Elsevier

Molecular Immunology

Volume 119, March 2020, Pages 35-45
Molecular Immunology

EZH2 inhibitors restore epigenetically silenced CD58 expression in B-cell lymphomas

https://doi.org/10.1016/j.molimm.2020.01.006Get rights and content

Highlights

  • Loss of CD58 is a common mechanism for immune evasion in lymphoid malignancies.

  • EZH2 inhibitors restore epigenetically suppressed CD58 expression of lymphoma cells.

  • EZH2 inhibitors enhance IFN-γ production of T and NK cells against lymphoma cells.

Abstract

Loss of CD58 is a common mechanism for tumor immune evasion in lymphoid malignancies. CD58 loss is known to occur due to both genetic and non-genetic causes; therefore, we hypothesized that restoring CD58 expression in lymphoma cells may be an effective treatment approach. To explore the potential for restoring CD58 expression, we first screened 11 B-cell lymphoma lines and found that 3 had decreased CD58 expression. Among these, CD58 was genetically damaged in two lines but not in the third line. Using the cell line with downregulated CD58 without a genetic abnormality, we performed epigenetic library screening and found that two EZH2 inhibitors, EPZ6438 and GSK126, specifically enhanced CD58 expression. By examining the effect of three EZH2 inhibitors with different selectivity profiles in different B-cell lines, EZH2 inhibition was shown to have a common activity in upregulating CD58 expression. Restoring the expression of CD58 in lymphoma cells using an EZH2 inhibitor was shown to enhance interferon-γ production of T and NK cells against lymphoma cells. H3K27 was shown to be highly trimethylated in the CD58 promoter region, and EZH2 inhibition induced its demethylation and activated transcription of the CD58 gene. These results indicated that EZH2 is involved in the epigenetic silencing of CD58 in lymphoma cells as a mechanism for tumor immune escape, and EZH2 inhibitors are able to restore epigenetically suppressed CD58 expression. Our findings provide a molecular basis for the combination of an EZH2 inhibitor and immunotherapy for lymphoma treatment.

Introduction

The tumor microenvironment plays a critical role in the development and progression of lymphoid malignancies. Lymphoma cell survival and proliferation in vivo are often dependent on interactions with the non-tumor cell components of the microenvironment to some degree. On the other hand, lymphoma cells also modulate these cellular interactions to escape from immune surveillance.

Understanding of these interactions has contributed to the development of novel immunotherapies. For instance, immune checkpoint inhibitors have been highly effective for the treatment of Hodgkin lymphoma (Ansell et al., 2015); Chen et al., 2017; Armand et al., 2018. Hodgkin lymphoma is characterized by malignant Hodgkin/Reed-Sternberg (H/RS) cells dispersed within the immune cell infiltrates, and a PD-1-blocking antibody inhibited the interaction between PD-1 and its ligands expressed on tumor-infiltrating T cells and H/RS cells, respectively (Ansell et al., 2015; Yamamoto et al., 2008; Carey et al., 2017). Although PD-1 signaling pathway is also suggested to be involved in other lymphoma subtypes, PD-1 blockade therapy is not as effective in Hodgkin lymphoma, at least when it is used as a single agent (Ansell et al., 2019; Lesokhin et al., 2016). One possible reason for the limited efficacy of immune checkpoint inhibitors in non-Hodgkin lymphomas is the frequent involvement of additional immune escape mechanisms. In the comprehensive genomic analysis of diffuse large B-cell lymphoma (DLBCL), molecules involved in antigen presentation and costimulatory signaling, such as HLA class I/II, CD58, B2M, CD70, and 4-1BBL, were shown to be recurrently mutated (Challa-Malladi et al., 2011); Dubois et al., 2016; Pasqualucci et al., 2011; Karube et al., 2018; Schmitz et al., 2018; Reddy et al., 2017; Chapuy et al., 2018. These findings suggested that defective immune synapse formation between lymphoma cells and immune effector cells is an important underlying mechanism of immune evasion in DLBCL.

The CD58 gene is one of the recurrent targets of genetic abnormalities in DLBCL and in other lymphoid malignancies such as peripheral T-cell lymphoma (Palomero et al., 2014) and adult T-cell leukemia/lymphoma (Kataoka et al., 2015; Yoshida et al., 2014). CD58 gene abnormality is also suggested to appear in the process of transformation of follicular lymphoma (FL) into DLBCL (Pasqualucci et al., 2014). CD58 is a member of the immunoglobulin superfamily, which has a function to bind to CD2 expressed on T and NK cells (Wang et al., 1999); Grakoui et al., 1999; Moingeon et al., 1989, and the CD2-CD58 interaction is especially important for tumor recognition by T and NK cells. Loss of CD58 expression is indicated to be associated with worse overall and event-free survivals in patients with DLBCL (Cao et al., 2016) and acute lymphoblastic leukemia (Li et al., 2016). CD58 expression is reported to be lost through both genetic and non-genetic mechanisms (Challa-Malladi et al., 2011); therefore, we hypothesized that restored expression of CD58 may facilitate T and NK cell-immune recognition of lymphoma cells and increase the efficacy of immune-targeted therapy.

To explore the non-genetic mechanism of defective CD58 expression in lymphoma cells, we performed epigenetic compound library screening using a B-cell lymphoma line with decreased CD58 expression without any CD58 gene abnormality. We found that EZH2 inhibitors specifically restored CD58 expression in these cells, and upregulation of CD58 by an EZH2 inhibitor enabled lymphoma cells to strongly stimulate T and NK cells. H3K27 was shown to be highly trimethylated in the CD58 promoter region, and EZH2 inhibition induced its demethylation and increased CD58 gene transcription. These results indicated that EZH2 is involved in the epigenetic silencing of CD58 in lymphoma cells as a mechanism for tumor immune escape, and an EZH2 inhibitor was effective for the restoration of epigenetically downregulated CD58 expression. Our findings provide a molecular basis for the effectiveness of EZH2 inhibitors and a rationale for their combination with immune-targeted therapies for the treatment of lymphomas.

Section snippets

Analysis of European Genome-phenome Archive (EGA) data sets

Whole exome sequencing and RNA-seq gene expression data derived from 1001 DLBCL samples and the core set of 624 DLBCL samples were obtained from EGA (dataset id: EGA00001003600) (Reddy et al., 2017). Gene expression was measured using terms of fragments per kilobase of exon per million fragments mapped and normalized using the Cufflinks package, version 2.2.1 (Trapnell et al., 2010). Quantile normalization was performed and the data were log2 normalized. To evaluate the correlation between

CD58 expression level correlates with T-cell activation signature in DLBCL

To examine the significance of CD58 expression in DLBCL, we analyzed a publicly available RNA-seq DLBCL database consisted of 278 cases of GCB-DLBCL and 252 cases of ABC-DLBCL (Fig. 1A). We found that the CD58 expression level significantly correlated with T-cell activation signature in both GCB-DLBCL and ABC-DLBCL. These results suggested that CD58 expression level is actually associated with T-cell function in DLBCL tissues, regardless of the cell-of-origin subtype.

Screening for decreased CD58 expression in B-cell lymphoma lines

To investigate the

Discussion

CD2-CD58 is an important intercellular signaling pathway for the immune reactions in cytotoxic T and NK cells against tumor cells (Challa-Malladi et al., 2011; Altomonte et al., 1993; Gwin et al., 1996). Loss of CD58 is one of the most frequent mechanisms of immune escape in lymphoid malignancies, and CD58 loss can be caused by both genetic and non-genetic mechanisms. In this report, we demonstrated that EZH2 is involved in the epigenetic silencing of CD58 in lymphoma cells, and EZH2 inhibitors

CRediT authorship contribution statement

Yasuyuki Otsuka: Investigation, Methodology, Writing - original draft. Momoko Nishikori: Conceptualization, Methodology, Writing - original draft, Writing - review & editing, Funding acquisition. Hiroshi Arima: Resources, Formal analysis. Kiyotaka Izumi: Resources. Toshio Kitawaki: Resources, Methodology. Masakatsu Hishizawa: Resources, Funding acquisition. Akifumi Takaori-Kondo: Supervision.

Declaration of Competing Interest

M.N. and A.T-K received honorarium and research funding from Eisai Co., Ltd. The other authors declare no potential conflicts of interest.

Acknowledgements

This work was supported by Japan Society for the Promotion of Science Grant Numbers 15K09474, 18K08324 (M.N.) and 17K09925 (M.H). We thank A. Reddy and S. S. Dave (Duke University, Durham, NC) for kindly providing access to the DLBCL database. We also thank Dr. H. Miyoshi, RIKEN Bioresource Center, Japan, for providing us the packaging plasmid pCAG-HIVgp and the VSV-G- and Rev-expressing plasmids (pCMV-VSV-G-RSV-Rev).

References (48)

  • D. Zingg et al.

    The histone methyltransferase Ezh2 controls mechanisms of adaptive resistance to tumor immunotherapy

    Cell Rep.

    (2017)
  • M. Altomonte et al.

    Differential expression of cell adhesion molecules CD54/CD11a and CD58/CD2 by human melanoma cells and functional role in their interaction with cytotoxic cells

    Cancer Res.

    (1993)
  • S.M. Ansell et al.

    PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma

    N. Engl. J. Med.

    (2015)
  • S.M. Ansell et al.

    Nivolumab for Relapsed/Refractory diffuse large B-Cell lymphoma in patients ineligible for or having failed autologous transplantation: a single-arm, phase II study

    J. Clin. Oncol.

    (2019)
  • P. Armand et al.

    Nivolumab for Relapsed/Refractory classic hodgkin lymphoma after failure of autologous hematopoietic cell transplantation: extended follow-up of the multicohort single-arm phase II CheckMate 205 trial

    J. Clin. Oncol.

    (2018)
  • R.L. Bolhuis et al.

    Induction and blocking of cytolysis in CD2+, CD3- NK and CD2+, CD3+ cytotoxic T lymphocytes via CD2 50 KD sheep erythrocyte receptor

    J. Immunol.

    (1986)
  • A.P. Bracken et al.

    Polycomb group proteins: navigators of lineage pathways led astray in cancer

    Nat. Rev. Cancer

    (2009)
  • M. Caganova et al.

    Germinal center dysregulation by histone methyltransferase EZH2 promotes lymphomagenesis

    J. Clin. Invest.

    (2013)
  • Y. Cao et al.

    Mutations or copy number losses of CD58 and TP53 genes in diffuse large B cell lymphoma are independent unfavorable prognostic factors

    Oncotarget

    (2016)
  • B. Chapuy et al.

    Molecular subtypes of diffuse large B cell lymphoma are associated with distinct pathogenic mechanisms and outcomes

    Nat. Med.

    (2018)
  • R. Chen et al.

    Phase II study of the efficacy and safety of Pembrolizumab for Relapsed/Refractory classic hodgkin lymphoma

    J. Clin. Oncol.

    (2017)
  • S. Dubois et al.

    Next-generation sequencing in diffuse large B-Cell lymphoma highlights molecular divergence and therapeutic opportunities: a LYSA study

    Clin. Cancer Res.

    (2016)
  • A.L. Epstein et al.

    Biology of the human malignant lymphomas. IV. Functional characterization of ten diffuse histiocytic lymphoma cell lines

    Cancer

    (1978)
  • S. Feske et al.

    Gene regulation mediated by calcium signals in T lymphocytes

    Nat. Immunol.

    (2001)
  • Cited by (28)

    • Anti-CD20 antibody treatment for diffuse large B cell lymphoma: Genetic alterations and signaling pathways

      2023, Resistance to Anti-CD20 Antibodies and approaches for their Reversal: Volume 2
    • CD58 loss in tumor cells confers functional impairment of CAR T cells

      2022, Blood Advances
      Citation Excerpt :

      CD58 is a member of the immunoglobulin superfamily and is a ligand for the costimulatory molecule CD2 expressed in T cells.41 Disruption of the CD58-CD2 axis by blocking antibodies results in decreased T-cell activation, reduced IFN-γ secretion, and reduced cytotoxicity.42,43 Several reports have shown that loss of CD58 in tumor cells is an unfavorable prognostic factor and a frequent genetic abnormality in patients with hematologic malignancies.44,45

    • Taking the EZ way: Targeting enhancer of zeste homolog 2 in B-cell lymphomas

      2022, Blood Reviews
      Citation Excerpt :

      However, the important immune effect of mutant EZH2 suggests that EZH2 inhibitors also may induce prominent anti-lymphoma immune effects in these patients, which would have been missed in preclinical studies published to date. Moreover, wild-type EZH2 plays a role in limiting differentiation and plasticity in T cells and may favor evasion from immune surveillance because of T-cell autonomous effects on T-regulatory cells (reviewed elsewhere [42,43]). These are important points to consider when interpreting the outcomes of EZH2 inhibitor clinical trials, and when designing correlative science protocols.

    • Role of EZH2 in bone marrow mesenchymal stem cells and immune–cancer interactions

      2022, Critical Reviews in Oncology/Hematology
      Citation Excerpt :

      EZH2 impacts NK cell-mediated antitumor effects in two ways: 1) deletion or functional inhibition of EZH2 significantly increases NK cell frequency and quality (Yin et al., 2015); 2) in the tumor microenvironment, the inhibition of EZH2 in tumor cells enhances the recognition and killing function of immune cells. For example, in B-cell lymphoma, EZH2 epigenetically silences the expression of the CD58 gene involved in presenting antigens to T cells and NK cells, which is one of the mechanisms of tumor immune evasion (Otsuka et al., 2020). In hepatocellular carcinoma, inhibition of EZH2 enhances NK cell killing function directly through upregulation of MHC class I polypeptide-related sequence A 1, the ligand of natural killer group 2D (NKG2D) on the surface of hepatocellular carcinoma cells (Bugide et al., 2018).

    View all citing articles on Scopus
    View full text