Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 7.246
Filtrer
1.
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
2.
Rinsho Ketsueki ; 65(6): 536-546, 2024.
Article de Japonais | MEDLINE | ID: mdl-38960654

RÉSUMÉ

Advances in understanding of the pathogenesis of B-cell lymphoma have led to development of various novel targeted therapies. Among them, CD19-targeted chimeric antigen receptor (CAR) T-cell therapies for relapsed and refractory B-cell lymphomas have shown remarkable efficacy in clinical trials, and three CAR T-cell products are now available in Japan. Real-world evidence (RWE) has shown that these products can provide comparable efficacy to clinical trials in clinical practice, where CAR T-cells were administered in patients with wider range of backgrounds. This finding will certainly broaden the role of CAR T-cell therapies in the treatment of B-cell lymphoma. However, since about half of the patients treated with CAR T-cell therapy progress thereafter, there is an urgent need for risk stratification and optimized management of refractory cases. Here, we review the results of clinical trials and RWE of CAR T-cell therapy in B-cell lymphoma.


Sujet(s)
Immunothérapie adoptive , Humains , Lymphomes/thérapie , Lymphomes/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Essais cliniques comme sujet , Lymphome B/thérapie , Lymphome B/immunologie
3.
Int J Immunopathol Pharmacol ; 38: 3946320241264369, 2024.
Article de Anglais | MEDLINE | ID: mdl-38886178

RÉSUMÉ

Background: Typically, lymphatic tissue proliferative lesions include either benign lesions or lymphoma. However, not all lymphatic lesions can currently be accurately classified into one category, particularly in mucosal areas that are in contact with the external environment.Aims: To explore the morphology, immunophenotype, and molecular changes of Non-neoplastic B-cell predominant lymphoid proliferations (NBPLP) in pathological areas that are exposed to external surroundings which mimicked lymphoma.Methods and Results: 18 cases of Atypical lymphoid hyperplasia (AtLP)  were retrieved in this study. The biopsy samples were mucosal samples obtained from areas exposed to external surroundings, including intestines, urethra, cervix, tonsils, and tongue. Microscopically, there is a different level of B cell hyperplasia accompanied by morphological atypia. We categorized the morphology into 4 groups: type A (7/18), type B (3/18), type C (3/18), type D (5/18). Part of the AtLP was found positive for BCR gene rearrangement (6/15), and TCR gene rearrangement (1/4). The follow-up period ranged from 14.2 to 70 months. No evidence of lymphoma was found. Therefore, we diagnosed all of the presented cases as NBPLP. We illustrated the key differential points and provided valuable diagnostic experience on each subtype.Conclusions: Areas exposed to the external environment are commonly exposed to antigen and easily present with AtLP of NBPLP, accompanying with positive IGH rearrangement. Therefore, a comprehensive evaluation of macroscopic, morphology, immunophenotype, and molecular diagnostics is required to prevent the overdiagnosis of lymphoma.


Sujet(s)
Lymphocytes B , Lymphomes , Humains , Femelle , Adulte d'âge moyen , Mâle , Lymphocytes B/immunologie , Lymphocytes B/anatomopathologie , Lymphomes/anatomopathologie , Lymphomes/diagnostic , Lymphomes/immunologie , Diagnostic différentiel , Adulte , Sujet âgé , Prolifération cellulaire , Immunophénotypage
4.
Am Soc Clin Oncol Educ Book ; 44(3): e433516, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38935881

RÉSUMÉ

This article endeavors to navigate the clinical journey of bispecific antibodies (BsAbs), from elucidating common toxicities and management strategies to examining novel agents and broadening access in community health care. These drugs, commonly through T-cell activation, result in shared adverse events such as cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome. Variations in target antigens and designs, however, might introduce unique toxicities for different BsAbs, warranting specific management approaches. Recent US Food and Drug Administration approvals of BsAbs targeting CD3+ T cells linked to CD20 for non-Hodgkin lymphoma and to B-cell maturation antigen or GPRC5D for multiple myeloma have transformed the treatment landscape for hematologic malignancies. Emerging new agents promise further enhancement and safety, exploring novel antigen targets, innovative structures such as trispecific antibodies, and the engagement of diverse immune cells. Simultaneously, the expansion of BsAbs into community practices is underway, demanding a multifaceted strategy that encompasses educational initiatives, operational adaptations, and collaborative frameworks. This ensures comprehensive treatment access, allowing every patient, irrespective of geographical or socioeconomic status, to benefit from these advancements in cancer therapy.


Sujet(s)
Anticorps bispécifiques , Myélome multiple , Humains , Anticorps bispécifiques/usage thérapeutique , Myélome multiple/traitement médicamenteux , Myélome multiple/immunologie , Lymphomes/traitement médicamenteux , Lymphomes/immunologie , Antinéoplasiques immunologiques/usage thérapeutique , Antinéoplasiques immunologiques/effets indésirables
5.
Zhonghua Bing Li Xue Za Zhi ; 53(6): 535-540, 2024 Jun 08.
Article de Chinois | MEDLINE | ID: mdl-38825896

RÉSUMÉ

The 5th edition of the World Health Organization (WHO) classification of haematolymphoid tumours used the hierarchical system to classify T-cell and NK-cell lymphoid proliferations and lymphomas (T/NK-LPD/LYM) based on research advances and clinicopathological characteristics of the diseases. In this edition of classification, tumour-like lesions were included, some tumors were added/deleted, the names or terms of certain diseases were refined, and the diagnostic criteria or subtypes of some diseases were revised. This group of diseases was reintegrated from non-clonal hyperplasia to highly aggressive lymphoma, which would further reflect the nature of T/NK-LPD/LYM and benefit to clinical application.


Sujet(s)
Cellules tueuses naturelles , Lymphomes , Lymphocytes T , Organisation mondiale de la santé , Humains , Cellules tueuses naturelles/anatomopathologie , Cellules tueuses naturelles/immunologie , Lymphocytes T/anatomopathologie , Lymphocytes T/immunologie , Lymphomes/anatomopathologie , Lymphomes/classification , Lymphomes/immunologie , Lymphome T/anatomopathologie , Lymphome T/classification , Lymphome T/immunologie , Syndromes lymphoprolifératifs/anatomopathologie , Syndromes lymphoprolifératifs/classification , Syndromes lymphoprolifératifs/immunologie
6.
Nat Commun ; 15(1): 5310, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38906867

RÉSUMÉ

Epstein-Barr virus (EBV) infects more than 95% of adults worldwide and is closely associated with various malignancies. Considering the complex life cycle of EBV, developing vaccines targeting key entry glycoproteins to elicit robust and durable adaptive immune responses may provide better protection. EBV gHgL-, gB- and gp42-specific antibodies in healthy EBV carriers contributed to sera neutralizing abilities in vitro, indicating that they are potential antigen candidates. To enhance the immunogenicity of these antigens, we formulate three nanovaccines by co-delivering molecular adjuvants (CpG and MPLA) and antigens (gHgL, gB or gp42). These nanovaccines induce robust humoral and cellular responses through efficient activation of dendritic cells and germinal center response. Importantly, these nanovaccines generate high levels of neutralizing antibodies recognizing vulnerable sites of all three antigens. IgGs induced by a cocktail vaccine containing three nanovaccines confer superior protection from lethal EBV challenge in female humanized mice compared to IgG elicited by individual NP-gHgL, NP-gB and NP-gp42. Importantly, serum antibodies elicited by cocktail nanovaccine immunization confer durable protection against EBV-associated lymphoma. Overall, the cocktail nanovaccine shows robust immunogenicity and is a promising candidate for further clinical trials.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , Infections à virus Epstein-Barr , Glycoprotéines , Herpèsvirus humain de type 4 , Animaux , Infections à virus Epstein-Barr/immunologie , Infections à virus Epstein-Barr/prévention et contrôle , Infections à virus Epstein-Barr/virologie , Anticorps neutralisants/immunologie , Herpèsvirus humain de type 4/immunologie , Humains , Femelle , Souris , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Glycoprotéines/immunologie , Glycoprotéines/administration et posologie , Nanoparticules/administration et posologie , Nanoparticules/composition chimique , Adjuvants immunologiques/administration et posologie , Lymphomes/immunologie , Lymphomes/virologie ,
7.
Clin Neurophysiol ; 163: 132-142, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38733703

RÉSUMÉ

BACKGROUND: Immune effector cell-associated neurotoxicity syndrome (ICANS) is common after chimeric antigen receptor T-cell (CAR-T) therapy. OBJECTIVE: This study aimed to assess the impact of preinfusion electroencephalography (EEG) abnormalities and EEG findings at ICANS onset for predicting ICANS risk and severity in 56 adult patients with refractory lymphoma undergoing CAR-T therapy. STUDY DESIGN: EEGs were conducted at the time of lymphodepleting chemotherapy and shortly after onset of ICANS. RESULTS: Twenty-eight (50%) patients developed ICANS at a median time of 6 days after CAR-T infusion. Abnormal preinfusion EEG was identified as a risk factor for severe ICANS (50% vs. 17%, P = 0.036). Following ICANS onset, EEG abnormalities were detected in 89% of patients [encephalopathy (n = 19, 70%) and/or interictal epileptiform discharges (IEDs) (n = 14, 52%)]. Importantly, IEDs seemed to be associated with rapid progression to higher grades of ICANS within 24 h. CONCLUSIONS: If confirmed in a large cohort of patients, these findings could establish the basis for modifying current management guidelines, enabling the identification of patients at risk of neurotoxicity, and providing support for preemptive corticosteroid use in patients with both initial grade 1 ICANS and IEDs at neurotoxicity onset, who are at risk of neurological impairment.


Sujet(s)
Électroencéphalographie , Immunothérapie adoptive , Syndromes neurotoxiques , Humains , Mâle , Femelle , Adulte d'âge moyen , Syndromes neurotoxiques/physiopathologie , Syndromes neurotoxiques/étiologie , Syndromes neurotoxiques/diagnostic , Adulte , Immunothérapie adoptive/effets indésirables , Immunothérapie adoptive/méthodes , Sujet âgé , Lymphomes/thérapie , Lymphomes/physiopathologie , Lymphomes/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Jeune adulte
8.
Nucl Med Commun ; 45(8): 718-726, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38726632

RÉSUMÉ

PURPOSE: The aim of this study was to evaluate metabolism change in reference organs (liver and mediastinum) and lymphoid cell-rich organs (spleen and bone marrow) during programmed cell death-1 immunotherapy in relapsed or refractory lymphoma patients. METHODS: A total of 66 patients with baseline and serial monitoring fluorodeoxyglucose (FDG) PET/computed tomography scans were retrospectively enrolled. Mean standardized uptake value (SUV) and maximum SUV of evaluated organs were obtained by two reviewers, and their association with tumor burden and clinical response were evaluated. Immune-related adverse events detected by FDG PET/computed tomography were also recorded. RESULTS: The SUV values of reference organs and lymphoid cell-rich organs did not change significantly during the immunotherapy process. The intersubject variability of these values ranged from 13.0 to 28.5%. Meanwhile, metabolism of reference organs was affected by neither the tumor burden nor clinical response. SUV change of lymphoid cell-rich organs was associated with clinical response to immunotherapy. Responders showed decreased metabolism, while nonresponders showed a reverse trend (spleen SUV max : -0.30 ±â€…0.47 vs. 0.18 ±â€…0.39, P  = 0.001, spleen SUV mean : -0.24 ±â€…0.39 vs. 0.14 ±â€…0.31, P  = 0.001; and bone marrow SUV max : -0.14 ±â€…0.37 vs. 0.07 ±â€…0.46, P  = 0.042, respectively). The influence of immune-related adverse events on the SUV change in evaluated organs was not significant. CONCLUSION: During programmed cell death-1 immunotherapy, metabolism change of reference organs is influenced neither by tumor burden nor by clinical response, while FDG uptake change of lymphoid cell-rich organs is significantly associated with clinical response.


Sujet(s)
Fluorodésoxyglucose F18 , Immunothérapie , Foie , Lymphomes , Médiastin , Tomographie par émission de positons couplée à la tomodensitométrie , Récepteur-1 de mort cellulaire programmée , Humains , Mâle , Femelle , Adulte d'âge moyen , Foie/imagerie diagnostique , Foie/métabolisme , Lymphomes/imagerie diagnostique , Lymphomes/thérapie , Lymphomes/métabolisme , Lymphomes/immunologie , Sujet âgé , Adulte , Récepteur-1 de mort cellulaire programmée/métabolisme , Études rétrospectives , Transport biologique , Sujet âgé de 80 ans ou plus , Lymphocytes/métabolisme , Jeune adulte
9.
Sci Rep ; 14(1): 11624, 2024 05 21.
Article de Anglais | MEDLINE | ID: mdl-38773194

RÉSUMÉ

The Nova Scotia Duck Tolling Retriever (NSDTR) is predisposed to immune mediated rheumatic disease (IMRD), steroid-responsive meningitis-arteritis (SRMA) and certain forms of cancer. Cytokines are the main regulators of the immune system. Interleukin 2 is a cytokine involved in activation of T regulatory cells, playing a role in central tolerance and tumor immunity. Interleukin 12 and interleukin 23 share the same subunit, p40, and are both pro-inflammatory cytokines. The aim of this study was to compare levels of IL-2 in healthy NSDTRs to those with cancer or autoimmune disease and to compare levels of IL-12/IL-23p40 in healthy NSDTRs and beagles versus NSDTRs with cancer or autoimmune disease. 62 dogs were included in the analysis of IL-12/IL-23p40; healthy NSDTRs (n = 16), healthy beagles (n = 16), NSDTRs autoimmune (n = 18) and NDSTRs lymphoma/mastocytoma (n = 12) and 68 dogs for IL-2; healthy (n = 20), autoimmune (n = 36) and lymphoma/mastocytoma/adenocarcinoma (n = 12). NSDTRs with autoimmune disease had higher levels of IL-12/IL-23p40 compared to healthy dogs (p = 0.008). NSDTRs with lymphoma also had higher levels of IL-12/IL-23p40 compared to healthy NSDTRs (p = 0.002). There was no difference in levels of IL-2 between healthy and diseased NSDTR. Statistical analysis was performed using Bonferroni corrections for multiple testing. These findings can contribute to the knowledge of autoimmune disease and cancer in dogs.


Sujet(s)
Maladies auto-immunes , Maladies des chiens , Interleukine-12 , Lymphomes , Animaux , Chiens , Maladies auto-immunes/médecine vétérinaire , Maladies auto-immunes/immunologie , Lymphomes/médecine vétérinaire , Lymphomes/immunologie , Maladies des chiens/immunologie , Femelle , Mâle , Interleukine-23 , Interleukine-2
10.
Front Immunol ; 15: 1389971, 2024.
Article de Anglais | MEDLINE | ID: mdl-38799440

RÉSUMÉ

Currently, therapies such as chimeric antigen receptor-T Cell (CAR-T) and immune checkpoint inhibitors like programmed cell death protein-1 (PD-1) blockers are showing promising results for numerous cancer patients. However, significant advancements are required before CAR-T therapies become readily available as off-the-shelf treatments, particularly for solid tumors and lymphomas. In this review, we have systematically analyzed the combination therapy involving engineered CAR-T cells and anti PD-1 agents. This approach aims at overcoming the limitations of current treatments and offers potential advantages such as enhanced tumor inhibition, alleviated T-cell exhaustion, heightened T-cell activation, and minimized toxicity. The integration of CAR-T therapy, which targets tumor-associated antigens, with PD-1 blockade augments T-cell function and mitigates immune suppression within the tumor microenvironment. To assess the impact of combination therapy on various tumors and lymphomas, we categorized them based on six major tumor-associated antigens: mesothelin, disialoganglioside GD-2, CD-19, CD-22, CD-133, and CD-30, which are present in different tumor types. We evaluated the efficacy, complete and partial responses, and progression-free survival in both pre-clinical and clinical models. Additionally, we discussed potential implications, including the feasibility of combination immunotherapies, emphasizing the importance of ongoing research to optimize treatment strategies and improve outcomes for cancer patients. Overall, we believe combining CAR-T therapy with PD-1 blockade holds promise for the next generation of cancer immunotherapy.


Sujet(s)
Inhibiteurs de points de contrôle immunitaires , Immunothérapie adoptive , Lymphomes , Récepteur-1 de mort cellulaire programmée , Récepteurs chimériques pour l'antigène , Humains , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/immunologie , Immunothérapie adoptive/méthodes , Lymphomes/thérapie , Lymphomes/immunologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/génétique , Animaux , Tumeurs/thérapie , Tumeurs/immunologie , Association thérapeutique , Microenvironnement tumoral/immunologie , Antigènes néoplasiques/immunologie , Lymphocytes T/immunologie , Lymphocytes T/métabolisme
11.
J Neurooncol ; 168(3): 487-494, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38658464

RÉSUMÉ

PURPOSE: The systemic inflammation response index (SIRI) and systemic immune-inflammation index (SII) are based on neutrophil, monocyte, platelet, and lymphocyte counts. The SIRI and SII are used to predict the survival of patients with malignant tumors. It is well known that the inflammatory immune response is closely related to cancer occurrence and progression. In the present study, we evaluated the potential prognostic significance of SIRI and SII in patients with primary central nervous system lymphoma (PCNSL). METHODS: Fifty-eight consecutive patients were enrolled in this study between November 2006 and May 2022. Among the 58 patients, 47 patients with sufficient blood test data and follow-up were analyzed. The patients with steroid intake at the time point of the blood test and higher C-reactive protein were excluded. RESULTS: The median follow-up and survival times were 31 and 36 months, respectively. The optimal cutoff SIRI value was based on the receiver operating characteristic curve (ROC) for overall survival (OS) and stratified patients into low (< 1.43 × 109/L, n = 22) and high (≥ 1.43 × 109/L, n = 25) SIRI groups. The optimal cutoff SII value based on the ROC for OS stratified patients into low (< 694.9, n = 28) and high (≥ 694.9, n = 19) SII groups. A low SIRI value was associated with longer OS (p = 0.006). Furthermore, a low SII value was associated with longer OS (p = 0.044). The prognostic factors associated with prolonged survival in univariate analysis using the Cox proportional hazard model were age < 65 years, low SIRI, and low SII. The multivariate analysis demonstrated that age < 65 years and low SIRI independently predicted longer OS. CONCLUSION: Simple, less expensive, and routinely ordered preoperative blood count assessments such as SIRI and SII predict the OS of patients with PCNSL. This study demonstrated that PCNSL is associated with pre-treatment systemic immune-inflammation states.


Sujet(s)
Tumeurs du système nerveux central , Inflammation , Lymphomes , Humains , Mâle , Femelle , Adulte d'âge moyen , Pronostic , Sujet âgé , Tumeurs du système nerveux central/immunologie , Tumeurs du système nerveux central/mortalité , Tumeurs du système nerveux central/sang , Adulte , Inflammation/immunologie , Inflammation/sang , Lymphomes/immunologie , Lymphomes/mortalité , Lymphomes/sang , Études de suivi , Sujet âgé de 80 ans ou plus , Études rétrospectives , Taux de survie , Jeune adulte , Courbe ROC , Granulocytes neutrophiles/immunologie
12.
Blood Adv ; 8(13): 3360-3371, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38640255

RÉSUMÉ

ABSTRACT: The remarkable efficacy of Epstein-Barr virus (EBV)-specific T cells for the treatment of posttransplant lymphomas has not been reproduced for EBV-positive (EBV+) malignancies outside the transplant setting. This is because of, in part, the heterogeneous expression and poor immunogenicity of the viral antigens expressed, namely latent membrane proteins 1 and 2, EBV nuclear antigen 1, and BamHI A rightward reading frame 1 (type-2 [T2] latency). However, EBV lytic cycle proteins are also expressed in certain EBV+ malignancies and, because several EBV lytic cycle proteins are abundantly expressed, have oncogenic activity, and likely contribute to malignancy, we sought and identified viral lytic-cycle transcripts in EBV+ Hodgkin lymphoma biopsies. This provided the rationale for broadening the target antigen-specific repertoire of EBV-specific T cells (EBVSTs) for therapy. We stimulated, peripheral blood mononuclear cells from healthy donors and patients with EBV+ lymphoma with both lytic and latent cycle proteins to produce broad repertoire (BR) EBVSTs. Compared with T2 antigen-specific EBVSTs, BR-EBVSTs more rapidly cleared autologous EBV+ tumors in NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ (NSG) mice and produced higher levels of proinflammatory cytokines that should reactivate the immunosuppressive tumor microenvironment leading to epitope spreading. Our results confirm that lytic cycle antigens are clinically relevant targets for EBV+ lymphoma and underpin the rationale for integrating BR-EBVSTs as a therapeutic approach for relapsed/refractory EBV+ lymphoma (www.clinicaltrials.gov identifiers: #NCT01555892 and #NCT04664179), as well as for other EBV-associated malignancies.


Sujet(s)
Antigènes viraux , Herpèsvirus humain de type 4 , Lymphocytes T , Humains , Herpèsvirus humain de type 4/immunologie , Animaux , Antigènes viraux/immunologie , Souris , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Infections à virus Epstein-Barr/immunologie , Infections à virus Epstein-Barr/complications , Lymphomes/immunologie , Lymphomes/thérapie , Maladie de Hodgkin/immunologie , Maladie de Hodgkin/thérapie , Maladie de Hodgkin/virologie , Latence virale
14.
In Vitro Cell Dev Biol Anim ; 60(4): 321-332, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38589736

RÉSUMÉ

Chimeric antigen receptors (CARs) offer a promising new approach for targeting B cell malignancies through the immune system. Despite the proven effectiveness of CAR T cells targeting CD19 and CD22 in hematological malignancies, it is imperative to note that their production remains a highly complex process. Unlike T cells, NK cells eliminate targets in a non-antigen-specific manner while avoiding graft vs. host disease (GvHD). CAR-NK cells are considered safer than CAR-T cells because they have a shorter lifespan and produce less toxic cytokines. Due to their unlimited ability to proliferate in vitro, NK-92 cells can be used as a source for CAR-engineered NK cells. We found that CARs created from the m971 anti-CD22 mAb, which specifically targets a proximal CD22 epitope, were more effective at anti-leukemic activity compared to those made with other binding domains. To further enhance the anti-leukemic capacity of NK cells, we used lentiviral transduction to generate the m971-CD28-CD3ζ NK-92. CD22 is highly expressed in B cell lymphoma. To evaluate the potential of targeting CD22, Raji cells were selected as CD22-positive cells. Our study aimed to investigate CD22 as a potential target for CAR-NK-92 therapy in the treatment of B cell lymphoma. We first generated m971-CD28-CD3ζ NK-92 that expressed a CAR for binding CD22 in vitro. Flow cytometric analysis was used to evaluate the expression of CAR. The 7AAD determined the cytotoxicity of the m971-CD28-CD3ζ NK-92 towards target lymphoma cell lines by flow cytometry assay. The ELISA assay evaluated cytokine production in CAR NK-92 cells in response to target cells. The m971-CD28-CD3ζ NK-92 cells have successfully expressed the CD22-specific CAR. m971-CD28-CD3ζ NK-92 cells efficiently lysed CD22-expressing lymphoma cell lines and produced large amounts of cytokines such as IFN-γ and GM-CSF but a lower level of IL-6 after coculturing with target cells. Based on our results, it is evident that transferring m971-CD28-CD3ζ NK-92 cells could be a promising immunotherapy for B cell lymphoma.


Sujet(s)
Cellules tueuses naturelles , Récepteurs chimériques pour l'antigène , Lectine-2 de type Ig liant l'acide sialique , Humains , Lectine-2 de type Ig liant l'acide sialique/immunologie , Cellules tueuses naturelles/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/métabolisme , Lignée cellulaire tumorale , Immunothérapie adoptive/méthodes , Lymphomes/thérapie , Lymphomes/immunologie , Lymphomes/anatomopathologie , Lymphome B/thérapie , Lymphome B/immunologie , Lymphome B/anatomopathologie , Cytotoxicité immunologique
15.
J Pediatr (Rio J) ; 100(4): 384-391, 2024.
Article de Anglais | MEDLINE | ID: mdl-38547930

RÉSUMÉ

OBJECTIVE: To investigate the incidence, clinical and genetic characteristics of pediatric lymphoma patients of China with inborn errors of immunity (IEI)-related gene mutations, which have not been fully studied. METHOD: From Jan. 2020 to Mar. 2023, IEI-related genetic mutations were retrospectively explored in 108 children with lymphomas admitted to Beijing Children's Hospital by NGS. Genetic rule and clinical characteristics as well as treatment outcomes were compared between patients with or without IEI-related gene mutations. RESULTS: A total of 17 patients (15.7 %) harbored IEI-associated mutations, including 4 cases with X-linked lymphoproliferative syndrome (XLP), 3 cases had mutations in tumor necrosis factor receptor superfamily 13B (TNFRSF13B), 2 cases with Activated p110 syndrome (APDS). Patients with IEI all had alteration of immunocompetence with decreased levels of immunoglobulin and lymphocyte subsets. Recurrent infection existed in 41.2 % of patients. The 18-month event-free survival (EFS) and the overall response rate (ORR) of patients with IEI are significantly lower than those without IEI (33.86% vs. 73.26 %, p = 0.011; 52.94% vs. 87.91 %, p = 0.002, respectively). In addition, patients with IEI had a higher progression disease (PD) rate of 23.5 % than those without IEI of 4.4 % (p = 0.006). CONCLUSION: The present study demonstrated that IEI-associated lymphomas were much more common than originally appreciated in pediatric lymphomas, and those were insensitive to treatment and more likely to progress or relapse. The genomic analysis and a thorough review of the medical history of IEI can be used to distinguish them from pediatric lymphomas without IEI, which are beneficial for the early diagnosis and direct intervention.


Sujet(s)
Lymphomes , Mutation , Humains , Mâle , Femelle , Études rétrospectives , Enfant , Chine/épidémiologie , Lymphomes/immunologie , Lymphomes/génétique , Enfant d'âge préscolaire , Nourrisson , Adolescent , Pertinence clinique
16.
Blood ; 143(18): 1816-1824, 2024 May 02.
Article de Anglais | MEDLINE | ID: mdl-38457360

RÉSUMÉ

ABSTRACT: Rituximab (RTX) and other monoclonal antibodies (mAbs) that bind directly to malignant cells are of great clinical value but are not effective for all patients. A major mechanism of action of RTX is antibody-dependent cellular cytotoxicity (ADCC) mediated by natural killer (NK) cells. Prior in vitro studies in our laboratory demonstrated that T cells contribute to maintaining the viability and cytotoxic potential of NK cells activated by anti-CD20-coated target B cells. Here, we conducted studies using a novel mouse model and clinical correlative analysis to assess whether T-cell help contribute to RTX-mediated NK-cell ADCC in the tumor microenvironment (TME) in vivo. A humanized mouse model was developed using Raji lymphoma cells and normal donor peripheral blood mononuclear cells that allows for control of T-cell numbers in the lymphoma TME. In this model, NK-cell viability and CD16 and CD25 expression dropped after RTX in the absence of T cells but increased in the presence of T cells. RTX therapy was more effective when T cells were present and was ineffective when NK cells were depleted. In patients with indolent lymphoma, fine needle aspirates were obtained before and ∼1 week after treatment with a RTX-containing regimen. There was a strong correlation between CD4+ T cells as well as total T cells in the pretherapy TME and an increase in NK-cell CD16 and CD25 expression after RTX. We conclude that T-cell help in the TME enhances RTX-mediated NK-cell viability and ADCC.


Sujet(s)
Cytotoxicité à médiation cellulaire dépendante des anticorps , Cellules tueuses naturelles , Rituximab , Microenvironnement tumoral , Rituximab/pharmacologie , Rituximab/usage thérapeutique , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/effets des médicaments et des substances chimiques , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie , Animaux , Cytotoxicité à médiation cellulaire dépendante des anticorps/effets des médicaments et des substances chimiques , Humains , Souris , Lymphocytes T/immunologie , Lymphocytes T/effets des médicaments et des substances chimiques , Antinéoplasiques immunologiques/pharmacologie , Antinéoplasiques immunologiques/usage thérapeutique , Lignée cellulaire tumorale , Souris SCID , Lymphomes/immunologie , Lymphomes/traitement médicamenteux , Lymphomes/anatomopathologie , Lymphomes/thérapie , Femelle
17.
J Neurosurg ; 140(6): 1549-1557, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38157532

RÉSUMÉ

OBJECTIVE: Malignancies of the CNS are difficult to treat because the blood-brain barrier (BBB) prevents most therapeutics from reaching the intracranial lesions at sufficiently high concentrations. This also applies to chimeric antigen receptor (CAR) T cells, for which systemic delivery is inferior to direct intratumoral or intraventricular injection of the cells. The authors previously reported on a novel approach to safely and reversibly open the BBB of mice by applying intra-arterial (IA) injections of NEO100, a pharmaceutical-grade version of the natural monoterpene perillyl alcohol. The authors hypothesized that this method would enable enhanced brain entry and therapeutic activity of intravenously delivered CAR T cells, which the authors tested in a mouse model of CNS lymphoma. METHODS: Human Raji lymphoma cells were implanted into the brains of immune-deficient mice. After tumor uptake was confirmed with bioluminescent imaging, 0.3% NEO100 was injected intra-arterially, which was followed by intravenous (IV) delivery of CD19-targeted CAR T cells. After this single intervention, tumor growth was monitored with imaging, long-term survival of mice was recorded, and select mice were euthanized to analyze the distribution of CAR T cells in brain tissue. RESULTS: Intravenously injected CAR T cells could be readily detected in brain tumor areas after IA injection of NEO100 but not after IA injection of the vehicle (without NEO100). Although all untreated control animals died within 3 weeks, all mice that received IA NEO100 followed by IV CAR T cells survived and thrived for 200 days, when the experiment was terminated. Of the mice that received IV CAR T cells without prior IA NEO100, 3 died within 3 weeks and 2 survived long-term. CONCLUSIONS: BBB opening by IA NEO100 facilitates brain entry of intravenously delivered CD19 CAR T cells. The long-term survival of all mice with CNS lymphoma, along with the disappearance of the tumor as determined with imaging, suggests that this one-time therapeutic intervention was curative. BBB opening by IA NEO100 may offer a novel option to increase brain access by CAR T cells.


Sujet(s)
Immunothérapie adoptive , Injections artérielles , Récepteurs chimériques pour l'antigène , Animaux , Souris , Immunothérapie adoptive/méthodes , Modèles animaux de maladie humaine , Barrière hémato-encéphalique , Humains , Tumeurs du cerveau/thérapie , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/immunologie , Lignée cellulaire tumorale/transplantation , Lymphomes/thérapie , Lymphomes/immunologie , Tumeurs du système nerveux central/thérapie , Tumeurs du système nerveux central/immunologie , Lymphocytes T/immunologie , Lymphocytes T/transplantation , Souris SCID
18.
J Neuroimmunol ; 382: 578156, 2023 09 15.
Article de Anglais | MEDLINE | ID: mdl-37556888

RÉSUMÉ

We reported a 61-year-old man presented with 10-month progressing left sciatic neuropathy and 10-day right facial neuropathy. Serum amphiphysin-IgG was positive. 18F-FDG PET/CT of the whole body showed no signs of malignancy. Treatment with plasma exchange and oral prednisone relieved the symptoms. Nine months later, right hemiparesis and seizure of right limbs developed. 18F-FDG and 18F-PBR06 (18 kDa translocator protein, TSPO) radioligand PET/MRI of the whole body revealed intense uptake in the intracranial lesions. Intracranial lymphoma was diagnosed by stereotactic needle brain biopsy. Mononeuropathies could be paraneoplastic syndromes. TSPO shows high uptake in intracranial lymphoma on 18F-PBR06 PET images.


Sujet(s)
Tumeurs du système nerveux central , Atteintes du nerf facial , Lymphomes , Neuropathie du nerf sciatique , Humains , Mâle , Adulte d'âge moyen , Encéphale/immunologie , Atteintes du nerf facial/étiologie , Atteintes du nerf facial/immunologie , Atteintes du nerf facial/thérapie , Fluorodésoxyglucose F18 , Immunoglobuline G/immunologie , Tomographie par émission de positons couplée à la tomodensitométrie/méthodes , Tomographie par émission de positons/méthodes , Radiopharmaceutiques , Récepteurs GABA/métabolisme , Neuropathie du nerf sciatique/étiologie , Neuropathie du nerf sciatique/immunologie , Neuropathie du nerf sciatique/thérapie , Tumeurs du système nerveux central/complications , Tumeurs du système nerveux central/imagerie diagnostique , Tumeurs du système nerveux central/immunologie , Maladies auto-immunes/étiologie , Maladies auto-immunes/immunologie , Lymphomes/complications , Lymphomes/imagerie diagnostique , Lymphomes/immunologie , Neuropathie paranéoplasique/étiologie , Neuropathie paranéoplasique/immunologie , Prednisone/usage thérapeutique , Glucocorticoïdes/usage thérapeutique , Échange plasmatique , Protéines de tissu nerveux/immunologie
19.
J Immunother Cancer ; 11(4)2023 04.
Article de Anglais | MEDLINE | ID: mdl-37072346

RÉSUMÉ

BACKGROUND: The wider application of T cells targeting viral tumor-antigens via their native receptors is hampered by the failure to expand potent tumor-specific T cells from patients. Here, we examine reasons for and solutions to this failure, taking as our model the preparation of Epstein-Barr virus (EBV)-specific T cells (EBVSTs) for the treatment of EBV-positive lymphoma. EBVSTs could not be manufactured from almost one-third of patients, either because they failed to expand, or they expanded, but lacked EBV specificity. We identified an underlying cause of this problem and established a clinically feasible approach to overcome it. METHODS: CD45RO+CD45RA- memory compartment residing antigen-specific T cells were enriched by depleting CD45RA positive (+) peripheral blood mononuclear cells (PBMCs) that include naïve T cells, among other subsets, prior to EBV antigen stimulation. We then compared the phenotype, specificity, function and T-cell receptor (TCR) Vß repertoire of EBVSTs expanded from unfractionated whole (W)-PBMCs and CD45RA-depleted (RAD)-PBMCs on day 16. To identify the CD45RA component that inhibited EBVST outgrowth, isolated CD45RA+ subsets were added back to RAD-PBMCs followed by expansion and characterization. The in vivo potency of W-EBVSTs and RAD-EBVSTs was compared in a murine xenograft model of autologous EBV+ lymphoma. RESULTS: Depletion of CD45RA+ PBMCs before antigen stimulation increased EBVST expansion, antigen-specificity and potency in vitro and in vivo. TCR sequencing revealed a selective outgrowth in RAD-EBVSTs of clonotypes that expanded poorly in W-EBVSTs. Inhibition of antigen-stimulated T cells by CD45RA+ PBMCs could be reproduced only by the naïve T-cell fraction, while CD45RA+ regulatory T cells, natural killer cells, stem cell memory and effector memory subsets lacked inhibitory activity. Crucially, CD45RA depletion of PBMCs from patients with lymphoma enabled the outgrowth of EBVSTs that failed to expand from W-PBMCs. This enhanced specificity extended to T cells specific for other viruses. CONCLUSION: Our findings suggest that naïve T cells inhibit the outgrowth of antigen-stimulated memory T cells, highlighting the profound effects of intra-T-cell subset interactions. Having overcome our inability to generate EBVSTs from many patients with lymphoma, we have introduced CD45RA depletion into three clinical trials: NCT01555892 and NCT04288726 using autologous and allogeneic EBVSTs to treat lymphoma and NCT04013802 using multivirus-specific T cells to treat viral infections after hematopoietic stem cell transplantation.


Sujet(s)
Herpèsvirus humain de type 4 , Cellules de la mémoire immunitaire , Immunothérapie , Lymphomes , Lymphocytes T , Lymphocytes T/immunologie , Humains , Lymphomes/immunologie , Lymphomes/thérapie , Antigènes CD45 , Cellules de la mémoire immunitaire/immunologie , Agranulocytes/immunologie , Cellules tueuses naturelles/immunologie , Immunothérapie/méthodes , Immunophénotypage , Femelle , Animaux , Souris , Hétérogreffes , Transplantation tumorale
20.
Nat Commun ; 13(1): 866, 2022 02 14.
Article de Anglais | MEDLINE | ID: mdl-35165282

RÉSUMÉ

Epstein-Barr virus (EBV) is reportedly the first identified human tumor virus, and is closely related to the occurrence and development of nasopharyngeal carcinoma (NPC), gastric carcinoma (GC), and several lymphomas. PD-L1 expression is elevated in EBV-positive NPC and GC tissues; however, the specific mechanisms underlying the EBV-dependent promotion of PD-L1 expression to induce immune escape warrant clarification. EBV encodes 44 mature miRNAs. In this study, we find that EBV-miR-BART11 and EBV-miR-BART17-3p upregulate the expression of PD-L1 in EBV-associated NPC and GC. Furthermore, EBV-miR-BART11 targets FOXP1, EBV-miR-BART17-3p targets PBRM1, and FOXP1 and PBRM1 bind to the enhancer region of PD-L1 to inhibit its expression. Therefore, EBV-miR-BART11 and EBV-miR-BART17-3p inhibit FOXP1 and PBRM1, respectively, and enhance the transcription of PD-L1 (CD274, http://www.ncbi.nlm.nih.gov/gene/29126 ), resulting in the promotion of tumor immune escape, which provides insights into potential targets for EBV-related tumor immunotherapy.


Sujet(s)
Herpèsvirus humain de type 4/génétique , microARN/génétique , Cancer du nasopharynx/immunologie , Tumeurs du rhinopharynx/immunologie , Tumeurs de l'estomac/immunologie , Échappement de la tumeur à la surveillance immunitaire/immunologie , Antigène CD274/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire , Protéines de liaison à l'ADN/antagonistes et inhibiteurs , Protéines de liaison à l'ADN/métabolisme , Infections à virus Epstein-Barr/virologie , Facteurs de transcription Forkhead/antagonistes et inhibiteurs , Facteurs de transcription Forkhead/métabolisme , Régulation de l'expression des gènes tumoraux/génétique , Herpèsvirus humain de type 4/immunologie , Humains , Lymphomes/immunologie , Lymphomes/virologie , Cancer du nasopharynx/génétique , Cancer du nasopharynx/virologie , Tumeurs du rhinopharynx/génétique , Tumeurs du rhinopharynx/virologie , Protéines de répression/antagonistes et inhibiteurs , Protéines de répression/métabolisme , Tumeurs de l'estomac/virologie , Facteurs de transcription/antagonistes et inhibiteurs , Facteurs de transcription/métabolisme , Échappement de la tumeur à la surveillance immunitaire/génétique , Microenvironnement tumoral/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...