defects of the immune system called primary immunodeficiency disorders (PID) describe a group of diseases characterized by a decrease, an absence, or a malfunction of at least one part of the immune system. As a result, PID patients are more prone to develop life-threatening complications, including cancer. PID currently include over 400 different disorders, however, the variety of PID-related cancers is narrow. We discuss here reasons for this clinical phenotype. Namely, PID can lead to cell intrinsic failure to control cell transformation, failure to activate tumor surveillance by cytotoxic cells or both. As the most frequent tumors seen among PID patients stem from faulty lymphocyte development leading to leukemia and lymphoma, we focus on the extensive genomic alterations needed to create the vast diversity of B and T lymphocytes with potential to recognize any pathogen and why defects in these processes lead to malignancies in the immunodeficient environment of PID patients. In the second part of the review, we discuss PID affecting tumor surveillance and especially membrane trafficking defects caused by altered exocytosis and regulation of the actin cytoskeleton. As an impairment of these membrane trafficking pathways often results in dysfunctional effector immune cells, tumor cell immune evasion is elevated in PID. By considering new anti-cancer treatment concepts, such as transfer of genetically engineered immune cells, restoration of anti-tumor immunity in PID patients could be an approach to complement standard therapies.


Sujet(s)
Leucémie B/étiologie , Lymphome B/étiologie , Maladies d'immunodéficience primaire/complications , Cytosquelette d'actine/génétique , Cytosquelette d'actine/immunologie , Lymphocytes B/immunologie , Lymphocytes B/anatomopathologie , Prolifération cellulaire , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/immunologie , Réparation de l'ADN/génétique , Réparation de l'ADN/immunologie , Exocytose/génétique , Exocytose/immunologie , Instabilité du génome , Humains , Synapses immunologiques/génétique , Leucémie B/génétique , Leucémie B/immunologie , Lymphome B/génétique , Lymphome B/immunologie , Modèles immunologiques , Maladies d'immunodéficience primaire/génétique , Maladies d'immunodéficience primaire/immunologie , Facteurs de risque , Échappement de la tumeur à la surveillance immunitaire/génétique
16.
Nat Med ; 26(10): 1569-1575, 2020 10.
Article de Anglais | MEDLINE | ID: mdl-33020647

RÉSUMÉ

Chimeric antigen receptor (CAR) T cells targeting CD19 are a breakthrough treatment for relapsed, refractory B cell malignancies1-5. Despite impressive outcomes, relapse with CD19- disease remains a challenge. We address this limitation through a first-in-human trial of bispecific anti-CD20, anti-CD19 (LV20.19) CAR T cells for relapsed, refractory B cell malignancies. Adult patients with B cell non-Hodgkin lymphoma or chronic lymphocytic leukemia were treated on a phase 1 dose escalation and expansion trial (NCT03019055) to evaluate the safety of 4-1BB-CD3ζ LV20.19 CAR T cells and the feasibility of on-site manufacturing using the CliniMACS Prodigy system. CAR T cell doses ranged from 2.5 × 105-2.5 × 106 cells per kg. Cell manufacturing was set at 14 d with the goal of infusing non-cryopreserved LV20.19 CAR T cells. The target dose of LV20.19 CAR T cells was met in all CAR-naive patients, and 22 patients received LV20.19 CAR T cells on protocol. In the absence of dose-limiting toxicity, a dose of 2.5 × 106 cells per kg was chosen for expansion. Grade 3-4 cytokine release syndrome occurred in one (5%) patient, and grade 3-4 neurotoxicity occurred in three (14%) patients. Eighteen (82%) patients achieved an overall response at day 28, 14 (64%) had a complete response, and 4 (18%) had a partial response. The overall response rate to the dose of 2.5 × 106 cells per kg with non-cryopreserved infusion (n = 12) was 100% (complete response, 92%; partial response, 8%). Notably, loss of the CD19 antigen was not seen in patients who relapsed or experienced treatment failure. In conclusion, on-site manufacturing and infusion of non-cryopreserved LV20.19 CAR T cells were feasible and therapeutically safe, showing low toxicity and high efficacy. Bispecific CARs may improve clinical responses by mitigating target antigen downregulation as a mechanism of relapse.


Sujet(s)
Antigènes CD19/immunologie , Antigènes CD20/immunologie , Immunothérapie adoptive/méthodes , Leucémie B/thérapie , Lymphome B/thérapie , Adulte , Sujet âgé , Relation dose-réponse (immunologie) , Femelle , Humains , Leucémie B/immunologie , Leucémie B/anatomopathologie , Numération des lymphocytes , Lymphome B/immunologie , Lymphome B/anatomopathologie , Mâle , Adulte d'âge moyen , Récepteurs aux antigènes des cellules T/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Récidive , Lymphocytes T/cytologie , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Lymphocytes T/transplantation
17.
Paediatr Drugs ; 22(5): 485-499, 2020 Oct.
Article de Anglais | MEDLINE | ID: mdl-32860590

RÉSUMÉ

Leukemia, most commonly B-cell acute lymphoblastic leukemia (B-ALL), accounts for about 30% of childhood cancer diagnoses. While there have been dramatic improvements in childhood ALL outcomes, certain subgroups-particularly those who relapse-fare poorly. In addition, cure is associated with significant short- and long-term side effects. Given these challenges, there is great interest in novel, targeted approaches to therapy. A number of new immunotherapeutic agents have proven to be efficacious in relapsed or refractory disease and are now being investigated in frontline treatment regimens. Blinatumomab (a bispecific T-cell engager that targets cluster of differentiation [CD]-19) and inotuzumab ozogamicin (a humanized antibody-drug conjugate to CD22) have shown the most promise. Chimeric antigen receptor T (CAR-T) cells, a form of adoptive immunotherapy, rely on the transfer of genetically modified effector T cells that have the potential to persist in vivo for years, providing ongoing long-term disease control. In this article, we discuss the clinical biology and treatment of B-ALL with an emphasis on the role of immunotherapy in overcoming the challenges of conventional cytotoxic therapy. As immunotherapy continues to move into the frontline of pediatric B-ALL therapy, we also discuss strategies to address unique side effects associated with these agents and efforts to overcome mechanisms of resistance to immunotherapy.


Sujet(s)
Antinéoplasiques immunologiques/usage thérapeutique , Immunothérapie , Leucémie B/thérapie , Enfant , Humains , Leucémie B/immunologie , Récepteurs chimériques pour l'antigène
18.
Leukemia ; 34(10): 2704-2707, 2020 10.
Article de Anglais | MEDLINE | ID: mdl-32594100

RÉSUMÉ

Chimeric antigen receptor (CAR)-T-cell is a safe and effective therapy of B-cell cancers but it is unknown if this is so in persons with prior hepatitis B virus (HBV) infection. We studied 70 subjects with advanced B-cell cancers receiving CAR-T-cell therapy, 12 of whom had chronic HBV-infection (HBsAg positive) and 29 with resolved HBV-infection (HBsAg negative and anti-HBc positive). Safety and efficacy were compared with 29 subjects without HBV-infection. HBV was reactivated in 2 subjects with chronic HBV-infection and 1 with resolved HBV-infection. There was no HBV-related hepatitis flare. Responses to CAR-T-cell therapy in the three cohorts were not significantly different. There was no significant difference in the incidence or severity of cytokine release syndrome (CRS) and neurologic toxicity between the cohorts. Our data suggest that chronic and resolved HBV-infection do not affect the safety and efficacy of CAR-T-cell therapy.


Sujet(s)
Hépatite B/immunologie , Hépatite B/thérapie , Immunothérapie adoptive , Leucémie B/immunologie , Leucémie B/thérapie , Lymphome B/immunologie , Lymphome B/thérapie , Adolescent , Adulte , Sujet âgé , Enfant , Enfant d'âge préscolaire , Femelle , Hépatite B/diagnostic , Hépatite B/virologie , Humains , Immunothérapie adoptive/effets indésirables , Immunothérapie adoptive/méthodes , Tests de la fonction hépatique , Numération des lymphocytes , Lymphome B/diagnostic , Mâle , Adulte d'âge moyen , Stadification tumorale , Récepteurs chimériques pour l'antigène/immunologie , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Résultat thérapeutique , Charge virale , Jeune adulte
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE