Skip to main content
Journal of Clinical and Experimental Hematopathology : JCEH logoLink to Journal of Clinical and Experimental Hematopathology : JCEH
. 2023 Jun 28;63(2):58–64. doi: 10.3960/jslrt.23003

Diagnostic approach for classic Hodgkin lymphoma in small samples with an emphasis on PD-L1 expression and EBV harboring in tumor cells: a brief review from morphology to biology

Taishi Takahara 1,, Ayako Sakakibara 1, Yuta Tsuyuki 1, Akira Satou 1, Seiichi Kato 1, Shigeo Nakamura 1
PMCID: PMC10410620  PMID: 37380470

Abstract

Classic Hodgkin lymphoma (CHL) was first described in 1832 by Thomas Hodgkin, and is characterized by a small number of Hodgkin and Reed–Sternberg cells in a rich inflammatory background. However, even in this modern era, due to the histological and biological overlap with CHL and other B-cell malignancies, including mediastinal grey zone lymphoma and other lymphomas accompanied by “Hodgkinoid cells”, their discrimination is challenging and sometimes impossible. The complexity and ambiguity of the boundaries of CHL and its related diseases make the definition of CHL unresolved. Our group has studied the significance of PD-L1 expression and infection of Epstein-Barr virus (EBV) in the diagnosis of CHL, emphasizing their pathological role, clinical significance, and high reproducibility even in daily clinical practice. In this review, we summarize the diagnostic strategy of CHL and its histological lookalikes based on neoplastic PD-L1 expression and infection of EBV, and attempt a reappraisal of the definition of CHL.

Keywords: classic Hodgkin lymphoma, Epstein-Barr virus, PD-L1

INTRODUCTION

Classic Hodgkin lymphoma (CHL) is biologically heterogeneous, exemplified by several clinic-pathologic differences between nodular sclerosis (NS) and mixed cellularity (MC). It is well known that this unique lymphoma encompasses four histologic subtypes (NS, MC, lymphocyte depleted [LD], and lymphocyte-rich [LR]) and often poses differential diagnostic problems due to morphologic lookalikes of other lymphoid neoplasia/proliferation. In this modern era, small-volume tissue sampling, such as core needle biopsy, is the preferred modality for diagnosis in routine practice, but does not allow for precise sub-classification or accurate assessment of the architecture due to the limited material collected. Of note, recent advances have clearly demonstrated that PD-L1 expression in tumor cells is a highly sensitive diagnostic marker and is regarded as a defining feature of CHL.1 EBV status is also an important prognostic indicator among the patients with CHL.2 Given their biological significance, the presence of PD-L1 and EBV in tumor cells should provide significant information for diagnosis. The reproducibility of their detection in tumor cells also aid accurate diagnosis, especially in small-volume samples. In this review, we provide an update on the diagnostic features of CHL with an emphasis on PD-L1 expression and EBV harboring in tumor cells.

Diagnostic dilemma and future perspective on CHL in small samples

CHL accounts for around 90% of all Hodgkin lymphoma.3 Hodgkin lymphomas (HLs) are historically defined by morphologic characteristics. CHL is characterized by tumor cells composed of mononuclear Hodgkin cells and multinucleated Reed-Stenberg (HRS) cells accompanied by a background containing a variable mixture of reactive immune cells including small lymphocytes, eosinophils, neutrophils, histiocytes, and plasma cells. HRS cells of CHL usually show loss of B-cell antigen, whereas germinal center (GC) B cells are currently regarded as the cellular origin of CHL in most cases. In the 2017 and 2022 WHO classifications, CHL is subdivided into four ‘histologic’ subtypes: NSCHL, MCCHL, LDCHL, and LRCHL. It is principally recommended to sub-classify CHL as one of these four histologic subtypes. It is emphasized that the paradigm for our understanding Hodgkin lymphomas is now rapidly shifting. Of note, in the international consensus classification (2022),4 the diagnostic title “nodular lymphocyte predominant B-cell lymphoma” (NLPBL) was recommended instead of NLPHL, with the term ‘Hodgkin’ being excised based on its distinctive biological and clinical features. Elaine S Jaffe clearly described that CHL is not a single disease, but likely consists of two or more entities, i.e., NSCHL and the spectrum encompassing MCCHL and LDCHL.5 She also indicated that LRCHL is still an enigma in terms of its biology and clinical features, and in some cases even shows overlap with NLPHL/NLPBL. We recently reported that typical cases of NSCHL and EBV+ MCCHL consistently feature neoplastic PD-L1 positivity in HRS cells, which is contrasted by the lack of PD-L1 expression in tumor cells of NLPHL/NLPBL and LRCHL of our studies.6 We believe that our observation is coincidental with that of Elaine S. Jaffe. Unfortunately, in the updated WHO classification (2022), the definitions of CHL and NLPHL remained obscure: in this content, CHL is a neoplasm derived from GC B-cells, characterized by a low fraction of tumor cells embedded in a reactive micro-environment rich in immune cells; and NLPHL is a GC-derived B-cell neoplasm composed of scattered large neoplastic B cells with multilobated nuclei (LP cells) within nodules dominated by mantle zone B cells and follicular dendritic cells (FDCs).

Never-the-less, a precise sub-classification is not always possible when the biopsy material inadequate, such as in the case of core needle biopsy. However, small-volume biopsy sampling is now the most favored clinical approach for node-based neoplastic diseases including CHL. Indeed, the 2022 WHO classification also described that the increasing trend to use core needle biopsies makes CHL diagnosis challenging due to the scarcity and uneven distribution of HRS cells, although most descriptions of the CHL histopathology have been made on the findings of whole node biopsies. In 2016, Roemer et al. indicated that PD-L1 and PD-L2 genetic alteration, which result in neoplastic PD-L1 expression in tumor cells, is a defining feature of CHL. An increasing number of the studies also provide additional support to the assertion that neoplastic PD-L1 expression in tumor cells is a highly sensitive diagnostic marker for the discrimination of CHL from other diagnostic targets, i.e., morphologic CHL lookalikes. In this diagnostic process, the association or not of large B-cells with EBV is also critical for our approach, because EBV-positive and –negative CHLs constitute quite distinct differential diagnostic lists. These situations led us to re-assess CHL from the morphologic basis to the biology, i.e., the high-frequent PD-L1 genetic alteration resulting in neoplastic PD-L1 overexpression and the EBV harboring (or not) in HRS cells, especially in small samples. Given the clinicopathological differences between CHL and its lookalikes in extranodal sites, CHL is hypothetically assumed to be a lymphoid organ (LN, thymus and spleen)-sited, immune escape-associated, lymphoid cell-driven (B-cell origin in most at least) neoplasm. From these standpoints, we propose a diagnostic strategy combing PD-L1 expression and EBV infection in HRS cells (Table 1).

Table 1. Differential diagnosis of CHL and its lookalikes.

Diagnosis AITL/FHTL with HRS-like large B-cells Age-related EBV+ B-LPD, i.e., EBV+ DLBCL CHL
Young
(<45 years)
Elderly EBV+GZL EBV+MCCHL CHL in patients with immune deficiency/dysregulation NSCHL LDCHL, syncytial variant EBV- MCCHL LRCHL
Clinical presentation Lymphadenopathy Lymphadenopathy Lymphadenopathy;
extranodal mass
Lymphadenopathy Lymphadenopathy Lymphadenopathy Lymphadenopathy Lymphadenopathy Lymphadenopathy
EBV + + + + + - Variable (25%) - -
PD-L1 IHC -(5%) + (77%) -/+ (11%) + (70%) +(100%) + (89%) + (100%) Heterogeneous -(0%) -(0%)
Other EBV+ large cells: CD20+/-, CD30+, PAX5+ CD20+, CD30+, PAX5+ (often strong), OCT2+, BOB1+ CD20+, PAX5+/-, CD30+, CD15+, OCT2+/-, BOB1+/- CD20-/+, PAX5+(dim), CD30+, CD15+, BOB1-, OCT2-
PD-1+ rosetting
T cells
+ - - - - - - NA -
Cytology Clear cells; immunoblasts, HRS-like cells; inflammatory background TCRBCL-like appearance Sheets of large lymphoma cells with HRS-like morphology; geographic necrosis Mononuclear Hodgkin cells; rich inflammatory background HRS and HRS-like cells, including lacunar cells; rich inflammatory background
Diagnosis NLPHL/
NLPBL/
TCRBCL
EBV+ NLPHL FHTL mimicking LRCHL* MGZL PMBCL ALK+ ALCL, HL-like DLBCL,
anaplastic variant
CHL-like PMBL-like
Clinical presentation Lymphadenopathy Lymphadenopathy Lymphadenopathy Mediastinal mass Mediastinal mass Lymphadenopathy
EBV - + - - - -
PD-L1 IHC -(0%) uncertain - NA + ~ - (50%) + (76%) -(0%)
Other CD20+, PAX5+, CD30-, CD15-, BOB1+, OCT2+ Large cells: CD20-/+, PAX5+(dim), CD30+, CD15+, BOB1-, OCT2- CD20+, PAX5+, CD30+, CD15+/-, OCT2+/-, BOB1+/- CD20-/+, PAX5-/+, CD30+, CD15+/-, OCT2-/+, BOB1-/+ CD20+, PAX5+, CD30+, CD15+/-, CD23+, OCT2+, BOB1+, MUM1+ CD2-, PAX5-, CD30+, CD15-, ALK+, granzyme B+ CD20+, PAX5+, CD30+, CD15-, OCT2+, BOB1+
PD-1+ rosetting
T cells
+ - - - - -
Cytology Scattered large cells, including popcorn cells; rich inflammatory background HRS and HRS-like cells, including lacunar cells; rich inflammatory background Sheets of large lymphoma cells with HRS-like morphology Sheets of large lymphoma cells; sclerosis HRS-like cells, including hallmark cells; rich inflammatory background Sheets of large lymphoma cells with HRS-like morphology; sinusoidal pattern

Abbreviations: AITL/FHTL, angioimmunoblastic T-cell lymphoma/follicular helper T-cell lymphoma; HRS, Hodgkin and Reed-Sternberg; age-related EBV+ B-LPD, age-related EBV-associated B-cell lymphoproliferative disorder; DLBCL, diffuse large B-cell lymphoma; CHL, Classic Hodgkin lymphoma; GZL, gray zone lymphoma; MCCHL, mixed cellularity classic Hodgkin lymphoma; NSCHL, nodular sclerosis classic Hodgkin lymphoma; LDCHL, lymphocyte depletion classic Hodgkin lymphoma; LRCHL, lymphocyte-rich classic Hodgkin lymphoma; NLPHL/NLPBL/TCRBCL, nodular lymphcyte predominant Hodgkin lymphoma/nodular lymphocyte predominant B-cell lymphoma/T-cell/histiocyte-rich large B-cell lymphoma; MGZL, mediastinal grayzone lymphoma; PMBL, primary mediastinal large B-cell lymphoma; ALK; ALCL, HL-like, anaplastic lymphoma kinase+ anaplastic large-cell lymphoma, Hodgkin lymphoma-like; and NA, not available

*The literature is limited, consisting of single case reports.

†Not expressed in the confluent sheet of tumor cells.

Genetic alterations of PD-L1

Repeatedly, the most frequent genetic alteration of CHL is copy number gain of 9p24.1 at the locus that includes PD-L1/L2 and JAK2, constituting up to 97% of all CHL.1 Copy number gain of PD-L1/L2 increases its transcripts in HRS cells,7 and copy number gain of JAK2, leading to constitutive activation of JAK/STAT signals, also induces neoplastic PD-L1 expression in HRS cells.8 In a minority of cases of CHL (4/200, 5%), unbalanced translocations involving 9p24.1 have been found, and these might upregulate PD-L1 expression by stabilizing PD-L1 mRNA.9 The inactivating mutation of the Beta 2 microglobulin gene (B2M), one of the most prevalent gene mutations in CHL (up to 40%), also contributes to escape from immune surveillance by CD8+ T cells by limiting expression of major histocompatibility complex class I (MHC class I) on the cell surface.10,11 These immune-evasion-associated genetic alterations are more frequently observed in EBV+ CHL than in EBV-CHL, and presumably inhibit the T cell response to the virus-derived antigen of EBV+ CHL tumor cells.11

PD-L1 Immunostaining

In almost all CHL cases, HRS cells express CD30 and CD15, historically traditional, but not specific, diagnostic markers for CHL.3,12,13 PD-L1 is a novel diagnostic marker of CHL, and its expression is observed in 73~96% of the CHL.1,14-17 PD-L1 immunostaining is now also highlighted to be helpful for recognizing neoplastic cells in small biopsy samples.17 Its expression in HRS cells indicates the significance of immune evasion in the tumorigenesis. More importantly, blocking the PD-1/PD-L1 axis has been proven to be an efficient therapeutic approach for CHL,18,19 and PD-L1 expression level is associated with a good response to PD-1/PD-L1 inhibitors.20 The prevalence of PD-L1 expression is largely dependent on the histologic subtype. Almost all NSCHL cases show PD-L1 expression in neoplastic cells, whereas other histological subtypes express PD-L1 much less frequently, especially in LRCHL.6,21 The PD-L1 expression rate if neoplastic cells may be affected by the antibody clone used in studies.22 SP142 has been reported to have lower sensitivity compared with other clones such as 22C3, 28-8, and SP263. However, our group and Menter et al. showed staining concordance between SP142 and E1L3N clones.23,24 These antibodies’ binding affinity are also affected by binding epitope and assay protocol.22 It is notable that the recognition sites of SP142 and E1L3N can be genetically impaired by structural variations involving PD-L1 3′-UTR in non-Hodgkin lymphomas, and the prevalence of such genetic alterations in classic Hodgkin lymphoma has not yet been elucidated.25 Some subtypes of classic Hodgkin lymphoma, such as syncytial variant Hodgkin lymphoma, show heterogeneous PD-L1 expression in neoplastic cells, whose biological backgrounds remain elusive.26 Under these circumstances, there is no consensus on the cutoff value for PD-L1 expression and which antibodies should be used. In our studies, we used an SP142 antibody for PD-L1 expression. The diagnostic efficacy of SP142 was also recently documented in a review article by Sakakibara et al. (Table 1).27

EBV infection

The prevalence of EBV positivity varies greatly depending on the histologic subtype of CHLs. EBV infection is most frequently detected in up to 75% and 65% of MCCHL and LDCHL cases, respectively.28 NSCHL shows a relatively low range of EBV-positivity (10–25%), and around half of LRCHL cases have EBV infection.29 Intriguingly, NSCHL in the elderly has a higher frequency of EBV positivity (40%) than NSCHL in the young (10%), suggesting that EBV+ NSCHL may be more associated with immunosenescence.30 EBV infection may play a substitute role in genetically altered intracellular pathways, as EBV+ CHL have a much lower number of somatic mutations compared with EBV-CHL.11,31 For example, EBV+ CHL cases express EBV-encoded latent membrane protein 1 (LMP1), and LMP1 interacts with tumor necrosis factor receptor-associated factors and activates NF-κB signaling.32 As a consequence, EBV+ CHL is less reliant on genetic aberrations inducing constitutive active NF-κB signaling.33 In addition, another EBV-coded latent membrane protein, LMP2a, mimics B-cell receptor (BCR) signaling, presumably rescuing BCR-deficient B-cells in the course of CHL development.34

PD-L1 overexpression is mediated by EBV-latent membrane protein 1 (LMP1), which activates the transcription factor AP-1, and JAK/STAT and NF-κB pathways,8,35 besides structural variations involving CD274 (PD-L1)/ PDCD1LG2 (PD-L2)  described above.25 Interestingly, Nicoale et al. reported neoplastic PD-L1 expression in 95% of nodal EBV+ DLBCL young patients with a unique pattern of T-cell/histiocyte-rich large B-cells.36 In contrast to this, Takahara et al. recently reported a much lower frequency (11%) of neoplastic PD-L1 expression among elderly patients, suggesting that immune escape may be more important in younger patients than in older patients.23 The discrepancy in the positive percentages of neoplastic PD-L1 expression among patients with EBV+ DLBCL has been highlighted by others, and should be clarified in the future.37,38

Cellular origin of Hodgkin lymphoma

The cellular origin of CHL has been controversial for a long time, because HRS cells of CHL have a morphology and immunophenotype that does not fit any type of immune cells. Genetic analysis has revealed that HRS cells have a clonally rearranged Immunoglobulin (IG) gene in almost all CHL cases.39 Furthermore, a significant number of HRS cells have a somatic hypermutation in the IG gene locus.39,40 In around 30% of cases, IG gene rearrangements were rendered nonfunctional by the introduction of stop codons, deletions, or destructive promoter mutations generated within the GC.40 In the other cases, HRS cells frequently lack Ig gene transcription ability due to functional defects in the Ig gene regulatory elements.39 These genetic analyses indicate that HRS cells originate from GC cells capable of surviving without BCR signaling.41 It should be noted that a minority of CHL cases have clonal TCR gene rearrangement and/or express T-cell markers.42 Moreover, Karube et al. also recently reported adult T-cell leukemia/lymphoma with HTLV-1-infected Hodgkin and Reed-Sternberg-like cells.43 Additionally, these HRS-like cells also share the neoplastic PD-L1 expression beyond their non-B-cell phenotype. Whether these minority cases represent T-cell derived Hodgkin lymphoma, T-cell lymphomas mimicking CHL, or ‘B-cell’ lymphomas with aberrant phenotypic/genetic changes still needs to be researched, although it might be dependent on our definition of “Hodgkin neoplasm”. Tsuyama et al. more recently reported unique cases of Hodgkinoid histiocytosis with CHL-like morphologic features and CD30 and PD-L1 expression in Hodgkinoid histiocytes.44 This diagnostic term, “Hodgkinoid”, was created by Tsuyama et al. in order to avoid confusion with CHL, which is now defined as GC-derived B-cell neoplasm in the 2022 WHO classification. An international consensus should be achieved for these issues.

Diagnostic approach in routine pathologic practice

It is well known that HRS cells are diagnostic hallmarks of CHL, but are not specific, because RS-like cells are often found in other non-Hodgkin lymphomas and lymphoproliferative disorders.45 A precise histologic sub-classification is very challenging in the setting of small biopsy specimens. Even if a diagnosis of CHL without further subtype classification can be made, we recommend to assess the presence or absence of neoplastic PD-L1 positivity and/or EBV harboring in HRS cells and their lookalikes, which can be delineated into 4 subgroups; i.e., PD-L1+ EBV-, PD-L1+EBV+, PD-L1-EBV-, and PD-L1-EBV+ large cell subtypes.

In our routine practice, neoplastic PD-L1+ EBV- cases correspond approximately to NSCHL. Nodular growth pattern and the presence of fibrosis are regarded as defining morphologic features of NSCHL, but may be also be obscured in small samples. Lacunar cells are also characteristic of NSCHL and typically have small, but not large, nucleoli, which may ironically be difficult to identify as HRS cells.45 Differential diagnosis includes mediastinal grey zone lymphoma and ALK+ anaplastic large cell lymphoma (ALCL) (especially with a Hodgkin-like pattern), which share the neoplastic PD-L1+ EBV-negative large cells. Their differential diagnostic approaches were well discussed in a recent review by Elaine S. Jaffe’s group.5 Cytotoxic molecule (CM)-positive CHL in our studies usually shows neoplastic PD-L1 expression (unpublished data).46 This combination of PD-L1 and CM positivity may be shared by ALK-negative ALCL, except for cases with DUSP22 rearrangement which lack CM expression.47 As another diagnostic pitfall, the syncytial variant of CHL may be accompanied by reduced expression of PD-L1 in tumor cells.26,48 Conversely, three cases of nodal diffuse large B-cell lymphoma with neoplastic PD-L1 positivity, but without EBV association, have been reported by Kohno et al.49

Neoplastic PD-L1+EBV+ CHL is almost equivalent to EBV+ MC and LD subtypes with prototypic HRS cells characterized by large distinct nucleoli and embedded in a reactive immune cell-rich background. Never-the-less, EBV+ MCCHL poses differential diagnostic problems from EBV+DLBCL, especially in young patients36 and EBV+ grey zone lymphoma.50 Interestingly, the latter two were characterized by strong expression of B-cell markers and a greater number of mononuclear tumor cells often with a T-cell/histiocyte-rich large B-cell lymphoma-like pattern, although their patients’ age distributions are quite distinct. Some exceptional cases of EBV+ nodular lymphocyte-predominant Hodgkin lymphoma (NLPHL) have been reported, but in which PD-L1 information was not available.51 Notably, neoplastic PD-L1+EBV+ CHL may be accompanied by primary extranodal disease among immunocompromised patients, and should be distinguished from EBV+ mucocutaneous ulcers, which usually lack PD-L1 positivity.52 Neoplastic PD-L1+EBV+ CHL-type lesions may be seen post-transplant and in other iatrogenic lymphoproliferative disorders, and detailed information of the patients’ background should be requested.

Neoplastic PD-L1-EBV- HL includes NLPHL, LRCHL, and MCCHL without EBV association in our preliminary studies. As is well-known, differential diagnosis based on morphology alone in small-volume samples is highly challenging or impossible among NLPHL, LRCHL, and T-cell/histiocyte-rich large B-cell lymphoma. An adequate tissue biopsy is recommended, if necessary. Anaplastic variant of DLBCL also possesses the same phenotype as PD-L1-EBV- type tumor cells, but is characterized by sheet-like proliferation of large cells and/or HRS-like cells, which is distinct from ordinal CHL.53

Neoplastic PD-L1-EBV+ HRS-like cells are frequently encountered as bystander cells linked with reactivation of EBV in normal B-cells, but are hardly seen in our studies of CHL. Indeed, Kume et al. also recently reported that all EBV+ CHL cases consistently expressed PD-L1 in tumor cells.54 It is emphasized that EBV+ HRS-like cells, which are often accompanied by nodal peripheral T-cell lymphoma of mainly follicular helper T-cell phenotype, i.e., angioimmunoblastic T-cell lymphoma in many, generally lack immunohistochemical PD-L1 positivity, which was first highlighted by Eladl et al. in 2017.55

CONCLUSION

Diagnosis of CHL is challenging in small-volume samples. However, our knowledge/understanding of CHLs’ biology is now rapidly expanding. This will prompt us to re-define the diagnosis CHL from morphology to biology based. Future discussion is warranted.

Footnotes

CONFLICT OF INTEREST

There is no conflict of interest.

REFERENCES

  • 1.Roemer MGM, Advani RH, Ligon AH, et al. PD-L1 and PD-L2 genetic alterations define classical Hodgkin lymphoma and predict outcome. J Clin Oncol. 2016; 34: 2690-2697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Elsayed AA, Asano N, Ohshima K, et al. Prognostic significance of CD20 expression and Epstein-Barr virus (EBV) association in classical Hodgkin lymphoma in Japan: a clinicopathologic study. Pathol Int. 2014; 64: 336-345. [DOI] [PubMed] [Google Scholar]
  • 3.Swerdlow SH. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. Lyon, International Agency for Research on Cancer. 2017; pp. 424-442. [Google Scholar]
  • 4.Campo E, Jaffe ES, Cook JR, et al. The International Consensus Classification of Mature Lymphoid Neoplasms: a report from the Clinical Advisory Committee. Blood. 2022; 140: 1229-1253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Armitage JO, Gale RP, Jaffe ES. Lymphoma Nomenclature ‐ What’s in a name? Br J Haematol. 2022; 197: 539-543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Sakakibara A, Kohno K, Eladl AE, et al. Immunohistochemical assessment of the diagnostic utility of PD-L1: a preliminary analysis of anti-PD-L1 antibody (SP142) for lymphoproliferative diseases with tumour and non-malignant Hodgkin-Reed-Sternberg (HRS)-like cells. Histopathology. 2018; 72: 1156-1163. [DOI] [PubMed] [Google Scholar]
  • 7.Green MR, Monti S, Rodig SJ, et al. Integrative analysis reveals selective 9p24.1 amplification, increased PD-1 ligand expression, and further induction via JAK2 in nodular sclerosing Hodgkin lymphoma and primary mediastinal large B-cell lymphoma. Blood. 2010; 116: 3268-3277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Green MR, Rodig S, Juszczynski P, et al. Constitutive AP-1 activity and EBV infection induce PD-L1 in Hodgkin lymphomas and posttransplant lymphoproliferative disorders: implications for targeted therapy. Clin Cancer Res. 2012; 18: 1611-1618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Van Roosbroeck K, Ferreiro JF, Tousseyn T, et al. Genomic alterations of the JAK2 and PDL loci occur in a broad spectrum of lymphoid malignancies. Genes Chromosomes Cancer. 2016; 55: 428-441. [DOI] [PubMed] [Google Scholar]
  • 10.Liu Y, Abdul Razak FR, Terpstra M, et al. The mutational landscape of Hodgkin lymphoma cell lines determined by whole-exome sequencing. Leukemia. 2014; 28: 2248-2251. [DOI] [PubMed] [Google Scholar]
  • 11.Wienand K, Chapuy B, Stewart C, et al. Genomic analyses of flow-sorted Hodgkin Reed-Sternberg cells reveal complementary mechanisms of immune evasion. Blood Adv. 2019; 3: 4065-4080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Dürkop H, Latza U, Hummel M, et al. Molecular cloning and expression of a new member of the nerve growth factor receptor family that is characteristic for Hodgkin’s disease. Cell. 1992; 68: 421-427. [DOI] [PubMed] [Google Scholar]
  • 13.von Wasielewski R, Mengel M, Fischer R, et al. Classical Hodgkin’s disease. Clinical impact of the immunophenotype. Am J Pathol. 1997; 151: 1123-1130. [PMC free article] [PubMed] [Google Scholar]
  • 14.Carbone A, Gloghini A, Carlo-Stella C. Are EBV-related and EBV-unrelated Hodgkin lymphomas different with regard to susceptibility to checkpoint blockade? Blood. 2018; 132: 17-22. [DOI] [PubMed] [Google Scholar]
  • 15.Panjwani PK, Charu V, DeLisser M, et al. Programmed death-1 ligands PD-L1 and PD-L2 show distinctive and restricted patterns of expression in lymphoma subtypes. Hum Pathol. 2018; 71: 91-99. [DOI] [PubMed] [Google Scholar]
  • 16.Gerhard-Hartmann E, Goergen H, Bröckelmann PJ, et al. 9p24.1 alterations and programmed cell death 1 ligand 1 expression in early stage unfavourable classical Hodgkin lymphoma: an analysis from the German Hodgkin Study Group NIVAHL trial. Br J Haematol. 2022; 196: 116-126. [DOI] [PubMed] [Google Scholar]
  • 17.Volaric A, Bacchi CE, Gru AA. PD-1 and PD-L1 Immunohistochemistry as a diagnostic tool for classic Hodgkin lymphoma in small-volume biopsies. Am J Surg Pathol. 2020; 44: 1353-1366. [DOI] [PubMed] [Google Scholar]
  • 18.Venkataraman G, Song JY, Tzankov A, et al. Aberrant T-cell antigen expression in classical Hodgkin lymphoma is associated with decreased event-free survival and overall survival. Blood. 2013; 121: 1795-1804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Ansell SM, Lesokhin AM, Borrello I, et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N Engl J Med. 2015; 372: 311-319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Chen R, Zinzani PL, Fanale MA, et al. Phase II study of the efficacy and safety of pembrolizumab for relapsed/refractory classic Hodgkin lymphoma. J Clin Oncol. 2017; 35: 2125-2132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Aoki T, Chong LC, Takata K, et al. Single-cell profiling reveals the importance of CXCL13/CXCR5 axis biology in lymphocyte-rich classic Hodgkin lymphoma. Proc Natl Acad Sci USA. 2021; 118: e2105822118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Lawson NL, Dix CI, Scorer PW, et al. Mapping the binding sites of antibodies utilized in programmed cell death ligand-1 predictive immunohistochemical assays for use with immuno-oncology therapies. Mod Pathol. 2020; 33: 518-530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Takahara T, Satou A, Ishikawa E, et al. Clinicopathological analysis of neoplastic PD-L1-positive EBV+ diffuse large B cell lymphoma, not otherwise specified, in a Japanese cohort. Virchows Arch. 2021; 478: 541-552. [DOI] [PubMed] [Google Scholar]
  • 24.Menter T, Bodmer-Haecki A, Dirnhofer S, Tzankov A. Evaluation of the diagnostic and prognostic value of PDL1 expression in Hodgkin and B-cell lymphomas. Hum Pathol. 2016; 54: 17-24. [DOI] [PubMed] [Google Scholar]
  • 25.Kataoka K, Miyoshi H, Sakata S, et al. Frequent structural variations involving programmed death ligands in Epstein-Barr virus-associated lymphomas. Leukemia. 2019; 33: 1687-1699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Kohno K, Sakakibara A, Iwakoshi A, et al. Syncytial variant of classic Hodgkin lymphoma: four cases diagnosed with the aid of CD274/programmed cell death ligand 1 immunohistochemistry. Pathol Int. 2020; 70: 108-115. [DOI] [PubMed] [Google Scholar]
  • 27.Sakakibara A, Kohno K, Ishikawa E, et al. Diagnostic utility of programmed cell death ligand 1 (clone SP142) immunohistochemistry for malignant lymphoma and lymphoproliferative disorders: A brief review. J Clin Exp Hematop. 2021; 61: 182-191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Chang KC, Khen NT, Jones D, Su IJ. Epstein-Barr virus is associated with all histological subtypes of Hodgkin lymphoma in Vietnamese children with special emphasis on the entity of lymphocyte predominance subtype. Hum Pathol. 2005; 36: 747-755. [DOI] [PubMed] [Google Scholar]
  • 29.Lee JH, Kim Y, Choi JW, Kim YS. Prevalence and prognostic significance of Epstein-Barr virus infection in classical Hodgkin’s lymphoma: a meta-analysis. Arch Med Res. 2014; 45: 417-431. [DOI] [PubMed] [Google Scholar]
  • 30.Asano N, Kinoshita T, Ohshima K, et al. Clinicopathological features of nodular sclerosis-type classical Hodgkin lymphoma in the elderly: multicenter study of Hodgkin lymphoma in Japan. Blood. 2010; 116: 2677. [Google Scholar]
  • 31.Tiacci E, Ladewig E, Schiavoni G, et al. Pervasive mutations of JAK-STAT pathway genes in classical Hodgkin lymphoma. Blood. 2018; 131: 2454-2465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Huen DS, Henderson SA, Croom-Carter D, Rowe M. The Epstein-Barr virus latent membrane protein-1 (LMP1) mediates activation of NF-kappa B and cell surface phenotype via two effector regions in its carboxy-terminal cytoplasmic domain. Oncogene. 1995; 10: 549-560. [PubMed] [Google Scholar]
  • 33.Schmitz R, Hansmann ML, Bohle V, et al. TNFAIP3 (A20) is a tumor suppressor gene in Hodgkin lymphoma and primary mediastinal B cell lymphoma. J Exp Med. 2009; 206: 981-989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Mancao C, Hammerschmidt W. Epstein-Barr virus latent membrane protein 2A is a B-cell receptor mimic and essential for B-cell survival. Blood. 2007; 110: 3715-3721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Montes-Moreno S, Odqvist L, Diaz-Perez JA, et al. EBV-positive diffuse large B-cell lymphoma of the elderly is an aggressive post-germinal center B-cell neoplasm characterized by prominent nuclear factor-kB activation. Mod Pathol. 2012; 25: 968-982. [DOI] [PubMed] [Google Scholar]
  • 36.Nicolae A, Pittaluga S, Abdullah S, et al. EBV-positive large B-cell lymphomas in young patients: a nodal lymphoma with evidence for a tolerogenic immune environment. Blood. 2015; 126: 863-872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Xing W, Dresser K, Zhang R, et al. PD-L1 expression in EBV-negative diffuse large B-cell lymphoma: clinicopathologic features and prognostic implications. Oncotarget. 2016; 7: 59976-59986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Bourbon E, Maucort-Boulch D, Fontaine J, et al. Clinicopathological features and survival in EBV-positive diffuse large B-cell lymphoma not otherwise specified. Blood Adv. 2021; 5: 3227-3239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Marafioti T, Hummel M, Foss HD, et al. Hodgkin and Reed-Sternberg cells represent an expansion of a single clone originating from a germinal center B-cell with functional immunoglobulin gene rearrangements but defective immunoglobulin transcription. Blood. 2000; 95: 1443-1450. [PubMed] [Google Scholar]
  • 40.Kanzler H, Küppers R, Hansmann ML, Rajewsky K. Hodgkin and Reed-Sternberg cells in Hodgkin’s disease represent the outgrowth of a dominant tumor clone derived from (crippled) germinal center B cells. J Exp Med. 1996; 184: 1495-1505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Huntington ND, Xu Y, Puthalakath H, et al. CD45 links the B cell receptor with cell survival and is required for the persistence of germinal centers. Nat Immunol. 2006; 7: 190-198. [DOI] [PubMed] [Google Scholar]
  • 42.Asano N, Kinoshita T, Tamaru J, et al. Cytotoxic molecule-positive classical Hodgkin’s lymphoma: a clinicopathological comparison with cytotoxic molecule-positive peripheral T-cell lymphoma of not otherwise specified type. Haematologica. 2011; 96: 1636-1643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Karube K, Takatori M, Sakihama S, et al. Clinicopathological features of adult T-cell leukemia/lymphoma with HTLV-1-infected Hodgkin and Reed-Sternberg-like cells. Blood Adv. 2021; 5: 198-206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Tsuyama N, Noguchi M, Asaka R, et al. Hodgkinoid histiocytosis: an atypical nodal CD30 and S100 ‐positive histiocytosis with eosinophilia. Histopathology. 2022; 81: 371-379. [DOI] [PubMed] [Google Scholar]
  • 45.Parente P, Zanelli M, Sanguedolce F, Mastracci L, Graziano P. Hodgkin Reed-Sternberg-like cells in non-Hodgkin lymphoma. Diagnostics (Basel). 2020; 10: 1019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Asano N, Oshiro A, Matsuo K, et al. Prognostic significance of T-cell or cytotoxic molecules phenotype in classical Hodgkin’s lymphoma: a clinicopathologic study. J Clin Oncol. 2006; 24: 4626-4633. [DOI] [PubMed] [Google Scholar]
  • 47.Shen J, Li S, Medeiros LJ, et al. PD-L1 expression is associated with ALK positivity and STAT3 activation, but not outcome in patients with systemic anaplastic large cell lymphoma. Mod Pathol. 2020; 33: 324-333. [DOI] [PubMed] [Google Scholar]
  • 48.Zhang Q, Kim DH, Xu Y, Wang W, Medeiros LJ. Clinicopathological features of syncytial variant nodular sclerosis Hodgkin lymphoma. Hum Pathol. 2022; 119: 105-113. [DOI] [PubMed] [Google Scholar]
  • 49.Kohno K, Suzuki Y, Harada T, et al. Nodal diffuse large B‐cell lymphoma with neoplastic PD‐L1 positivity, but without EBV association: three cases highlighting an aspect of gray zone lymphoma. Pathol Int. 2020; 70: 695-697. [DOI] [PubMed] [Google Scholar]
  • 50.Elsayed AA, Satou A, Eladl AE, et al. Grey zone lymphoma with features intermediate between diffuse large B-cell lymphoma and classical Hodgkin lymphoma: a clinicopathological study of 14 Epstein-Barr virus-positive cases. Histopathology. 2017; 70: 579-594. [DOI] [PubMed] [Google Scholar]
  • 51.Huppmann AR, Nicolae A, Slack GW, et al. EBV may be expressed in the LP cells of nodular lymphocyte-predominant Hodgkin lymphoma (NLPHL) in both children and adults. Am J Surg Pathol. 2014; 38: 316-324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Daroontum T, Kohno K, Eladl AE, et al. Comparison of Epstein-Barr virus-positive mucocutaneous ulcer associated with treated lymphoma or methotrexate in Japan. Histopathology. 2018; 72: 1115-1127. [DOI] [PubMed] [Google Scholar]
  • 53.Megahed NA, Kohno K, Sakakibara A, et al. Anaplastic variant of diffuse large B‐cell lymphoma: reappraisal as a nodal disease with sinusoidal involvement. Pathol Int. 2019; 69: 697-705. [DOI] [PubMed] [Google Scholar]
  • 54.Kume A, Shinozaki-Ushiku A, Kunita A, Kondo A, Ushiku T. Enhanced PD-L1 expression in LMP1-positive cells of Epstein-Barr virus-associated malignant lymphomas and lymphoproliferative disorders: a single-cell resolution analysis with multiplex fluorescence immunohistochemistry and in situ hybridization. Am J Surg Pathol. 2022; 46: 1386-1396. [DOI] [PubMed] [Google Scholar]
  • 55.Eladl AE, Satou A, Elsayed AA, et al. Clinicopathological study of 30 cases of peripheral T-cell lymphoma with Hodgkin and Reed-Sternberg-like B-cells from Japan. Am J Surg Pathol. 2017; 41: 506-516. [DOI] [PubMed] [Google Scholar]

Articles from Journal of Clinical and Experimental Hematopathology : JCEH are provided here courtesy of Japanese Society for Lymphoreticular Tissue Research

RESOURCES