Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27.876
Filter
1.
J Clin Invest ; 134(11)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38828721

ABSTRACT

The adoptive transfer of T cell receptor-engineered (TCR-engineered) T cells (ACT) targeting the HLA-A2-restricted cancer-testis epitope NY-ESO-1157-165 (A2/NY) has yielded favorable clinical responses against several cancers. Two approaches to improve ACT are TCR affinity optimization and T cell coengineering to express immunomodulatory molecules that can exploit endogenous immunity. By computational design we previously developed a panel of binding-enhanced A2/NY-TCRs including A97L, which augmented the in vitro function of gene-modified T cells as compared with WT. Here, we demonstrated higher persistence and improved tumor control by A97L-T cells. In order to harness macrophages in tumors, we further coengineered A97L-T cells to secrete a high-affinity signal regulatory protein α (SiRPα) decoy (CV1) that blocks CD47. While CV1-Fc-coengineered A97L-T cells mediated significantly better control of tumor outgrowth and survival in Winn assays, in subcutaneous xenograft models the T cells, coated by CV1-Fc, were depleted. Importantly, there was no phagocytosis of CV1 monomer-coengineered T cells by human macrophages. Moreover, avelumab and cetuximab enhanced macrophage-mediated phagocytosis of tumor cells in vitro in the presence of CV1 and improved tumor control upon coadministration with A97L-T cells. Taken together, our study indicates important clinical promise for harnessing macrophages by combining CV1-coengineered TCR-T cells with targeted antibodies to direct phagocytosis against tumor cells.


Subject(s)
Macrophages , Phagocytosis , Receptors, Immunologic , Humans , Animals , Mice , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Macrophages/immunology , Macrophages/metabolism , T-Lymphocytes/immunology , Antigens, Differentiation/immunology , HLA-A2 Antigen/immunology , HLA-A2 Antigen/genetics , Antigens, Neoplasm/immunology , Cell Line, Tumor , Xenograft Model Antitumor Assays , CD47 Antigen/immunology , Immunotherapy, Adoptive , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism
2.
Commun Biol ; 7(1): 684, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38834836

ABSTRACT

Identifying interactions between T-cell receptors (TCRs) and immunogenic peptides holds profound implications across diverse research domains and clinical scenarios. Unsupervised clustering models (UCMs) cannot predict peptide-TCR binding directly, while supervised predictive models (SPMs) often face challenges in identifying antigens previously unencountered by the immune system or possessing limited TCR binding repertoires. Therefore, we propose HeteroTCR, an SPM based on Heterogeneous Graph Neural Network (GNN), to accurately predict peptide-TCR binding probabilities. HeteroTCR captures within-type (TCR-TCR or peptide-peptide) similarity information and between-type (peptide-TCR) interaction insights for predictions on unseen peptides and TCRs, surpassing limitations of existing SPMs. Our evaluation shows HeteroTCR outperforms state-of-the-art models on independent datasets. Ablation studies and visual interpretation underscore the Heterogeneous GNN module's critical role in enhancing HeteroTCR's performance by capturing pivotal binding process features. We further demonstrate the robustness and reliability of HeteroTCR through validation using single-cell datasets, aligning with the expectation that pMHC-TCR complexes with higher predicted binding probabilities correspond to increased binding fractions.


Subject(s)
Neural Networks, Computer , Peptides , Receptors, Antigen, T-Cell , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/chemistry , Peptides/chemistry , Peptides/metabolism , Peptides/immunology , Protein Binding , Humans , Computational Biology/methods
3.
Haematologica ; 109(6): 1656-1667, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38832421

ABSTRACT

Recurrent and/or refractory (R/R) pediatric acute myeloid leukemia (AML) remains a recalcitrant disease with poor outcomes. Cell therapy with genetically modified immune effector cells holds the promise to improve outcomes for R/R AML since it relies on cytotoxic mechanisms that are distinct from chemotherapeutic agents. While T cells expressing chimeric antigen receptors (CAR T cells) showed significant anti-AML activity in preclinical models, early phase clinical studies have demonstrated limited activity, irrespective of the targeted AML antigen. Lack of efficacy is most likely multifactorial, including: (i) a limited array of AML-specific targets and target antigen heterogeneity; (ii) the aggressive nature of R/R AML and heavy pretreatment of patients; (iii) T-cell product manufacturing, and (iv) limited expansion and persistence of the CAR T cells, which is in part driven by the immunosuppressive AML microenvironment. Here we review the results of early phase clinical studies with AML-specific CAR T cells, and avenues investigators are exploring to improve their effector function.


Subject(s)
Immunotherapy, Adoptive , Leukemia, Myeloid, Acute , Receptors, Chimeric Antigen , Humans , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/immunology , Receptors, Chimeric Antigen/immunology , Immunotherapy, Adoptive/methods , Child , Clinical Trials as Topic , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Treatment Outcome , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/genetics , Tumor Microenvironment/immunology , Animals
4.
Haematologica ; 109(6): 1677-1688, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38832423

ABSTRACT

Chimeric antigen receptor (CAR) T-cell therapy is a new and effective treatment for patients with hematologic malignancies. Clinical responses to CAR T cells in leukemia, lymphoma, and multiple myeloma have provided strong evidence of the antitumor activity of these cells. In patients with refractory or relapsed B-cell acute lymphoblastic leukemia (ALL), the infusion of autologous anti-CD19 CAR T cells is rapidly gaining standard-of-care status and might eventually be incorporated into frontline treatment. In T-ALL, however, leukemic cells generally lack surface molecules recognized by established CAR, such as CD19 and CD22. Such deficiency is particularly important, as outcome is dismal for patients with T-ALL that is refractory to standard chemotherapy and/or hematopoietic stem cell transplant. Recently, CAR T-cell technologies directed against T-cell malignancies have been developed and are beginning to be tested clinically. The main technical obstacles stem from the fact that malignant and normal T cells share most surface antigens. Therefore, CAR T cells directed against T-ALL targets might be susceptible to self-elimination during manufacturing and/or have suboptimal activity after infusion. Moreover, removing leukemic cells that might be present in the cell source used for CAR T-cell manufacturing might be problematic. Finally, reconstitution of T cells and natural killer cells after CAR T-cell infusion might be impaired. In this article, we discuss potential targets for CAR T-cell therapy of T-ALL with an emphasis on CD7, and review CAR configurations as well as early clinical results.


Subject(s)
Immunotherapy, Adoptive , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Receptors, Chimeric Antigen , Humans , Receptors, Chimeric Antigen/immunology , Immunotherapy, Adoptive/methods , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation , Animals , Treatment Outcome , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology
5.
Cancer Immunol Immunother ; 73(8): 150, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38832948

ABSTRACT

Hotspot driver mutations presented by human leukocyte antigens might be recognized by anti-tumor T cells. Based on their advantages of tumor-specificity and immunogenicity, neoantigens derived from hotspot mutations, such as PIK3CAH1047L, may serve as emerging targets for cancer immunotherapies. NetMHCpan V4.1 was utilized for predicting neoepitopes of PIK3CA hotspot mutation. Using in vitro stimulation, antigen-specific T cells targeting the HLA-A*11:01-restricted PIK3CA mutation were isolated from healthy donor-derived peripheral blood mononuclear cells. T cell receptors (TCRs) were cloned using single-cell PCR and sequencing. Their functionality was assessed through T cell activation markers, cytokine production and cytotoxic response to cancer cell lines pulsed with peptides or transduced genes of mutant PIK3CA. Immunogenic mutant antigens from PIK3CA and their corresponding CD8+ T cells were identified. These PIK3CA mutation-specific CD8+ T cells were subsequently enriched, and their TCRs were isolated. The TCR clones exhibited mutation-specific and HLA-restricted reactivity, demonstrating varying degrees of functional avidity. Identified TCR genes were transferred into CD8+ Jurkat cells and primary T cells deficient of endogenous TCRs. TCR-expressing cells demonstrated specific recognition and reactivity against the PIK3CAH1047L peptide presented by HLA-A*11:01-expressing K562 cells. Furthermore, mutation-specific TCR-T cells demonstrated an elevation in cytokine production and profound cytotoxic effects against HLA-A*11:01+ malignant cell lines harboring PIK3CAH1047L. Our data demonstrate the immunogenicity of an HLA-A*11:01-restricted PIK3CA hotspot mutation and its targeting therapeutic potential, together with promising candidates of TCR-T cell therapy.


Subject(s)
Class I Phosphatidylinositol 3-Kinases , Mutation , Neoplasms , Receptors, Antigen, T-Cell , Humans , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/genetics , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/genetics , Immunotherapy/methods , HLA-A11 Antigen/genetics , HLA-A11 Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Antigens, Neoplasm/immunology , Antigens, Neoplasm/genetics , Cell Line, Tumor
6.
Front Immunol ; 15: 1372658, 2024.
Article in English | MEDLINE | ID: mdl-38827740

ABSTRACT

Background: Persistent radiological lung abnormalities are evident in many survivors of acute coronavirus disease 2019 (COVID-19). Consolidation and ground glass opacities are interpreted to indicate subacute inflammation whereas reticulation is thought to reflect fibrosis. We sought to identify differences at molecular and cellular level, in the local immunopathology of post-COVID inflammation and fibrosis. Methods: We compared single-cell transcriptomic profiles and T cell receptor (TCR) repertoires of bronchoalveolar cells obtained from convalescent individuals with each radiological pattern, targeting lung segments affected by the predominant abnormality. Results: CD4 central memory T cells and CD8 effector memory T cells were significantly more abundant in those with inflammatory radiology. Clustering of similar TCRs from multiple donors was a striking feature of both phenotypes, consistent with tissue localised antigen-specific immune responses. There was no enrichment for known SARS-CoV-2-reactive TCRs, raising the possibility of T cell-mediated immunopathology driven by failure in immune self-tolerance. Conclusions: Post-COVID radiological inflammation and fibrosis show evidence of shared antigen-specific T cell responses, suggesting a role for therapies targeting T cells in limiting post-COVID lung damage.


Subject(s)
COVID-19 , SARS-CoV-2 , Single-Cell Analysis , Humans , COVID-19/immunology , COVID-19/pathology , SARS-CoV-2/immunology , Male , Female , Middle Aged , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/genetics , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/pathology , CD8-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Lung/immunology , Lung/pathology , Lung/diagnostic imaging , Aged , Adult , Inflammation/immunology , Inflammation/pathology , Bronchoalveolar Lavage Fluid/immunology , Bronchoalveolar Lavage Fluid/cytology , Memory T Cells/immunology , Transcriptome
7.
Autoimmunity ; 57(1): 2360490, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38836341

ABSTRACT

The heterogeneity of the T cell receptor (TCR) repertoire critically influences the autoimmune response in obstetric antiphospholipid syndrome (OAPS) and is intimately associated with the prophylaxis of autoimmune disorders. Investigating the TCR diversity patterns in patients with OAPS is thus of paramount clinical importance. This investigation procured peripheral blood specimens from 31 individuals with OAPS, 21 patients diagnosed with systemic lupus erythematosus (SLE), and 22 healthy controls (HC), proceeding with TCR repertoire sequencing. Concurrently, adverse pregnancy outcomes in the OAPS cohort were monitored and documented over an 18-month timeframe. We paid particular attention to disparities in V/J gene utilisation and the prevalence of shared clonotypes amongst OAPS patients and the comparative groups. When juxtaposed with observations from healthy controls and SLE patients, immune repertoire sequencing disclosed irregular T- and B-cell profiles and a contraction of diversity within the OAPS group. Marked variances were found in the genomic rearrangements of the V gene, J gene, and V/J combinations. Utilising a specialised TCRß repertoire, we crafted a predictive model for OAPS classification with robust discriminative capability (AUC = 0.852). Our research unveils alterations in the TCR repertoire among OAPS patients for the first time, positing potential covert autoimmune underpinnings. These findings nominate the TCR repertoire as a prospective peripheral blood biomarker for the clinical diagnosis of OAPS and may offer valuable insights for advancing the understanding of OAPS immunologic mechanisms and prognostic outcomes.


Subject(s)
Antiphospholipid Syndrome , Biomarkers , Receptors, Antigen, T-Cell , Humans , Antiphospholipid Syndrome/immunology , Antiphospholipid Syndrome/diagnosis , Antiphospholipid Syndrome/genetics , Antiphospholipid Syndrome/blood , Female , Pregnancy , Adult , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/diagnosis , Lupus Erythematosus, Systemic/blood , Pregnancy Complications/immunology , Pregnancy Complications/genetics , Pregnancy Complications/diagnosis
9.
J Biomed Sci ; 31(1): 58, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824576

ABSTRACT

BACKGROUND: A vaccine against Trypanosoma cruzi, the agent of Chagas disease, would be an excellent additional tool for disease control. A recombinant vaccine based on Tc24 and TSA1 parasite antigens was found to be safe and immunogenic in naïve macaques. METHODS: We used RNA-sequencing and performed a transcriptomic analysis of PBMC responses to vaccination of naïve macaques after each vaccine dose, to shed light on the immunogenicity of this vaccine and guide the optimization of doses and formulation. We identified differentially expressed genes and pathways and characterized immunoglobulin and T cell receptor repertoires. RESULTS: RNA-sequencing analysis indicated a clear transcriptomic response of PBMCs after three vaccine doses, with the up-regulation of several immune cell activation pathways and a broad non-polarized immune profile. Analysis of the IgG repertoire showed that it had a rapid turnover with novel IgGs produced following each vaccine dose, while the TCR repertoire presented several persisting clones that were expanded after each vaccine dose. CONCLUSIONS: These data suggest that three vaccine doses may be needed for optimum immunogenicity and support the further evaluation of the protective efficacy of this vaccine.


Subject(s)
Chagas Disease , Macaca mulatta , Protozoan Vaccines , Receptors, Antigen, T-Cell , Animals , Chagas Disease/immunology , Chagas Disease/prevention & control , Receptors, Antigen, T-Cell/immunology , Protozoan Vaccines/immunology , Trypanosoma cruzi/immunology , Immunoglobulins/immunology
10.
Cell Chem Biol ; 31(5): 920-931.e6, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38759618

ABSTRACT

T cell receptor (TCR) plays a fundamental role in adaptive immunity, and TCR-T cell therapy holds great promise for treating solid tumors and other diseases. However, there is a noticeable absence of chemical tools tuning TCR activity. In our study, we screened natural sterols for their regulatory effects on T cell function and identified 7-alpha-hydroxycholesterol (7a-HC) as a potent inhibitor of TCR signaling. Mechanistically, 7a-HC promoted membrane binding of CD3ε cytoplasmic domain, a crucial signaling component of the TCR-CD3 complex, through alterations in membrane physicochemical properties. Enhanced CD3ε membrane binding impeded the condensation between CD3ε and the key kinase Lck, thereby inhibiting Lck-mediated TCR phosphorylation. Transient treatments of TCR-T cells with 7a-HC resulted in reduced signaling strength, increased memory cell populations, and superior long-term antitumor functions. This study unveils a chemical regulation of TCR signaling, which can be exploited to enhance the long-term efficacy of TCR-T cell therapy.


Subject(s)
Hydroxycholesterols , Receptors, Antigen, T-Cell , Signal Transduction , Signal Transduction/drug effects , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , Humans , Hydroxycholesterols/chemistry , Hydroxycholesterols/metabolism , Hydroxycholesterols/pharmacology , Animals , Mice , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Mice, Inbred C57BL , Phosphorylation/drug effects
11.
Lancet Neurol ; 23(6): 615-624, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38760099

ABSTRACT

BACKGROUND: Neuroimmunology research and development has been marked by substantial advances, particularly in the treatment of neuroimmunological diseases, such as multiple sclerosis, myasthenia gravis, neuromyelitis optica spectrum disorders, and myelin oligodendrocyte glycoprotein antibody disease. With more than 20 drugs approved for multiple sclerosis alone, treatment has become more personalised. The approval of disease-modifying therapies, particularly those targeting B cells, has highlighted the role of immunotherapeutic interventions in the management of these diseases. Despite these successes, challenges remain, particularly for patients who do not respond to conventional therapies, underscoring the need for innovative approaches. RECENT DEVELOPMENTS: The approval of monoclonal antibodies, such as ocrelizumab and ofatumumab, which target CD20, and inebilizumab, which targets CD19, for the treatment of various neuroimmunological diseases reflects progress in the understanding and management of B-cell activity. However, the limitations of these therapies in halting disease progression or activity in patients with multiple sclerosis or neuromyelitis optica spectrum disorders have prompted the exploration of cell-based therapies, particularly chimeric antigen receptor (CAR) T cells. Initially successful in the treatment of B cell-derived malignancies, CAR T cells offer a novel therapeutic mechanism by directly targeting and eliminating B cells, potentially overcoming the shortcomings of antibody-mediated B cell depletion. WHERE NEXT?: The use of CAR T cells in autoimmune diseases and B cell-driven neuroimmunological diseases shows promise as a targeted and durable option. CAR T cells act autonomously, penetrating deep tissue and effectively depleting B cells, especially in the CNS. Although the therapeutic potential of CAR T cells is substantial, their application faces hurdles such as complex logistics and management of therapy-associated toxic effects. Ongoing and upcoming clinical trials will be crucial in determining the safety, efficacy, and applicability of CAR T cells. As research progresses, CAR T cell therapy has the potential to transform treatment for patients with neuroimmunological diseases. It could offer extended periods of remission and a new standard in the management of autoimmune and neuroimmunological disorders.


Subject(s)
B-Lymphocytes , Receptors, Chimeric Antigen , Humans , B-Lymphocytes/immunology , Receptors, Chimeric Antigen/immunology , Immunotherapy, Adoptive/methods , T-Lymphocytes/immunology , Receptors, Antigen, T-Cell/immunology , Animals , Autoimmune Diseases of the Nervous System/therapy , Autoimmune Diseases of the Nervous System/immunology
12.
Cell Immunol ; 399-400: 104827, 2024.
Article in English | MEDLINE | ID: mdl-38733699

ABSTRACT

The need to contrive interventions to curb the rise in cancer incidence and mortality is critical for improving patients' prognoses. Adoptive cell therapy is challenged with quality large-scale production, heightening its production cost. Several cancer types have been associated with the expression of highly-immunogenic CTAG1 and CTAG2 antigens, which share common epitopes. Targeting two antigens on the same cancer could improve the antitumor response of TCR-T cells. In this study, we exploited an efficient way to generate large-fold quality TCR-T cells and also demonstrated that the common epitopes of CTAG1 and CTAG2 antigens provide an avenue for improved cancer-killing via dual-antigen-epitope targeting. Our study revealed that xeno/sera-free medium could expand TCR-T cells to over 500-fold, posing as a better replacement for FBS-supplemented media. Human AB serum was also shown to be a good alternative in the absence of xeno/sera-free media. Furthermore, TCR-T cells stimulated with beads-coated T-activator showed a better effector function than soluble T-activator stimulated TCR-T cells. Additionally, TCR-T cells that target multiple antigens in the same cancer yield better anticancer activity than those targeting a single antigen. This showed that targeting multiple antigens with a common epitope may enhance the antitumor response efficacy of T cell therapies.


Subject(s)
Antigens, Neoplasm , Immunotherapy, Adoptive , Receptors, Antigen, T-Cell , Antigens, Neoplasm/immunology , Humans , Immunotherapy, Adoptive/methods , Receptors, Antigen, T-Cell/immunology , Animals , Epitopes, T-Lymphocyte/immunology , Neoplasms/immunology , Neoplasms/therapy , Mice , Cell Line, Tumor , T-Lymphocytes/immunology , Epitopes/immunology
13.
Sheng Wu Gong Cheng Xue Bao ; 40(5): 1338-1351, 2024 May 25.
Article in Chinese | MEDLINE | ID: mdl-38783801

ABSTRACT

Chimeric antigen receptor T cells (CAR-T) immunotherapy, which activates immunity specific to the system in order to achieve antitumor effects, has experienced exciting progress in recent years. mRNA nano-delivery systems, which encapsulate tumor immunotherapy-related antigen mRNA with nanoparticles, have shown great potential in CAR-T tumor immunotherapy. On one hand, these systems can directly target T cells to generate CAR-T cells that directly act upon the corresponding tumor cells. On the other hand, they can be delivered to antigen-presenting cells through targeting, thereby enhancing the function of CAR-T cells and further inducing specific immune responses against tumor cells. This review summarizes the synthesis of mRNA nano-delivery systems and their application in CAR-T tumor immunotherapy.


Subject(s)
Immunotherapy, Adoptive , Nanoparticles , Neoplasms , RNA, Messenger , Receptors, Chimeric Antigen , Humans , Neoplasms/therapy , Neoplasms/immunology , RNA, Messenger/genetics , RNA, Messenger/immunology , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/genetics , Nanoparticles/chemistry , Immunotherapy , T-Lymphocytes/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/genetics , Animals
14.
Sheng Wu Gong Cheng Xue Bao ; 40(5): 1365-1379, 2024 May 25.
Article in Chinese | MEDLINE | ID: mdl-38783803

ABSTRACT

Globally, colorectal cancer (CRC) ranks as the third most common cancer and the second leading cause of cancer-related fatalities. According to the World Health Organization, there are over 1.9 million annual cases of CRC diagnosed worldwide, resulting in more than 900 000 deaths. In recent years, chimeric antigen receptor T (CAR-T) cell therapy has shown clinical success in treating certain hematological malignancies and is now being explored for its potential in targeting solid tumors like CRC. Currently, CAR-T cell therapies targeting carcinoembryonic antigen (CEA), natural killer group 2, member D ligand (NKG2DL), and other markers have achieved remarkable results in clinical trials, albeit encountering significant challenges. This review summarizes the promising targets of CAR-T cell therapy for CRC and highlights progress made in clinical trials and preclinical studies. Additionally, the review discusses the challenges faced by CAR-T cell therapy in CRC treatment, including a shortage of tumor-specific antigens, cytokine release syndrome, adverse tumor microenvironment, and limited infiltration of CAR-T cells. In summary, this review provides an overview of the latest research progress and challenges in CAR-T cell therapy for CRC, aiming to contribute fresh insights for the clinical treatment of this disease.


Subject(s)
Colorectal Neoplasms , Immunotherapy, Adoptive , Receptors, Chimeric Antigen , Humans , Colorectal Neoplasms/therapy , Colorectal Neoplasms/immunology , Receptors, Chimeric Antigen/immunology , Carcinoembryonic Antigen/immunology , Tumor Microenvironment , Antigens, Neoplasm/immunology , T-Lymphocytes/immunology , Receptors, Antigen, T-Cell/immunology , Animals
15.
Cell Death Dis ; 15(5): 359, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38789450

ABSTRACT

Chimeric antigen receptor engineered T (CAR T) cell therapy has developed rapidly in recent years, leading to profound developments in oncology, especially for hematologic malignancies. However, given the pressure of immunosuppressive tumor microenvironments, antigen escape, and diverse other factors, its application in solid tumors is less developed. Urinary system tumors are relatively common, accounting for approximately 24% of all new cancers in the United States. CAR T cells have great potential for urinary system tumors. This review summarizes the latest developments of CAR T cell therapy in urinary system tumors, including kidney cancer, bladder cancer, and prostate cancer, and also outlines the various CAR T cell generations and their pathways and targets that have been developed thus far. Finally, the current advantages, problems, and side effects of CAR T cell therapy are discussed in depth, and potential future developments are proposed in view of current shortcomings.


Subject(s)
Immunotherapy, Adoptive , Receptors, Chimeric Antigen , Urologic Neoplasms , Humans , Immunotherapy, Adoptive/methods , Urologic Neoplasms/therapy , Urologic Neoplasms/immunology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Animals , Treatment Outcome , Tumor Microenvironment/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism
17.
Cell Rep Med ; 5(5): 101567, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38744277

ABSTRACT

Bispecific T cell engagers (TCEs) show promising clinical efficacy in blood tumors, but their application to solid tumors remains challenging. Here, we show that Fc-fused IL-7 (rhIL-7-hyFc) changes the intratumoral CD8 T cell landscape, enhancing the efficacy of TCE immunotherapy. rhIL-7-hyFc induces a dramatic increase in CD8 tumor-infiltrating lymphocytes (TILs) in various solid tumors, but the majority of these cells are PD-1-negative tumor non-responsive bystander T cells. However, they are non-exhausted and central memory-phenotype CD8 T cells with high T cell receptor (TCR)-recall capacity that can be triggered by tumor antigen-specific TCEs to acquire tumoricidal activity. Single-cell transcriptome analysis reveals that rhIL-7-hyFc-induced bystander CD8 TILs transform into cycling transitional T cells by TCE redirection with decreased memory markers and increased cytotoxic molecules. Notably, TCE treatment has no major effect on tumor-reactive CD8 TILs. Our results suggest that rhIL-7-hyFc treatment promotes the antitumor efficacy of TCE immunotherapy by increasing TCE-sensitive bystander CD8 TILs in solid tumors.


Subject(s)
CD8-Positive T-Lymphocytes , Immunotherapy , Interleukin-7 , Lymphocytes, Tumor-Infiltrating , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , CD8-Positive T-Lymphocytes/immunology , Interleukin-7/immunology , Interleukin-7/metabolism , Humans , Animals , Immunotherapy/methods , Mice , Neoplasms/immunology , Neoplasms/therapy , Cell Line, Tumor , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Bystander Effect/immunology
18.
Front Endocrinol (Lausanne) ; 15: 1377322, 2024.
Article in English | MEDLINE | ID: mdl-38800484

ABSTRACT

Type 1 diabetes (T1D) is an organ-specific autoimmune disease caused by pancreatic ß cell destruction and mediated primarily by autoreactive CD8+ T cells. It has been shown that only a small number of stem cell-like ß cell-specific CD8+ T cells are needed to convert normal mice into T1D mice; thus, it is likely that T1D can be cured or significantly improved by modulating or altering self-reactive CD8+ T cells. However, stem cell-type, effector and exhausted CD8+ T cells play intricate and important roles in T1D. The highly diverse T-cell receptors (TCRs) also make precise and stable targeted therapy more difficult. Therefore, this review will investigate the mechanisms of autoimmune CD8+ T cells and TCRs in T1D, as well as the related single-cell RNA sequencing (ScRNA-Seq), CRISPR/Cas9, chimeric antigen receptor T-cell (CAR-T) and T-cell receptor-gene engineered T cells (TCR-T), for a detailed and clear overview. This review highlights that targeting CD8+ T cells and their TCRs may be a potential strategy for predicting or treating T1D.


Subject(s)
CD8-Positive T-Lymphocytes , Diabetes Mellitus, Type 1 , Receptors, Antigen, T-Cell , Single-Cell Analysis , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/genetics , CD8-Positive T-Lymphocytes/immunology , Humans , Animals , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Single-Cell Analysis/methods , Sequence Analysis, RNA/methods , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/metabolism , Autoimmunity , Mice
19.
Proc Natl Acad Sci U S A ; 121(19): e2318003121, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38691588

ABSTRACT

Peptides presented by HLA-E, a molecule with very limited polymorphism, represent attractive targets for T cell receptor (TCR)-based immunotherapies to circumvent the limitations imposed by the high polymorphism of classical HLA genes in the human population. Here, we describe a TCR-based bispecific molecule that potently and selectively binds HLA-E in complex with a peptide encoded by the inhA gene of Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis in humans. We reveal the biophysical and structural bases underpinning the potency and specificity of this molecule and demonstrate its ability to redirect polyclonal T cells to target HLA-E-expressing cells transduced with mycobacterial inhA as well as primary cells infected with virulent Mtb. Additionally, we demonstrate elimination of Mtb-infected cells and reduction of intracellular Mtb growth. Our study suggests an approach to enhance host T cell immunity against Mtb and provides proof of principle for an innovative TCR-based therapeutic strategy overcoming HLA polymorphism and therefore applicable to a broader patient population.


Subject(s)
Histocompatibility Antigens Class I , Mycobacterium tuberculosis , Receptors, Antigen, T-Cell , T-Lymphocytes , Mycobacterium tuberculosis/immunology , Humans , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , T-Lymphocytes/immunology , HLA-E Antigens , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Tuberculosis/immunology
20.
Immunol Lett ; 267: 106861, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38697225

ABSTRACT

Hematopoietic precursors (HPCs) entering into the thymus undergo a sequential process leading to the generation of a variety of T cell subsets. This developmental odyssey unfolds in distinct stages within the thymic cortex and medulla, shaping the landscape of T cell receptor (TCR) expression and guiding thymocytes through positive and negative selection. Initially, early thymic progenitors (ETPs) take residence in the thymic cortex, where thymocytes begin to express their TCR and undergo positive selection. Subsequently, thymocytes transition to the thymic medulla, where they undergo negative selection. Both murine and human thymocyte development can be broadly classified into distinct stages based on the expression of CD4 and CD8 coreceptors, resulting in categorizations as double negative (DN), double positive (DP) or single positive (SP) cells. Thymocyte migration to the appropriate thymic microenvironment at the right differentiation stage is pivotal for the development and the proper functioning of T cells, which is critical for adaptive immune responses. The journey of lymphoid progenitor cells into the T cell developmental pathway hinges on an ongoing dialogue between the differentiating cell and the signals emanating from the thymus niche. Herein, we review the contribution of the key factors mentioned above for the localization, migration and emigration of thymocytes.


Subject(s)
Cell Differentiation , Cell Movement , Thymocytes , Thymus Gland , Thymocytes/immunology , Thymocytes/cytology , Thymocytes/metabolism , Animals , Humans , Thymus Gland/cytology , Thymus Gland/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Receptors, Antigen, T-Cell/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...