1.CAR-based cell therapies for systemic lupus erythematosus.
Yiyang WANG ; Liangjing LU ; Shuang YE ; Qiong FU
Chinese Medical Journal 2025;138(5):523-530
The remarkable efficacy of chimeric antigen receptor (CAR) T cell therapy in hematological malignancies has provided a solid basis for the therapeutic concept, wherein specific pathogenic cell populations can be eradicated by means of targeted recognition. During the past few years, CAR-based cell therapies have been extensively investigated in preclinical and clinical research across various non-tumor diseases, with particular emphasis in the treatment of autoimmune diseases (ADs), yielding significant advancements. The recent deployment of CD19-directed CAR T cells has induced long-lasting, drug-free remission in patients with systemic lupus erythematosus (SLE) and other systemic ADs, alongside a more profound immune reconstruction of B cell repertoire compared with conventional immunosuppressive agents and B cell-targeting biologics. Despite the initial success achieved by CAR T cell therapy, it is critical to acknowledge the divergences in its application between cancer and ADs. Through examining recent clinical studies and ongoing research, we highlight the transformative potential of this therapeutic approach in the treatment of SLE, while also addressing the challenges and future directions necessary to enhance the long-term efficacy and safety of CAR-based cell therapies in clinical practice.
Humans
;
Lupus Erythematosus, Systemic/immunology*
;
Receptors, Chimeric Antigen/metabolism*
;
Immunotherapy, Adoptive/methods*
;
Cell- and Tissue-Based Therapy/methods*
;
Animals
;
T-Lymphocytes/immunology*
2.c-Met-targeted chimeric antigen receptor T cells inhibit human serous ovarian cancer cell SKOV-3 in vitro.
Na-Na DU ; Yan-Jun ZHANG ; Yan-Qiu LI ; Lu ZHANG ; Ran AN ; Xiang-Cheng ZHEN ; Jing-Ting MIN ; Zheng-Hong LI
Acta Physiologica Sinica 2025;77(2):241-254
The study aimed to construct the second and third generation chimeric antigen receptor T cells (CAR-T) targeting the c-mesenchymal-epithelial transition factor (c-Met) protein, and observe their killing effect on human serous ovarian cancer cell SKOV-3. The expression of MET gene in ovarian serous cystadenocarcinoma, the correlation between MET gene expression and the abundance of immune cell infiltration, and the effect of MET gene expression on the tissue function of ovarian serous cystadenocarcinoma were analyzed by bioinformatics. The expression of c-Met in ovarian cancer tissues and adjacent tissues was detected by immunohistochemical staining. The second and third generation c-Met CAR-T cells, namely c-Met CAR-T(2G/3G), were prepared by lentivirus infection, and the cell subsets and infection efficiency were detected by flow cytometry. Using CD19 CAR-T and activated T cells as control groups and A2780 cells with c-Met negative expression as Non target groups, the kill efficiency on SKOV-3 cells with c-Met positive expression, cytokine release and cell proliferation of c-Met CAR-T(2G/3G) were explored by lactate dehydrogenase (LDH) release, ELISA and CCK-8 respectively. The results showed that MET gene expression was significantly up-regulated in ovarian cancer tissues compared with normal tissues, which was consistent with the immunohistochemistry results. However, in all pathological stages, there was no obvious difference in MET expression and no correlation between MET gene expression and the race and age of ovarian cancer patients. The second generation and third generation c-Met CAR-T cells were successfully constructed. After lentivirus infection, the proportion of CD8+ T cells in c-Met CAR-T(2G) was upregulated, while there was no significant change in the cell subsets of c-Met CAR-T(3G). The LDH release experiment showed that the kill efficiency of c-Met CAR-T(2G/3G) on SKOV-3 increased with the increase of effect-target ratio. When the effect-target ratio was 20:1, the kill efficiency of c-Met CAR-T(2G) reached (42.02 ± 5.17)% (P < 0.05), and the kill efficiency of c-Met CAR-T(3G) reached (51.40 ± 2.71)% (P < 0.05). ELISA results showed that c-Met CAR-T released more cytokine compared to CD19 CAR-T and activated T cells (P < 0.05). Moreover, the cytokine release of c-Met CAR-T(3G) was higher than c-Met CAR-T(2G) (P < 0.01). The CCK-8 results showed that after 48 h, the cell number of c-Met CAR-T(2G) was higher than that of c-Met CAR-T(3G) (P < 0.01). In conclusion, both the second and third generation c-Met CAR-T can target and kill c-Met-positive SKOV-3 cells, with no significant difference. c-Met CAR-T(2G) has stronger proliferative ability, and c-Met CAR-T(3G) releases more cytokines.
Humans
;
Female
;
Ovarian Neoplasms/immunology*
;
Proto-Oncogene Proteins c-met/metabolism*
;
Receptors, Chimeric Antigen/immunology*
;
Cell Line, Tumor
;
Cystadenocarcinoma, Serous/immunology*
;
T-Lymphocytes/immunology*
3.Screening of Anti-Tumor Drugs that Enhance Antigen Presentation of AML Cells with TCR-Like Antibody.
Xiao-Ying YANG ; Bo TANG ; Hui-Hui LIU ; Wei-Wei XIE ; Shuang-Lian XIE ; Wen-Qiong WANG ; Jin WANG ; Shan ZHAO ; Yu-Jun DONG
Journal of Experimental Hematology 2025;33(5):1305-1311
OBJECTIVE:
To screen anti-tumor drugs that improve antigen processing and presentation in acute myeloid leukemia (AML) cells.
METHODS:
A TCR-like or TCR mimic antibody that can specifically recognize HLA-A*0201:WT1126-134 ( RMFPNAPYL) complex (hereafter referred to as HLA-A2:WT1) was synthesized to evaluate the function of antigen processing and presentation machinery (APM) in AML cells. AML cell line THP1 was incubated with increasing concentrations of IFN-γ, hypomethylating agents (HMA), immunomodulatory drugs (IMiD), proteasome inhibitors (PI) and γ-secretase inhibitors (GSI), followed by measuring of HLA-ABC, HLA-A2 and HLA-A2:WT1 levels by flow cytometry at consecutive time points.
RESULTS:
The TCR-like antibody we generated only binds to HLA-A*0201+WT1+ cells, indicating the specificity of the antibody. HLA-A2:WT1 level of THP-1 cells detected with the TCR-like antibody was increased significantly after co-incubation with IFN-γ, showing that the HLA-A2:WT1 TCR like antibody could evaluate the function of APM. Among the anti-tumor agents screened in this study, GSI (LY-411575) and HMA (decitabine and azacitidine) could significantly increase the HLA-A2:WT1 level. The IMiD lenalidomide and pomalidomide could aslo upregulate the expression of HLA-A2:WT1 complex under certain concentrations of the drugs and incubation time. As proteasome inhibitors, carfilzomib could significantly decreased the expression of HLA-A2:WT1, while bortezomib had no significant effect on HLA-A2:WT1 expression.
CONCLUSION
HLA-A2:WT1 TCR-like antibody can effectively reflect the APM function. Some of the anti-tumor drugs can affect the APM function and immunogenicity of tumor cells.
Humans
;
Leukemia, Myeloid, Acute/immunology*
;
Antineoplastic Agents/pharmacology*
;
Antigen Presentation/drug effects*
;
HLA-A2 Antigen/immunology*
;
Receptors, Antigen, T-Cell/immunology*
;
Cell Line, Tumor
;
Interferon-gamma
4.Prediction of MHC II antigen peptide-T cell receptors binding based on foundation model.
Minrui XU ; Siwen ZHANG ; Manman LU ; Yuan GAO ; Menghuan ZHANG ; Yong LIN ; Lu XIE
Journal of Biomedical Engineering 2024;41(6):1243-1249
The specific binding of T cell receptors (TCRs) to antigenic peptides plays a key role in the regulation and mediation of the immune process and provides an essential basis for the development of tumour vaccines. In recent years, studies have mainly focused on TCR prediction of major histocompatibility complex (MHC) class I antigens, but TCR prediction of MHC class II antigens has not been sufficiently investigated and there is still much room for improvement. In this study, the combination of MHC class II antigen peptide and TCR prediction was investigated using the ProtT5 grand model to explore its feature extraction capability. In addition, the model was fine-tuned to retain the underlying features of the model, and a feed-forward neural network structure was constructed for fusion to achieve the prediction model. The experimental results showed that the method proposed in this study performed better than the traditional methods, with a prediction accuracy of 0.96 and an AUC of 0.93, which verifies the effectiveness of the model proposed in this paper.
Receptors, Antigen, T-Cell/immunology*
;
Histocompatibility Antigens Class II/metabolism*
;
Humans
;
Neural Networks, Computer
;
Peptides/metabolism*
;
Protein Binding
5.Anti-tumor therapy strategy of CAR-T cells based on stem cell memory and central memory cells.
Weihua LIU ; Yifei WANG ; Xiaoting SUN ; Zhibin WANG
Chinese Journal of Cellular and Molecular Immunology 2024;40(12):1121-1126
Cancer immunotherapy including immune checkpoint inhibitors and adoptive cell therapy has gained revolutionary success in the treatment of hematologic tumors; however, it only gains limited success in solid tumors. For example, chimeric antigen receptor T (CAR-T) cell therapy has shown significant effects and potential for curing patients with B-cell malignancies. In contrast, it remains a challenge for CAR-T cell therapy to gain similar success in solid tumors. The anti-tumor effect of endogenous or adoptively transferred tumor-specific T cells depends largely on their differentiation status. T cells at early differentiation stage show better anti-tumor therapeutic effects than fully differentiated effector T cells. In cancer patients, the persistence of tumor-specific T cells with the stem cell memory or precursor phenotype is significantly associated with improved therapeutic outcomes; therefore, adoptively transfered CAR-T cells with stem cell memory and/or central memory is expected to gain better anti-tumor effects. Herein we focused on the in vitro optimized culture and expansion system to obtain CAR-T cells with stem cell memory or central memory phenotype for the review.
Humans
;
Immunotherapy, Adoptive/methods*
;
Receptors, Chimeric Antigen/genetics*
;
Neoplasms/immunology*
;
Immunologic Memory
;
T-Lymphocytes/immunology*
;
Memory T Cells/immunology*
;
Animals
;
Stem Cells/cytology*
;
Cell Differentiation
6.T cell characteristics in individuals with different immune responses after hepatitis B vaccination.
Zhiyong HAN ; Dan WANG ; Xiaoyan HE ; Qiang XIA
Journal of Central South University(Medical Sciences) 2024;49(12):1983-1990
Hepatitis B is a global public health concern. Inducing hepatitis B surface antibody (HBsAb) through vaccination is a crucial preventive strategy. However, individuals show varying immune responses to the hepatitis B vaccine. Based on HBsAb levels, individuals can be categorized as high responders, low responders, or non-responders. T cells and their subsets play critical roles in modulating this response, and the composition of the T cell receptor (TCR) repertoire also influences immune responsiveness. Investigating the characteristics of T cells, their subsets, and TCR repertoires in individuals with differential responses post-vaccination may provide theoretical guidance for optimizing vaccine design and immunization strategies.
Humans
;
Hepatitis B Vaccines/immunology*
;
Hepatitis B/immunology*
;
Vaccination
;
Hepatitis B Antibodies/blood*
;
T-Lymphocytes/immunology*
;
Receptors, Antigen, T-Cell/immunology*
;
Hepatitis B Surface Antigens/immunology*
;
T-Lymphocyte Subsets/immunology*
7.Short-chain fatty acids ameliorate spinal cord injury recovery by regulating the balance of regulatory T cells and effector IL-17+ γδ T cells.
Pan LIU ; Mingfu LIU ; Deshuang XI ; Yiguang BAI ; Ruixin MA ; Yaomin MO ; Gaofeng ZENG ; Shaohui ZONG
Journal of Zhejiang University. Science. B 2023;24(4):312-325
Spinal cord injury (SCI) causes motor, sensory, and autonomic dysfunctions. The gut microbiome has an important role in SCI, while short-chain fatty acids (SCFAs) are one of the main bioactive mediators of microbiota. In the present study, we explored the effects of oral administration of exogenous SCFAs on the recovery of locomotor function and tissue repair in SCI. Allen's method was utilized to establish an SCI model in Sprague-Dawley (SD) rats. The animals received water containing a mixture of 150 mmol/L SCFAs after SCI. After 21 d of treatment, the Basso, Beattie, and Bresnahan (BBB) score increased, the regularity index improved, and the base of support (BOS) value declined. Spinal cord tissue inflammatory infiltration was alleviated, the spinal cord necrosis cavity was reduced, and the numbers of motor neurons and Nissl bodies were elevated. Enzyme-linked immunosorbent assay (ELISA), real-time quantitative polymerase chain reaction (qPCR), and immunohistochemistry assay revealed that the expression of interleukin (IL)-10 increased and that of IL-17 decreased in the spinal cord. SCFAs promoted gut homeostasis, induced intestinal T cells to shift toward an anti-inflammatory phenotype, and promoted regulatory T (Treg) cells to secrete IL-10, affecting Treg cells and IL-17+ γδ T cells in the spinal cord. Furthermore, we observed that Treg cells migrated from the gut to the spinal cord region after SCI. The above findings confirm that SCFAs can regulate Treg cells in the gut and affect the balance of Treg and IL-17+ γδ T cells in the spinal cord, which inhibits the inflammatory response and promotes the motor function in SCI rats. Our findings suggest that there is a relationship among gut, spinal cord, and immune cells, and the "gut-spinal cord-immune" axis may be one of the mechanisms regulating neural repair after SCI.
Animals
;
Rats
;
Interleukin-17
;
Rats, Sprague-Dawley
;
Recovery of Function
;
Spinal Cord Injuries/drug therapy*
;
T-Lymphocytes, Regulatory
;
Receptors, Antigen, T-Cell, gamma-delta/immunology*
8.Heterogeneity of Human γδ T Cells and Their Role in Cancer Immunity
Hye Won LEE ; Yun Shin CHUNG ; Tae Jin KIM
Immune Network 2020;20(1):5-
The γδ T cells are unconventional lymphocytes that function in both innate and adaptive immune responses against various intracellular and infectious stresses. The γδ T cells can be exploited as cancer-killing effector cells since γδ TCRs recognize MHC-like molecules and growth factor receptors that are upregulated in cancer cells, and γδ T cells can differentiate into cytotoxic effector cells. However, γδ T cells may also promote tumor progression by secreting IL-17 or other cytokines. Therefore, it is essential to understand how the differentiation and homeostasis of γδ T cells are regulated and whether distinct γδ T cell subsets have different functions. Human γδ T cells are classified into Vδ2 and non-Vδ2 γδ T cells. The majority of Vδ2 γδ T cells are Vγ9δ2 T cells that recognize pyrophosphorylated isoprenoids generated by the dysregulated mevalonate pathway. In contrast, Vδ1 T cells expand from initially diverse TCR repertoire in patients with infectious diseases and cancers. The ligands of Vδ1 T cells are diverse and include the growth factor receptors such as endothelial protein C receptor. Both Vδ1 and Vδ2 γδ T cells are implicated to have immunotherapeutic potentials for cancers, but the detailed elucidation of the distinct characteristics of 2 populations will be required to enhance the immunotherapeutic potential of γδ T cells. Here, we summarize recent progress regarding cancer immunology of human γδ T cells, including their development, heterogeneity, and plasticity, the putative mechanisms underlying ligand recognition and activation, and their dual effects on tumor progression in the tumor microenvironment.
Allergy and Immunology
;
Communicable Diseases
;
Cytokines
;
Homeostasis
;
Humans
;
Interleukin-17
;
Ligands
;
Lymphocytes
;
Mevalonic Acid
;
Plastics
;
Population Characteristics
;
Protein C
;
Receptors, Antigen, T-Cell, gamma-delta
;
Receptors, Growth Factor
;
T-Lymphocyte Subsets
;
T-Lymphocytes
;
Terpenes
;
Tumor Microenvironment
9.Current advances in chimeric antigen receptor T-cell therapy for refractory/relapsed multiple myeloma.
He HUANG ; Heng-Wei WU ; Yong-Xian HU
Journal of Zhejiang University. Science. B 2020;21(1):29-41
Multiple myeloma (MM), considered an incurable hematological malignancy, is characterized by its clonal evolution of malignant plasma cells. Although the application of autologous stem cell transplantation (ASCT) and the introduction of novel agents such as immunomodulatory drugs (IMiDs) and proteasome inhibitors (PIs) have doubled the median overall survival to eight years, relapsed and refractory diseases are still frequent events in the course of MM. To achieve a durable and deep remission, immunotherapy modalities have been developed for relapsed/refractory multiple myeloma (RRMM). Among these approaches, chimeric antigen receptor (CAR) T-cell therapy is the most promising star, based on the results of previous success in B-cell neoplasms. In this immunotherapy, autologous T cells are engineered to express an artificial receptor which targets a tumor-associated antigen and initiates the T-cell killing procedure. Tisagenlecleucel and Axicabtagene, targeting the CD19 antigen, are the two pacesetters of CAR T-cell products. They were approved by the US Food and Drug Administration (FDA) in 2017 for the treatment of acute lymphocytic leukemia (ALL) and diffuse large B-cell lymphoma (DLBCL). Their development enabled unparalleled efficacy in combating hematopoietic neoplasms. In this review article, we summarize six promising candidate antigens in MM that can be targeted by CARs and discuss some noteworthy studies of the safety profile of current CAR T-cell therapy.
ADP-ribosyl Cyclase 1/immunology*
;
B-Cell Maturation Antigen/immunology*
;
Humans
;
Immunotherapy, Adoptive/methods*
;
Multiple Myeloma/therapy*
;
Receptors, Chimeric Antigen/immunology*
;
Receptors, G-Protein-Coupled/immunology*
;
Signaling Lymphocytic Activation Molecule Family/immunology*
;
Syndecan-1/immunology*
;
T-Lymphocytes/immunology*
10.Single-cell Analysis of CAR-T Cell Activation Reveals A Mixed T1/T2 Response Independent of Differentiation.
Iva XHANGOLLI ; Burak DURA ; GeeHee LEE ; Dongjoo KIM ; Yang XIAO ; Rong FAN
Genomics, Proteomics & Bioinformatics 2019;17(2):129-139
The activation mechanism of chimeric antigen receptor (CAR)-engineered T cells may differ substantially from T cells carrying native T cell receptor, but this difference remains poorly understood. We present the first comprehensive portrait of single-cell level transcriptional and cytokine signatures of anti-CD19/4-1BB/CD28/CD3ζ CAR-T cells upon antigen-specific stimulation. Both CD4 helper T (T) cells and CD8 cytotoxic CAR-T cells are equally effective in directly killing target tumor cells and their cytotoxic activity is associated with the elevation of a range of T1 and T2 signature cytokines, e.g., interferon γ, tumor necrotic factor α, interleukin 5 (IL5), and IL13, as confirmed by the expression of master transcription factor genes TBX21 and GATA3. However, rather than conforming to stringent T1 or T2 subtypes, single-cell analysis reveals that the predominant response is a highly mixed T1/T2 function in the same cell. The regulatory T cell activity, although observed in a small fraction of activated cells, emerges from this hybrid T1/T2 population. Granulocyte-macrophage colony stimulating factor (GM-CSF) is produced from the majority of cells regardless of the polarization states, further contrasting CAR-T to classic T cells. Surprisingly, the cytokine response is minimally associated with differentiation status, although all major differentiation subsets such as naïve, central memory, effector memory, and effector are detected. All these suggest that the activation of CAR-engineered T cells is a canonical process that leads to a highly mixed response combining both type 1 and type 2 cytokines together with GM-CSF, supporting the notion that polyfunctional CAR-T cells correlate with objective response of patients in clinical trials. This work provides new insights into the mechanism of CAR activation and implies the necessity for cellular function assays to characterize the quality of CAR-T infusion products and monitor therapeutic responses in patients.
Antigens
;
metabolism
;
CTLA-4 Antigen
;
metabolism
;
Cell Differentiation
;
drug effects
;
Cell Line
;
Cytokines
;
metabolism
;
Cytotoxicity, Immunologic
;
drug effects
;
Granulocyte-Macrophage Colony-Stimulating Factor
;
pharmacology
;
Humans
;
Lymphocyte Activation
;
drug effects
;
immunology
;
Lymphocyte Subsets
;
drug effects
;
metabolism
;
Phenotype
;
Proteomics
;
Receptors, Chimeric Antigen
;
metabolism
;
Single-Cell Analysis
;
methods
;
T-Lymphocytes, Regulatory
;
drug effects
;
metabolism
;
Th1 Cells
;
cytology
;
drug effects
;
Th2 Cells
;
cytology
;
drug effects
;
Transcription, Genetic
;
drug effects
;
Up-Regulation
;
drug effects

Result Analysis
Print
Save
E-mail