Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 5.084
Filtrer
1.
Oncol Res ; 32(6): 1109-1118, 2024.
Article de Anglais | MEDLINE | ID: mdl-38827326

RÉSUMÉ

Background: Chimeric antigen receptor T (CAR-T) cell therapy has achieved marked therapeutic success in ameliorating hematological malignancies. However, there is an extant void in the clinical guidelines concerning the most effective chemotherapy regimen prior to chimeric antigen receptor T (CAR-T) cell therapy, as well as the optimal timing for CAR-T cell infusion post-chemotherapy. Materials and Methods: We employed cell-derived tumor xenograft (CDX) murine models to delineate the optimal pre-conditioning chemotherapy regimen and timing for CAR-T cell treatment. Furthermore, transcriptome sequencing was implemented to identify the therapeutic targets and elucidate the underlying mechanisms governing the treatment regimen. Results: Our preclinical in vivo evaluation determined that a combination of cyclophosphamide and fludarabine, followed by the infusion of CD19 CAR-T cells five days subsequent to the chemotherapy, exerts the most efficacious therapeutic effect in B-cell hematological malignancies. Concurrently, RNA-seq data indicated that the therapeutic efficacy predominantly perturbs tumor cell metabolism, primarily through the inhibition of key mitochondrial targets, such as C-Jun Kinase enzyme (C-JUN). Conclusion: In summary, the present study offers critical clinical guidance and serves as an authoritative reference for the deployment of CD19 CAR-T cell therapy in the treatment of B-cell hematological malignancies.


Sujet(s)
Antigènes CD19 , Cyclophosphamide , Immunothérapie adoptive , Récepteurs chimériques pour l'antigène , Vidarabine , Tests d'activité antitumorale sur modèle de xénogreffe , Vidarabine/analogues et dérivés , Vidarabine/pharmacologie , Cyclophosphamide/usage thérapeutique , Cyclophosphamide/pharmacologie , Animaux , Souris , Humains , Immunothérapie adoptive/méthodes , Antigènes CD19/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Tumeurs hématologiques/thérapie , Tumeurs hématologiques/traitement médicamenteux , Lignée cellulaire tumorale , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Association thérapeutique
2.
Neurol Neuroimmunol Neuroinflamm ; 11(4): e200264, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38870458

RÉSUMÉ

OBJECTIVES: To report CD19+ B-cell counts and possible adverse effects on infants of mothers exposed to anti-CD20 mAbs ≤6 months before/during pregnancy or lactation. METHODS: We conducted a retrospective study using data from the German nationwide neuroimmunologic pregnancy registry. Inclusion criteria involved infants whose mothers received anti-CD20 mAbs ≤6 months before/during pregnancy or lactation, with ≥1 postnatal CD19+ B-cell count. Main outcomes were absolute and relative CD19+ B-cell counts. Comparison with reference values was performed conservatively in a subgroup with maternal exposure ≤3 months before/during pregnancy. Additional outcomes included pregnancy results, severe infections, and lymphocyte counts. RESULTS: The cohort comprised 49 infants (F:M 25:24) exposed to anti-CD20 mAbs ≤6 months before/during pregnancy or lactation. CD19+ B-cell and lymphocyte counts in 40 infants with maternal exposure ≤3 months before/during pregnancy were comparable with normative values. Only 2 cases of complete CD19+ B-cell depletion occurred after second-trimester and third-trimester ocrelizumab exposure, with repopulation observed within 2 months. Exclusive lactation exposure had no significant effect on infants' absolute CD19+ B-cell counts. DISCUSSION: Administering anti-CD20 mAbs before or at the pregnancy onset, or during lactation, seems safe without significant impact on infant B-cell development. However, second-trimester or third-trimester exposure can cause CD19+ B-cell depletion due to placental transfer, necessitating monitoring and postponing live vaccines.


Sujet(s)
Antigènes CD20 , Lymphocytes B , Lactation , Humains , Femelle , Grossesse , Nouveau-né , Lymphocytes B/immunologie , Lymphocytes B/effets des médicaments et des substances chimiques , Études rétrospectives , Lactation/immunologie , Mâle , Adulte , Antigènes CD20/immunologie , Santé infantile , Effets différés de l'exposition prénatale à des facteurs de risque/immunologie , Effets différés de l'exposition prénatale à des facteurs de risque/induit chimiquement , Antigènes CD19/immunologie , Numération des lymphocytes , Rituximab/effets indésirables , Rituximab/administration et posologie , Rituximab/pharmacologie , Facteurs immunologiques/effets indésirables , Facteurs immunologiques/administration et posologie , Facteurs immunologiques/pharmacologie , Nourrisson
3.
Proc Natl Acad Sci U S A ; 121(26): e2403227121, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38885382

RÉSUMÉ

Treatment with autologous chimeric antigen receptor (CAR) T cells has emerged as a highly effective approach in neuroimmunological disorders such as myasthenia gravis. We report a case of successful anti-CD19 CAR T cell use in treatment-refractory stiff-person syndrome (SPS). To investigate clinical and immunological effects of anti-CD19 CAR T cell use in treatment-refractory SPS, a 69-y-old female with a 9-y history of treatment-refractory SPS with deteriorating episodes of stiffness received an infusion of autologous anti-CD19 CAR T cells (KYV-101) and was monitored clinically and immunologically for more than 6 mo. CAR T cell infusion resulted in reduced leg stiffness, drastic improvement in gait, walking speed increase over 100%, and daily walking distance improvement from less than 50 m to over 6 km within 3 mo. GABAergic medication (benzodiazepines) was reduced by 40%. KYV-101 CAR T cells were well tolerated with only low-grade cytokine release syndrome. This report of successful use of anti-CD19 CAR T cells in treatment-refractory SPS supports continued exploration of this approach in SPS and other B cell-related autoimmune disorders.


Sujet(s)
Antigènes CD19 , Immunothérapie adoptive , Syndrome de l'homme raide , Humains , Syndrome de l'homme raide/thérapie , Syndrome de l'homme raide/immunologie , Femelle , Sujet âgé , Immunothérapie adoptive/méthodes , Antigènes CD19/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Lymphocytes T/immunologie , Résultat thérapeutique
5.
Front Immunol ; 15: 1401683, 2024.
Article de Anglais | MEDLINE | ID: mdl-38868778

RÉSUMÉ

Introduction: Chimeric antigen receptor-expressing T cells (CAR T cells) have revolutionized cancer treatment, particularly in B cell malignancies. However, the use of autologous T cells for CAR T therapy presents several limitations, including high costs, variable efficacy, and adverse effects linked to cell phenotype. Methods: To overcome these challenges, we developed a strategy to generate universal and safe anti-CD19 CAR T cells with a defined memory phenotype. Our approach utilizes CRISPR/Cas9 technology to target and eliminate the B2M and TRAC genes, reducing graft-versus-host and host-versus-graft responses. Additionally, we selected less differentiated T cells to improve the stability and persistence of the universal CAR T cells. The safety of this method was assessed using our CRISPRroots transcriptome analysis pipeline, which ensures successful gene knockout and the absence of unintended off-target effects on gene expression or transcriptome sequence. Results: In vitro experiments demonstrated the successful generation of functional universal CAR T cells. These cells exhibited potent lytic activity against tumor cells and a reduced cytokine secretion profile. The CRISPRroots analysis confirmed effective gene knockout and no unintended off-target effects, validating it as a pioneering tool for on/off-target and transcriptome analysis in genome editing experiments. Discussion: Our findings establish a robust pipeline for manufacturing safe, universal CAR T cells with a favorable memory phenotype. This approach has the potential to address the current limitations of autologous CAR T cell therapy, offering a more stable and persistent treatment option with reduced adverse effects. The use of CRISPRroots enhances the reliability and safety of gene editing in the development of CAR T cell therapies. Conclusion: We have developed a potent and reliable method for producing universal CAR T cells with a defined memory phenotype, demonstrating both efficacy and safety in vitro. This innovative approach could significantly improve the therapeutic landscape for patients with B cell malignancies.


Sujet(s)
Antigènes CD19 , Systèmes CRISPR-Cas , Édition de gène , Mémoire immunologique , Immunothérapie adoptive , Récepteurs chimériques pour l'antigène , Transcriptome , Humains , Immunothérapie adoptive/méthodes , Immunothérapie adoptive/effets indésirables , Antigènes CD19/immunologie , Antigènes CD19/génétique , Édition de gène/méthodes , Récepteurs chimériques pour l'antigène/génétique , Récepteurs chimériques pour l'antigène/immunologie , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Phénotype , Lignée cellulaire tumorale
6.
Immunohorizons ; 8(6): 404-414, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38864817

RÉSUMÉ

T cell activation is an essential step in chimeric Ag receptor (CAR) T (CAR T) cell manufacturing and is accomplished by the addition of activator reagents that trigger the TCR and provide costimulation. We explore several T cell activation reagents and examine their effects on key attributes of CAR T cell cultures, such as activation/exhaustion markers, cell expansion, gene expression, and transduction efficiency. Four distinct activators were examined, all using anti-CD3 and anti-CD28, but incorporating different mechanisms of delivery: Dynabeads (magnetic microspheres), TransAct (polymeric nanomatrix), Cloudz (alginate hydrogel), and Microbubbles (lipid membrane containing perfluorocarbon gas). Clinical-grade lentiviral vector was used to transduce cells with a bivalent CD19/CD22 CAR, and cell counts and flow cytometry were used to monitor the cells throughout the culture. We observed differences in CD4/CD8 ratio when stimulating with the Cloudz activator, where there was a significant skewing toward CD8 T cells. The naive T cell subset expressing CD62L+CCR7+CD45RA+ was the highest in all donors when stimulating with Dynabeads, whereas effector/effector memory cells were highest when using the Cloudz. Functional assays demonstrated differences in killing of target cells and proinflammatory cytokine secretion, with the highest killing from the Cloudz-stimulated cells among all donors. This study demonstrates that the means by which these stimulatory Abs are presented to T cells contribute to the activation, resulting in differing effects on CAR T cell function. These studies highlight important differences in the final product that should be considered when manufacturing CAR T cells for patients in the clinic.


Sujet(s)
Activation des lymphocytes , Récepteurs chimériques pour l'antigène , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/génétique , Récepteurs chimériques pour l'antigène/métabolisme , Humains , Activation des lymphocytes/immunologie , Immunothérapie adoptive/méthodes , Lymphocytes T CD8+/immunologie , Lymphocytes T/immunologie , Phénotype , Récepteurs aux antigènes des cellules T/immunologie , Récepteurs aux antigènes des cellules T/métabolisme , Récepteurs aux antigènes des cellules T/génétique , Antigènes CD19/immunologie , Antigènes CD19/métabolisme
7.
Haematologica ; 109(6): 1668-1676, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38832422

RÉSUMÉ

Immunotherapy has revolutionized treatment for a wide variety of cancers yet its use has been relatively limited in childhood malignancies. With the introduction of bispecific T-cell engagers (BiTE®) and chimeric antigen T-cell receptor technologies, previously refractory patients have attained remission, including molecularly negative states of disease, thus providing the possibility of long-term cure. Blinatumomab is a widely available CD3-CD19 BiTE that has dramatically changed the landscape of therapy for some children with precursor-B acute lymphoblastic leukemias (B-ALL) and lymphoblastic lymphomas. Challenges remain with using BiTE in a broader population although the appeal of now-confirmed reduced toxicity and deeper molecular remissions suggests that this approach will be an essential part of future treatment of childhood B-ALL. Herein, we review some of the pertinent literature covering clinical trials with blinatumomab and address future approaches and combination trials including BiTE.


Sujet(s)
Anticorps bispécifiques , Leucémie-lymphome lymphoblastique à précurseurs B , Lymphocytes T , Humains , Anticorps bispécifiques/usage thérapeutique , Leucémie-lymphome lymphoblastique à précurseurs B/thérapie , Leucémie-lymphome lymphoblastique à précurseurs B/immunologie , Leucémie-lymphome lymphoblastique à précurseurs B/traitement médicamenteux , Enfant , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Essais cliniques comme sujet , Antigènes CD19/immunologie , Résultat thérapeutique , Antinéoplasiques immunologiques/usage thérapeutique
8.
Med ; 5(6): 482-484, 2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38878763

RÉSUMÉ

The efficacy of CD19 chimeric antigen receptor (CAR) T cells in B cell malignancies has generated recent interest in their application to other B cell-related pathologies, such as autoimmune diseases. Fischbach et al.1 report on the use of CD19 CAR T cells in two patients with progressive multiple sclerosis, demonstrating feasibility and safety for the first time in this disease process.


Sujet(s)
Antigènes CD19 , Immunothérapie adoptive , Sclérose en plaques , Récepteurs chimériques pour l'antigène , Humains , Antigènes CD19/immunologie , Sclérose en plaques/immunologie , Sclérose en plaques/thérapie , Immunothérapie adoptive/méthodes , Récepteurs chimériques pour l'antigène/immunologie , Lymphocytes T/immunologie , Récepteurs aux antigènes des cellules T/immunologie
9.
Eur J Cancer ; 204: 114071, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38691878

RÉSUMÉ

Systemic sclerosis, a severe inflammatory autoimmune disease, shares a common thread with cancer through the underlying mechanism of inflammation. This inflammatory milieu not only drives the immune dysregulation characteristic of autoimmune diseases but also plays a pivotal role in the pathogenesis of cancer. Among the cellular components involved, B cells have emerged as key players in hematologic tumor and autoimmune disease, contributing to immune dysregulation and persistent tissue fibrosis in systemic sclerosis, as well as tumor progression and immune evasion in cancer. Consequently, novel therapeutic strategies targeting B cells hold promise in both conditions. Recent exploration of CD19 CAR T cells in severe systemic sclerosis patients has shown great potential, but also introduced possible risks and drawbacks associated with viral vectors, prolonged CAR T cell persistence, lengthy production timelines, high costs, and the necessity of conditioning patients with organotoxic and fertility-damaging chemotherapy. Given these challenges, alternative CD19-depleting approaches are of high interest for managing severe systemic autoimmune diseases. Here, we present the pioneering use of blinatumomab, a bispecific anti-CD3/anti-CD19 T cell engager in a patient with progressive, severe systemic sclerosis, offering a promising alternative for such challenging cases.


Sujet(s)
Anticorps bispécifiques , Antigènes CD19 , Sclérodermie systémique , Humains , Anticorps bispécifiques/usage thérapeutique , Sclérodermie systémique/traitement médicamenteux , Sclérodermie systémique/immunologie , Antigènes CD19/immunologie , Lymphocytes T/immunologie , Lymphocytes T/effets des médicaments et des substances chimiques , Femelle , Antigènes CD3/immunologie , Antigènes CD3/métabolisme , Adulte d'âge moyen , Immunothérapie adoptive/méthodes
10.
Dtsch Med Wochenschr ; 149(11): 630-637, 2024 May.
Article de Allemand | MEDLINE | ID: mdl-38749439

RÉSUMÉ

The introduction of immunologically targeted therapies has represented a significant advancement in the treatment of B-cell lymphomas, particularly aggressive B-cell lymphoma. CD19 CAR-T cells such as Axicabtagen-Ciloleucel (Axi-cel) and Lisocabtagen Maraleucel (Liso-cel) have been approved since 2022 and 2023, respectively, for second-line therapy of Diffuse Large B-Cell Lymphomas (DLBCL), when there is primary refractory disease or relapse within 12 months after the end of first-line therapy. These therapies result in a significant improvement in progression-free survival compared to the previous standard therapy (salvage chemotherapy followed by high-dose chemotherapy and autologous stem cell transplantation). Especially in elderly patients or patients with underlying medical conditions, CAR-T cell therapies like Axi-cel and Liso-cel demonstrate acceptable tolerability and high efficacy.Furthermore, bispecific T-cell-engaging antibodies ("bispecifics") such as Glofitamab, Epcoritamab, and Mosunetuzumab also represent promising treatment options for patients with relapsed disease after failure of second- or later line therapy and show efficacy even in a subset of patients relapsing after CD19 CAR-T cells. However, randomized study results for these substances are not yet available. They are expected to be used in earlier lines of therapy in the future, especially in combination with standard chemotherapy regimens. Common side effects of bispecific antibody therapies are cytokine release syndrome (CRS) and immune-mediated cytopenias, whereas immune-cell associated neurotoxicity syndrome (ICANS) is relatively rare compared to CD19 CAR T cells. In summary, bispecifics represent a novel, highly effective immunotherapy for the treatment of lymphomas with a very favourable toxicity profile.


Sujet(s)
Immunothérapie adoptive , Humains , Immunothérapie adoptive/effets indésirables , Lymphocytes T/immunologie , Immunothérapie/méthodes , Lymphome B/thérapie , Lymphome B/immunologie , Anticorps bispécifiques/usage thérapeutique , Lymphome B diffus à grandes cellules/thérapie , Lymphome B diffus à grandes cellules/immunologie , Antigènes CD19/immunologie
12.
Nat Commun ; 15(1): 4182, 2024 May 17.
Article de Anglais | MEDLINE | ID: mdl-38755157

RÉSUMÉ

Bone marrow plasma cells (BMPC) are the correlate of humoral immunity, consistently releasing antibodies into the bloodstream. It remains unclear if BMPC reflect different activation environments or maturation of their precursors. Here we define human BMPC heterogeneity and track the recruitment of antibody-secreting cells (ASC) from SARS-CoV-2 vaccine immune reactions to the bone marrow (BM). Trajectories based on single-cell transcriptomes and repertoires of peripheral and BM ASC reveal sequential colonisation of BMPC compartments. In activated B cells, IL-21 suppresses CD19 expression, indicating that CD19low-BMPC are derived from follicular, while CD19high-BMPC originate from extrafollicular immune reactions. In primary immune reactions, both CD19low- and CD19high-BMPC compartments are populated. In secondary immune reactions, most BMPC are recruited to CD19high-BMPC compartments, reflecting their origin from extrafollicular reactivations of memory B cells. A pattern also observable in vaccinated-convalescent individuals and upon diphtheria/tetanus/pertussis recall-vaccination. Thus, BMPC diversity reflects the evolution of a given humoral immune response.


Sujet(s)
Antigènes CD19 , Moelle osseuse , Interleukines , Plasmocytes , Humains , Plasmocytes/immunologie , Interleukines/immunologie , Interleukines/métabolisme , Moelle osseuse/immunologie , Antigènes CD19/immunologie , Antigènes CD19/métabolisme , Immunité humorale/immunologie , COVID-19/immunologie , COVID-19/virologie , SARS-CoV-2/immunologie , Cellules de la moelle osseuse/immunologie , Cellules de la moelle osseuse/cytologie , Analyse sur cellule unique , Adulte , Lymphocytes B/immunologie , Cellules productrices d'anticorps/immunologie , Femelle , Mâle , Vaccination , Adulte d'âge moyen , Vaccin diphtérie-tétanos-coqueluche/immunologie
14.
Mol Biol Rep ; 51(1): 629, 2024 May 08.
Article de Anglais | MEDLINE | ID: mdl-38717637

RÉSUMÉ

It has been rediscovered in the last fifteen years that B-cells play an active role in autoimmune etiology rather than just being spectators. The clinical success of B-cell depletion therapies (BCDTs) has contributed to this. BCDTs, including those that target CD20, CD19, and BAFF, were first developed to eradicate malignant B-cells. These days, they treat autoimmune conditions like multiple sclerosis and systemic lupus erythematosus. Particular surprises have resulted from the use of BCDTs in autoimmune diseases. For example, even in cases where BCDT is used to treat the condition, its effects on antibody-secreting plasma cells and antibody levels are restricted, even though these cells are regarded to play a detrimental pathogenic role in autoimmune diseases. In this Review, we provide an update on our knowledge of the biology of B-cells, examine the outcomes of clinical studies employing BCDT for autoimmune reasons, talk about potential explanations for the drug's mode of action, and make predictions about future approaches to targeting B-cells other than depletion.


Sujet(s)
Maladies auto-immunes , Lymphocytes B , Déplétion lymphocytaire , Humains , Lymphocytes B/immunologie , Maladies auto-immunes/immunologie , Maladies auto-immunes/thérapie , Déplétion lymphocytaire/méthodes , Antigènes CD20/immunologie , Antigènes CD19/immunologie , Animaux , Facteur d'activation des lymphocytes B/immunologie , Sclérose en plaques/immunologie , Sclérose en plaques/thérapie , Lupus érythémateux disséminé/immunologie , Lupus érythémateux disséminé/thérapie
15.
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
16.
Nat Commun ; 15(1): 4327, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38773088

RÉSUMÉ

The antitumor efficacy of adoptively transferred T cells is limited by their poor persistence, in part due to exhaustion, but the underlying mechanisms and potential interventions remain underexplored. Here, we show that targeting histone demethylase LSD1 by chemical inhibitors reshapes the epigenome of in vitro activated and expanded CD8+ T cells, and potentiates their antitumor efficacy. Upon T cell receptor activation and IL-2 signaling, a timely and transient inhibition of LSD1 suffices to improve the memory phenotype of mouse CD8+ T cells, associated with a better ability to produce multiple cytokines, resist exhaustion, and persist in both antigen-dependent and -independent manners after adoptive transfer. Consequently, OT1 cells primed with LSD1 inhibitors demonstrate an enhanced antitumor effect in OVA-expressing solid tumor models implanted in female mice, both as a standalone treatment and in combination with PD-1 blockade. Moreover, priming with LSD1 inhibitors promotes polyfunctionality of human CD8+ T cells, and increases the persistence and antitumor efficacy of human CD19-CAR T cells in both leukemia and solid tumor models. Thus, pharmacological inhibition of LSD1 could be exploited to improve adoptive T cell therapy.


Sujet(s)
Lymphocytes T CD8+ , Histone Demethylases , Histone Demethylases/antagonistes et inhibiteurs , Histone Demethylases/métabolisme , Animaux , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Souris , Humains , Femelle , Souris de lignée C57BL , Immunothérapie adoptive/méthodes , Lignée cellulaire tumorale , Activation des lymphocytes/effets des médicaments et des substances chimiques , Transfert adoptif , Tumeurs/immunologie , Tumeurs/thérapie , Tumeurs/traitement médicamenteux , Récepteurs aux antigènes des cellules T/métabolisme , Récepteurs aux antigènes des cellules T/immunologie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/métabolisme , Interleukine-2/métabolisme , Antigènes CD19/métabolisme , Antigènes CD19/immunologie , Mémoire immunologique/effets des médicaments et des substances chimiques
17.
J Transl Med ; 22(1): 482, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38773607

RÉSUMÉ

BACKGROUND: Cellular immunotherapy, represented by the chimeric antigen receptor T cell (CAR-T), has exhibited high response rates, durable remission, and safety in vitro and in clinical trials. Unfortunately, anti-CD19 CAR-T (CART-19) treatment alone is prone to relapse and has a particularly poor prognosis in relapsed/refractory (r/r) B-ALL patients. To date, addressing or reducing relapse remains one of the research priorities to achieve broad clinical application. METHODS: We manufactured second generation CART-19 cells and validated their efficacy and safety in vitro and in vivo. Through co-culture of Nalm-6 cells with short-term cultured CART-19 cells, CD19-negative Nalm-6 cells were detected by flow cytometry, and further investigation of the relapsed cells and their resistance mechanisms was evaluated in vitro. RESULTS: In this study, we demonstrated that CART-19 cells had enhanced and specific antileukemic activities, and the survival of B-ALL mouse models after CART-19 treatment was significantly prolonged. We then shortened the culture time and applied the serum-free culture to expand CAR-T cells, followed by co-culturing CART-19 cells with Nalm-6 cells. Surprisingly, we observed the proliferation of CD19-negative Nalm-6 cells around 28 days. Identification of potential resistance mechanisms showed that the relapsed cells express truncated CD19 proteins with decreased levels and, more importantly, CAR expression was detected on the relapsed cell surface, which may ultimately keep them antigen-negative. Furthermore, it was validated that CART-22 and tandem CART-22/19 cells could effectively kill the relapsed cells, but neither could completely eradicate them. CONCLUSIONS: We successfully generated CART-19 cells and obtained a CD19-negative refractory relapsed B-ALL cell line, providing new insights into the underlying mechanisms of resistance and a new in vitro model for the treatment of r/r B-ALL patients with low antigen density.


Sujet(s)
Antigènes CD19 , Récepteurs chimériques pour l'antigène , Antigènes CD19/métabolisme , Antigènes CD19/immunologie , Animaux , Humains , Récepteurs chimériques pour l'antigène/métabolisme , Récepteurs chimériques pour l'antigène/immunologie , Lignée cellulaire tumorale , Immunothérapie adoptive/méthodes , Résistance aux médicaments antinéoplasiques , Souris , Techniques de coculture , Tests d'activité antitumorale sur modèle de xénogreffe , Leucémie-lymphome lymphoblastique à précurseurs B/thérapie , Leucémie-lymphome lymphoblastique à précurseurs B/immunologie
18.
Front Immunol ; 15: 1412002, 2024.
Article de Anglais | MEDLINE | ID: mdl-38779668

RÉSUMÉ

Chimeric Antigen Receptor T-cell (CAR-T) therapy has transformed the treatment landscape for hematological malignancies, showing high efficacy in patients with relapsed or refractory (R/R) disease and otherwise poor prognosis in the pre-CAR-T era. These therapies have been usually administered in the inpatient setting due to the risk of cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). However, there is a growing interest in the transition to outpatient administration due to multiple reasons. We review available evidence regarding safety and feasibility of outpatient administration of CD19 targeted and BCMA targeted CAR T-cell therapy with an emphasis on the implementation of outpatient CAR-T programs in community-based centers.


Sujet(s)
Immunothérapie adoptive , Humains , Immunothérapie adoptive/effets indésirables , Immunothérapie adoptive/méthodes , Patients en consultation externe , Tumeurs hématologiques/thérapie , Tumeurs hématologiques/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Soins ambulatoires , Syndrome de libération de cytokines/thérapie , Syndrome de libération de cytokines/étiologie , Antigènes CD19/immunologie , Centres de santé communautaires
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...