Cancer Letters

Cancer Letters

Volume 485, 10 August 2020, Pages 14-26
Cancer Letters

Insulin-like growth factor-1 receptor induces immunosuppression in lung cancer by upregulating B7–H4 expression through the MEK/ERK signaling pathway

https://doi.org/10.1016/j.canlet.2020.04.013Get rights and content

Highlights

  • IGF1R is associated with CD8+ T cell inhibition in lung cancer.

  • IGF1R upregulates B7–H4 expression.

  • IGF1R downstream MEK/ERK activation contributes to the B7–H4 upregulation.

  • B7–H4 knockdown inhibits IGF1R-overexpression tumor growth and reverse the tumor infiltrated CD8+ T cell inhibition.

Abstract

The Insulin-like growth factor-1/Insulin-like growth factor-1 receptor (IGF1/IGF1R) axis contributes to immunosuppression during tumor progression; however, the underlying mechanism remains unclear. In the present study, we found that IGF1 stimulation or IGF1R overexpression (IGF1R-OE) could upregulate the expression of B7–H4, while IGF1R inhibition downregulated B7–H4 in both A549 and SPC-A-1 lung cancer cell lines. IGF1R-OE conferred the inhibition of CD8+ T cells by cancer cells in vitro, and induction of B7–H4 expression was mediated by the activation of the MEK/ERK1/2 signaling pathway. The in vitro findings were further confirmed in vivo using a Lewis lung cancer mouse model. IGF1R-OE promoted tumor growth and inhibited tumor infiltration by CD8+ T cells in the mouse model. However, this effect was suppressed when B7–H4 was knocked down in IGF1R-OE cells. Our findings suggest that IGF1R could induce immunosuppression in lung cancer by upregulating the expression of B7–H4 through the MEK/ERK pathway. B7–H4 may therefore be a potential therapeutic target for lung cancer immunotherapy.

Introduction

The IGF axis, which is composed of ligands (IGF-1, IGF-2), IGF receptors (IGF1R, IGF2R), and six IGF binding proteins (IGFBPs 1–6), has a pivotal role in tumor progression [1]. IGF1R activation results in the activation of the PI3K/Akt and MEK/ERK signaling pathways [2]. IGF signaling can promote immunosuppressive and anti-inflammatory responses that facilitate tumor growth [[3], [4], [5]]. By disrupting PI3K/Akt signaling, which is downstream of IGF1R, tumor cells are sensitized toward cytotoxicity by antigen-specific CTL both in vitro and in vivo [6]. Activation of MEK/ERK, the other IGF1R downstream signaling pathway, is associated with reduced tumor-infiltrated lymphocytes (TILs) [7]. Taken together, these findings suggest that IGF1R signaling activation might affect the tumor immunity by regulating TILs. However, the exact molecular mechanism by which IGF1R regulates the tumor immune microenvironment remains largely unknown.

Immune checkpoints are the T-cell regulatory molecules that regulate the activation and effector functions of TILs through costimulatory and coinhibitory signals [8,9]. Tumor cells can deploy co-inhibitory molecules to escape immune surveillance and resist the cytotoxic effect of host T cells [10]. Among these molecules, B7–H4 (VTCN1), which is expressed by tumor cells and a variety of immune cells, is known to inhibit T cell function [11,12]. B7–H4 expression is regulated by lipopolysaccharides (LPS), phytohemagglutinin (PHA), phorbol 12-myristate 13-acetate (PMA), and inflammatory cytokines such as IL-6, IL-10, IL-2, IFN-α, IFN-γ, and TNF-α [[13], [14], [15], [16], [17], [18]]. B7–H4 inhibits T cell activation and proliferation through binding with unknown receptor on T cells [12]. The B7–H4 antibody blockade can reverse the inhibition of tumor-specific T-cells in vivo [[19], [20], [21]]. Increasing numbers of studies have shown that B7–H4 is highly expressed in different tumor types [22], and its expression levels correlate with clinical tumor progression and pathological characteristics [21,[23], [24], [25], [26]]. Blocking with a B7–H4 antibody synergizes with anti-PD-1 in suppression of tumor growth [12,20]. B7–H4 is therefore a promising immunotherapeutic target [23,26,27]. However, the exact signaling pathways that regulate B7–H4 expression remain largely unclear [22]. The association between B7–H4 and IGF1R has not previously been reported.

In the present study, we found that IGF1R was associated with increased B7–H4 expression, and decreased CD8, IFN-γ, and Granzyme B (GzmB) expression in lung cancer by analyzing the Cancer Genome Atlas (TCGA) datasets and performing immunohistochemistry (IHC). IGF1R expression in lung cancer cells mediated CD8+ T cell inhibition through B7–H4 in vitro. The MEK/ERK1/2 pathway was involved in IGF1R-induced B7–H4 upregulation. B7–H4 knockdown could inhibit IGF1R-overexpression-induced tumor growth and restore tumor infiltrated CD8+ T cells in vivo. These findings demonstrate that IGF1R contributes to tumor immunosuppression through upregulating B7–H4 in lung cancer. B7–H4 might therefore be an immunotherapeutic target for lung cancers overexpressing IGF1R.

Section snippets

Cell lines and culture

Human lung cancer A549 and SPC-A-1 cell lines and the human embryo kidney 293 (HEK-293) cell line were purchased from the Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences. The mouse Lewis lung cancer (LLC) and mouse embryonic fibroblast NIH3T3 cell lines were kindly provided by Dr. Xuefeng Wang from the School of Biology and Basic Medical Sciences, Soochow University. A549, HEK-293, NIH3T3, and LLC were cultured in DMEM (Gibco) supplemented with 10% fetal bovine serum

IGF1R is associated with decreased CD8 and increased VTCN1 in lung cancer in TCGA datasets

IGF1R is thought to be associated with the immunosuppressive tumor microenvironment. We analyzed the correlation between the levels of IGF1R and CD8A using the TCGA lung cancer dataset of 1325 samples on the XENA website [32]. IGF1R levels are negatively associated with the abundance of CD8 (R = −0.2701, p = 2.559e-10), IFN-γ (R = −0.1890, p = 1.548E-10) and Granzyme B (GzmB) (R = −0.1507, p = 3.618e-7) in lung cancer (Fig. 1A), suggesting that IGF1R is correlated with CD8+ T cell inhibition in

Discussion

Lung cancer is the most common cancer worldwide and was the leading cause of cancer mortality in 2018 [42]. Multiple oncogene-driven mutations have been identified in lung cancer, such as EGFR mutations, ALK rearrangements, and MET amplification [43,44]. Therapeutic strategies targeting these genetic abnormalities have been successfully developed, and include EGFR tyrosine kinase inhibitors (EGFR-TKIs), ALK inhibitors, and MET inhibitors [43]. Subsets of patients with known mutations and gene

Author contributions

FW and LZ designed the experiments and research plan. ZZ, AL, NZ, and YC conducted experiments. BZ and FW provided patient specimens and acquired patient clinical data. MH performed the bioinformatic analysis. MH and ZZ analyzed data. FW and LZ wrote the manuscript.

Funding

This work was supported by National Nature Science Foundation of China (Grant No. 31370872; No. 81402381; No. 81502454).

Declaration of competing interest

None.

Acknowledgement

We are particularly grateful to Dr. Jinlei Chen and Ying Mou for their valuable assistance in flow cytometry.

References (60)

  • V. Sriraman et al.

    Identification of ERK and JNK as signaling mediators on protein kinase C activation in cultured granulosa cells

    Mol. Cell. Endocrinol.

    (2008)
  • M.C. Mendoza et al.

    The Ras-ERK and PI3K-mTOR pathways: cross-talk and compensation

    Trends Biochem. Sci.

    (2011)
  • K.Z. Peng et al.

    OP16, a novel ent-kaurene diterpenoid, potentiates the antitumor effect of rapamycin by inhibiting rapamycin-induced feedback activation of Akt signaling in esophageal squamous cell carcinoma

    Biochem. Pharmacol.

    (2017)
  • A.K. Roof et al.

    Consider the context: Ras/ERK and PI3K/AKT/mTOR signaling outcomes are pituitary cell type-specific

    Mol. Cell. Endocrinol.

    (2018)
  • L. Jerome et al.

    Deregulation of the IGF axis in cancer: epidemiological evidence and potential therapeutic interventions

    Endocr. Relat. Canc.

    (2003)
  • I. Bruchim et al.

    Targeting IGF-1 signaling pathways in gynecologic malignancies

    Expert Opin. Ther. Targets

    (2013)
  • N.T. Xuan et al.

    Regulation of dendritic cell function by insulin/IGF-1/PI3K/Akt signaling through klotho expression

    J. Recept. Signal Transduct. Res.

    (2017)
  • D. Bilbao et al.

    Insulin-like growth factor-1 stimulates regulatory T cells and suppresses autoimmune disease

    EMBO Mol. Med.

    (2014)
  • P.S. Hahnel et al.

    Targeting AKT signaling sensitizes cancer to cellular immunotherapy

    Canc. Res.

    (2008)
  • M. Sobral-Leite et al.

    Cancer-immune interactions in ER-positive breast cancers: PI3K pathway alterations and tumor-infiltrating lymphocytes

    Breast Cancer Res.: BCR

    (2019)
  • S. Loi et al.

    RAS/MAPK activation is associated with reduced tumor-infiltrating lymphocytes in triple-negative breast cancer: therapeutic cooperation between MEK and PD-1/PD-L1 immune checkpoint inhibitors

    Clin. Canc. Res.

    (2016)
  • W. Liu et al.

    Structures of immune checkpoints: an overview on the CD28-B7 family

    Adv. Exp. Med. Biol.

    (2019)
  • L. Ni et al.

    New B7 family checkpoints in human cancers

    Mol. Canc. Therapeut.

    (2017)
  • J. Li et al.

    Co-inhibitory molecule B7 superfamily member 1 expressed by tumor-infiltrating myeloid cells induces dysfunction of anti-tumor CD8(+) T Cells

    Immunity

    (2018)
  • K.A. Schalper et al.

    Differential expression and significance of PD-L1, Ido-1, and B7-H4 in human lung cancer

    Clin. Canc. Res.

    (2017)
  • Y. Xu et al.

    B7-H4 expression and its role in interleukin-2/interferon treatment of clear cell renal cell carcinoma

    Oncol. Lett.

    (2014)
  • I. Kryczek et al.

    B7-H4 expression identifies a novel suppressive macrophage population in human ovarian carcinoma

    J. Exp. Med.

    (2006)
  • Y. Yao et al.

    B7-H4(B7x)-mediated cross-talk between glioma-initiating cells and macrophages via the IL6/JAK/STAT3 pathway lead to poor prognosis in glioma patients

    Clin. Canc. Res.

    (2016)
  • H. El-Osta et al.

    Immune checkpoint inhibitors: the new frontier in non-small-cell lung cancer treatment

    OncoTargets Ther.

    (2016)
  • I. Kryczek et al.

    Relationship between B7-H4, regulatory T cells, and patient outcome in human ovarian carcinoma

    Canc. Res.

    (2007)
  • Cited by (0)

    1

    These authors contributed equally to the paper.

    View full text