1.Construction of NKG2D CAR-NK92 cells and its killing effect on multiple myeloma cells.
Jing LONG ; Rong ZHENG ; Sishi YE ; Shanwen KE ; Deming DUAN ; Cheng WEI ; Jimin GAO
Chinese Journal of Cellular and Molecular Immunology 2023;39(7):577-585
Objective This study aims to construct and identify the chimeric antigen receptor NK92 (CAR-NK92) cells targeting NKG2D ligand (NKG2DL) (secreting IL-15Ra-IL-15) and verify the killing activity of NKG2D CAR-NK92 cells against multiple myeloma cells. Methods The extracellular segment of NKG2D was employed to connect 4-1BB and CD3Z, as well as IL-15Ra-IL-15 sequence to obtain a CAR expression framework. The lentivirus was packaged and transduced into NK92 cells to obtain NKG2D CAR-NK92 cells. The proliferation of NKG2D CAR-NK92 cells was detected by CCK-8 assay, IL-15Ra secretion was detected by ELISA and killing efficiency was detected by lactate dehydrogenase (LDH) assay. The molecular markers of NKp30, NKp44, NKp46, the ratio of apoptotic cell population, CD107a, and the secretion level of granzyme B and perforin were detected using flow cytometry. In addition, the cytotoxic mechanism of NKG2D CAR-NK92 cells on the tumor was verified by measuring the degranulation ability. Moreover, after NKG2D antibody inhibited effector cells and histamine inhibited tumor cells, LDH assay was utilized to detect the effect on cell-killing efficiency. Finally, the multiple myeloma tumor xenograft model was constructed to verify its anti-tumor activity in vivo. Results Lentiviral transduction significantly increased NKG2D expression in NK92 cells. Compared with NK92 cells, the proliferation ability of NKG2D CAR-NK92 cells was weaker. The early apoptotic cell population of NKG2D CAR-NK92 cells was less, and NKG2D CAR-NK92 cells had stronger cytotoxicity to multiple myeloma cells. Additionally, IL-15Ra secretion could be detected in its culture supernatant. NKp44 protein expression in NKG2D CAR-NK92 cells was clearly increased, demonstrating an enhanced activation level. Inhibition test revealed that the cytotoxicity of CAR-NK92 cells to MHC-I chain-related protein A (MICA) and MICB-positive tumor cells was more dependent on the interaction between NKG2D CAR and NKG2DL. After stimulating NKG2D CAR-NK92 cells with tumor cells, granzyme B and perforin expression increased, and NK cells obviously upregulated CD107α. Furthermore, multiple myeloma tumor xenograft model revealed that the tumors of mice treated with NKG2D CAR-NK92 cells were significantly reduced, and the cell therapy did not sensibly affect the weight of the mice. Conclusion A type of CAR-NK92 cell targeting NKG2DL (secreting IL-15Ra-IL-15) is successfully constructed, indicating the effective killing of multiple myeloid cells.
Humans
;
Mice
;
Animals
;
Receptors, Chimeric Antigen/genetics*
;
Interleukin-15
;
NK Cell Lectin-Like Receptor Subfamily K/metabolism*
;
Granzymes
;
Cell Line, Tumor
;
Multiple Myeloma/therapy*
;
Perforin
2.Successful treatment of refractory pure red cell aplasia with eltrombopag after ABO-incompatible allogeneic hematopoietic stem cell transplantation.
Yang GAO ; Fei GAO ; Jimin SHI ; Huarui FU ; He HUANG ; Yanmin ZHAO
Journal of Zhejiang University. Science. B 2021;22(8):695-700
Pure red cell aplasia (PRCA) is a well-recognized complication of ABO major mismatched allogeneic hematopoietic stem cell transplantation (allo-HSCT), with a reported incidence of 10%-20% (Zhidong et al., 2012; Busca et al., 2018). It is clinically characterized by anemia, reticulocytopenia, and the absence of erythroblasts in a normal-appearing bone marrow biopsy (Shahan and Hildebrandt, 2015). The mechanism for PRCA has been presumed to be persistence of recipient isoagglutinins, produced by residual host B lymphocytes or plasma cells, which can interfere with the engraftment of donor erythroid cells (Zhidong et al., 2012). Several risk factors of PRCA at presentation are known, such as presence of anti-A isoagglutinins before transplantation, reduced intensity conditioning, absence of acute graft-versus-host disease (GVHD), sibling donors, and cyclosporin A (CsA) as GVHD prophylaxis (Hirokawa et al., 2013). PRCA is not considered to be a barrier to HSCT, as some patients can recover spontaneously or benefit from various approaches including high-dose steroids, erythropoietin (EPO), plasma exchange, immunoadsorption, donor lymphocyte infusion (DLI), treatment with rituximab, bortezomib, or daratumumab, and tapering or discontinuation of immunosuppression (Hirokawa et al., 2013; Bathini et al., 2019). However, there are still some patients who fail to respond even to aggressive treatment; they become red cell transfusion-dependent and iron-overloaded, and their life quality is impaired.
3.Preliminary study of the fourth-generation CAR-T cells targeting CS1 in the treatment of refractory and recurrent multiple myeloma
Dudu FENG ; Xinhua CHEN ; Jingjing GUO ; Keke WANG ; Xiaoman ZHANG ; Jimin GAO
Chinese Journal of Oncology 2021;43(6):657-665
Objective:To design the fourth-generation chimeric antigen receptor-T (CAR-T) cells that secrete interleukin-7 (IL7) and chemokine C legend 19 (CCL19) on the basis of the second-generation CAR, and to analyze and compare the differences in proliferation, chemotaxis, tumor cell clearance and persistence in the microenvironment of multiple myeloma (MM) between them.Methods:The fourth-generation CAR vector plasmid was constructed by using 2A self-cleaving peptide technology. The third-generation lentiviral packaging system was used to prepare high-titer lentivirus. Flow cytometry was used to monitor the transduction efficiency of lentivirus and the subtype changes of CAR-T cells. The enzyme-linked immunosorbent assay (ELISA) was used to quantify the IL7 and CCL19 secreted by CAR-T cells.The calculation of absolute number of CAR-T cells during culture was used to analysis cell proliferation activity. Transwell migration assay was used to verify the chemotactic ability of CAR-T cells. The specific killing activity of CAR-T cells was detected by using the luciferase bioluminescence method. The NOD-Prkdcem26Cd52Il2rgem26Cd22/Nju (NOD) mouse xenograft model was used to verify the anti-myeloma activity and safety of CAR-T cells in vivo.Results:Flow cytometry results showed that the stable CAR expression rates of the second-generation anti-CS1 CAR-T and fourth-generation anti-CS1-IL7-CCL19 CAR-T cells were (91.50±0.29)% and (46.7±0.12)%, respectively. CAR-T cells were successfully constructed. Subtype analysis demonstrated that the ratio of stem memory T cell (TSCM) in anti-CS1-IL7-CCL19 CAR-T cells was (67.58±0.59)%, which was significantly higher than (50.74 ± 1.01)% of anti-CS1 CAR-T ( P=0.000 1), with more strong immune memory function and better durability. Anti-CS1-IL7-CCL19 CAR-T cells can continuously secrete IL7 and CCL19 compared to MOCK-T and anti-CS1 CAR-T ( P<0.000 1). The number of anti-CS1-IL7-CCL19 CAR-T cells reached (22.77±0.79)×10 6 on the 9th day after lentivirus transduction, which was significantly higher than (9.40±0.79)×10 6 of anti-CS1 CAR-T cells ( P=0.000 1), with stronger proliferation ability. The number of chemotaxis cells of anti-CS1-IL7-CCL19 CAR-T cells to reactive T cells was (109.0±4.04), which was significantly higher than (9.33±1.20) of MOCK-T ( P<0.000 1) and (7.33±0.88) of anti-CS1 CAR-T ( P<0.000 1), with stronger chemotactic ability. The specific killing activity showed that both anti-CS1-IL7-CCL19 CAR-T and anti-CS1 CAR-T cells had specific killing efficacies when compared with the MOCK-T cells ( P<0.000 1). Animal experiment indicated that anti-CS1-IL7-CCL19 CAR-T cells significantly reduced the tumor burden ( P<0.000 1) and extended the overall survival time ( P=0.006 1) of tumor-bearing mice. Conclusions:The anti-CS1-IL7-CCL19 CAR-T cells designed in this study show stronger proliferative activity, chemotactic ability, and durability without affecting the anti-myeloma activity in vivo and in vivo, which provides strategies for overcoming the defects of low survival rate, poor durability and inhibition by tumor microenvironment of traditional CAR-T cells, and offers preliminary experimental basis for the clinical application of the fourth-generation CAR-T cells.
4.Preliminary study of the fourth-generation CAR-T cells targeting CS1 in the treatment of refractory and recurrent multiple myeloma
Dudu FENG ; Xinhua CHEN ; Jingjing GUO ; Keke WANG ; Xiaoman ZHANG ; Jimin GAO
Chinese Journal of Oncology 2021;43(6):657-665
Objective:To design the fourth-generation chimeric antigen receptor-T (CAR-T) cells that secrete interleukin-7 (IL7) and chemokine C legend 19 (CCL19) on the basis of the second-generation CAR, and to analyze and compare the differences in proliferation, chemotaxis, tumor cell clearance and persistence in the microenvironment of multiple myeloma (MM) between them.Methods:The fourth-generation CAR vector plasmid was constructed by using 2A self-cleaving peptide technology. The third-generation lentiviral packaging system was used to prepare high-titer lentivirus. Flow cytometry was used to monitor the transduction efficiency of lentivirus and the subtype changes of CAR-T cells. The enzyme-linked immunosorbent assay (ELISA) was used to quantify the IL7 and CCL19 secreted by CAR-T cells.The calculation of absolute number of CAR-T cells during culture was used to analysis cell proliferation activity. Transwell migration assay was used to verify the chemotactic ability of CAR-T cells. The specific killing activity of CAR-T cells was detected by using the luciferase bioluminescence method. The NOD-Prkdcem26Cd52Il2rgem26Cd22/Nju (NOD) mouse xenograft model was used to verify the anti-myeloma activity and safety of CAR-T cells in vivo.Results:Flow cytometry results showed that the stable CAR expression rates of the second-generation anti-CS1 CAR-T and fourth-generation anti-CS1-IL7-CCL19 CAR-T cells were (91.50±0.29)% and (46.7±0.12)%, respectively. CAR-T cells were successfully constructed. Subtype analysis demonstrated that the ratio of stem memory T cell (TSCM) in anti-CS1-IL7-CCL19 CAR-T cells was (67.58±0.59)%, which was significantly higher than (50.74 ± 1.01)% of anti-CS1 CAR-T ( P=0.000 1), with more strong immune memory function and better durability. Anti-CS1-IL7-CCL19 CAR-T cells can continuously secrete IL7 and CCL19 compared to MOCK-T and anti-CS1 CAR-T ( P<0.000 1). The number of anti-CS1-IL7-CCL19 CAR-T cells reached (22.77±0.79)×10 6 on the 9th day after lentivirus transduction, which was significantly higher than (9.40±0.79)×10 6 of anti-CS1 CAR-T cells ( P=0.000 1), with stronger proliferation ability. The number of chemotaxis cells of anti-CS1-IL7-CCL19 CAR-T cells to reactive T cells was (109.0±4.04), which was significantly higher than (9.33±1.20) of MOCK-T ( P<0.000 1) and (7.33±0.88) of anti-CS1 CAR-T ( P<0.000 1), with stronger chemotactic ability. The specific killing activity showed that both anti-CS1-IL7-CCL19 CAR-T and anti-CS1 CAR-T cells had specific killing efficacies when compared with the MOCK-T cells ( P<0.000 1). Animal experiment indicated that anti-CS1-IL7-CCL19 CAR-T cells significantly reduced the tumor burden ( P<0.000 1) and extended the overall survival time ( P=0.006 1) of tumor-bearing mice. Conclusions:The anti-CS1-IL7-CCL19 CAR-T cells designed in this study show stronger proliferative activity, chemotactic ability, and durability without affecting the anti-myeloma activity in vivo and in vivo, which provides strategies for overcoming the defects of low survival rate, poor durability and inhibition by tumor microenvironment of traditional CAR-T cells, and offers preliminary experimental basis for the clinical application of the fourth-generation CAR-T cells.
5.Construction of a novel lentiviral vector knocking down PD-1 via microRNA and its application in CAR-T cells.
Hui CHEN ; Xi JIN ; Xiaoman ZHANG ; Jimin GAO
Chinese Journal of Biotechnology 2020;36(7):1395-1404
By inserting microRNAs into the intron of EF1α promoter, we constructed a novel lentiviral vector knocking down PD-1 gene via microRNA and applied it to CAR-T cells. Lentiviral transduction efficiency and PD-1-silencing efficiency were detected by flow cytometry. PD-1 expression was detected by Western blotting. Relative expression of microRNA was measured by Q-PCR. Cytotoxicity of CAR-T cells based on this vector was tested by luciferase bioluminescence and flow cytometry. Compared with lentiviral vector with microRNA transcribed by U6 promotor, the transduction efficiency of lentiviral vector with microRNA which was inserted into the intron of EF1α promoter was more significant, and the knockdown rate of PD-1 was more than 90%, which was validated by flow cytometry and Western blotting. And the relative expression level of microRNA in Jurkat cells transduced with this novel lentiviral vector was shown by Q-PCR. Compared with normal CAR-T cells, CAR-T cells based on this vector showed stronger cytotoxicity against PD-L1 positive Raji cells. We successfully constructed a novel lentiviral vector that knocked down PD-1 via microRNA and verified the superiority of its transduction efficiency and knockdown efficiency of PD-1. CAR-T cells based on this vector can exert a more powerful cytotoxicity, thus providing theoretical support for the subsequent treatment of PD-L1 positive tumors.
Cell Line, Tumor
;
Gene Knockdown Techniques
;
Genetic Vectors
;
genetics
;
Humans
;
Lentivirus
;
genetics
;
MicroRNAs
;
metabolism
;
Programmed Cell Death 1 Receptor
;
Promoter Regions, Genetic
;
genetics
6.Construction and function of Glypican-3-targeted fourth-generation chimeric antigen receptor T cells (secreting IL-7 and CCL19).
Wanli HUANG ; Yu LIU ; Yaodi HU ; Jimin GAO
Chinese Journal of Biotechnology 2020;36(5):979-991
Adoptive immunotherapy based on chimeric antigen receptor-modified T cells (CAR-T) is one of the most promising strategies to treat malignant tumors, but its application in solid tumors is still limited. Glypican-3 (GPC3) is a meaningful diagnostic, therapeutic, and prognostic biomarker for hepatocellular carcinoma (HCC). The second/third generation GPC3-targeted CAR-T cells are generated to treat HCC. In order to improve the therapeutic effect, we constructed a fourth-generation lentiviral vector to express GPC3 CAR, human interleukin-7 (IL-7) and CCL19. Then the lentiviral vector and packaging plasmids were co-transfected into HEK293T cells to generate CAR lentiviral particles. Human T lymphocyte cells were transduced with CAR lentiviral to develop the fourth-generation GPC3-targeted CAR-T cells (GPC3-BBZ-7×19). In vitro, we used cell counting, transwell assay, luciferase bioluminescence assay and flow cytometry to compare the proliferation, chemotaxis, cytotoxicity and subtype distribution between GPC3-BBZ-7×19 CAR-T cells and the second generation GPC3-targeted CAR-T cells (GPC3-BBZ). In vivo, we established GPC3-positive HCC xenograft model in immunodeficient mice, then untransduced T cells (non-CAR-T) or GPC3-BBZ-7×19 CAR-T cells were injected. Tumor growth in mice was observed by bioluminescence imaging. Results showed that compared with GPC3-BBZ CAR-T, GPC3-BBZ-7×19 CAR-T cells had stronger proliferation, chemotactic ability, and higher composition of memory stem T cells (Tscm) (P values<0.05). However, there were no significant difference in cytotoxicity and cytokine secretion between them. In addition, GPC3-BBZ-7×19 CAR-T cells could significantly eliminate GPC3-positive HCC xenografts established in immunodeficient mice. Therefore, the fourth-generation GPC3-targeted CAR-T cells (secreting IL-7 and CCL19) are expected to be more durable and effective against HCC and produce tumor-specific memory, to provide a preclinical research basis for future clinical trials.
Animals
;
Carcinoma, Hepatocellular
;
Cell Line, Tumor
;
Chemokine CCL19
;
metabolism
;
Glypicans
;
metabolism
;
HEK293 Cells
;
Humans
;
Interleukin-7
;
metabolism
;
Lentivirus
;
genetics
;
Liver Neoplasms
;
Mice
;
Receptors, Chimeric Antigen
;
metabolism
;
T-Lymphocytes
;
metabolism
;
Xenograft Model Antitumor Assays
7.Eukaryotic expression, protein purification and biological effects research of human CS1-Fc fusion protein.
Ruzhang CHEN ; Xitong WANG ; Yanchen LI ; Jimin GAO
Chinese Journal of Biotechnology 2020;36(1):122-132
Signaling lymphocyte activation family 7 (SLAMF7/CS1) is a cell surface glycoprotein that is highly expressed in multiple myeloma cells. CS1 is a sensitive and specific biomarker for multiple myeloma. CAR-T cell immunotherapy is a new method for the treatment of multiple myeloma. CS1 CAR-T cell immunotherapy has good effect on relapsed refractory multiple myeloma. To detect the expression efficiency of CS1 CAR on CS1 CAR-T cells and to find an auxiliary means to CAR-T cell immunotherapy, we prepared a CS1-Fc fusion protein. First, the extracellular domain of CS1 was amplified from the existing plasmid by PCR and ligated with human IgG1-Fc fragment by overlap extension PCR. The recombinant fragment was ligated into pMH3 eukaryotic expression vector. After restriction enzyme digestion and DNA sequencing, the pMH3-CS1-Fc-his recombinant plasmid was successfully constructed. The recombinant plasmid was transfected into Chinese hamster ovary cell (CHO-S) by liposome. The expression of the CS1-Fc fusion protein in CHO-S cells was identified by flow cytometry after G418 pressure screening. Next, the CS1-Fc fusion protein was purified by nickel column. Western-blot analysis showed that molecular weight of the fusion protein was about 70 kDa was identified by Western blotting. The CS1-Fc fusion protein couldeffectively detect the expression rate of CS1 CAR and promote the activation, proliferation andcytokines secretion of the CS1 CAR-T cells. The results will lay the experimental foundation for the in vitro detection and potentiation of CAR-T cells in multiple myeloma treated with CS1 CAR-T cell.
8.Construction of CAR-T cells targeting CS1 and analysis of their antitumor activity in vitro.
Weiguang ZHANG ; Chunling WANG ; Zhibo TAO ; Changlin YIN ; Jimin GAO
Chinese Journal of Biotechnology 2020;36(10):2162-2170
We constructed the CS1-targeted second- and third-generation CAR-T cells with genetic engineered 4-1BB or/and ICOS as a costimulatory signaling molecule by use of lentiviral platform. The CS1-targeted second-generation CAR-T cells with ICOS or 4-1BB had similar anti-neoplastic activity. When effector/target ratio was 1:1, the CAR-T cells with ICOS showed better killing effect on IM9-lucgfp cells than those with 4-1BB. However, The CS1-targeted third-generation CAR-T cells exihibited lower cytolytic capacity against IM9-lucgfp cells than the CS1-targeted second-generation CAR-T cells when the ratio of effector/target was 1:1, 2:1 or 5:1. When the ratio of effector/target was 10:1, the killing efficacy of both the second- and third-generation CAR-T cells against IM9-lucgfp cells was more than 85%, significantly higher than that of the control T cells. Taken together, both the CS1-targeted second- and third-generation CAR-T cells with ICOS or/and 4-1BB could efficiently kill CS1-positive multiple myeloma cells, but the CS1-targeted second-generation CAR-T cells had more potent killing effect on CS1-positive multiple myeloma cells than the CS1-targeted third-generation CAR-T cells.
4-1BB Ligand/metabolism*
;
Cell Line, Tumor
;
Genetic Engineering
;
Humans
;
Inducible T-Cell Co-Stimulator Protein/metabolism*
;
Multiple Myeloma/therapy*
;
Signal Transduction
;
T-Lymphocytes/chemistry*
;
Xenograft Model Antitumor Assays
9.Construction and characterization of the fourth-generation CAR-T cells targeting fibroblast activation protein (FAP)
Fanfan LI ; Hui CHEN ; Dudu FENG ; Jimin GAO
Chinese Journal of Microbiology and Immunology 2020;40(5):372-381
Objective:To construct the second and fourth generations of CAR-T cells targeting fibroblast activation protein (FAP) on the surface of stromal carcinoma-associated fibroblasts and compare their characteristics in vitro and in vivo. Methods:ELISA was used to detect the cytokines secreted by CAR-T cells. Cell proliferation and viability were analyzed by counting. Chemotactic ability was tested by Transwell migration assay. Distribution of T cell subsets was analyzed by flow cytometry. Cytotoxicity was assessed by luciferase bioluminescence. The safety and therapeutic effects were evaluated in a NOG mouse model of metastatic human lung cancer.Results:The expression rates of the second and fourth generations of CAR-T cells (h4BBz CAR-T and h4BBz-7.19 CAR-T) were (74.280±4.384)% and (67.220±4.013)%, respectively. The h4BBz-7.19 CAR-T cells had better in vitro proliferation and chemotactic activity than h4BBz CAR-T cells as they were able to secrete IL-7 and CCL19, while the viability of h4BBz-7.19 CAR-T cells was comparable to that of h4BBz CAR-T cells. There was no significant difference in the expression rate of h4BBz CAR or h4BBz-7.19 CAR between CD4 + T and CD8 + T cells. The proportions of both Naive cells and T memory stem cells (TSCM) in CD4 + and CD8 + T cells were higher in h4BBz-7.19 CAR-T cells than in h4BBz CAR-T cells. Moreover, h4BBz-7.19 CAR-T cells possessed stronger specific cytotoxicity on the target cancer cells than h4BBz CAR-T cells when the ratio of effectors/targets was low ( P1∶1=0.004, P2∶1=0.000 6, P5∶1<0.000 1, P10∶1=0.022, P20∶1=0.116), while the expression of PD-1 on the surface of h4BBz-7.19 CAR-T cells was lower than that on h4BBz CAR-T cells. In the NOG mouse model of metastatic human lung cancer, h4BBz-7.19 CAR-T cells could slow the tumor growth and prolong the survival time of mice without causing weight loss or pathological changes in the organs. Conclusions:The fourth-generation CAR-T cells targeting FAP were shown to have stronger proliferation, better penetration and more potent specific cytotoxicity by secreting IL-7 and CCL19 and could slow the tumor growth and prolong survival by improving tumor immunosuppressive microenvironment. This study provided reference for the clinical application of the fourth generation of CAR-T cells.
10.Metabolomic analysis of human cerebrospinal fluid in patients with tuberculous meningitis using ultra high performance liquid chromatography tandem mass spectrometry
Jingyun YE ; Aihua TONG ; Yanfei HAO ; Xingwang JIA ; Xinyu WEN ; Ximeng CHEN ; Chengbin WANG ; Jimin GAO
Chinese Journal of Laboratory Medicine 2020;43(6):646-652
Objective:To study the non-target metabolomics analysis and to analyze the metabolomic changesof cerebrospinal fluid (CSF) in patients with tuberculous meningitis.Methods:Case-control study. From July 2018 to July 2019, 20 cerebrospinal fluid specimens of diagnosed patients with tuberculous meningitis were collectedin the department of neurology from the first medical center of the PLA general hospital and the eighth medical center of the PLA general hospital and 20 CSF without tuberculous meningitis as the control. Among them, there were 12 males and 8 femalesin the tuberculous meningitis group, aged (37.9±16.1) years; there were 13 males and 7 femalesin the control group, aged (34.7±14.8) years. Using ultra-high performance liquid chromatography-mass spectrometry (UPLC-MS) technology with three different mode, namely reverse phase chromatography positive ion mode, reverse phase chromatography negative ion mode and hydrophilic chromatography positive ion mode,to detectthe metabolic fingerprints of patients′CSF and analyzed by SIMCA software for orthogonal partial least squares discriminant analysis (OPLS-DA). The variable importance projection value of OPLS-DA model (threshold value>1) plus the P value of t-test (P<0.05) was applied to find the differential metabolites in the cerebrospinal fluid of the two groups of patients.Results:Ten differential metabolites were found in CSF, including L-isoleucine, L-phenylalanine, L-kynurenine, L-methionine, L-tyrosine acid, dimethylglycine, L-alanine, L-threonine, L-histidine and L-lysine, and all of them were up-regulated in the tuberculous meningitis group.Conclusion:Changesof the amino acid metabolism found in the cerebrospinal fluid of tuberculous meningitis patients can provide basis for differential diagnosis and basic molecular research of tuberculous meningitis.

Result Analysis
Print
Save
E-mail