Cargando…

ILT4 inhibition prevents TAM- and dysfunctional T cell-mediated immunosuppression and enhances the efficacy of anti-PD-L1 therapy in NSCLC with EGFR activation

Rationale: Immune checkpoint inhibitors (ICIs) against the PD-1/PD-L1 pathway showed limited success in non-small cell lung cancer (NSCLC) patients, especially in those with activating epidermal growth factor receptor (EGFR) mutations. Elucidation of the mechanisms underlying EGFR-mediated tumor imm...

Descripción completa

Detalles Bibliográficos
Autores principales: Chen, Xiaozheng, Gao, Aiqin, Zhang, Fang, Yang, Zijiang, Wang, Shuyun, Fang, Yuying, Li, Juan, Wang, Jingnan, Shi, Wenjing, Wang, Linlin, Zheng, Yan, Sun, Yuping
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7847666/
https://www.ncbi.nlm.nih.gov/pubmed/33537094
http://dx.doi.org/10.7150/thno.52435
_version_ 1783644969434611712
author Chen, Xiaozheng
Gao, Aiqin
Zhang, Fang
Yang, Zijiang
Wang, Shuyun
Fang, Yuying
Li, Juan
Wang, Jingnan
Shi, Wenjing
Wang, Linlin
Zheng, Yan
Sun, Yuping
author_facet Chen, Xiaozheng
Gao, Aiqin
Zhang, Fang
Yang, Zijiang
Wang, Shuyun
Fang, Yuying
Li, Juan
Wang, Jingnan
Shi, Wenjing
Wang, Linlin
Zheng, Yan
Sun, Yuping
author_sort Chen, Xiaozheng
collection PubMed
description Rationale: Immune checkpoint inhibitors (ICIs) against the PD-1/PD-L1 pathway showed limited success in non-small cell lung cancer (NSCLC) patients, especially in those with activating epidermal growth factor receptor (EGFR) mutations. Elucidation of the mechanisms underlying EGFR-mediated tumor immune escape and the development of effective immune therapeutics are urgently needed. Immunoglobulin-like transcript (ILT) 4, a crucial immunosuppressive molecule initially identified in myeloid cells, is enriched in solid tumor cells and promotes the malignant behavior of NSCLC. However, the upstream regulation of ILT4 overexpression and its function in tumor immunity of NSCLC with EGFR activation remains unclear. Methods: ILT4 expression and EGFR phosphorylation in human NSCLC tissues and cell lines were analyzed using immunohistochemistry (IHC), real-time PCR, Western blotting, immunofluorescence, and flow cytometry. The molecular signaling for EGFR-regulated ILT4 expression was investigated using mRNA microarray and The Cancer Genome Atlas (TCGA) database analyses and then confirmed by Western blotting. The regulation of tumor cell proliferation and apoptosis by ILT4 was examined by CCK8 proliferation and apoptosis assays. The impact of ILT4 and PD-L1 on tumor-associated macrophage (TAM) recruitment and polarization was evaluated using Transwell migration assay, flow cytometry, enzyme linked immunosorbent assay (ELISA) and real-time PCR, while their impact on T cell survival and cytotoxicity was analyzed by CFSE proliferation assay, apoptotic assay, flow cytometry, ELISA and cytolytic assay. Tumor immunotherapy models targeting at paired Ig-like receptor B (PIR-B, an ortholog of ILT4 in mouse)/ILT4 and/or PD-L1 were established in C57BL/6 mice inoculated with stable EGFR- overexpressing Lewis lung carcinoma (LLC) cells and in humanized NSG mice inoculated with EGFR mutant, gefitinib-resistant PC9 (PC9-GR) or EGFR-overexpressing wild type H1299 cells. PIR-B and ILT4 inhibition was implemented by infection of specific knockdown lentivirus and PD-L1 was blocked using human/mouse neutralizing antibodies. The tumor growth model was established in NSG mice injected with PIR-B-downregulated LLC cells to evaluate the effect of PIR-B on tumor proliferation. The frequencies and phenotypes of macrophages and T cells in mouse spleens and blood were detected by flow cytometry while those in tumor tissues were determined by IHC and immunofluorescence. Results: We found that ILT4 expression in tumor cells was positively correlated with EGFR phosphorylation in human NSCLC tissues. Using NSCLC cell lines, we demonstrated that ILT4 was upregulated by both tyrosine kinase mutation-induced and epidermal growth factor (EGF)-dependent EGFR activation and subsequent AKT/ERK1/2 phosphorylation. Overexpressed ILT4 in EGFR-activated tumor cells induced TAM recruitment and M2-like polarization, which impaired T cell function. ILT4 also directly inhibited T cell proliferation, cytotoxicity, and IFN-γ expression and secretion. In EGFR-activated cell lines in vitro and in wild-type EGFR-activated C57BL/6 and humanized NSG immunotherapy models in vivo, either ILT4 (PIR-B) or PD-L1 inhibition enhanced anti-tumor immunity and suppressed tumor progression by counteracting TAM- and dysfunctional T cell- induced immuno-suppressive TME; the combined inhibition of both molecules showed the most dramatic tumor retraction. Surprisingly, in EGFR mutant, TKI resistant humanized NSG immunotherapy model, ILT4 inhibition alone rather than in combination with a PD-L1 inhibitor suppressed tumor growth and immune evasion. Conclusions: ILT4 was induced by activation of EGFR-AKT and ERK1/2 signaling in NSCLC cells. Overexpressed ILT4 suppressed tumor immunity by recruiting M2-like TAMs and impairing T cell response, while ILT4 inhibition prevented immunosuppression and tumor promotion. Furthermore, ILT4 inhibition enhanced the efficacy of PD-L1 inhibitor in EGFR wild-type but not in EGFR mutant NSCLC. Our study identified novel mechanisms for EGFR-mediated tumor immune escape, and provided promising immunotherapeutic strategies for patients with EGFR-activated NSCLC.
format Online
Article
Text
id pubmed-7847666
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-78476662021-02-02 ILT4 inhibition prevents TAM- and dysfunctional T cell-mediated immunosuppression and enhances the efficacy of anti-PD-L1 therapy in NSCLC with EGFR activation Chen, Xiaozheng Gao, Aiqin Zhang, Fang Yang, Zijiang Wang, Shuyun Fang, Yuying Li, Juan Wang, Jingnan Shi, Wenjing Wang, Linlin Zheng, Yan Sun, Yuping Theranostics Research Paper Rationale: Immune checkpoint inhibitors (ICIs) against the PD-1/PD-L1 pathway showed limited success in non-small cell lung cancer (NSCLC) patients, especially in those with activating epidermal growth factor receptor (EGFR) mutations. Elucidation of the mechanisms underlying EGFR-mediated tumor immune escape and the development of effective immune therapeutics are urgently needed. Immunoglobulin-like transcript (ILT) 4, a crucial immunosuppressive molecule initially identified in myeloid cells, is enriched in solid tumor cells and promotes the malignant behavior of NSCLC. However, the upstream regulation of ILT4 overexpression and its function in tumor immunity of NSCLC with EGFR activation remains unclear. Methods: ILT4 expression and EGFR phosphorylation in human NSCLC tissues and cell lines were analyzed using immunohistochemistry (IHC), real-time PCR, Western blotting, immunofluorescence, and flow cytometry. The molecular signaling for EGFR-regulated ILT4 expression was investigated using mRNA microarray and The Cancer Genome Atlas (TCGA) database analyses and then confirmed by Western blotting. The regulation of tumor cell proliferation and apoptosis by ILT4 was examined by CCK8 proliferation and apoptosis assays. The impact of ILT4 and PD-L1 on tumor-associated macrophage (TAM) recruitment and polarization was evaluated using Transwell migration assay, flow cytometry, enzyme linked immunosorbent assay (ELISA) and real-time PCR, while their impact on T cell survival and cytotoxicity was analyzed by CFSE proliferation assay, apoptotic assay, flow cytometry, ELISA and cytolytic assay. Tumor immunotherapy models targeting at paired Ig-like receptor B (PIR-B, an ortholog of ILT4 in mouse)/ILT4 and/or PD-L1 were established in C57BL/6 mice inoculated with stable EGFR- overexpressing Lewis lung carcinoma (LLC) cells and in humanized NSG mice inoculated with EGFR mutant, gefitinib-resistant PC9 (PC9-GR) or EGFR-overexpressing wild type H1299 cells. PIR-B and ILT4 inhibition was implemented by infection of specific knockdown lentivirus and PD-L1 was blocked using human/mouse neutralizing antibodies. The tumor growth model was established in NSG mice injected with PIR-B-downregulated LLC cells to evaluate the effect of PIR-B on tumor proliferation. The frequencies and phenotypes of macrophages and T cells in mouse spleens and blood were detected by flow cytometry while those in tumor tissues were determined by IHC and immunofluorescence. Results: We found that ILT4 expression in tumor cells was positively correlated with EGFR phosphorylation in human NSCLC tissues. Using NSCLC cell lines, we demonstrated that ILT4 was upregulated by both tyrosine kinase mutation-induced and epidermal growth factor (EGF)-dependent EGFR activation and subsequent AKT/ERK1/2 phosphorylation. Overexpressed ILT4 in EGFR-activated tumor cells induced TAM recruitment and M2-like polarization, which impaired T cell function. ILT4 also directly inhibited T cell proliferation, cytotoxicity, and IFN-γ expression and secretion. In EGFR-activated cell lines in vitro and in wild-type EGFR-activated C57BL/6 and humanized NSG immunotherapy models in vivo, either ILT4 (PIR-B) or PD-L1 inhibition enhanced anti-tumor immunity and suppressed tumor progression by counteracting TAM- and dysfunctional T cell- induced immuno-suppressive TME; the combined inhibition of both molecules showed the most dramatic tumor retraction. Surprisingly, in EGFR mutant, TKI resistant humanized NSG immunotherapy model, ILT4 inhibition alone rather than in combination with a PD-L1 inhibitor suppressed tumor growth and immune evasion. Conclusions: ILT4 was induced by activation of EGFR-AKT and ERK1/2 signaling in NSCLC cells. Overexpressed ILT4 suppressed tumor immunity by recruiting M2-like TAMs and impairing T cell response, while ILT4 inhibition prevented immunosuppression and tumor promotion. Furthermore, ILT4 inhibition enhanced the efficacy of PD-L1 inhibitor in EGFR wild-type but not in EGFR mutant NSCLC. Our study identified novel mechanisms for EGFR-mediated tumor immune escape, and provided promising immunotherapeutic strategies for patients with EGFR-activated NSCLC. Ivyspring International Publisher 2021-01-19 /pmc/articles/PMC7847666/ /pubmed/33537094 http://dx.doi.org/10.7150/thno.52435 Text en © The author(s) This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Chen, Xiaozheng
Gao, Aiqin
Zhang, Fang
Yang, Zijiang
Wang, Shuyun
Fang, Yuying
Li, Juan
Wang, Jingnan
Shi, Wenjing
Wang, Linlin
Zheng, Yan
Sun, Yuping
ILT4 inhibition prevents TAM- and dysfunctional T cell-mediated immunosuppression and enhances the efficacy of anti-PD-L1 therapy in NSCLC with EGFR activation
title ILT4 inhibition prevents TAM- and dysfunctional T cell-mediated immunosuppression and enhances the efficacy of anti-PD-L1 therapy in NSCLC with EGFR activation
title_full ILT4 inhibition prevents TAM- and dysfunctional T cell-mediated immunosuppression and enhances the efficacy of anti-PD-L1 therapy in NSCLC with EGFR activation
title_fullStr ILT4 inhibition prevents TAM- and dysfunctional T cell-mediated immunosuppression and enhances the efficacy of anti-PD-L1 therapy in NSCLC with EGFR activation
title_full_unstemmed ILT4 inhibition prevents TAM- and dysfunctional T cell-mediated immunosuppression and enhances the efficacy of anti-PD-L1 therapy in NSCLC with EGFR activation
title_short ILT4 inhibition prevents TAM- and dysfunctional T cell-mediated immunosuppression and enhances the efficacy of anti-PD-L1 therapy in NSCLC with EGFR activation
title_sort ilt4 inhibition prevents tam- and dysfunctional t cell-mediated immunosuppression and enhances the efficacy of anti-pd-l1 therapy in nsclc with egfr activation
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7847666/
https://www.ncbi.nlm.nih.gov/pubmed/33537094
http://dx.doi.org/10.7150/thno.52435
work_keys_str_mv AT chenxiaozheng ilt4inhibitionpreventstamanddysfunctionaltcellmediatedimmunosuppressionandenhancestheefficacyofantipdl1therapyinnsclcwithegfractivation
AT gaoaiqin ilt4inhibitionpreventstamanddysfunctionaltcellmediatedimmunosuppressionandenhancestheefficacyofantipdl1therapyinnsclcwithegfractivation
AT zhangfang ilt4inhibitionpreventstamanddysfunctionaltcellmediatedimmunosuppressionandenhancestheefficacyofantipdl1therapyinnsclcwithegfractivation
AT yangzijiang ilt4inhibitionpreventstamanddysfunctionaltcellmediatedimmunosuppressionandenhancestheefficacyofantipdl1therapyinnsclcwithegfractivation
AT wangshuyun ilt4inhibitionpreventstamanddysfunctionaltcellmediatedimmunosuppressionandenhancestheefficacyofantipdl1therapyinnsclcwithegfractivation
AT fangyuying ilt4inhibitionpreventstamanddysfunctionaltcellmediatedimmunosuppressionandenhancestheefficacyofantipdl1therapyinnsclcwithegfractivation
AT lijuan ilt4inhibitionpreventstamanddysfunctionaltcellmediatedimmunosuppressionandenhancestheefficacyofantipdl1therapyinnsclcwithegfractivation
AT wangjingnan ilt4inhibitionpreventstamanddysfunctionaltcellmediatedimmunosuppressionandenhancestheefficacyofantipdl1therapyinnsclcwithegfractivation
AT shiwenjing ilt4inhibitionpreventstamanddysfunctionaltcellmediatedimmunosuppressionandenhancestheefficacyofantipdl1therapyinnsclcwithegfractivation
AT wanglinlin ilt4inhibitionpreventstamanddysfunctionaltcellmediatedimmunosuppressionandenhancestheefficacyofantipdl1therapyinnsclcwithegfractivation
AT zhengyan ilt4inhibitionpreventstamanddysfunctionaltcellmediatedimmunosuppressionandenhancestheefficacyofantipdl1therapyinnsclcwithegfractivation
AT sunyuping ilt4inhibitionpreventstamanddysfunctionaltcellmediatedimmunosuppressionandenhancestheefficacyofantipdl1therapyinnsclcwithegfractivation