A PD-L1-targeting chimeric switch receptor enhances efficacy of CAR-T cell for pleural and peritoneal metastasis

Qizhi Ma, Xia He, Benxia Zhang, Fuchun Guo, Xuejin Ou, Qiyu Yang, Pei Shu, Yue Chen, Kai Li, Ge Gao, Yajuan Zhu, Diyuan Qin, Jie Tang, Xiaoyu Li, Meng Jing, Jian Zhao, Zeming Mo, Ning Liu, Yao Zeng, Kexun Zhou, Mingyang Feng, Weiting Liao, Wanting Lei, Qiu Li, Dan Li, Yongsheng Wang, Qizhi Ma, Xia He, Benxia Zhang, Fuchun Guo, Xuejin Ou, Qiyu Yang, Pei Shu, Yue Chen, Kai Li, Ge Gao, Yajuan Zhu, Diyuan Qin, Jie Tang, Xiaoyu Li, Meng Jing, Jian Zhao, Zeming Mo, Ning Liu, Yao Zeng, Kexun Zhou, Mingyang Feng, Weiting Liao, Wanting Lei, Qiu Li, Dan Li, Yongsheng Wang

Abstract

Pleural and peritoneal metastasis accompanied by malignant pleural effusion (MPE) or malignant ascites (MA) is frequent in patients with advanced solid tumors that originate from the lung, breast, gastrointestinal tract and ovary. Regional delivery of CAR-T cells represents a new strategy to control tumor dissemination in serous cavities. However, malignant effusions constitute an immune-suppressive environment that potentially induces CAR-T cell dysfunction. Here, we demonstrated that the anti-tumor cytotoxicity of conventional 2nd-generation CAR-T cells was significantly inhibited by both the cellular and non-cellular components of MPE/MA, which was primarily attributed to impaired CAR-T cell proliferation and cytokine production in MPE/MA environment. Interestingly, we found that PD-L1 was widely expressed on freshly-isolated MPE/MA cells. Based on this feature, a novel PD-L1-targeting chimeric switch receptor (PD-L1.BB CSR) was designed, which can bind to PD-L1, switching the inhibitory signal into an additional 4-1BB signal. When co-expressed with a 2nd-generation CAR, PD-L1.BB CSR-modified CAR-T cells displayed superior fitness and enhanced functions in both culture medium and MPE/MA environment, causing rapid and durable eradication of pleural and peritoneal metastatic tumors in xenograft models. Further investigations revealed elevated expressions of T-cell activation, proliferation, and cytotoxicity-related genes, and we confirmed that PD-L1 scFv and 4-1BB intracellular domain, the two important components of PD-L1.BB CSR, were both necessary for the functional improvements of CAR-T cells. Overall, our study shed light on the clinical application of PD-L1.BB CSR-modified dual-targeting CAR-T cells. Based on this study, a phase I clinical trial was initiated in patients with pleural or peritoneal metastasis (NCT04684459).

Conflict of interest statement

The authors declare that they have no competing interests. Yongsheng Wang is the editorial board member of Signal Transduction and Targeted Therapy, but he has not been involved in the process of manuscript handling.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
The functions of 2nd-generation CAR-T cells are significantly impaired in MPE/MA environment, and it is partially related to MPE/MA-induced PD-L1 expression. a The cytotoxicity of HER CAR-T cells (HER2.28ζ and HER2.BBζ) that incubated with SKOV3 or A549 cells for 24 h in cytokine-free medium (culture medium) was analyzed by CCK-8 assay. b Summary of IFN-γ, IL-2 and TNF-α release by HER2.28ζ and HER2.BBζ CAR-T cells after 24 h of co-incubation with SKOV3 or A549 (E: T = 2: 1) as measured by ELISA assay. c The cytotoxicity of HER2.28ζ CAR-T cells was analyzed by CCK-8 assay, after 48 h of co-culture with SKOV3 cells (E: T = 2: 1) in different components of patient-derived MPE or MA samples (Supplementary Table. S1). d The cytotoxicity of HER2.28ζ CAR-T cells was analyzed by CCK-8 assay, after 24 h of co-culture with SKOV3 cells in culture medium or MPE-supernatant (Pt1, as Supplementary Table. S1). e The levels of cytokines (IFN-γ, IL-2 and TNF-α) in the supernatants from (c) were determined by ELISA. f The fold expansion of HER2.28ζ CAR-T cells were analyzed through FCM-counting beads, after stimulated by irradiated SKOV3 cells (E: T = 2: 1) in culture medium or MPE-supernatant (Pt1, as Supplementary Table. S1), and the portion of live CAR-T cells (Annexin-V-/PI-) on the 3rd day after stimulation was shown in g. h The cytotoxicity of ROR1.28ζ and CD19.28ζ CAR-T cells were co-cultured with SKOV3 and Raji cells respectively in culture medium or MPE-supernatant (Pt1, as Supplementary Table. S1) for 24 h, and analyzed by CCK-8 assay. i The mRNA expression of PD-L1 in cancer cells, fibroblasts, macrophages and T cells of human ovarian cancer ascites analyzed by single-cell RNA-sequencing (scRNA-seq) data deposited in public Gene Expression Omnibus (GSE146026). j PD-L1 mRNA expression levels in MPE/MA-cells were shown as fold change to 293 T, HUVEC and MDA-MB-468 cells (as controls). k The statistical analysis of PD-L1 expression on clinically derived MPE/MA-cells (Supplementary Table. S1) was analyzed by flow cytometry. l Representative flow cytometry histogram of PD-L1 expression on CD45- and CD45+ cells from MPE/MA samples (Supplementary Table. S1). m Representative flow cytometry histogram of PD-L1 expression on healthy donor-derived CD3+ T cells that cultured in MPE/MA-supernatant (Supplementary Table. S1) continuously for 7 days. (Pre: pre-culturing; Post: post-culturing) Pt, patient; MPE, malignant pleural effusion; MA, malignant ascites. P-values were determined by unpaired two-tailed t-test a, b, d, f, g, h or one-way ANOVA with Tukey’s multiple comparison test adjusted P value c, e. *P <0.05; **P <0.01; ns, not significant. Data show the mean ± SD from three independent experiments
Fig. 2
Fig. 2
PD-L1.BB CSR engagement enhances the cytotoxicity and CAR activation-induced cytokine release of HER2.28ζ CAR-T cells. a Schematic illustration of lentiviral vectors encoding HER2.28ζ, PD-L1.BB and HER2.28ζ/PD-L1.BB. b Representative flow cytometry plots of CAR expressions on healthy donor derived CD3+ T cells. c Representative flow cytometry histograms of HER2 and PD-L1 expressions on various human tumor cell lines. d The cytotoxicities of CAR-T cells were analyzed by CCK-8 assay, after 24 h of co-culture with HER2+PD-L1+ (SKOV3PD-L1, A549 or H1975) or HER2-PD-L1- (MDA-MB-468) cancer cells at various E:T ratios, and IFN-γ e, IL-2, and TNF-α f released by CAR-T cells (E: T = 2: 1) in the culture supernatant were determined by ELISA. g Continuous cytotoxicity of CAR-T cells was assessed by an xCELLigene Real-Time Cell Analysis (RTCA) system. Left: Untransduced (UTD) T cells, HER2.28ζ, PD-L1.BB and HER2.28ζ/PD-L1.BB CAR-T cells that co-incubated with SKOV3PD-L1 at E: T of 2: 1. Right: HER2.28ζ/PD-L1.BB CAR-T cells that co-incubated with SKOV3PD-L1 at various E: T ratios. P-values were determined by one-way ANOVA with Tukey’s multiple comparison test adjusted P value df. *P <0.05, **P <0.01, ns, not significant. Data show the mean ± SD from three independent experiments
Fig. 3
Fig. 3
PD-L1.BB CSR engagement improves the functions of HER2.28ζ CAR-T cells and resists MPE/MA-mediated immunosuppression. a Representative microscopy images of CAR-T cell cluster formation after 24 h of co-culture with SKOV3PD-L1 cells (E: T = 2: 1) in culture medium. Magnification: 100×. b Proliferation of HER2.28ζ, PD-L1.BB and HER2.28ζ/PD-L1.BB CAR-T after activation. CFSE-labeled CAR-T stimulated by irradiated SKOV3PD-L1 or MDA-MB-468 cells (as control) on day 0 at E: T ratio of 2:1. CFSE dilution was measured by flow cytometry on days 2, 3, 4 and 5 after stimulation. c The fold expansion of CAR-T cells after stimulated by irradiated SKOV3PD-L1 cells (E: T = 2: 1) in culture medium was measured by FCM-counting beads. d The ratio of central memory T cells (CD45RO+CCR7+) in each group of CAR-T cells that cultured in culture medium or MPE (Pt1, as Supplementary Table. S1) was determined on day 3 post-stimulation by irradiated SKOV3PD-L1 cells (E: T = 2: 1). e Representative microscopy images of CAR-T cell cluster formation after 24 h of co-culture with SKOV3PD-L1 (E: T = 2: 1) in MPE-supernatant (Pt1, as Supplementary Table. S1). Magnification: 100×. f The cytotoxicity of CAR-T cells was analyzed by CCK-8 assay after 24 h of co-culture with SKOV3PD-L1 cells in MPE-supernatant (Pt1, as Supplementary Table. S1). g The fold expansion of CAR-T cells after stimulated by irradiated SKOV3PD-L1 cells (E: T = 2: 1) in MPE-supernatant (Pt1, as Supplementary Table. S1). h Summary of IFN-γ, IL-2 and TNF-α released by CAR-T cells after 24 h of co-culture with SKOV3PD-L1 or MA-treated SKOV3PD-L1 cells (E: T = 2: 1) in MA-supernatant (Pt4, as Supplementary Table. S1). MA-treated SKOV3PD-L1 cells: SKOV3PD-L1 cells that cultured continuously in MA-supernatant for 1 month. i Representative flow cytometry histograms of HER2 and PD-L1 expressions on MPE-isolated primary small-cell lung cancer (SCLC) cells (Pt10, as Supplementary Table. S1). j The representative microscopy images of CAR-T cell cluster formation after 48 h of co-incubation with primary SCLC cells (E: T = 2: 1) in MPE-supernatant from the same patient (Pt10, as Supplementary Table. S1). Magnification: 100×. k The primary SCLC cells viability was determined by CCK-8 assay. IFN-γ, IL-2 and TNF-α released by CAR-T cells in the supernatant were measured by ELISA. P-values were determined by one-way ANOVA with Tukey’s multiple comparison test adjusted P value (c, d, f, g, h, k). *P <0.05, **P <0.01, ns, not significant. Data show the mean ± SD from three independent experiments
Fig. 4
Fig. 4
PD-L1.BB CSR engagement enhances 4-1BB expression and inhibits PD-1/PD-L1signals. a, b Statistical analysis of 4-1BB expression on HER2.28ζ and HER2.28ζ/PD-L1.BB CAR-T cells after stimulation. CAR-T cells were stimulated by either a irradiated SKOV3PD-L1 or b SKOV3 cells (E: T = 2: 1) in culture medium, and 4-1BB expression was assessed by flow cytometry at the indicated time points. c Representative flow cytometry plots showed PD-1 expression on CD4+ and CD8+ CAR-T cells on day 5 after stimulation (E: T = 2: 1). d Statistical analysis of PD-1 expression on CD4+ and CD8+ CAR-T cells. e PD-L1 expression on CAR-T cells. CAR-T cells were incubated with irradiated SKOV3PD-L1 cells (E: T = 2: 1) for 5 days, either in culture medium or MPE-supernatant (Pt1, as Supplementary Table. S1). f The cytotoxicity and cytokine release of HER2.28ζ and HER2.28ζ/PD-L1BB CAR-T cells before and after PD-1 blockade. CAR-T cells were stimulated with irradiated SKOV3PD-L1 cells, and treated with PD-1 blocking antibody (Tislelizumab, 10 μg/ml) for 3 days. Then the post-stimulated CAR-T cells were co-incubated with SKOV3PD-L1 (E: T = 2: 1) for 24 h. The cytotoxic activity of CAR-T cells was analyzed by CCK-8, and effector cytokine (IFN-γ, IL-2 and TNF-α) in culture supernatant were measured by ELISA. P-values were determined by one-way ANOVA with Tukey’s multiple comparison test adjusted P value (a, b, f) or unpaired two-tailed t-test d. *P <0.05; **P <0.01, ns, not significant. Data show the mean ± SD from three independent experiments
Fig. 5
Fig. 5
HER2.28ζ/PD-L1.BB CAR-T cells have prolonged antitumor activity in vivo, with rapid and long-lasting eradication of pleural and peritoneal metastasis. a The schematic diagram of A549 and SKOV3PD-L1 metastatic xenograft models in NSG mice inoculated via intrapleural or intraperitoneal (i.p.) injection, and treated 10 days later with CAR-T cells. b Representative bioluminescence (BLI) images of A549-Luc tumor growth in the pleural metastasis xenograft model. Mice were intrapleurally given 3 × 106 CAR+ T or UTD cells, and tumor progression was monitored by in vivo imaging system (IVIS). c The BLI kinetics of A549-Luc tumor growth in the pleural metastasis xenograft model (n = 5). d Kaplan–Meier survival curve of mice after CAR-T cell treatment (n = 5). e Representative BLI images of SKOV3PDL1-Luc tumor growth in the peritoneal metastasis xenograft model. Mice were intraperitoneally given 1 × 106 CAR+ T or UTD cells, and tumor progression was monitored by IVIS. f The BLI kinetics of SKOV3PDL1-Luc tumor growth in the peritoneal metastasis xenograft model (n = 5). g Kaplan–Meier survival curve of mice after CAR-T cell treatment (n = 5). h, i Summary of CAR-T cell (CD45+CD3+) number in the peritoneal cavity h and peripheral blood i of SKOV3PD-L1-Luc peritoneal metastasis mice analyzed by flow cytometry 7 days after CAR-T cell treatment (n = 5). j The ratios of central memory T cells (CD45RO+CCR7+), PD-1+k and PD-L1+l CAR-T cells (n = 5). m The levels of effector cytokines (IFN-γ, IL-2 and TNF-α) in serum and peritoneal lavage fluid were measured by ELISA 7 days after CAR-T cells treatment (n = 5). n Quantification of CAR copy number by q-PCR to evaluate CAR-T cell persistence in the pleural cavity 2 weeks after SKOV3PD-L1-Luc tumor regression (n = 3). P-values were determined by one-way ANOVA with Tukey’s multiple comparison test adjusted P value (c, f, h, i, j, k, l, m, n) or long-rank test d, g. *P < 0.05, **P < 0.01, ns, not significant. Data are expressed as the mean ± SD of each group
Fig. 6
Fig. 6
PD-L1.BB CSR engagement upregulates T-cell activation, cytotoxicity, and proliferation related gene expressions in HER2.28ζ/PD-L1.BB CAR-T cells. a RNA-seq analysis of HER2.28ζ/PD-L1.BB and HER2.28ζ CAR-T cells at 12 h, 24 h and 48 h after stimulation with irradiated SKOV3PD-L1 cells, and the differentially expressed genes (DEGs) were shown in volcano plot (FC ≥ 2, Q ≤ 0.05). FDR, false discovery rate; FC, fold change. b Significant enriched KEGG pathway terms of DEGs between HER2.28ζ/PD-L1.BB and HER2.28ζ CAR-T cells at 24 h post-stimulation (FC ≥ 2, Q ≤ 0.001). c Heatmap of selected DEGs with different expression related to cytokine-cytokine receptor interaction, chemokine and T cell receptor (TCR) signaling pathways. d Analysis of protein-protein interaction (PPI) networks of DEGs between HER2.28ζ/PD-L1.BB and HER2.28ζ CAR-T cells 24 h after stimulation. ej Gene-set enrichment analysis (GSEA) of IFN-γ e, IL-2 f, TNF-α g signaling pathways, as well as mitosis genes h, G2M checkpoint genes i and stem cell memory pathway j was performed on all gene sets at 24 h post-stimulation. Data shown are representative of 2 replicates
Fig. 7
Fig. 7
The roles of PD-L1 scFv and 4-1BB intracellular domain on the functional improvements of PD-L1.BB.CSR-modified CAR-T cells. a Heatmap of selected DEGs in HER2.28ζ/PD-L1.BB, HER2.28ζ/PD-L1, HER2.28ζ, PD-L1.BB CAR-T and UTD cells at 24 h post-stimulation with irradiated SKOV3PD-L1 cells. b Heatmap of selected DEGs between PD-L1.BB CSR-T and UTD cells at 12 h and 24 h post-stimulation. c GSEA of IFN-γ, TNF-α, and mTORC1 signaling pathway, as well as mitosis genes in PD-L1.BB CSR-T and UTD cells at 24 h post-stimulation. d Heatmap of DEGs between HER2.28ζ/PD-L1 and HER2.28ζ CAR-T cells at 24 h post-stimulation. e GSEA of IFN-γ, IL-2, TNF-α, IL-6 signaling pathways in HER2.28ζ/PD-L1 and HER2.28ζ CAR-T cells at 24 h post-stimulation. f Heatmap of selected DEGs between HER2.28ζ/PD-L1.BB and HER2.28ζ/PD-L1 CAR-T cells at 24 h post-stimulation. g GSEA of IFN-γ signaling pathway, T cell activation genes, E2F target genes and stem cell memory pathway in HER2.28ζ/PD-L1.BB and HER2.28ζ/PD-L1 CAR-T cells at 24 h post-stimulation. h The schematic diagram of repeated-stimulation experiment. At days 0 and 2, CAR-T cells were stimulated with irradiated SKOV3PD-L1 cells at E: T ratio of 2:1, and at day 6, CAR-T cells were sorted by human CD3 magnetic beads for functional test i, j, m, n. The cytotoxicity of post-stimulated CAR-T cells was analyzed by CCK-8, after 24 h of co-culture with SKOV3PD-L1 cells at indicated E: T ratios i, and effector cytokine (IFN-γ, IL-2 and TNF-α) secretion in culture supernatant was detected by ELISA j. k At days 1, 2, 4 and 6 of repeated-stimulation experiment, the proliferation folds of CAR-T cells were quantified by flow cytometry using absolute counting bead. l Statistical analysis of 4-1BB expression on CD4+ and CD8+ CAR-T cells on day 1 of repeated-stimulation experiment. m, n Statistical analysis of CD45RO+CCR7+ TCM cells m, PD-1+ and PD-L1+ expressions on CD4+ and CD8+ CAR-T cells n at day 6 of repeated-stimulation experiment. Representative RNA-seq pictures shown in 2 replicates. P-values were determined by unpaired two-tailed t-test in. *P <0.05, **P <0.01, ns, not significant. Data are expressed as the mean ± SD from three independent experiments
Fig. 8
Fig. 8
A virtual treatment scenario of CAR-T cells in pleural/peritoneal cavity. ac The therapeutic pattern and potential mechanism of regional delivery of HER2.28ζ a and HER2.28ζ/PD-L1.BB CAR-T cells b, c to treat pleural or peritoneal metastasis accompanied by MPE/MA

References

    1. Hodge C, Badgwell BD. Palliation of malignant ascites. J. Surg. Oncol. 2019;120:67–73. doi: 10.1002/jso.25453.
    1. Walker S, Mercer R, Maskell N, Rahman NM. Malignant pleural effusion management: keeping the flood gates shut. Lancet Respir. Med. 2020;8:609–618. doi: 10.1016/S2213-2600(19)30373-X.
    1. Bibby AC, et al. ERS/EACTS statement on the management of malignant pleural effusions. Eur. Respir. J. 2018;52:1800349. doi: 10.1183/13993003.00349-2018.
    1. Clive AO, et al. Predicting survival in malignant pleural effusion: development and validation of the LENT prognostic score. Thorax. 2014;69:1098–1104. doi: 10.1136/thoraxjnl-2014-205285.
    1. Psallidas I, et al. Malignant pleural effusion: from bench to bedside. Eur. Respir. Rev. 2016;25:189–198. doi: 10.1183/16000617.0019-2016.
    1. Aggarwal C, et al. Phase I study of intrapleural gene-mediated cytotoxic immunotherapy in patients with malignant pleural effusion. Mol. Ther. 2018;26:1198–1205. doi: 10.1016/j.ymthe.2018.02.015.
    1. Adusumilli PS, et al. A phase I trial of regional mesothelin-targeted CAR T-cell therapy in patients with malignant pleural disease, in combination with the anti-PD-1 agent pembrolizumab. Cancer Disco. 2021;11:2748–2763. doi: 10.1158/-21-0407.
    1. Hou AJ, Chen LC, Chen YY. Navigating CAR-T cells through the solid-tumour microenvironment. Nat. Rev. Drug. Discov. 2021;20:531–550. doi: 10.1038/s41573-021-00189-2.
    1. Brown CE, et al. Regression of glioblastoma after chimeric antigen receptor T-cell therapy. N. Engl. J. Med. 2016;375:2561–2569. doi: 10.1056/NEJMoa1610497.
    1. Adusumilli PS, et al. Regional delivery of mesothelin-targeted CAR T cell therapy generates potent and long-lasting CD4-dependent tumor immunity. Sci. Transl. Med. 2014;6:261ra151. doi: 10.1126/scitranslmed.3010162.
    1. Kostron A, et al. Relapse pattern and second-line treatment following multimodality treatment for malignant pleural mesothelioma. Eur. J. Cardiothorac. Surg. 2016;49:1516–1523. doi: 10.1093/ejcts/ezv398.
    1. Jiwangga D, Cho S, Kim K, Jheon S. Recurrence pattern of pathologic stage I lung adenocarcinoma with visceral pleural invasion. Ann. Thorac. Surg. 2017;103:1126–1131. doi: 10.1016/j.athoracsur.2016.09.052.
    1. Yoo CH, et al. Recurrence following curative resection for gastric carcinoma. Br. J. Surg. 2000;87:236–242. doi: 10.1046/j.1365-2168.2000.01360.x.
    1. Esselen KM, et al. Patterns of recurrence in advanced epithelial ovarian, fallopian tube and peritoneal cancers treated with intraperitoneal chemotherapy. Gynecol. Oncol. 2012;127:51–54. doi: 10.1016/j.ygyno.2012.05.026.
    1. Horikawa N, et al. Expression of vascular endothelial growth factor in ovarian cancer inhibits tumor immunity through the accumulation of myeloid-derived suppressor cells. Clin. Cancer Res. 2017;23:587–599. doi: 10.1158/1078-0432.CCR-16-0387.
    1. Yin T, et al. Malignant pleural effusion and ascites induce epithelial-mesenchymal transition and cancer stem-like cell properties via the vascular endothelial growth factor (VEGF)/phosphatidylinositol 3-Kinase (PI3K)/Akt/mechanistic target of rapamycin (mTOR) pathway. J. Biol. Chem. 2016;291:26750–26761. doi: 10.1074/jbc.M116.753236.
    1. Giuntoli RL, 2nd, et al. Ovarian cancer-associated ascites demonstrates altered immune environment: implications for antitumor immunity. Anticancer Res. 2009;29:2875–2884.
    1. Abiko K, et al. PD-L1 on tumor cells is induced in ascites and promotes peritoneal dissemination of ovarian cancer through CTL dysfunction. Clin. Cancer Res. 2013;19:1363–1374. doi: 10.1158/1078-0432.CCR-12-2199.
    1. Tseng YH, et al. PD-L1 expression of tumor cells, macrophages, and immune cells in non-small cell lung cancer patients with malignant pleural effusion. J. Thorac. Oncol. 2018;13:447–453. doi: 10.1016/j.jtho.2017.10.034.
    1. Xu J, et al. PD-L1 expression in pleural effusions of pulmonary adenocarcinoma and survival prediction: a controlled study by pleural biopsy. Sci. Rep. 2018;8:11206. doi: 10.1038/s41598-018-29156-5.
    1. Pasello G, et al. Malignant pleural mesothelioma immune microenvironment and checkpoint expression: correlation with clinical-pathological features and intratumor heterogeneity over time. Ann. Oncol. 2018;29:1258–1265. doi: 10.1093/annonc/mdy086.
    1. Shibaki R, et al. Malignant pleural effusion as a predictor of the efficacy of anti-PD-1 antibody in patients with non-small cell lung cancer. Thorac. Cancer. 2019;10:815–822. doi: 10.1111/1759-7714.13004.
    1. Fuca G, et al. Ascites and resistance to immune checkpoint inhibition in dMMR/MSI-H metastatic colorectal and gastric cancers. J. Immunother. Cancer. 2022;10:e004001. doi: 10.1136/jitc-2021-004001.
    1. Prado-Garcia H, Romero-Garcia S, Puerto-Aquino A, Rumbo-Nava U. The PD-L1/PD-1 pathway promotes dysfunction, but not “exhaustion”, in tumor-responding T cells from pleural effusions in lung cancer patients. Cancer Immunol. Immunother. 2017;66:765–776. doi: 10.1007/s00262-017-1979-x.
    1. Textor A, et al. CD28 co-stimulus achieves superior CAR T cell effector function against solid tumors than 4-1BB co-stimulus. Cancers. 2021;13:1050. doi: 10.3390/cancers13051050.
    1. Cappell KM, Kochenderfer JN. A comparison of chimeric antigen receptors containing CD28 versus 4-1BB costimulatory domains. Nat. Rev. Clin. Oncol. 2021;18:715–727. doi: 10.1038/s41571-021-00530-z.
    1. Rafiq S, Hackett CS, Brentjens RJ. Engineering strategies to overcome the current roadblocks in CAR T cell therapy. Nat. Rev. Clin. Oncol. 2020;17:147–167. doi: 10.1038/s41571-019-0297-y.
    1. Etxeberria I, Glez-Vaz J, Teijeira A, Melero I. New emerging targets in cancer immunotherapy: CD137/4-1BB costimulatory axis. Esmo. Open. 2020;4:e000733. doi: 10.1136/esmoopen-2020-000733.
    1. Roth TL, et al. Pooled knockin targeting for genome engineering of cellular immunotherapies. Cell. 2020;181:728–744. doi: 10.1016/j.cell.2020.03.039.
    1. Zhao Z, et al. Structural design of engineered costimulation determines tumor rejection kinetics and persistence of CAR T cells. Cancer Cell. 2015;28:415–428. doi: 10.1016/j.ccell.2015.09.004.
    1. Nami B, Ghanaeian A, Black C, Wang Z. Epigenetic silencing of HER2 expression during epithelial-mesenchymal transition leads to trastuzumab resistance in breast cancer. Life. 2021;11:868. doi: 10.3390/life11090868.
    1. Bitra A, Doukov T, Croft M, Zajonc DM. Crystal structures of the human 4-1BB receptor bound to its ligand 4-1BBL reveal covalent receptor dimerization as a potential signaling amplifier. J. Biol. Chem. 2018;293:9958–9969. doi: 10.1074/jbc.RA118.003176.
    1. Diskin B, et al. PD-L1 engagement on T cells promotes self-tolerance and suppression of neighboring macrophages and effector T cells in cancer. Nat. Immunol. 2020;21:442–454. doi: 10.1038/s41590-020-0620-x.
    1. Cherkassky L, et al. Human CAR T cells with cell-intrinsic PD-1 checkpoint blockade resist tumor-mediated inhibition. J. Clin. Invest. 2016;126:3130–3144. doi: 10.1172/JCI83092.
    1. Principe N, et al. Malignant pleural effusions-a window into local anti-tumor T cell immunity? Front. Oncol. 2021;11:672747. doi: 10.3389/fonc.2021.672747.
    1. Hong M, Clubb JD, Chen YY. Engineering CAR-T Cells for Next-Generation Cancer Therapy. Cancer Cell. 2020;38:473–488. doi: 10.1016/j.ccell.2020.07.005.
    1. Khanna S, et al. Malignant mesothelioma effusions are infiltrated by CD3(+) T cells highly expressing PD-L1 and the PD-L1(+) tumor cells within these effusions are susceptible to ADCC by the anti-PD-L1 antibody avelumab. J. Thorac. Oncol. 2016;11:1993–2005. doi: 10.1016/j.jtho.2016.07.033.
    1. Peng J, et al. Chemotherapy induces programmed cell death-ligand 1 overexpression via the nuclear factor-kappaB to foster an immunosuppressive tumor microenvironment in ovarian cancer. Cancer Res. 2015;75:5034–5045. doi: 10.1158/0008-5472.CAN-14-3098.
    1. Ikematsu Y, et al. Immune checkpoint protein and cytokine expression by T lymphocytes in pleural effusion of cancer patients receiving anti-PD-1 therapy. Lung Cancer. 2019;138:58–64. doi: 10.1016/j.lungcan.2019.10.011.
    1. Hirabayashi K, et al. Dual-targeting CAR-T cells with optimal co-stimulation and metabolic fitness enhance antitumor activity and prevent escape in solid tumors. Nat. Cancer. 2021;2:904–918. doi: 10.1038/s43018-021-00244-2.
    1. Wei F, et al. Strength of PD-1 signaling differentially affects T-cell effector functions. Proc. Natl Acad. Sci. USA. 2013;110:E2480–E2489. doi: 10.1073/pnas.1305394110.
    1. Kawalekar OU, et al. Distinct signaling of coreceptors regulates specific metabolism pathways and impacts memory development in CAR T cells. Immunity. 2016;44:380–390. doi: 10.1016/j.immuni.2016.01.021.
    1. Burr ML, et al. CMTM6 maintains the expression of PD-L1 and regulates anti-tumour immunity. Nature. 2017;549:101–105. doi: 10.1038/nature23643.
    1. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science. 2018;359:1350–1355. doi: 10.1126/science.aar4060.
    1. Lee HT, et al. Molecular mechanism of PD-1/PD-L1 blockade via anti-PD-L1 antibodies atezolizumab and durvalumab. Sci. Rep. 2017;7:5532. doi: 10.1038/s41598-017-06002-8.
    1. Ding G, et al. IFN-gamma down-regulates the PD-1 expression and assist nivolumab in PD-1-blockade effect on CD8+ T-lymphocytes in pancreatic cancer. Bmc. Cancer. 2019;19:1053. doi: 10.1186/s12885-019-6145-8.
    1. Curiel TJ, et al. Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity. Nat. Med. 2003;9:562–567. doi: 10.1038/nm863.
    1. Liu Y, et al. Immune cell PD-L1 colocalizes with macrophages and is associated with outcome in PD-1 pathway blockade therapy. Clin. Cancer Res. 2020;26:970–977. doi: 10.1158/1078-0432.CCR-19-1040.
    1. Pulko V, et al. B7-h1 expressed by activated CD8 T cells is essential for their survival. J. Immunol. 2011;187:5606–5614. doi: 10.4049/jimmunol.1003976.
    1. Chapman NM, Chi H. mTOR links environmental signals to T cell fate decisions. Front. Immunol. 2014;5:686.
    1. Tang T, et al. Advantages of targeting the tumor immune microenvironment over blocking immune checkpoint in cancer immunotherapy. Signal Transduct. Target Ther. 2021;6:72. doi: 10.1038/s41392-020-00449-4.
    1. Taube JM, et al. Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin. Cancer Res. 2014;20:5064–5074. doi: 10.1158/1078-0432.CCR-13-3271.

Source: PubMed

3
Předplatit