Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 15.162
Filtrer
1.
Mol Imaging ; 23: 15353508241257924, 2024.
Article de Anglais | MEDLINE | ID: mdl-38952399

RÉSUMÉ

Chimeric antigen receptor (CAR)-T cell-based immunotherapy has emerged as a path-breaking strategy for certain hematological malignancies. Assessment of the response to CAR-T therapy using quantitative imaging techniques such as positron emission tomography/computed tomography (PET/CT) has been broadly investigated. However, the definitive role of PET/CT in CAR-T therapy remains to be established. [18F]FDG PET/CT has demonstrated high sensitivity and specificity for differentiating patients with a partial and complete response after CAR-T therapy in lymphoma. The early therapeutic response and immune-related adverse effects such as cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome can also be detected on [18F]FDG PET images. In otherwise asymptomatic lymphoma patients with partial response following CAR-T therapy, the only positive findings could be abnormal PET/CT results. In multiple myeloma, a negative [18F]FDG PET/CT after receiving B-cell maturation antigen-directed CAR-T therapy has been associated with a favorable prognosis. In leukemia, [18F]FDG PET/CT can detect extramedullary metastases and treatment responses after therapy. Hence, PET/CT is a valuable imaging tool for patients undergoing CAR-T therapy for pretreatment evaluation, monitoring treatment response, assessing safety, and guiding therapeutic strategies. Developing guidelines with standardized cutoff values for various PET parameters and tumor cell-specific tracers may improve the efficacy and safety of CAR-T therapy.


Sujet(s)
Tumeurs hématologiques , Tomographie par émission de positons couplée à la tomodensitométrie , Humains , Tomographie par émission de positons couplée à la tomodensitométrie/méthodes , Tumeurs hématologiques/thérapie , Tumeurs hématologiques/imagerie diagnostique , Tumeurs hématologiques/immunologie , Immunothérapie adoptive/méthodes , Immunothérapie/méthodes , Récepteurs chimériques pour l'antigène/usage thérapeutique , Fluorodésoxyglucose F18
2.
Front Immunol ; 15: 1400177, 2024.
Article de Anglais | MEDLINE | ID: mdl-38953027

RÉSUMÉ

Background: Chimeric antigen receptor T (CAR-T) cell therapies have achieved remarkable success in the treatment of hematological tumors. However, given the distinct features of solid tumors, particularly heterogeneity, metabolic aggressiveness, and fewer immune cells in tumor microenvironment (TME), the practical utility of CAR-T cells for solid tumors remains as a challenging issue. Meanwhile, although anti-PD-1 monoclonal antibody (mAb) has shown clinical efficacy, most mAbs also show limited clinical benefits for solid tumors due mainly to the issues associated with the lack of immune cells in TME. Thus, the infiltration of targeted immunological active cells into TME could generate synergistic efficacy for mAbs. Methods: We present a combinational strategy for solid tumor treatment, which combines armored-T cells to express Fc-gamma receptor I (FcγRI) fragment on the surfaces for targeting various tumors with therapeutically useful mAbs. Choosing CD20 and HER-2 as the targets, we characterized the in vitro and in vivo efficacy and latent mechanism of the combination drug by using flow cytometry, ELISA and other methods. Results: The combination and preprocessing of armored T-cells with corresponding antibody of Rituximab and Pertuzumab exerted profound anti-tumor effects, which is demonstrated to be mediated by synergistically produced antibody-dependent cellular cytotoxicity (ADCC) effects. Meanwhile, mAb was able to carry armored-T cell by preprocessing for the infiltration to TME in cell derived xenograft (CDX) model. Conclusions: This combination strategy showed a significant increase of safety profiles from the reduction of antibody doses. More importantly, the present strategy could be a versatile tool for a broad spectrum of cancer treatment, with a simple pairing of engineered T cells and a conventional antibody.


Sujet(s)
Tumeurs , Récepteurs du fragment Fc des IgG , Lymphocytes T , Microenvironnement tumoral , Récepteurs du fragment Fc des IgG/immunologie , Récepteurs du fragment Fc des IgG/métabolisme , Humains , Animaux , Souris , Tumeurs/immunologie , Tumeurs/thérapie , Tumeurs/traitement médicamenteux , Lymphocytes T/immunologie , Microenvironnement tumoral/immunologie , Anticorps monoclonaux/usage thérapeutique , Anticorps monoclonaux/immunologie , Lignée cellulaire tumorale , Tests d'activité antitumorale sur modèle de xénogreffe , Immunothérapie adoptive/méthodes , Récepteur ErbB-2/immunologie , Récepteur ErbB-2/antagonistes et inhibiteurs , Antinéoplasiques immunologiques/pharmacologie , Antinéoplasiques immunologiques/usage thérapeutique , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/génétique , Femelle , Antigènes CD20/immunologie
3.
Front Immunol ; 15: 1411393, 2024.
Article de Anglais | MEDLINE | ID: mdl-38962002

RÉSUMÉ

Chimeric antigen receptor (CAR) T-cell therapy has proven a breakthrough in cancer treatment in the last decade, giving unprecedented results against hematological malignancies. All approved CAR T-cell products, as well as many being assessed in clinical trials, are generated using viral vectors to deploy the exogenous genetic material into T-cells. Viral vectors have a long-standing clinical history in gene delivery, and thus underwent iterations of optimization to improve their efficiency and safety. Nonetheless, their capacity to integrate semi-randomly into the host genome makes them potentially oncogenic via insertional mutagenesis and dysregulation of key cellular genes. Secondary cancers following CAR T-cell administration appear to be a rare adverse event. However several cases documented in the last few years put the spotlight on this issue, which might have been underestimated so far, given the relatively recent deployment of CAR T-cell therapies. Furthermore, the initial successes obtained in hematological malignancies have not yet been replicated in solid tumors. It is now clear that further enhancements are needed to allow CAR T-cells to increase long-term persistence, overcome exhaustion and cope with the immunosuppressive tumor microenvironment. To this aim, a variety of genomic engineering strategies are under evaluation, most relying on CRISPR/Cas9 or other gene editing technologies. These approaches are liable to introduce unintended, irreversible genomic alterations in the product cells. In the first part of this review, we will discuss the viral and non-viral approaches used for the generation of CAR T-cells, whereas in the second part we will focus on gene editing and non-gene editing T-cell engineering, with particular regard to advantages, limitations, and safety. Finally, we will critically analyze the different gene deployment and genomic engineering combinations, delineating strategies with a superior safety profile for the production of next-generation CAR T-cell.


Sujet(s)
Édition de gène , Immunothérapie adoptive , Récepteurs chimériques pour l'antigène , Lymphocytes T , Humains , Immunothérapie adoptive/méthodes , Immunothérapie adoptive/effets indésirables , Récepteurs chimériques pour l'antigène/génétique , Récepteurs chimériques pour l'antigène/immunologie , Édition de gène/méthodes , Lymphocytes T/immunologie , Animaux , Tumeurs/thérapie , Tumeurs/immunologie , Tumeurs/génétique , Vecteurs génétiques/génétique , Vecteurs génétiques/immunologie , Génie génétique , Systèmes CRISPR-Cas , Récepteurs aux antigènes des cellules T/génétique , Récepteurs aux antigènes des cellules T/immunologie , Microenvironnement tumoral/immunologie
4.
Front Immunol ; 15: 1383894, 2024.
Article de Anglais | MEDLINE | ID: mdl-38962014

RÉSUMÉ

Chimeric antigen receptor (CAR) T cell therapy has effectively complemented the treatment of advanced relapsed and refractory hematological cancers. The remarkable achievements of CD19- and BCMA-CAR T therapies have raised high expectations within the fields of hematology and oncology. These groundbreaking successes are propelling a collective aspiration to extend the reach of CAR therapies beyond B-lineage malignancies. Advanced CAR technologies have created a momentum to surmount the limitations of conventional CAR concepts. Most importantly, innovations that enable combinatorial targeting to address target antigen heterogeneity, using versatile adapter CAR concepts in conjunction with recent transformative next-generation CAR design, offer the promise to overcome both the bottleneck associated with CAR manufacturing and patient-individualized treatment regimens. In this comprehensive review, we delineate the fundamental prerequisites, navigate through pivotal challenges, and elucidate strategic approaches, all aimed at paving the way for the future establishment of multitargeted immunotherapies using universal CAR technologies.


Sujet(s)
Immunothérapie adoptive , Récepteurs chimériques pour l'antigène , Humains , Immunothérapie adoptive/méthodes , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/génétique , Animaux , Lymphocytes T/immunologie , Antigènes CD19/immunologie , Tumeurs hématologiques/thérapie , Tumeurs hématologiques/immunologie , Récepteurs aux antigènes des cellules T/immunologie , Récepteurs aux antigènes des cellules T/génétique , Tumeurs/thérapie , Tumeurs/immunologie
6.
Zhonghua Xue Ye Xue Za Zhi ; 45(5): 436-444, 2024 May 14.
Article de Chinois | MEDLINE | ID: mdl-38964917

RÉSUMÉ

Objective: To construct a novel chimeric antigen receptor T (CAR-T) cell targeting CD138 and to investigate its cytotoxicity against myeloma cells. Methods: The hybridoma strain that can stably secrete the CD138 monoclonal antibody (mAb) was prepared and obtained through monoclonal antibody screening technology. The hybridoma strain cells were intraperitoneally injected into mice to produce ascites containing monoclonal antibodies, which were then collected and purified to obtain pure CD138 mAb. Further examinations were performed to assess the biological characteristics of CD138 mAb. The variable region sequence of this antibody was amplified through reverse transcription polymerase chain reaction and was used as the antigen recognition domain of CD138 CAR, which was subsequently expressed on the surface of T cells by lentiviral infection. Flow cytometry was employed to assess the phenotype of CD138 CAR-T cells. In vitro cytotoxicity and degranulation assays were performed to evaluate their antitumor effects. Results: ① We successfully prepared anti-human CD138 antibody hybridoma cell lines and screened a hybridoma cell strain, 5G2, which could persistently and stably secrete the anti-CD138 antibody. ② The purified CD138 (5G2) mAb can especially recognize CD138(+) cells with a binding affinity constant (K(D)) of 6.011×10(-9) mol/L and showed no significant binding activity with CD138(-) cells. ③The variable region sequence of the CD138 (5G2) antibody was obtained using molecular cloning technology, and CD138 (5G2) CAR was successfully constructed and expressed on T cells through lentivirus infection and, concurrently, demonstrated effective binding to recombinant human CD138 protein.④ The proliferation of T cells transduced with the CD138 (5G2) CAR was highly efficient. The phenotype analysis revealed that CD138 (5G2) CAR-T cells exhibited a greater tendency to differentiate into central memory T cells and memory stem T cells, with a reduced proportion of terminally differentiated effector memory subsets. ⑤CD138 (5G2) CAR-T cells demonstrated specific cytotoxicity against CD138(+) myeloma cell line H929, whereas CD138(-) cell line K562 remained unaffected. The percentage of residual H929 cells was (12.92±8.02) % after co-culturing with CD138 (5G2) CAR-T cells, while (54.25±15.79) % was left in the Vector-T group (E∶T=1∶2; P<0.001). ⑥Results of degranulation assays demonstrated a significant activation of CD138 (5G2) CAR-T cells after co-culture with the H929 cell line, whereas no significant activation was observed in Vector-T cells [ (25.78±3.35) % vs (6.13±1.30) %, P<0.001]. ⑦After co-culturing with CD138(+) cells, CD138 (5G2) CAR-T cells exhibited a significant increase in cytokine secretion compared to the Vector-T group [interleukin-2: (1 697.52±599.05) pg/ml vs (5.07±1.17) pg/ml, P<0.001; interferon-γ: (3 312.20±486.38) pg/ml vs (9.28±1.46) pg/ml, P<0.001; and tumor necrosis factor-α: (1 837.43±640.49) pg/ml vs (8.75±1.65) pg/ml, P<0.001]. However, no significant difference was observed in cytokine secretion levels between the two groups after co-culturing with CD138(-) cells. Conclusion: This study successfully prepared a novel monoclonal antibody against CD138, and CAR-T cells constructed with the antigen recognition domain derived from this 5G2 mAb demonstrated effective antitumor activity against myeloma cells. This can be used as a new option for the detection of the CD138 antigen and proposes a novel strategy for multiple myeloma immunotherapy.


Sujet(s)
Myélome multiple , Récepteurs chimériques pour l'antigène , Syndécane-1 , Lymphocytes T , Myélome multiple/thérapie , Myélome multiple/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Souris , Animaux , Humains , Syndécane-1/immunologie , Lymphocytes T/immunologie , Hybridomes , Immunothérapie adoptive/méthodes , Lignée cellulaire tumorale , Récepteurs aux antigènes des cellules T/immunologie , Récepteurs aux antigènes des cellules T/génétique , Anticorps monoclonaux/immunologie
7.
Rinsho Ketsueki ; 65(6): 597-602, 2024.
Article de Japonais | MEDLINE | ID: mdl-38960662

RÉSUMÉ

CAR-T cell therapy targeting CD19 and BCMA for relapsed or refractory hematopoietic tumors has been adopted in routine practice and has shown dramatic results. However, half of patients who achieve remission with CAR-T therapy eventually relapse, and thus efforts to improve the efficacy of CAR-T therapy are gaining momentum. Notably, studies have described innovative technologies that enable control of cell kinetics after infusion, which is not possible with conventional CAR-T therapies. In this article, we review the challenges of CAR-T cell therapy and the development of new technologies.


Sujet(s)
Immunothérapie adoptive , Humains , Immunothérapie adoptive/méthodes , Tumeurs hématologiques/thérapie , Antigènes CD19/immunologie , Résultat thérapeutique , Récepteurs chimériques pour l'antigène
8.
Cancer Immunol Immunother ; 73(9): 179, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38960949

RÉSUMÉ

Adoptive cellular therapy (ACT) using memory-like (ML) natural killer (NK) cells, generated through overnight ex vivo activation with IL-12, IL-15, and IL-18, has shown promise for treating hematologic malignancies. We recently reported that a multifunctional fusion molecule, HCW9201, comprising IL-12, IL-15, and IL-18 domains could replace individual cytokines for priming human ML NK cell programming ("Prime" step). However, this approach does not include ex vivo expansion, thereby limiting the ability to test different doses and schedules. Here, we report the design and generation of a multifunctional fusion molecule, HCW9206, consisting of human IL-7, IL-15, and IL-21 cytokines. We observed > 300-fold expansion for HCW9201-primed human NK cells cultured for 14 days with HCW9206 and HCW9101, an IgG1 antibody, recognizing the scaffold domain of HCW9206 ("Expand" step). This expansion was dependent on both HCW9206 cytokines and interactions of the IgG1 mAb with CD16 receptors on NK cells. The resulting "Prime and Expand" ML NK cells exhibited elevated metabolic capacity, stable epigenetic IFNG promoter demethylation, enhanced antitumor activity in vitro and in vivo, and superior persistence in NSG mice. Thus, the "Prime and Expand" strategy represents a simple feeder cell-free approach to streamline manufacturing of clinical-grade ML NK cells to support multidose and off-the-shelf ACT.


Sujet(s)
Mémoire immunologique , Cellules tueuses naturelles , Protéines de fusion recombinantes , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Humains , Animaux , Protéines de fusion recombinantes/génétique , Souris , Thérapie cellulaire et tissulaire/méthodes , Immunothérapie adoptive/méthodes , Interleukine-15/métabolisme
9.
J Vis Exp ; (208)2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38949315

RÉSUMÉ

Extensive studies have proven the promise of chimeric antigen receptor T (CAR-T) cell therapy in treating hematological malignancies. However, treating solid tumors remains challenging, as exemplified by the safety concerns that arise when CAR-T cells attack normal cells expressing the target antigens. Researchers have explored various approaches to enhance the tumor selectivity of CAR-T cell therapy. One representative strategy along this line is the construction of hypoxia-sensitive CAR-T cells, which are designed by fusing an oxygen-dependent degradation domain to the CAR moiety and are strategized to attain high CAR expression only in a hypoxic environment-the tumor microenvironment (TME). This paper presents a protocol for the generation of such CAR-T cells and their functional characterization, including methods to analyze the changes in CAR expression and killing capacity in response to different oxygen levels established by a mobile incubator chamber. The constructed CAR-T cells are anticipated to demonstrate CAR expression and cytotoxicity in an oxygen-sensitive manner, thus supporting their capability to distinguish between hypoxic TME and normoxic normal tissues for selective activation.


Sujet(s)
Récepteurs chimériques pour l'antigène , Lymphocytes T , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/génétique , Humains , Lymphocytes T/immunologie , Immunothérapie adoptive/méthodes , Hypoxie cellulaire/physiologie , Microenvironnement tumoral/immunologie
12.
Cancer Immunol Immunother ; 73(9): 168, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38953939

RÉSUMÉ

For advanced therapy medicinal products, the development and validation of potency assays are required, in accordance with international guidelines, to characterise the product and obtain reliable and consistent data. Our purpose was to validate the killing assay for the evaluation of autologous anti-CD19 chimeric antigen receptor (CAR) T potency. We used CD4 + and CD8 + lymphocytes or anti-CD19 CAR-T cells as effector cells and REH (CD19 +) or MOLM-13 (CD19 -) cell lines as target cells. After co-culturing target and effector cells (1:1 ratio) for 24 h, samples were labelled with 7-AAD, anti-CD3 and anti-CD19 antibodies and the frequency of CD19 + dead cells was evaluated by flow cytometry. In order to verify the CAR-T specificity for the CD19 + target, the co-culture between CAR-T and REH or MOLM-13 at different effector-to-target ratios was scheduled. Moreover, not transduced CD4 + and CD8 + lymphocytes were tested in comparison with CAR-T from the same donor to demonstrate the assay specificity. Linearity and accuracy were evaluated, and established acceptance criteria were compiled for both parameters (r2 ≥ 0.97 for linearity and average relative error ≤ 10% for accuracy). Furthermore, the method was considered robust when performed between 23 and 25 h of co-culture, and the intra-assay, inter-assay and inter-day precision was obtained. Finally, in order to verify the inter-analyst precision, the test was executed by three different operators and the intra-class correlation coefficient was > 0.4 in both cases. In conclusion, we consider this CAR-T potency assay as validated and usable in all steps of product development and quality control.


Sujet(s)
Antigènes CD19 , Immunothérapie adoptive , Récepteurs chimériques pour l'antigène , Humains , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/métabolisme , Immunothérapie adoptive/méthodes , Antigènes CD19/immunologie , Techniques de coculture , Lymphocytes T CD8+/immunologie , Cytotoxicité immunologique , Lignée cellulaire tumorale , Lymphocytes T CD4+/immunologie
13.
Cancer Immunol Immunother ; 73(9): 163, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38954005

RÉSUMÉ

In addition to their immunosuppressive effect, cytostatics conditioning prior to adoptive therapy such as chimeric antigen receptor (CAR) T cells may play a role in debulking and remodeling the tumor microenvironment. We investigated in vitro the killing efficacy and impact of treosulfan and fludarabine on ovarian cancer cells expressing mesothelin (MSLN) and effect on MSLN-targeting CAR T cells. Treosulfan and fludarabine had a synergetic effect on killing of SKOV3 and OVCAR4 cells. Sensitivity to the combination of treosulfan and fludarabine was increased when SKOV3 cells expressed MSLN and when OVCAR4 cells were tested in hypoxia, while MSLN cells surface expression by SKOV3 and OVCAR4 cells was not altered after treosulfan or fludarabine exposure. Exposure to treosulfan or fludarabine (10 µM) neither impacted MSLN-CAR T cells degranulation, cytokines production upon challenge with MSLN + OVCAR3 cells, nor induced mitochondrial defects. Combination of treosulfan and fludarabine decreased MSLN-CAR T cells anti-tumor killing in normoxia but not hypoxia. In conclusion, treosulfan and fludarabine killed MSLN + ovarian cancer cells without altering MSLN-CAR T cells functions (at low cytostatics concentration) even in hypoxic conditions, and our data support the use of treosulfan and fludarabine as conditioning drugs prior to MSLN-CAR T cell therapy.


Sujet(s)
Busulfan , Protéines liées au GPI , Immunothérapie adoptive , Mésothéline , Tumeurs de l'ovaire , Récepteurs chimériques pour l'antigène , Vidarabine , Vidarabine/analogues et dérivés , Vidarabine/pharmacologie , Humains , Femelle , Tumeurs de l'ovaire/immunologie , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/thérapie , Récepteurs chimériques pour l'antigène/immunologie , Busulfan/analogues et dérivés , Busulfan/pharmacologie , Immunothérapie adoptive/méthodes , Lignée cellulaire tumorale , Lymphocytes T/immunologie , Lymphocytes T/effets des médicaments et des substances chimiques , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie
14.
Cancer Immunol Immunother ; 73(9): 173, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38953982

RÉSUMÉ

Recent studies have indicated that combining oncolytic viruses with CAR-T cells in therapy has shown superior anti-tumor effects, representing a promising approach. Nonetheless, the localized delivery method of intratumoral injection poses challenges for treating metastatic tumors or distal tumors that are difficult to reach. To address this obstacle, we employed HSV-1-infected CAR-T cells, which systemically delivery HSV into solid tumors. The biological function of CAR-T cells remained intact after loading them with HSV for a period of three days. In both immunocompromised and immunocompetent GBM orthotopic mouse models, B7-H3 CAR-T cells effectively delivered HSV to tumor lesions, resulting in enhanced T-cell infiltration and significantly prolonged survival in mice. We also employed a bilateral subcutaneous tumor model and observed that the group receiving intratumoral virus injection exhibited a significant reduction in tumor volume on the injected side, while the group receiving intravenous infusion of CAR-T cells carrying HSV displayed suppressed tumor growth on both sides. Hence, CAR-THSV cells offer notable advantages in the systemic delivery of HSV to distant tumors. In conclusion, our findings emphasize the potential of CAR-T cells as carriers for HSV, presenting significant advantages for oncolytic virotherapy targeting distant tumors.


Sujet(s)
Immunothérapie adoptive , Thérapie virale de cancers , Virus oncolytiques , Récepteurs chimériques pour l'antigène , Animaux , Souris , Thérapie virale de cancers/méthodes , Humains , Virus oncolytiques/immunologie , Virus oncolytiques/génétique , Immunothérapie adoptive/méthodes , Récepteurs chimériques pour l'antigène/immunologie , Herpèsvirus humain de type 1/immunologie , Tests d'activité antitumorale sur modèle de xénogreffe , Lignée cellulaire tumorale , Lymphocytes T/immunologie , Femelle , Glioblastome/thérapie , Glioblastome/immunologie
15.
J Immunother Cancer ; 12(7)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38955420

RÉSUMÉ

BACKGROUND: Fludarabine in combination with cyclophosphamide (FC) is the standard lymphodepletion regimen for CAR T-cell therapy (CAR T). A national fludarabine shortage in 2022 necessitated the exploration of alternative regimens with many centers employing single-agent bendamustine as lymphodepletion despite a lack of clinical safety and efficacy data. To fill this gap in the literature, we evaluated the safety, efficacy, and expansion kinetics of bendamustine as lymphodepletion prior to axicabtagene ciloleucel (axi-cel) therapy. METHODS: 84 consecutive patients with relapsed or refractory large B-cell lymphoma treated with axi-cel and managed with a uniform toxicity management plan at Stanford University were studied. 27 patients received alternative lymphodepletion with bendamustine while 57 received FC. RESULTS: Best complete response rates were similar (73.7% for FC and 74% for bendamustine, p=0.28) and there was no significant difference in 12-month progression-free survival or overall survival estimates (p=0.17 and p=0.62, respectively). The frequency of high-grade cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome was similar in both the cohorts. Bendamustine cohort experienced lower proportions of hematological toxicities and antibiotic use for neutropenic fever. Immune reconstitution, as measured by quantitative assessment of cellular immunity, was better in bendamustine cohort as compared with FC cohort. CAR T expansion as measured by peak expansion and area under the curve for expansion was comparable between cohorts. CONCLUSIONS: Bendamustine is a safe and effective alternative lymphodepletion conditioning for axi-cel with lower early hematological toxicity and favorable immune reconstitution.


Sujet(s)
Chlorhydrate de bendamustine , Produits biologiques , Lymphome B diffus à grandes cellules , Humains , Chlorhydrate de bendamustine/usage thérapeutique , Chlorhydrate de bendamustine/administration et posologie , Mâle , Femelle , Adulte d'âge moyen , Sujet âgé , Lymphome B diffus à grandes cellules/traitement médicamenteux , Produits biologiques/usage thérapeutique , Produits biologiques/effets indésirables , Adulte , Immunothérapie adoptive/méthodes , Immunothérapie adoptive/effets indésirables , Antigènes CD19/immunologie , Antigènes CD19/usage thérapeutique
16.
J Immunother Cancer ; 12(7)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38955421

RÉSUMÉ

BACKGROUND: Adoptive cell therapy using genetically modified T cells to express chimeric antigen receptors (CAR-T) has shown encouraging results, particularly in certain blood cancers. Nevertheless, over 40% of B cell malignancy patients experience a relapse after CAR-T therapy, likely due to inadequate persistence of the modified T cells in the body. IL15, known for its pro-survival and proliferative properties, has been suggested for incorporation into the fourth generation of CAR-T cells to enhance their persistence. However, the potential systemic toxicity associated with this cytokine warrants further evaluation. METHODS: We analyzed the persistence, antitumor efficacy and potential toxicity of anti-mouse CD19 CAR-T cells which express a membrane-bound IL15-IL15Rα chimeric protein (CD19/mbIL15q CAR-T), in BALB/c mice challenged with A20 tumor cells as well as in NSG mice. RESULTS: Conventional CD19 CAR-T cells showed low persistence and poor efficacy in BALB/c mice treated with mild lymphodepletion regimens (total body irradiation (TBI) of 1 Gy). CD19/mbIL15q CAR-T exhibits prolonged persistence and enhanced in vivo efficacy, effectively eliminating established A20 B cell lymphoma. However, this CD19/mbIL15q CAR-T displays important long-term toxicities, with marked splenomegaly, weight loss, transaminase elevations, and significant inflammatory findings in some tissues. Mice survival is highly compromised after CD19/mbIL15q CAR-T cell transfer, particularly if a high TBI regimen is applied before CAR-T cell transfer. CONCLUSION: Tethered IL15-IL15Rα augments the antitumor activity of CD19 CAR-T cells but displays long-term toxicity in immunocompetent mice. Inducible systems to regulate IL15-IL15Rα expression could be considered to control this toxicity.


Sujet(s)
Antigènes CD19 , Immunothérapie adoptive , Interleukine-15 , Animaux , Souris , Antigènes CD19/immunologie , Immunothérapie adoptive/méthodes , Humains , Modèles animaux de maladie humaine , Lignée cellulaire tumorale , Femelle , Sous-unité alpha du récepteur à l'interleukine-15 , Récepteurs chimériques pour l'antigène/immunologie , Lymphomes/thérapie , Lymphomes/immunologie , Souris de lignée BALB C , Lymphocytes T/immunologie , Lymphocytes T/transplantation
17.
Sci Adv ; 10(28): eadn0881, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38996027

RÉSUMÉ

Epithelial ovarian cancer (EOC) remains one of the most lethal gynecological cancers. Cytokine-induced memory-like (CIML) natural killer (NK) cells have shown promising results in preclinical and early-phase clinical trials. In the current study, CIML NK cells demonstrated superior antitumor responses against a panel of EOC cell lines, increased expression of activation receptors, and up-regulation of genes involved in cell cycle/proliferation and down-regulation of inhibitory/suppressive genes. CIML NK cells transduced with a chimeric antigen receptor (CAR) targeting the membrane-proximal domain of mesothelin (MSLN) further improved the antitumor responses against MSLN-expressing EOC cells and patient-derived xenograft tumor cells. CAR arming of the CIML NK cells subtanstially reduced their dysfunction in patient-derived ascites fluid with transcriptomic changes related to altered metabolism and tonic signaling as potential mechanisms. Lastly, the adoptive transfer of MSLN-CAR CIML NK cells demonstrated remarkable inhibition of tumor growth and prevented metastatic spread in xenograft mice, supporting their potential as an effective therapeutic strategy in EOC.


Sujet(s)
Cellules tueuses naturelles , Mésothéline , Tumeurs de l'ovaire , Récepteurs chimériques pour l'antigène , Tests d'activité antitumorale sur modèle de xénogreffe , Humains , Animaux , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Femelle , Souris , Tumeurs de l'ovaire/métabolisme , Tumeurs de l'ovaire/anatomopathologie , Tumeurs de l'ovaire/immunologie , Tumeurs de l'ovaire/thérapie , Lignée cellulaire tumorale , Récepteurs chimériques pour l'antigène/métabolisme , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/génétique , Protéines liées au GPI/métabolisme , Protéines liées au GPI/génétique , Immunothérapie adoptive/méthodes , Carcinome épithélial de l'ovaire/métabolisme , Carcinome épithélial de l'ovaire/anatomopathologie , Carcinome épithélial de l'ovaire/immunologie , Carcinome épithélial de l'ovaire/thérapie , Mémoire immunologique , Domaines protéiques
18.
Cancer Immunol Immunother ; 73(9): 170, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38954079

RÉSUMÉ

Chimeric antigen receptor (CAR) T cell therapy has shown promising results in hematologic malignancies, but its effectiveness in solid cancers remains challenging. Macrophages are immune cells residing within the tumor microenvironment. They can phagocytose tumor cells. Recently, CAR macrophages (CAR-M) have been a promising candidate for treating solid cancers. One of the common cancer antigens overexpressed in various types of cancer is CD147. CAR-T and NK cells targeting CD147 antigen have shown significant efficacy against hepatocellular carcinoma. Nevertheless, CAR-M targeting the CD147 molecule has not been investigated. In this study, we generated CAR targeting the CD147 molecule using the THP-1 monocytic cell line (CD147 CAR-M). The CD147 CAR-M exhibited typical macrophage characteristics, including phagocytosis of zymosan bioparticles and polarization ability toward M1 and M2 phenotypes. Furthermore, the CD147 CAR-M demonstrated enhanced anti-tumor activity against K562 and MDA-MB-231 cells without exhibiting off-target cytotoxicity against normal cells. Our research provides valuable insights into the potential of CD147 CAR-M as a promising platform for cancer immunotherapy, with applications in both hematologic malignancies and solid cancers.


Sujet(s)
Antigènes CD147 , Immunothérapie adoptive , Macrophages , Phagocytose , Récepteurs chimériques pour l'antigène , Humains , Phagocytose/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/génétique , Récepteurs chimériques pour l'antigène/métabolisme , Macrophages/immunologie , Macrophages/métabolisme , Immunothérapie adoptive/méthodes , Antigènes CD147/immunologie , Antigènes CD147/métabolisme , Tumeurs/immunologie , Tumeurs/thérapie , Souris , Animaux , Lignée cellulaire tumorale , Microenvironnement tumoral/immunologie
19.
Blood Cancer J ; 14(1): 108, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38977682

RÉSUMÉ

In patients with relapsed DLBCL in complete remission (CR), autologous hematopoietic cell transplantation (auto-HCT) and CAR-T therapy are both effective, but it is unknown which modality provides superior outcomes. We compared the efficacy of auto-HCT vs. CAR-T in patients with DLBCL in a CR. A retrospective observational study comparing auto-HCT (2015-2021) vs. CAR-T (2018-2021) using the Center for International Blood & Marrow Transplant Research registry. Median follow-up was 49.7 months for the auto-HCT and 24.7 months for the CAR-T cohort. Patients ages 18 and 75 with a diagnosis of DLBCL were included if they received auto-HCT (n = 281) or commercial CAR-T (n = 79) while in a CR. Patients undergoing auto-HCT with only one prior therapy line and CAR-T patients with a previous history of auto-HCT treatment were excluded. Endpoints included Progression-free survival (PFS), relapse rate, non-relapse mortality (NRM) and overall survival (OS). In univariate analysis, treatment with auto-HCT was associated with a higher rate of 2-year PFS (66.2% vs. 47.8%; p < 0.001), a lower 2-year cumulative incidence of relapse (27.8% vs. 48% ; p < 0.001), and a superior 2-year OS (78.9% vs. 65.6%; p = 0.037). In patients with early (within 12 months) treatment failure, auto-HCT was associated with a superior 2-year PFS (70.9% vs. 48.3% ; p < 0.001), lower 2-year cumulative incidence of relapse (22.8% vs. 45.9% ; p < 0.001) and trend for higher 2-year OS (82.4% vs. 66.1% ; p = 0.076). In the multivariable analysis, treatment with auto-HCT was associated with a superior PFS (hazard ratio 1.83; p = 0.0011) and lower incidence of relapse (hazard ratio 2.18; p < 0.0001) compared to CAR-T. In patients with relapsed LBCL who achieve a CR, treatment with auto-HCT is associated with improved clinical outcomes compared to CAR-T. These data support the consideration of auto-HCT in select patients with LBCL achieving a CR in the relapsed setting.


Sujet(s)
Transplantation de cellules souches hématopoïétiques , Immunothérapie adoptive , Lymphome B diffus à grandes cellules , Transplantation autologue , Humains , Lymphome B diffus à grandes cellules/thérapie , Lymphome B diffus à grandes cellules/mortalité , Adulte d'âge moyen , Femelle , Mâle , Adulte , Études rétrospectives , Sujet âgé , Transplantation de cellules souches hématopoïétiques/méthodes , Immunothérapie adoptive/méthodes , Jeune adulte , Induction de rémission , Adolescent , Résultat thérapeutique ,
20.
J Transl Med ; 22(1): 633, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38978106

RÉSUMÉ

BACKGROUND: Pancreatic cancer is one of the most lethal malignancies and the lack of treatment options makes it more deadly. Chimeric Antigen Receptor T-cell (CAR-T) immunotherapy has revolutionized cancer treatment and made great breakthroughs in treating hematological malignancies, however its success in treating solid cancers remains limited mainly due to the lack of tumor-specific antigens. On the other hand, the prolonged traditional manufacturing process poses challenges, taking 2 to 6 weeks and impacting patient outcomes. CD276 has recently emerged as a potential therapeutic target for anti-solid cancer therapy. Here, we investigated the efficacy of CD276 CAR-T and rapidly-manufactured CAR-T against pancreatic cancer. METHODS: In the present study, CD276 CAR-T was prepared by CAR structure carrying 376.96 scFv sequence, CD8 hinge and transmembrane domain, 4-1BB and CD3ζ intracellular domains. Additionally, CD276 rapidly-manufactured CAR-T (named CD276 Dash CAR-T) was innovatively developed by shortening the duration of ex vitro culture to reduce CAR-T manufacturing time. We evaluated the anti-tumor efficacy of CD276 CAR-T and further compared the functional assessment of Dash CAR-T and conventional CAR-T in vitro and in vivo by detecting the immunophenotypes, killing ability, expansion capacity and tumor-eradicating effect of CAR-T. RESULTS: We found that CD276 was strongly expressed in multiple solid cancer cell lines and that CD276 CAR-T could efficiently kill these solid cancer cells. Moreover, Dash CAR-T was successfully manufactured within 48-72 h and the functional validation was carried out subsequently. In vitro, CD276 Dash CAR-T possessed a less-differentiated phenotype and robust proliferative ability compared to conventional CAR-T. In vivo xenograft mouse model, CD276 Dash CAR-T showed enhanced anti-pancreatic cancer efficacy and T cell expansion. Besides, except for the high-dose group, the body weight of mice was maintained stable, and the state of mice was normal. CONCLUSIONS: In this study, we proved CD276 CAR-T exhibited powerful activity against pancreatic cancer cells in vitro and in vivo. More importantly, we demonstrated the manufacturing feasibility, acceptable safety and superior anti-tumor efficacy of CD276 Dash CAR-T generated with reduced time. The results of the above studies indicated that CD276 Dash CAR-T immunotherapy might be a novel and promising strategy for pancreatic cancer treatment.


Sujet(s)
Antigènes B7 , Immunothérapie adoptive , Tumeurs du pancréas , Récepteurs chimériques pour l'antigène , Tumeurs du pancréas/thérapie , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/anatomopathologie , Humains , Animaux , Lignée cellulaire tumorale , Récepteurs chimériques pour l'antigène/métabolisme , Récepteurs chimériques pour l'antigène/immunologie , Immunothérapie adoptive/méthodes , Antigènes B7/métabolisme , Antigènes B7/immunologie , Tests d'activité antitumorale sur modèle de xénogreffe , Souris , Prolifération cellulaire , Lymphocytes T/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE