Blockade of myeloid-derived suppressor cell function by valproic acid enhanced anti-PD-L1 tumor immunotherapy

https://doi.org/10.1016/j.bbrc.2019.11.155Get rights and content

Highlights

  • VPA-treatment promoted the polarization of bone marrow-derived precursor cells into M-MDSCs.

  • VPA combined with anti-PD-L1 antibody block the immunosuppressive function of MDSCs by downregulating the expression of IL-10, IL-6, and ARG1.

  • Combination therapy of VPA and anti-PD-L1 antibody promoted the activation of IRF1/IRF8 transcriptional axis in MDSCs.

  • The combined treatment enhanced PD-L1 blockade-mediated melanoma regression through the secretion of TNFα.

Abstract

Regardless of the remarkable clinical success of immune checkpoint blockade (ICB) against PD-1/PD-L1 pathway, this approach has encountered drawbacks in most patients due to the activation of tumor immunosuppressive factors such as myeloid-derived suppressor cells (MDSCs). Histone deacetylase (HDAC) inhibitors combat ICB resistance by attenuating the immunosuppressive function of MDSCs and increasing PD-L1 expression on tumor cells. However, whether an HDAC inhibitor - valproic acid (VPA) suppression of MDSCs function could enhance PD-L1 blockade-mediated tumor immunotherapy remains unknown. Here we report that VPA and anti-PD-L1 antibody combined treatment promoted the polarization of bone marrow-derived precursor cells into M-MDSCs. Interestingly, the combination treatment of VPA and anti-PD-L1 antibody activated IRF1/IRF8 transcriptional axis in MDSCs leading to blockade of their immunosuppressive function by downregulating the expression of IL-10, IL-6, and ARG1 while re-activating CD8+ T-cells for the production of TNFα to further enhance anti-tumor immunity. These observations provide further rationale for the combination therapy of VPA with anti-PD-L1 antibody in preclinical settings.

Introduction

Immune checkpoint inhibitors (ICIs) targeting programmed cell death 1/programmed cell death 1 ligand 1 (PD-1/PD-L1) axis generate durable clinical responses in a sizeable minority of tumor patients partly through reinvigoration of CD8+ T cells [1]. However, several cancer patients have shown overwhelming resistance to immune checkpoint blockades (ICB). The major obstacle to ICB in cancer immunotherapy is activation of different immunosuppressive factors in tumor microenvironment (TME), which inhibit T-cell effector and decrease infiltration of T cells into the tumor tissue.

Myeloid-derived suppressor cells (MDSCs), a major component of pathologically activated cells displaying an exceptional immunosuppressive ability against anti-tumor T-cell response contribute to resistance of ICB. MDSCs are further divided into two subsets: monocytic MDSCs (M-MDSCs) and granulocytic MDSCs (G-MDSCs), respectively [2,3]. Generally, the cell surface markers for MDSCs include Gr1 and CD11b in mice [4,5]. The M-MDSCs are CD11b+Ly6ChighLy6G whereas G-MDSCs are CD11b+Ly6ClowLy6G+ [3,4]. More importantly, MDSCs exert its functions through the production of IL-6, IL-10, arginase 1 (ARG1), reactive oxygen species (ROS), and nitric oxide (NO). The differentiation of myeloid cell subsets is coordinated by several transcriptional regulators; interferon regulatory factors (IRF), IRF1 and IRF8 have been identified to be involved in the production of myeloid progenitors [6]. They control various functions; modulation of immune responses, host defense mechanism, cell proliferation, hematopoietic development, and cytokine signaling [7]. IRF8 interacts with other transcription factors such as IRF1 through its association domain to confer immunity against tumor and infectious diseases [8,9]. Blocking immunosuppressive functions of MDSCs leads to markedly enhanced anti-tumor immunity. Interestingly, cancer patients who had a higher level of MDSCs were resistant to anti-PD-L1 therapy. But whether inhibition of MDSCs could relieve resistance to anti-PD-L1 therapy still needs to be investigated.

Histone deacetylases (HDACs) have demonstrated potent anticancer activities, regulation of immune cells function through epigenetic modification of multiple genes [10]. Remarkably, valproic acid (VPA), which is a histone deacetylase inhibitor (HDACi) targeting HDAC class I enzymes (HDAC1, 2 and 3), was reported to have a potential in attenuating the immunosuppressive function of MDSCs [11]. VPA promoted tumor-induced M-MDSCs differentiation into dendritic cells (DCs) and macrophages in-vitro [12]. However, whether VPA suppression of MDSCs function could enhance PD-L1 blockade-mediated tumor immunotherapy remains unknown. In this study, we provide a rationale for combined therapy of VPA and anti-PD-L1 antibody which could be explored in clinical settings.

Section snippets

Mice

Six-to eight-week-old wild-type (WT) C57BL/6 mice were used in this study. Mouse handling and experimental procedures was in accordance with established institute’s guidance and approved protocols from the animal facility committee of Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS).

Cell line

B16F10 or LLC cell line was obtained from CAS cell bank and cultured in DMEM supplemented with 10% FBS, 1% penicillin-streptomycin (PS), 37 °C, 5% CO2.

Reagent and antibodies

DMEM, RPMI 1640, FBS, PBS and

In vitro

Studies had demonstrated GM-CSF could be used to generate MDSCs in-vitro from bone marrow (BM) precursor cells [13,14]. To investigate the effect of VPA treatment on GM-CSF-induced MDSCs in vitro, we isolated BM cells from mice stimulated with GM-CSF for 3 days, and then treated with either VPA, or anti-PD-L1 antibody, or both VPA and anti-PD-L1 antibody for 24 h, the percentages of MDSCs and its subset were evaluated by flow cytometry. We observed that VPA promoted the differentiation of CD11b+

Discussion

This study demonstrated that VPA augments PD-L1 blockade therapy to impair melanoma growth via activation of IRF1/IRF8 transcriptional axis of MDSCs, thus inhibiting MDSCs immunosuppressive function through downregulating IL-10, IL-6, and ARG1 while re-activating CD8+ T-cells.

HDACi has been identified as anti-cancer agent by regulating MDSCs generation and function [19]. Class I and II non-selective pan-HDACi (SAHA and TSA) expanded M-MDSCs in in-vitro culture system and in tumor-bearing mice [

Author contributions

Conceptualization, D.Y; Data curation, A.O.A; Formal analysis, A.O.A; Funding acquisition, D.Y and X.W; Investigation, A.O.A; Methodology, W.L; Project administration, X.W; Resources, L.W; Supervision, D.Y and X.W; Validation, M.Z; Visualization, F.O.A; Writing – original draft, A.O.A; Writing – review & editing, D.Y.

Funding

This work was supported by the National Natural Science Foundation of China (Grant 81501356 and Grant 81373112), the Shenzhen Basic Science Research Project (Grants JCYJ20170413153158716 and JCYJ20170818164619194), Nanshan pilot team project (LHTD20160004), Start-up funding (CYZZ20180307154657923), Guangdong Provincial Research Award for Thousand Talents Program Scholars.

Declaration of competing interest

The authors declare no competing financial interests.

Acknowledgements

We appreciate the technical contributions from Lukman. O. Afolabi, Ruiling Liu (Renee) and Jiang Bian during this research.

References (28)

  • T. Taniguchi et al.

    IRF family of transcription factors as regulators of host defense

    Annu. Rev. Immunol.

    (2001)
  • D. Langlais et al.

    The macrophage IRF8/IRF1 regulome is required for protection against infections and is associated with chronic inflammation

    J. Exp. Med.

    (2016)
  • A. Villagra et al.

    Histone deacetylases and the immunological network: implications in cancer and inflammation

    Oncogene

    (2010)
  • J.I. Youn et al.

    Epigenetic silencing of retinoblastoma gene regulates pathologic differentiation of myeloid cells in cancer

    Nat. Immunol.

    (2013)
  • Cited by (29)

    • Combining histone deacetylase inhibitors (HDACis) with other therapies for cancer therapy

      2021, European Journal of Medicinal Chemistry
      Citation Excerpt :

      HDACis are promosing to reverse the resistance of cancer cells to immune checkpoint blockade therapy by reducing the immunosuppressive activity of MDSCs and downregulating the expression of immunosuppresive proteins[163]. In order to achieve enhanced antitumor efficacy, Adeshakin et al. combined VPA and anti-PD-L1 antibody, aiming at promoting the polarization of bone marrow-derived precursor cells into monocytic MDSCs (M-MDSCs) [164]. Both the in vitro and in vivo results demonstrated that VPA promoted the reactivation of CD8+ T cells response and generated TNFα, which further converted M-MDSCs to a stimulatory phenotype from an immunosuppressive phenotype.

    • Deep and Durable Response to Nivolumab and Temozolomide in Small-Cell Lung Cancer Associated With an Early Decrease in Myeloid-Derived Suppressor Cells

      2021, Clinical Lung Cancer
      Citation Excerpt :

      MDSC have also been associated with decreased responsiveness to ICI through the inhibition of specific antitumor immunity and promotion of an immunosuppressive tumor microenvironment. Preclinical studies have shown that modulation of MDSC through a variety of mechanisms can enhance T-cell responsiveness to immunotherapy.37-39 MDSC are divided into two major subsets, monocytic (M) and granulocytic or PMN-MDSC, according to their phenotypic and morphologic features.40-42

    • Lipidomics data showing the effect of lipofermata on myeloid-derived suppressor cells in the spleens of tumor-bearing mice

      2021, Data in Brief
      Citation Excerpt :

      The cell pellet was re-suspended in PBS to obtain a single-cell suspension. The single-cell suspension was subjected to mojosort magnetic cell separation according to the manufacturer's procedure and followed by cell surface staining with CD11b and Gr1 monoclonal antibodies for cell sorting on BD FACS Aria III cell sorter (BD Biosciences) as previously described [5]. CD11b+ Gr1+ flow cytometry sorted MDSCs from the spleens of B16F10 tumor-bearing mice treated with or without FATP2 inhibitor (lipofermata) were resuspended in 200μl of PBS and lipid was extracted according to a previously described method [4].

    • Regulation of ROS in myeloid-derived suppressor cells through targeting fatty acid transport protein 2 enhanced anti-PD-L1 tumor immunotherapy

      2021, Cellular Immunology
      Citation Excerpt :

      Phenotypically, these cells are similar to neutrophils and monocytes; however, biochemically and functionally, they are different from the aforementioned cell subsets [5]. In mice, MDSCs are characterized as cells co-expressing CD11b and Gr1 markers, which can be further categorized into two populations: polymorphonuclear (PMN)-MDSCs (CD11b+Ly6G+Ly6Clow) and monocytic (M)-MDSCs (CD11b+Ly6G−Ly6Chigh) [8–10]. Functionally, MDSCs use different mechanisms such as induction of arginase 1 (Arg1), inducible nitric oxide synthase (iNOS), and reactive oxygen species (ROS) production to suppress anti-tumor immune responses [11].

    • Interferon regulatory factor 8 governs myeloid cell development

      2020, Cytokine and Growth Factor Reviews
      Citation Excerpt :

      In tumor xenograft model, ectopic expression of IRF8 leads to regression of tumor nodules and notable reduction of MDSCs [111]. Interestingly, combination treatment of histone deacetylase (HDAC) inhibitor VPA with anti-PD-L1 antibody can significantly block the immunosuppressive activity of MDSCs by activating IRF1/IRF8 axis that further weaken the production of downstream suppressive molecules, which suggests the great target potential of IRF8 in anti-tumor strategy [112]. Moreover, IRF8 as a susceptibility factor in MS patients has been widely recognized and also aberrant expression of IRF8 transcripts is independent adverse prognostic factors for patients with Acute myeloid leukemia (AML) [113,114].

    View all citing articles on Scopus
    1

    These authors contributed equally.

    View full text