Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

Postal Subscription Code 80-967

2018 Impact Factor: 1.847

Front. Med.    2024, Vol. 18 Issue (1) : 128-146    https://doi.org/10.1007/s11684-023-1010-1
Tumor-derived exosomes induce initial activation by exosomal CD19 antigen but impair the function of CD19-specific CAR T-cells via TGF-β signaling
Yuanyuan Hao1,2, Panpan Chen1, Shanshan Guo1, Mengyuan Li1, Xueli Jin1, Minghuan Zhang1, Wenhai Deng3, Ping Li4, Wen Lei1(), Aibin Liang4(), Wenbin Qian1()
1. Department of Hematology, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China
2. Department of Hematology, Henan Provincial People’s Hospital; Zhengzhou University People’s Hospital, Zhengzhou 450003, China
3. Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325000, China
4. Department of Hematology, Tongji Hospital of Tongji University, Shanghai 200065, China
 Download: PDF(7321 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Tumor-derived exosomes (TEXs) enriched in immune suppressive molecules predominantly drive T-cell dysfunction and impair antitumor immunity. Chimeric antigen receptor (CAR) T-cell therapy has emerged as a promising treatment for refractory and relapsed hematological malignancies, but whether lymphoma TEXs have the same impact on CAR T-cell remains unclear. Here, we demonstrated that B-cell lymphoma-derived exosomes induce the initial activation of CD19–CAR T-cells upon stimulation with exosomal CD19. However, lymphoma TEXs might subsequently induce CAR T-cell apoptosis and impair the tumor cytotoxicity of the cells because of the upregulated expression of the inhibitory receptors PD-1, TIM3, and LAG3 upon prolonged exposure. Similar results were observed in the CAR T-cells exposed to plasma exosomes from patients with lymphoma. More importantly, single-cell RNA sequencing revealed that CAR T-cells typically showed differentiated phenotypes and regulatory T-cell (Treg) phenotype conversion. By blocking transforming growth factor β (TGF-β)–Smad3 signaling with TGF-β inhibitor LY2109761, the negative effects of TEXs on Treg conversion, terminal differentiation, and immune checkpoint expression were rescued. Collectively, although TEXs lead to the initial activation of CAR T-cells, the effect of TEXs suppressed CAR T-cells, which can be rescued by LY2109761. A treatment regimen combining CAR T-cell therapy and TGF-β inhibitors might be a novel therapeutic strategy for refractory and relapsed B-cell lymphoma.

Keywords tumor-derived exosome      chimeric antigen receptor T-cell      lymphoma      TGF-β     
Corresponding Author(s): Wen Lei,Aibin Liang,Wenbin Qian   
About author:

Li Liu and Yanqing Liu contributed equally to this work.

Just Accepted Date: 13 September 2023   Online First Date: 20 October 2023    Issue Date: 22 April 2024
 Cite this article:   
Yuanyuan Hao,Panpan Chen,Shanshan Guo, et al. Tumor-derived exosomes induce initial activation by exosomal CD19 antigen but impair the function of CD19-specific CAR T-cells via TGF-β signaling[J]. Front. Med., 2024, 18(1): 128-146.
 URL:  
https://academic.hep.com.cn/fmd/EN/10.1007/s11684-023-1010-1
https://academic.hep.com.cn/fmd/EN/Y2024/V18/I1/128
Fig.1  Characterization and surface markers of exosomes from B-cell malignancies. (A) Size distribution of exosomes from lymphoma cell lines was determined by dynamic light scattering (DLS). (B) Transmission electron micrographs of the exosomes. Scale bars = 200 nm. (C) Western blot for exosome markers (ALIX, TSG101, and CD63) and cellular proteins in Raji–Exo or OCI-LY3–Exo. (D) Flow cytometry analysis of CD19, CD20, and PD-L1 expression on the surfaces of exosomes and their parental tumor cells. The green histograms represent the isotype-matched control mAbs for indicated fluorescent antibodies, whereas the red histograms indicate positive fluorescence. (E) Western blot for determining exosome markers (ALIX, TSG101, and CD63) and TGF-β in exosomes from six patients with B-cell lymphoma. (F) CD19 expression on the surface of exosomes from 18 patients with newly-diagnosed or refractory/relapsed B-cell lymphoma was analyzed through flow cytometry. Data are mean ± S.D.
Fig.2  Priming with CD19-contained lymphoma exosomes activates CD19-specific CAR T-cells. (A) CD19-specific CAR T-cells stained with Hoechst 33 258 (1 μg/mL) were cultured with PKH67-labeled exosomes (100 μg/mL) derived from Raji or OCI-LY3 cells, and the uptake of exosomes was photographed by confocal laser scanning microscopy at 0 and 6 h. Green represents PKH67-labeled exosomes; blue represents CAR T-cells’ nuclei. (B) Killing activity of CAR T-cells that primed with 100 μg/mL Raji–Exo or OCI-LY3–Exo in response to tumor cells. The cytotoxic activity of CAR-T against tumor cells Raji–Luc–GFP was assessed by a luminescence-mediated cytotoxic assay at effector-to-target (E:T) ratios of 10:1, 5:1, 2.5:1, 1.25:1, and 0.625:1 for 4 h. (C) CAR T-cells were cocultured with or without exosomes (100 μg/mL) for 48 h. The concentration of IFN-?, TNF-α, IL-10, IL-6, and IL-4 cytokines in the coculture system were measured by cytometric bead array (CBA) human Th1/Th2 cytokine kit. (D) Flow cytometry analyses the expression of granzyme B in Raji or OCI-LY3 exosome–pretreated CAR T-cells. (E) (a) CAR T-cells (1 × 105 cells) were stained with 1 μmol/L CFSE dye at 37 °C in the dark for 30 min and then treated with Raji or OCI-LY3-exosomes (100 μg/mL) for 48 h. The dilution of CFSE was evaluated through flow cytometry analysis in each group. Flow cytometry analyses the cell cycle (b) and expression of Ki-67 (c) and CD25 and CD69 (d) in the CAR T-cells, which were exposed to indicated exosomes for 48 h. Data are represented as mean ± S.D of three independent biological replicates. P values were obtained from unpaired two-tailed Student’s t-tests. *P < 0.05, **P < 0.01, ***P < 0.001.
Fig.3  Initial activation of CAR T-cells is dependent on exosomal CD19 antigen. (A) The expression of CD19 on the surface of wide-type (Wt) Raji, CD19-KO Raji cells, or their derived exosomes. Red histogram indicates PE-conjugated CD19 antibody fluorescence. The green histogram represents PE isotype Ctrl antibody fluorescence. (B) (a) CFSE-mediated proliferation of CAR-T cocultured with Wt Raji or CD19-KO Raji–Exo was analyzed. Spared CAR-T served as the control. CAR T-cells were exposed to Wt Raji or CD19-KO Raji–Exo for 48 h, and the expression of Ki-67 (b), cell cycle (c), cytotoxicity (d), and the secretion of granzyme B (e) were analyzed. (C) Western blot for exosome marker protein CD63 in Raji–Exo or Raji–Rab27a KO cells (left). Transmission electron micrographs of exosomes in Raji–Exo or Raji–Rab27aKO cells (right). Scale bars = 200 nm. (D) Long-term cytotoxicity of CAR T-cells exposed to Raji–Exo on Raji–Rab27a KO-GFP cells. CAR T-cells were pulsed with the Raji–Exo or not for 48 h and cocultured with Raji–Rab27a KO-GFP cells at an E:T ratio of 1:4 for 5, 10, and 13 days. Flow cytometry analyses the expression of residual GFP+ tumor cells at indicated times. (E) In vivo antitumor effect of Raji–Exo-pretreated CAR T-cells. NSG mice were injected intravenously with Raji–Rab27a KO-GFP-luciferase cells (1 × 105) on day −5 and treated with CD19–CAR-T or Raji–Exo-primed CD19–CAR T-cells (1 × 106) on day 0. PBS was injected into the control group. (a) The tumor burden was measured using bioluminescence imaging; (b) Total bioluminescence was traced in CAR-T, Raji–Exo-primed CAR T-cells, and control group; (c) Kaplan–Meier analysis of mice survival curve. Error bars represent SEM. P values were calculated using unpaired two-tailed Student’s t-tests. *P < 0.05, **P < 0.01, ***P < 0.001.
Fig.4  TEXs and plasma exosomes from patients with lymphoma impair the effector functions of CAR T-cells. (A) CAR T-cells were exposed to a dose escalation of the Raji–Exo for 6 h, and cell viability was determined using MTT assay. The expression of 7AAD. (B) TIM-3, LAG-3, and PD-1. (C) and the differentiation of TCM (CD45RACD62L+), TNaïve (CD45RA+CD62L+), TEFF (CD45RA+CD62L), and TEM (CD45RACD62L). (D) Analysis by flow cytometry after CAR T-cells were exposed to Raji–Exo for 48 h. (E) CAR T-cells primed with or without Raji–Exo were cocultured in the presence or absence of Raji TEXs with Raji–Luc–GFP cells for 6 h, and a luminescence-mediated cytotoxic assay was performed for cytotoxicity evaluation. CAR T-cells were exposed to plasma exosomes (100 μg/mL) from two patients for 48 h, the expression of TIM-3, LAG-3, and PD-1 (F), and the differentiation of TCM (CD45RACD62L+), TNaïve (CD45RA+CD62L+), TEFF (CD45RA+CD62L), and TEM (CD45RACD62L) (G) were analyzed by flow cytometry. (H) CAR T-cells derived from three different patients were cocultured with plasma exosomes (100 μg/mL) from two patients for 48 h and then analyzed through flow cytometry to determine the percent of Tregs (CD4+CD25+FOXP3+). P values were obtained through unpaired two-tailed Student’s t-tests. *P < 0.05, **P < 0.01, ***P < 0.001.
Fig.5  Single-cell transcriptional profiling of CAR T-cells after Raji TEX exposure. (A) The UMAP plot shows the annotation and color codes for cell types of the CAR T-cells derived from three different patients. (B) Heatmap showing the expression of marker genes in the indicated cell types. The top genes are shown on the left. (C) Plot depicts the differences in the proportion of clusters between the CAR T primed with Raji–Exo and untreated CAR T-cells. Red dots indicate that the proportions of these clusters increased in CAR T-cells primed with the Raji–Exo group, and the blue dots mean a decrease. (D) The graphs show the results of a GSEA performed for the indicated gene sets in Raji–Exo-exposed CAR T-cells. The normalized enrichment Score (NES) and false discovery rate (FDR) are indicated.
Fig.6  Analysis of differentially expressed gene related to the T-cell function and phenotype of CAR T-cells after Raji–Exo exposure by single-cell transcriptional profiling. (A) Dot plot illustrates the expression level of genes associated with differentiation, activation, cytotoxicity, proliferation, immune inhibitory, transactions, and chemotaxis in Raji–Exo-exposed CAR T-cell (experiment) and untreated CAR T-cells (control). The size of the circle represents the percentage of cells expressing the gene in each cluster, and the color depicts how highly expressed the gene is within that cluster. (B) Monocle3 trajectory analysis of CAR T-cells. The color represents the stage of cell growth. The black arrow shows the pseudo-time trajectory of the cell growth. (C) The expression of genes (CD69, GZMB, HAVCR2, and LAG3) during the pseudo-time trajectory.
Fig.7  Exosomal transforming growth factor β contributes to the conversion of CAR T-cells into Tregs and terminal differentiated phenotype. (A) UMAP plot shows the expression pattern of FOXP3. Color intensity represents expression level (left). The percentage of FOXP3+ cells was shown in the experimental and control groups (Right). (B) Real-time PCR (left) and flow cytometry (right) were performed for the analysis of FOXP3 expression after CAR T-cells were primed with Raji–Exo for 48 h. (C) Expression of TGF-β in Raji–Exo and OCI-LY3–Exo by Western blots analysis. (D) Western blot was performed for the analysis of SMAD3 and p-SMAD3 expression in CAR T-cells cocultured with Raji–Exo in the presence or absence of the TGF-β inhibitor LY2109761 (100 μmol/L). CAR T-cells alone or CAR T-cells cocultured with Raji–Exo in the presence of 0, 25, 50, and 100 μmol/L LY2109761 for 48 h, and then flow cytometry analyses were performed to determine the percentages of Tregs (CD4+CD25+FOXP3+) (E), TEFF (CD62LCD45RA+) (F), and the expression of LAG-3, TIM-3, and PD-1 (G). CAR T-cells were cocultured with Raji–Exo and PD-1 inhibitor (IBI308, 30 μg/mL) or LAG3 inhibitor (SHR-1702, 40 μg/mL) for 48 h, and the expression levels of LAG3, TIM3, and PD-1 on the surfaces of CAR T-cells (H) and differentiated phenotypes (I) were analyzed by flow cytometry. P values were from unpaired two-tailed Student’s t-tests. *P < 0.05, **P < 0.01, ***P < 0.001.
Fig.8  CD19–CAR T-cells combined with TGF-β inhibitor eliminate B-cell lymphoma. (A) Schematic of experimental design. Briefly, mice were transplanted with Raji–Luc–EGFP cells (1 × 105) on day 7. Mice began to receive combination treatment of TGF-β inhibitor (LY2109761, 50 mg or 100 mg/kg twice daily by oral) on Day −2. On Day 0, mice were treated with PBS, CAR T-cells (1 × 106 cells) or CAR T-cells combined with LY2109761. In the combinatorial groups, LY2109761 (50 or 100 mg/kg twice daily orally, 5 days per week for 8 weeks) was administrated. (B) The tumor burden was measured using bioluminescence imaging. Total bioluminescence was traced in the vehicle, CAR T-cells combined with the LY2109761 group, and CAR T-cells. Error bars represent SEM. (C) Kaplan–Meier analysis of mice survival curve. P values were calculated using unpaired two-tailed Student’s t-tests. *P < 0.05.
1 EA Chong, M Ruella, SJ; Lymphoma Program Investigators at the University of Pennsylvania Schuster. Five-year outcomes for refractory b-cell lymphomas with car t-cell therapy. N Engl J Med 2021; 384(7): 673–674
https://doi.org/10.1056/NEJMc2030164
2 JN Kochenderfer, ME Dudley, SH Kassim, RP Somerville, RO Carpenter, M Stetler-Stevenson, JC Yang, GQ Phan, MS Hughes, RM Sherry, M Raffeld, S Feldman, L Lu, YF Li, LT Ngo, A Goy, T Feldman, DE Spaner, ML Wang, CC Chen, SM Kranick, A Nath, DA Nathan, KE Morton, MA Toomey, SA Rosenberg. Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. J Clin Oncol 2015; 33(6): 540–549
https://doi.org/10.1200/JCO.2014.56.2025
3 EJ Orlando, X Han, C Tribouley, PA Wood, RJ Leary, M Riester, JE Levine, M Qayed, SA Grupp, M Boyer, B De Moerloose, ER Nemecek, H Bittencourt, H Hiramatsu, J Buechner, SM Davies, MR Verneris, K Nguyen, JL Brogdon, H Bitter, M Morrissey, P Pierog, S Pantano, JA Engelman, W Winckler. Genetic mechanisms of target antigen loss in CAR19 therapy of acute lymphoblastic leukemia. Nat Med 2018; 24(10): 1504–1506
https://doi.org/10.1038/s41591-018-0146-z
4 E Sotillo, DM Barrett, KL Black, A Bagashev, D Oldridge, G Wu, R Sussman, C Lanauze, M Ruella, MR Gazzara, NM Martinez, CT Harrington, EY Chung, J Perazzelli, TJ Hofmann, SL Maude, P Raman, A Barrera, S Gill, SF Lacey, JJ Melenhorst, D Allman, E Jacoby, T Fry, C Mackall, Y Barash, KW Lynch, JM Maris, SA Grupp, A Thomas-Tikhonenko. Convergence of acquired mutations and alternative splicing of cd19 enables resistance to cart-19 immunotherapy. Cancer Discov 2015; 5(12): 1282–1295
https://doi.org/10.1158/2159-8290.CD-15-1020
5 C Tong, Y Zhang, Y Liu, X Ji, W Zhang, Y Guo, X Han, D Ti, H Dai, C Wang, Q Yang, W Liu, Y Wang, Z Wu, W Han. Optimized tandem CD19/CD20 CAR-engineered T cells in refractory/relapsed B-cell lymphoma. Blood 2020; 136(14): 1632–1644
https://doi.org/10.1182/blood.2020005278
6 W Deng, P Chen, W Lei, Y Xu, N Xu, JJ Pu, A Liang, W Qian. CD70-targeting CAR-T cells have potential activity against CD19-negative B-cell Lymphoma. Cancer Commun (Lond) 2021; 41(9): 925–929
https://doi.org/10.1002/cac2.12201
7 JY Spiegel, S Patel, L Muffly, NM Hossain, J Oak, JH Baird, MJ Frank, P Shiraz, B Sahaf, J Craig, M Iglesias, S Younes, Y Natkunam, MG Ozawa, E Yang, J Tamaresis, H Chinnasamy, Z Ehlinger, W Reynolds, R Lynn, MC Rotiroti, N Gkitsas, S Arai, L Johnston, R Lowsky, RG Majzner, E Meyer, RS Negrin, AR Rezvani, S Sidana, J Shizuru, WK Weng, C Mullins, A Jacob, I Kirsch, M Bazzano, J Zhou, S Mackay, SJ Bornheimer, L Schultz, S Ramakrishna, KL Davis, KA Kong, NN Shah, H Qin, T Fry, S Feldman, CL Mackall, DB Miklos. CAR T cells with dual targeting of CD19 and CD22 in adult patients with recurrent or refractory B cell malignancies: a phase 1 trial. Nat Med 2021; 27(8): 1419–1431
https://doi.org/10.1038/s41591-021-01436-0
8 S Cordoba, S Onuoha, S Thomas, DS Pignataro, R Hough, S Ghorashian, A Vora, D Bonney, P Veys, K Rao, G Lucchini, R Chiesa, J Chu, L Clark, MM Fung, K Smith, C Peticone, M Al-Hajj, V Baldan, M Ferrari, S Srivastava, R Jha, F Arce Vargas, K Duffy, W Day, P Virgo, L Wheeler, J Hancock, F Farzaneh, S Domning, Y Zhang, NZ Khokhar, VGR Peddareddigari, R Wynn, M Pule, PJ Amrolia. CAR T cells with dual targeting of CD19 and CD22 in pediatric and young adult patients with relapsed or refractory B cell acute lymphoblastic leukemia: a phase 1 trial. Nat Med 2021; 27(10): 1797–1805
https://doi.org/10.1038/s41591-021-01497-1
9 H Liu, W Lei, C Zhang, C Yang, J Wei, Q Guo, X Guo, Z Chen, Y Lu, KH Young, Z Lu, W Qian. CD19-specific CAR T cells that express a PD-1/CD28 chimeric switch-receptor are effective in patients with PD-L1-positive B-cell lymphoma. Clin Cancer Res 2021; 27(2): 473–484
https://doi.org/10.1158/1078-0432.CCR-20-1457
10 X Yan, D Chen, Y Wang, Y Guo, C Tong, J Wei, Y Zhang, Z Wu, W Han. Identification of NOXA as a pivotal regulator of resistance to CAR T-cell therapy in B-cell malignancies. Signal Transduct Target Ther 2022; 7(1): 98
https://doi.org/10.1038/s41392-022-00915-1
11 L Milane, A Singh, G Mattheolabakis, M Suresh, MM Amiji. Exosome mediated communication within the tumor microenvironment. J Control Release 2015; 219: 278–294
https://doi.org/10.1016/j.jconrel.2015.06.029
12 L Mashouri, H Yousefi, AR Aref, AM Ahadi, F Molaei, SK Alahari. Exosomes: composition, biogenesis, and mechanisms in cancer metastasis and drug resistance. Mol Cancer 2019; 18(1): 75
https://doi.org/10.1186/s12943-019-0991-5
13 TL Whiteside. Tumor-derived exosomes and their role in cancer progression. Adv Clin Chem 2016; 74: 103–141
https://doi.org/10.1016/bs.acc.2015.12.005
14 E Yang, X Wang, Z Gong, M Yu, H Wu, D Zhang. Exosome-mediated metabolic reprogramming: the emerging role in tumor microenvironment remodeling and its influence on cancer progression. Signal Transduct Target Ther 2020; 5(1): 242
https://doi.org/10.1038/s41392-020-00359-5
15 AK Ghosh, CR Secreto, TR Knox, W Ding, D Mukhopadhyay, NE Kay. Circulating microvesicles in B-cell chronic lymphocytic leukemia can stimulate marrow stromal cells: implications for disease progression. Blood 2010; 115(9): 1755–1764
https://doi.org/10.1182/blood-2009-09-242719
16 F Ma, J Vayalil, G Lee, Y Wang, G Peng. Emerging role of tumor-derived extracellular vesicles in T cell suppression and dysfunction in the tumor microenvironment. J Immunother Cancer 2021; 9(10): e003217
https://doi.org/10.1136/jitc-2021-003217
17 MJ Szczepanski, M Szajnik, A Welsh, TL Whiteside, M Boyiadzis. Blast-derived microvesicles in sera from patients with acute myeloid leukemia suppress natural killer cell function via membrane-associated transforming growth factor-beta1. Haematologica 2011; 96(9): 1302–1309
https://doi.org/10.3324/haematol.2010.039743
18 EU Wieckowski, C Visus, M Szajnik, MJ Szczepanski, WJ Storkus, TL Whiteside. Tumor-derived microvesicles promote regulatory T cell expansion and induce apoptosis in tumor-reactive activated CD8+ T lymphocytes. J Immunol 2009; 183(6): 3720–3730
https://doi.org/10.4049/jimmunol.0900970
19 Q Hao, Y Wu, Y Wu, P Wang, JV Vadgama. Tumor-derived exosomes in tumor-induced immune suppression. Int J Mol Sci 2022; 23(3): 1461
https://doi.org/10.3390/ijms23031461
20 Z Chen, L You, L Wang, X Huang, H Liu, JY Wei, L Zhu, W Qian. Dual effect of DLBCL-derived EXOs in lymphoma to improve DC vaccine efficacy in vitro while favor tumorgenesis in vivo. J Exp Clin Cancer Res 2018; 37(1): 190
https://doi.org/10.1186/s13046-018-0863-7
21 Y Hao, P Chen, X Zhang, Y Shao, Y Xu, W Qian. The effects of tumor-derived exosomes on T-cell function and efficacy of cancer immunotherapy. ImmunoMedicine 2021; 1(2): e1029
https://doi.org/10.1002/imed.1029
22 M Poggio, T Hu, CC Pai, B Chu, CD Belair, A Chang, E Montabana, UE Lang, Q Fu, L Fong, R Blelloch. Suppression of exosomal PD-L1 induces systemic anti-tumor immunity and memory. Cell 2019; 177(2): 414–427.e13
https://doi.org/10.1016/j.cell.2019.02.016
23 T Stüber, R Monjezi, L Wallstabe, J Kühnemundt, SL Nietzer, G Dandekar, A Wöckel, H Einsele, J Wischhusen, M Hudecek. Inhibition of TGF-β-receptor signaling augments the antitumor function of ROR1-specific CAR T-cells against triple-negative breast cancer. J Immunother Cancer 2020; 8(1): e000676
https://doi.org/10.1136/jitc-2020-000676
24 T Aung, B Chapuy, D Vogel, D Wenzel, M Oppermann, M Lahmann, T Weinhage, K Menck, T Hupfeld, R Koch, L Trümper, GG Wulf. Exosomal evasion of humoral immunotherapy in aggressive B-cell lymphoma modulated by ATP-binding cassette transporter A3. Proc Natl Acad Sci USA 2011; 108(37): 15336–15341
https://doi.org/10.1073/pnas.1102855108
25 M Aitamer, H Akil, C Vignoles, M Branchaud, J Abraham, N Gachard, J Feuillard, MO Jauberteau, H Shirvani, D Troutaud, H Bentayeb. CD20 expression, TrkB activation and functional activity of diffuse large B cell lymphoma-derived small extracellular vesicles. Br J Cancer 2021; 125(12): 1687–1698
https://doi.org/10.1038/s41416-021-01611-7
26 MJ Cox, F Lucien, R Sakemura, JC Boysen, Y Kim, P Horvei, C Manriquez Roman, MJ Hansen, EE Tapper, EL Siegler, C Forsman, SB Crotts, KJ Schick, M Hefazi, MW Ruff, I Can, M Adada, E Bezerra, LA Kankeu Fonkoua, WK Nevala, E Braggio, W Ding, SA Parikh, NE Kay, SS Kenderian. Leukemic extracellular vesicles induce chimeric antigen receptor T cell dysfunction in chronic lymphocytic leukemia. Mol Ther 2021; 29(4): 1529–1540
https://doi.org/10.1016/j.ymthe.2020.12.033
27 I Xhangolli, B Dura, G Lee, D Kim, Y Xiao, R Fan. Single-cell analysis of CAR-T cell activation reveals a mixed TH1/TH2 response independent of differentiation. Genom Proteom Bioinf 2019; 17(2): 129–139
https://doi.org/10.1016/j.gpb.2019.03.002
28 K Nakamura, A Kitani, W Strober. Cell contact-dependent immunosuppression by CD4+CD25+ regulatory T cells is mediated by cell surface-bound transforming growth factor beta. J Exp Med 2001; 194(5): 629–644
https://doi.org/10.1084/jem.194.5.629
29 S Huber, C Schramm, HA Lehr, A Mann, S Schmitt, C Becker, M Protschka, PR Galle, MF Neurath, M Blessing. TGF-beta signaling is required for the in vivo expansion and immunosuppressive capacity of regulatory CD4+CD25+ T cells. J Immunol 2004; 173(11): 6526–6531
https://doi.org/10.4049/jimmunol.173.11.6526
30 N Tang, C Cheng, X Zhang, M Qiao, N Li, W Mu, XF Wei, W Han, H Wang. TGF-β inhibition via CRISPR promotes the long-term efficacy of CAR T cells against solid tumors. JCI Insight 2020; 5(4): e133977
https://doi.org/10.1172/jci.insight.133977
31 W Chen, JE Konkel. TGF-beta and ‘adaptive’ Foxp3+ regulatory T cells. J Mol Cell Biol 2010; 2(1): 30–36
https://doi.org/10.1093/jmcb/mjp004
32 Y Tone, K Furuuchi, Y Kojima, ML Tykocinski, MI Greene, M Tone. Smad3 and NFAT cooperate to induce Foxp3 expression through its enhancer. Nat Immunol 2008; 9(2): 194–202
https://doi.org/10.1038/ni1549
33 BV Park, ZT Freeman, A Ghasemzadeh, MA Chattergoon, A Rutebemberwa, J Steigner, ME Winter, TV Huynh, SM Sebald, SJ Lee, F Pan, DM Pardoll, AL Cox. TGFβ1-mediated smad3 enhances pd-1 expression on antigen-specific t cells in cancer. Cancer Discov 2016; 6(12): 1366–1381
https://doi.org/10.1158/2159-8290.CD-15-1347
34 MP Oksvold, A Kullmann, L Forfang, B Kierulf, M Li, A Brech, AV Vlassov, EB Smeland, A Neurauter, KW Pedersen. Expression of B-cell surface antigens in subpopulations of exosomes released from B-cell lymphoma cells. Clin Ther 2014; 36(6): 847–862.e1
https://doi.org/10.1016/j.clinthera.2014.05.010
35 S Ali, K Toews, S Schwiebert, A Klaus, A Winkler, L Grunewald, L Oevermann, HE Deubzer, A Tüns, MC Jensen, AG Henssen, A Eggert, JH Schulte, E Schwich, V Rebmann, A Schramm, A Künkele. Tumor-derived extracellular vesicles impair cd171-specific cd4+ car t cell efficacy. Front Immunol 2020; 11: 531
https://doi.org/10.3389/fimmu.2020.00531
36 SC Lin, K Haga, XL Zeng, MK Estes. Generation of CRISPR-Cas9-mediated genetic knockout human intestinal tissue-derived enteroid lines by lentivirus transduction and single-cell cloning. Nat Protoc 2022; 17(4): 1004–1027
https://doi.org/10.1038/s41596-021-00669-0
37 DG Tantalo, AJ Oliver, B von Scheidt, AJ Harrison, SN Mueller, MH Kershaw, CY Slaney. Understanding T cell phenotype for the design of effective chimeric antigen receptor T cell therapies. J Immunother Cancer 2021; 9(5): e002555
https://doi.org/10.1136/jitc-2021-002555
38 JA Fraietta, SF Lacey, EJ Orlando, I Pruteanu-Malinici, M Gohil, S Lundh, AC Boesteanu, Y Wang, RS O’Connor, WT Hwang, E Pequignot, DE Ambrose, C Zhang, N Wilcox, F Bedoya, C Dorfmeier, F Chen, L Tian, H Parakandi, M Gupta, RM Young, FB Johnson, I Kulikovskaya, L Liu, J Xu, SH Kassim, MM Davis, BL Levine, NV Frey, DL Siegel, AC Huang, EJ Wherry, H Bitter, JL Brogdon, DL Porter, CH June, JJ Melenhorst. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat Med 2018; 24(5): 563–571
https://doi.org/10.1038/s41591-018-0010-1
39 GM Chen, C Chen, RK Das, P Gao, CH Chen, S Bandyopadhyay, YY Ding, Y Uzun, W Yu, Q Zhu, RM Myers, SA Grupp, DM Barrett, K Tan. Integrative bulk and single-cell profiling of premanufacture t-cell populations reveals factors mediating long-term persistence of CAR T-cell therapy. Cancer Discov 2021; 11(9): 2186–2199
https://doi.org/10.1158/2159-8290.CD-20-1677
40 C Yang, SH Kim, NR Bianco, PD Robbins. Tumor-derived exosomes confer antigen-specific immunosuppression in a murine delayed-type hypersensitivity model. PLoS One 2011; 6(8): e22517
https://doi.org/10.1371/journal.pone.0022517
41 CC Kloss, J Lee, A Zhang, F Chen, JJ Melenhorst, SF Lacey, MV Maus, JA Fraietta, Y Zhao, CH June. Dominant-negative tgf-β receptor enhances psma-targeted human car t cell proliferation and augments prostate cancer eradication. Mol Ther 2018; 26(7): 1855–1866
https://doi.org/10.1016/j.ymthe.2018.05.003
42 X Chen, S Yang, S Li, Y Qu, HY Wang, J Liu, ZS Dunn, GE Cinay, MA MacMullan, F Hu, X Zhang, P Wang. Secretion of bispecific protein of anti-PD-1 fused with TGF-β trap enhances antitumor efficacy of CAR-T cell therapy. Mol Ther Oncolytics 2021; 21: 144–157
https://doi.org/10.1016/j.omto.2021.03.014
43 J Proff, CU Brey, A Ensser, W Holter, M Lehner. Turning the tables on cytomegalovirus: targeting viral Fc receptors by CARs containing mutated CH2-CH3 IgG spacer domains. J Transl Med 2018; 16(1): 26
https://doi.org/10.1186/s12967-018-1394-x
44 M Boyiadzis, TL Whiteside. Plasma-derived exosomes in acute myeloid leukemia for detection of minimal residual disease: are we ready?. Expert Rev Mol Diagn 2016; 16(6): 623–629
https://doi.org/10.1080/14737159.2016.1174578
[1] FMD-23029-OF-QWB_suppl_1 Download
[1] Xianggui Yuan, Teng Yu, Jianzhi Zhao, Huawei Jiang, Yuanyuan Hao, Wen Lei, Yun Liang, Baizhou Li, Wenbin Qian. Analysis of the genomic landscape of primary central nervous system lymphoma using whole-genome sequencing in Chinese patients[J]. Front. Med., 2023, 17(5): 889-906.
[2] Zixun Yan, Li Li, Di Fu, Wen Wu, Niu Qiao, Yaohui Huang, Lu Jiang, Depei Wu, Yu Hu, Huilai Zhang, Pengpeng Xu, Shu Cheng, Li Wang, Sahin Lacin, Muharrem Muftuoglu, Weili Zhao. Immunosuppressive tumor microenvironment contributes to tumor progression in diffuse large B-cell lymphoma upon anti-CD19 chimeric antigen receptor T therapy[J]. Front. Med., 2023, 17(4): 699-713.
[3] Suning Chen, Weili Zhao, Jianyong Li, Depei Wu, on behalf of Lymphoid Disease Group, Chinese Society of Hematology, Chinese Medical Association. Chinese expert consensus on oral drugs for the treatment of mature B-cell lymphomas (2020 edition)[J]. Front. Med., 2022, 16(5): 815-826.
[4] Lei Fan, Li Wang, Lei Cao, Huayuan Zhu, Wei Xu, Jianyong Li. Phase I study of CBM.CD19 chimeric antigen receptor T cell in the treatment of refractory diffuse large B-cell lymphoma in Chinese patients[J]. Front. Med., 2022, 16(2): 285-294.
[5] Maura Massimino, Marta Podda, Lorenza Gandola, Emanuele Pignoli, Ettore Seregni, Carlo Morosi, Filippo Spreafico, Andrea Ferrari, Emilia Pecori, Monica Terenziani. Long-term results of suppressing thyroid-stimulating hormone during radiotherapy to prevent primary hypothyroidism in medulloblastoma/PNET and Hodgkin lymphoma: a prospective cohort study[J]. Front. Med., 2021, 15(1): 101-107.
[6] Houli Zhao, Yiyun Wang, Elaine Tan Su Yin, Kui Zhao, Yongxian Hu, He Huang. A giant step forward: chimeric antigen receptor T-cell therapy for lymphoma[J]. Front. Med., 2020, 14(6): 711-725.
[7] Lili Zhou, Ping Li, Shiguang Ye, Xiaochen Tang, Junbang Wang, Jie Liu, Aibin Liang. Different sites of extranodal involvement may affect the survival of patients with relapsed or refractory non-Hodgkin lymphoma after chimeric antigen receptor T cell therapy[J]. Front. Med., 2020, 14(6): 786-791.
[8] Ping Li, Ningxin Dong, Yu Zeng, Jie Liu, Xiaochen Tang, Junbang Wang, Wenjun Zhang, Shiguang Ye, Lili Zhou, Alex Hongsheng Chang, Aibin Liang. Chimeric antigen receptor T-cell therapy: a promising treatment modality for relapsed/refractory mantle cell lymphoma[J]. Front. Med., 2020, 14(6): 811-815.
[9] Yiwen Cao, Zhenhua Liu, Wen Wu, Ying Qian, Qin Shi, Rong Shen, Binshen Ouyang, Pengpeng Xu, Shu Cheng, Jin Ye, Yiming Lu, Chaofu Wang, Chengde Yang, Li Wang, Weili Zhao. Presence of multiple abnormal immunologic markers is an independent prognostic factor of diffuse large B-cell lymphoma[J]. Front. Med., 2019, 13(1): 94-103.
[10] Xuemei Zhao, Donghe Li, Qingsong Qiu, Bo Jiao, Ruihong Zhang, Ping Liu, Ruibao Ren. Zfyve16 regulates the proliferation of B-lymphoid cells[J]. Front. Med., 2018, 12(5): 559-565.
[11] Kohei Miyazono, Yoko Katsuno, Daizo Koinuma, Shogo Ehata, Masato Morikawa. Intracellular and extracellular TGF-β signaling in cancer: some recent topics[J]. Front. Med., 2018, 12(4): 387-411.
[12] Sumedha Roy, Yuan Zhuang. Paradoxical role of Id proteins in regulating tumorigenic potential of lymphoid cells[J]. Front. Med., 2018, 12(4): 374-386.
[13] Xiaofeng Shi, Rong Ba, Haiyan You, Qian Jiang, Jiansong Huang, Jianhua Mao, Lanxiu Han, Shuo Zhang, Qin Zhuang, Xianqiu Yu, Lixia Wang, Yun Wang, Dongya Li, Wei Zhu, Yong Zhang, Yan Zhu, Xiaodong Xi. A rare case of B-lymphoproliferative disorder with villous lymphocytes harboring t(8;14)(q24;q32) translocation[J]. Front. Med., 2018, 12(3): 324-329.
[14] Muhammad Furqan,Yamei Chen,Akintunde Akinleye,Judy Sarungbam,Alan Gass,Karen Seiter,Delong Liu. Management of mantle cell leukemia with cardiac involvement leading to cardiogenic shock[J]. Front. Med., 2014, 8(2): 254-258.
[15] Miao Xu, Yu Hou, Lei Sheng, Jun Peng. Therapeutic effects of thalidomide in hematologic disorders: a review[J]. Front Med, 2013, 7(3): 290-300.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed