Elsevier

Human Pathology

Volume 54, August 2016, Pages 17-24
Human Pathology

In this issue
Evaluation of the diagnostic and prognostic value of PDL1 expression in Hodgkin and B-cell lymphomas,☆☆

https://doi.org/10.1016/j.humpath.2016.03.005Get rights and content

Summary

Activation of the programmed death 1 (PD1)/PD1 ligand (PDL1) pathway is important for tumor cells to escape from immune control. The clinical efficacy of therapeutic modulation of the PD1-PDL1 pathway has been recently shown in classical Hodgkin lymphoma (cHL), but little is known about the frequency and diagnostic and prognostic importance of PDL1 expression in lymphomas. The available anti-PDL1 antibody clones E1L3N and SP142 were compared, and a large cohort of Hodgkin lymphomas (n = 280) and B-cell lymphomas (n = 619) was examined for PDL1 using E1L3N. The results were correlated with the expression of other phenotypic markers, interphase fluorescence in situ hybridization data of the 9p24.1 region (PDL1 locus), and the clinical outcome. PDL1 was expressed on more than 5% of tumor cells in 70% of cHL, 54% of nodular lymphocyte-predominant Hodgkin lymphoma, and 35% of primary mediastinal B-cell lymphomas; in the latter, PDL1 expression correlated with PDL1 gains (ρ = 0.573). PDL1 was expressed in 31% of primary diffuse large B-cell lymphomas (DLBCLs), whereas most other entities did not express PDL1. In cHL, expression of PDL1 correlated with increased numbers of granzyme + T cells (ρ = 0.251) and CD68 + macrophages (ρ = 0.221) but with decreased numbers of FoxP3 + T cells (ρ = 0.145). In activated B-cell–like DLBCL, PDL1 positively correlated with PD1 + T cells, whereas an inverse correlation with FoxP3 + T cells was seen in the germinal center B-cell–like DLBCL. PDL1 expression can be diagnostically valuable in some gray zones around DLBCL and cHL; it identifies an “immune escape” cluster of cHL and activated B-cell–like DLBCL with increased granzyme + and PD1 + T cells and macrophages and decreased regulatory T cells.

Introduction

The programmed death 1 (PD1)/PD ligand (PDL) pathway is an important checkpoint for the regulation of T-cell–mediated immune responses [1]. It consists of the transmembrane protein PD1/CD279 itself and its 2 ligands PDL1 (B7-H1, CD274) and PDL2 (B7-DC, CD273). These PDLs activate PD1, which results in a reversible inhibition of T-cell activity and proliferation also known as T-cell exhaustion or anergy. This mechanism plays an important physiological role in preventing placenta infiltration by T cells [2] and maintaining self-tolerance [3], which has been verified in animal studies of pd1 knock-out mice developing several autoimmune diseases [4].

Unfortunately, malignancies also make use of the immunosuppressive effects of the PD1-PDL pathway [5], which is to a part reflected by high levels of PD1-positive tumor infiltrating T-cells. Besides, several tumors are known to express PDL1, being one of the mechanisms of building up a defense line against tumor-infiltrating lymphocytes (TILs) [6]. This applies not only to solid tumors such as lung or breast cancer but also to hematolymphoid neoplasms such as angioimmunoblastic T-cell lymphoma, follicular lymphoma (FL), diffuse large B-cell lymphoma (DLBCL), primary mediastinal B-cell lymphoma (PMBCL), and classical Hodgkin lymphoma (cHL), especially the nodular sclerosis subtype [7], [8]. Amplifications of the PDL1 gene locus on 9p24.1 are recurrent in the last 2 entities [9] further underscoring the importance of that pathway. PDL1 expression by immunohistochemistry has been demonstrated in some of the lymphomas listed above only in smaller cohorts or with antibodies shown to more poorly perform so far [10], [11]. Because of the fact that intrinsic cancer-specific T cells might be thwarted by PD1-PDL1 interactions [12], targeting PDL1 has become a new approach in tumor therapy [13]. Recently, first very promising results of PD1-PDL1 blockade have been reported in cHL, melanoma, and non–small cell lung cancer [14], [15], [16].

So far, the question whether expression of PD1 and PDL1 by immunohistochemistry might also be of diagnostic or prognostic importance in lymphomas has not been answered on larger collectives. It was our aim to investigate the expression of PDL1 at large scale in cHL and various B-cell lymphoma subtypes to draw conclusions both for diagnostic and potential clinicopathological applications.

Section snippets

Selection and tissue microarray construction

A total number of 899 cases encompassing different lymphoma entities were selected and examined on tissue microarrays (TMAs) and whole slides (Table 1); 15 nodular lymphocyte-predominant Hodgkin lymphomas (NLPHLs), 7 Burkitt lymphomas (BLs), 35 mantle cell lymphomas (MCLs), and 11 T-cell and histiocyte-rich B-cell lymphomas (THRBCLs) were analyzed on whole mount slides. The study was approved by the ethics committee of Northwestern and Central Switzerland (EKNZ 2014-252).

Immunohistochemistry

To assess the optimal

Staining results

The Cronbach α value was 0.896 for overall PDL1 staining evaluation; thus, the staining assessment was regarded reproducible. The PDL1 staining was strong to moderate, membranous, and submembranous, occasionally dotted (for explanation of the dots see Materials and methods/Immunohistochemistry). Fig. 2 shows representative examples of PDL1 staining results in several lymphoma entities. In THRBCL and Hodgkin lymphoma (HL), there were many reactive nontumor cells expressing PDL1. In THRBCL, a

Discussion

In this study, we comprehensively analyzed the expression of PDL1 in both Hodgkin and B-cell (non-Hodgkin) lymphomas. To this end, we selected an optimally working antibody giving best signal-to-noise staining results and assessed the reproducibility of staining evaluation, which showed excellent results. PDL1 expression was studied in a large cohort of TMA cases as well as on whole mount slides. Besides, a double staining for BCL6 and PDL1 was established for THRBCL to facilitate the

Acknowledgments

The authors would like to thank Mrs Petra Hirschmann for performing the immunohistochemical stainings.

References (41)

  • J. Krugmann et al.

    Longer failure-free survival interval of Epstein-Barr virus–associated classical Hodgkin's lymphoma: a single-institution study

    Mod Pathol

    (2003)
  • M.E. Keir et al.

    PD-1 and its ligands in tolerance and immunity

    Annu Rev Immunol

    (2008)
  • H. Jin et al.

    Role of PD-1 in regulating T-cell immunity

    Curr Top Microbiol Immunol

    (2011)
  • B.T. Fife et al.

    The role of the PD-1 pathway in autoimmunity and peripheral tolerance

    Ann N Y Acad Sci

    (2011)
  • S. Muenst et al.

    The PD-1/PD-L1 pathway: biological background and clinical relevance of an emerging treatment target in immunotherapy

    Expert Opin Ther Targets

    (2015)
  • S. Muenst et al.

    Diagnostic and prognostic utility of PD-1 in B cell lymphomas

    Dis Markers

    (2010)
  • A. Rosenwald et al.

    Molecular diagnosis of primary mediastinal B cell lymphoma identifies a clinically favorable subgroup of diffuse large B cell lymphoma related to Hodgkin lymphoma

    J Exp Med

    (2003)
  • C.B. Hutchinson et al.

    Primary mediastinal (thymic) large B-cell lymphoma: a short review with brief discussion of mediastinal gray zone lymphoma

    Arch Pathol Lab Med

    (2011)
  • W. Yamamoto et al.

    Clinicopathological analysis of mediastinal large B-cell lymphoma and classical Hodgkin lymphoma of the mediastinum

    Leuk Lymphoma

    (2013)
  • C. Blank et al.

    Interaction of PD-L1 on tumor cells with PD-1 on tumor-specific T cells as a mechanism of immune evasion: implications for tumor immunotherapy

    Cancer Immunol Immunother

    (2005)
  • Cited by (125)

    • Visible- and near-infrared hyperspectral imaging for the quantitative analysis of PD-L1+ cells in human lymphomas: Comparison with fluorescent multiplex immunohistochemistry

      2023, Spectrochimica Acta - Part A: Molecular and Biomolecular Spectroscopy
      Citation Excerpt :

      In human Non-Hodgkin B-cell-lymphomas, PD-L1 expression is rarely, if ever, seen in lymphoma cells themselves but has been found in tumor-associated immune cells, mainly macrophages [33,34]. In follicular lymphoma (FL), PD-L1 expression was described primarily on macrophages, and higher levels of intrafollicular PD-L1+ macrophages were associated with a shorter time to transformation, but overall and progression-free survival were not affected [35,36]. In diffuse large B-cell lymphoma (DLBCL) PD-L1 is found rarely in tumor cells and is related to non-germinal centre (non-GCB) subtype, Epstein-Barr virus (EBV) positivity, and a worse prognosis [33,34,37].

    View all citing articles on Scopus

    Competing interests: The authors declare to have no competing interests.

    ☆☆

    Funding/Support: Thomas Menter is supported by the Nuovo-Soldati Cancer Research Foundation, Vaduz, Liechtenstein.

    View full text