Home
JournalsCollections
For Authors For Reviewers For Editorial Board Members
Article Processing Charges Open Access
Ethics Advertising Policy
Editorial Policy Resource Center
Company Information Contact Us
OPEN ACCESS

Histopathologic Features and Differential Diagnosis in Challenging Cases of Nodular Lymphocyte Predominant B-cell Lymphoma/Nodular Lymphocyte Predominant Hodgkin Lymphoma

  • Yanna Ding and
  • Elaine S. Jaffe* 
Journal of Clinical and Translational Pathology   2024;4(2):61-69

doi: 10.14218/JCTP.2024.00015

Received:

Revised:

Accepted:

Published online:

 Author information

Citation: Ding Y, Jaffe ES. Histopathologic Features and Differential Diagnosis in Challenging Cases of Nodular Lymphocyte Predominant B-cell Lymphoma/Nodular Lymphocyte Predominant Hodgkin Lymphoma. J Clin Transl Pathol. 2024;4(2):61-69. doi: 10.14218/JCTP.2024.00015.

Abstract

Nodular lymphocyte predominant Hodgkin lymphoma was termed “nodular lymphocyte predominant B-cell lymphoma” in the International Consensus Classification (ICC), to emphasize clinical and biological differences from classic Hodgkin lymphoma (CHL). The abbreviation “NLP” represents both terms in the ICC and World Health Organization classifications. Variations in the growth pattern, originally reported as Fan patterns A-F, are designated as either grade 1 or grade 2 in the ICC. NLP is uncommon, and in some cases an accurate diagnosis is challenging. The objectives of this article were to review the histopathologic features of NLP and the differential diagnosis from other key entities including de novo T-cell/histiocyte-rich large B-cell lymphoma (THRLBL) and lymphocyte-rich classic Hodgkin lymphoma (LRCHL). Histologically, NLP Fan pattern E (THRLBL-like) can be indistinguishable from de novo THRLBL. However, focal nodular areas, clustering of tumor cells, presence of few admixed small B-cells or FDC meshworks, and T-cell rosettes favor NLP Fan pattern E and argue against de novo THRLBL. NLP may also be confused with LRCHL. Patients with NLP are younger than those with LRCHL, and LRCHL may show mediastinal involvement. In LRCHL, the nodular pattern often contains eccentrically located small regressed germinal centers and intact small dense FDC meshworks, in contrast to the expanded, and fragmented FDC meshworks in NLP. Neoplastic cells that are positive for CD30 and CD15 but negative for CD20 and CD79a are characteristic of LRCHL. Additionally, Fascin and Gata3 are commonly positive in LRCHL but usually negative in NLP.

Keywords

Nodular lymphocyte predominant Hodgkin lymphoma, Nodular lymphocyte predominant B-cell lymphoma, T-cell/Histiocyte rich large B-cell lymphoma, Lymphocyte-rich classic Hodgkin lymphoma, Immunohistochemistry, Epidemiology

Introduction

Nodular lymphocyte predominant Hodgkin lymphoma, alternatively termed nodular lymphocyte predominant B-cell lymphoma in the International Consensus Classification (ICC), is uncommon. Some cases can present challenges in diagnosis and may be difficult to differentiate from other entities with similar histologic features.1,2 The objectives of this article were to review the histopathologic features of this disease, abbreviated as NLP, and to provide some clues in its differentiation from de novo T-cell/histiocyte-rich large B-cell lymphoma (THRLBL) and lymphocyte-rich classic Hodgkin lymphoma (LRCHL).

History and terminology

Early on, NLP was identified as a B-cell neoplasm and later shown to be of germinal center derivation.3,4 It is characterized by either nodular or nodular and diffuse proliferation, containing scattered large mononucleated or multilobated neoplastic cells known as lymphocyte-predominant (LP) cells or popcorn cells.5,6 This entity was previously categorized as one of the variants of Hodgkin lymphoma, most likely due to the histologic features, including sparse neoplastic cells and abundant non-neoplastic cells in the background, similar to classic Hodgkin lymphoma (CHL). A proposal from the International Lymphoma Study Group first suggested a more formal separation of NLP from CHL in 1994.7 Subsequently, based on data indicating greater differences in clinical behavior and treatment approaches, the ICC took this a step further by recommending the removal of the eponymic term “Hodgkin”, from the disease, proposing the term “nodular lymphocyte predominant B-cell lymphoma” to emphasize emerging differences in clinical management for these two lymphomas of B-cell lineage.8 Currently, the term “nodular lymphocyte predominant Hodgkin lymphoma” remains unchanged in the 5th Edition of the World Health Organization classification. In the following discussion, NLP will be the abbreviation for both nodular lymphocyte predominant Hodgkin lymphoma and nodular lymphocyte predominant B-cell lymphoma.

Epidemiology and clinical features

NLP is more common in young patients (aged 30–50 years) with a male predominance and comprises 10% of all Hodgkin lymphomas.9 Patients usually present with localized lymphadenopathy initially, most commonly involving cervical, axillary, and inguinal lymph nodes or rarely mesenteric lymph nodes. Mediastinal and extranodal involvements are uncommon. Overall, the disease demonstrates an indolent clinical course and an excellent outcome with standard therapy.10,11 Systemic B symptoms are uncommon in the absence of advanced disease. Approximately 20% of patients can present with advanced-stage disease at diagnosis with involvement of the spleen, liver, or more rarely bone marrow and bone.12 Transformation of NLP into diffuse large B-cell lymphoma occurs in 5–10% of cases.11,13

Epstein-Barr virus (EBV) has been thought to play a significant role in the pathogenesis of some CHL subtypes. However, it is only rarely positive in NLP (< 5%).14,15 LP cells can be positive for IgD in a subgroup of patients, particularly young male patients with isolated neck or axillary lymphadenopathy.16–18 A subset of IgD+ cases were found to be associated with Moraxella catarrhalis and Rothia mucilaginosa in recent studies,19,20 suggesting a potential role for bacterial antigens in the stimulation of the B-cells that ultimately give rise to the LP cells of NLP.

Microscopy

NLP has a nodular growth pattern in most cases and a diffuse pattern in fewer cases (Figs. 1 and 2a). The initial lesion appears to arise within pre-existing reactive follicles. The follicular structures are usually larger than normal follicles and are associated with expanded follicular dendritic cell (FDC) meshworks. LP cells show a rim of pale cytoplasm, large twisted or multilobated nuclei with thin nuclear membranes, vesicular chromatin, and basophilic nucleoli, which are usually smaller than those in Hodgkin/Reed-Sternberg (HRS) cells (Fig. 2a). Multilobated LP cells have been likened to a “popped kernel of corn”, termed “popcorn cells”. Multinucleated cells may be observed. LP cells reside in an immune niche in a background of predominantly small lymphocytes with a variable number of histiocytes. Occasionally, significantly increased histiocytes, clusters of epithelioid histiocytes, or granulomas can be appreciated.

Schematic diagram of histopathologic patterns in NLP and relevant ICC grades.
Fig. 1  Schematic diagram of histopathologic patterns in NLP and relevant ICC grades.

Fan pattern A, so-called classical B-cell-rich nodular pattern: Clusters of single LP cells are embedded within enlarged follicles/nodules predominantly composed of small B-cells. Reactive T-cells are mainly in the interfollicular areas. The FDC meshworks are expanded and associated with the atypical follicles/nodules. Fan pattern B, so-called serpiginous/interconnected pattern: Similar to Fan pattern A, LP cells are mainly within B-cells rich atypical follicles/nodules, which interconnect with each other. Fan pattern C, so-called prominent extra-nodular LP cells pattern: LP cells extend beyond the follicular compartment with an internodular distribution. Fan pattern D, so-called T-cell-rich nodular pattern: LP cells distribute in a vaguely nodular pattern, but normal follicular B-cells are lost, and the atypical nodules are rich in T-cells. FDC meshworks are usually still present. Fan pattern E, so-called diffuse pattern (THRLBL or DLBCL-like): The growth pattern is diffuse. LP cells are widely scattered in a background rich in T-cells and histiocytes. FDC meshworks are lost and normal admixed B-cells are largely absent. Fan Pattern F, the so-called diffuse moth-eaten, B-cell-rich pattern: The LP cells are scattered in a background that is rich in both small B-cells and T-cells. The FDC meshworks are absent. Per ICC classification, Fan Patterns A, B, and C are categorized as grade 1, and Fan patterns D, E, and F are categorized as grade 2. DLBCL, diffuse large B-cell lymphoma; FDC, follicular dendritic cells; NLP, nodular lymphocyte predominant Hodgkin lymphoma/nodular lymphocyte predominant B-cell lymphoma; ICC, International Consensus Classification; LP, lymphocyte-predominant; THRLBL, T-cell/histiocytes-rich large B-cell lymphoma.

The histologic and immunohistochemical features of NLP.
Fig. 2  The histologic and immunohistochemical features of NLP.

(a–c) NLP Fan pattern A. (a) H&E stain section shows a vaguely nodular pattern with expanded atypical follicles lacking normal germinal centers. LP cells are largely confined to the follicular remnants (original magnification 4×); the insets show LP cells at high power (original magnification 80×). (b) The abnormal follicles are rich in CD20+ small B-cells (original magnification 4×); the inset shows LP cells at higher power (original magnification 40×). (c) Oct-2 highlights LP cells. Staining is strong in LP cells and weak in background small B-cells (original magnification 10×); the inset shows LP cells at higher power (original magnification 30×). (d) This case of NLP involving the tonsil contains LP cells that are positive for IgD (original magnification 10×); the inset at higher power (original magnification 30×). The background small B-cells are also positive for IgD. (e–g) NLP, Fan pattern A (same case as a–c). (e) CD21 highlights the nodular pattern delineated by the FDC meshworks (original magnification 2×). (f) CD3 and (g) PD-1 highlight T-cell rosettes, which surround the LP cells (arrows) (original magnification 30×). FDC, follicular dendritic cells; H&E, hematoxylin and eosin; LP, lymphocyte-predominant; NLP, nodular lymphocyte predominant Hodgkin lymphoma/nodular lymphocyte predominant B-cell lymphoma; Oct-2, Octamer-binding protein 2; PD-1, programmed cell death protein 1.

Various growth patterns of LP cells and the associated lymphocytic background were initially described by Fan et al. (Fig. 1).21 The most common growth patterns in NLP are Fan patterns A and B, comprising about 75% of cases. LP cells are scattered singly but commonly show vague clustering within the follicular structures/nodules of small B-cells with a few reactive T-cells. As the disease progresses, normal small B-cells decrease, while admixed T-cells become more abundant. In addition, Fan pattern C shows LP cells extending beyond the follicular compartment. Further loss of normal B-cells is evident in Fan pattern D. Evidence of follicular remnants disappears in Fan pattern E, which may be entirely diffuse. This THRLBL-like transformation is difficult to differentiate from de novo THRLBL. Fan pattern F also shows loss of follicular structures with a vaguely nodular background containing both B-cells and T-cells, lacking FDC meshworks (Fig. 1).

Immunohistochemical (IHC) stains are crucial to identify the various growth patterns, and more than one pattern can be observed in a single lymph node biopsy. The significance of the various growth patterns was recognized in ICC grading, with Fan patterns A, B, and C being grade 1, while Fan patterns D, E, and F considered grade 2.8,21 Patients with grade 2 disease may warrant treatment as diffuse large B-cell lymphoma, especially with advanced clinical stage.22

Reactive follicular hyperplasia, often with foci of progressive transformation of germinal centers (PTGC), may be seen in adjacent uninvolved nodal tissue. PTGC was reported to precede, follow, or occur concurrently with NLP. However, isolated PTGC is not a significant risk for subsequent NLP.23,24 The follicular structures in PTGC are markedly enlarged, largely as a consequence of the mantle zone expansion. The resultant follicle contains clusters of germinal center cells surrounded by IgD-positive cells. However, typical LP cells are not present. In addition, PTGC follicles are usually enriched for T follicular helper (TFH) cells stained by programmed cell death protein 1 (PD-1) and CD57. Therefore, an increase in TFH cells should not raise significant concern for involvement by NLP.

Immunophenotype

LP cells typically demonstrate strong expression of transcription factors Octamer-binding protein 2 (Oct-2) and B cell Oct binding protein 1 (Bob1), which are critical for functional immunoglobulin (IG) expression. They also maintain other B-cell associated markers such as CD20, CD79a, paired box protein 5 (Pax5), PU-1, and B-cell lymphoma 6 (Bcl-6) but are usually negative for multiple myeloma 1/interferon regulatory factor 4 (MUM-1/IRF4) and CD10 (Fig. 2b). In our experience, Oct-2 facilitates the detection of the neoplastic cells, especially when there are abundant small background B-cells. The background non-neoplastic B-cells show weak staining for Oct-2, as they are typically derived from the residual mantle cuffs (Fig. 2c). Pax5 is positive, but more weakly expressed in tumor cells than background B-cells. In addition, myocyte enhancer binding factor 2B (MEF2B) is expressed in LP cells.25–27 IgD was reported to be positive in 27% of cases, particularly affecting young male patients (Fig. 2d).16–18 LP cells are generally negative for EBV-encoded small RNA (EBER), with EBER rarely positive in 3–5% of cases.14,15,28–30 In some cases LP cells show downregulation or absence of some B-cell markers, such as CD20, Pax5, or CD79a,31 particularly in EBV+ cases.14

FDC markers such as CD21 and CD23 highlight the expanded and often fragmented FDC meshworks in the atypical nodules (Fig. 2e). NLP arises in preexistent follicles. Over time, the background normal B-cells disappear and are replaced by infiltrating T-cells. Most of the T-cells associated with the neoplastic LP cells exhibit a TFH phenotype and are strongly positive for PD-1/CD279. They form tight rosettes around the LP cells, which can help facilitate their detection (Fig. 2f–g). Prolonged close contact between LP cells and the surrounding PD-1+ T-cells was observed and may play a role in tumorigenesis.32,33 However, rosette formation with PD-1+ T-cells is not specific for NLP; it can be found in CHL but is usually less conspicuous.34,35 CD57+ cells are also common in the background of NLP.

Genetic findings

The LP cells possess functional IG rearrangements and express IG transcripts, which are more easily detected in DNA from isolated tumor cells or in tumor-rich samples. However, IG rearrangements are challenging to detect in whole tissue sections with sparse neoplastic cells and rich non-neoplastic cells in the microenvironment. LP cells typically exhibit a high load of somatic hypermutation in IGH variant regions with ongoing mutations, indicating the derivation from germinal center B-cells.4 FISH (fluorescence in situ hybridization) analysis may reveal gene rearrangement of BCL6 with partners including IG genes, IKAROS family genes, ABR, and others.36–38 Next generation sequencing study found mutations in PAX5, PIM1, RHOH (TTF), and MYC,39 as well as somatic mutations in SGK1, DUSP2, and JUNB in some cases of NLP.40 LP cells share mutations in JUNB, DUSP2, SGK1, and SOCS1 with THRLBL which suggests a close relationship between these two entities.41 Aberrations in TNFAIP3 or NFkBIA are rare in LP cells,42 unlike in HRS cells of CHL. Instead, the active NFkB signaling gene signature in LP cells results from different mechanisms.43–45 Familial NLP has been reported in patients with Hermansky-Pudlak syndrome type 2 and autoimmune lymphoproliferative syndrome with mutations in FAS.46,47

Challenging cases of NLP and differential diagnosis

Genetically, the tumor cells of NLP and THRLBL exhibit marked similarity, suggesting they are part of a single entity. The differential diagnosis between LRCHL and NLP can also be challenging in some cases. Herein, we discuss some helpful tips for accurate diagnosis in challenging cases of NLP.

NLP, Fan pattern E vs. THRLBL

NLP Fan pattern E shows sparse tumor cells dispersed in a rich inflammatory background composed largely of T-cells and histiocytes. Background FDC meshworks and nodular areas may be absent or only focally seen. The background T-cells are predominantly CD4+ T-cells, while CD8+ T-cells have been reported to be more frequent in THRLBL (Figs. 1 and 3).

The histologic and immunohistochemical features of THRLBL vs. NLP Fan pattern E.
Fig. 3  The histologic and immunohistochemical features of THRLBL vs. NLP Fan pattern E.

(a–c) Representative images of THRLBL. (a) H&E stain section of THRLBL involving the spleen (original magnification 20×); the insets show neoplastic cells with pleomorphism (original magnification 80×). (b) CD20 highlights the widely scattered large neoplastic cells with the absence of background small B-cells in a case of THRLBL involving the bone marrow (original magnification 20×). (c) CD3 shows the T-cell-rich background (original magnification 20×). (d–f) Representative images of NLP Fan pattern E. (d) NLP Fan pattern E involving a lymph node (H&E) (original magnification 20×). (e) LP cells forming loose clusters are highlighted by CD20 (original magnification 4×); the inset shows LP cells at high power; a few small B-cells are present in the background (original magnification 30×). (f) CD4 highlights background T-cells forming rosettes around the LP cells (arrows) (original magnification 30×). H&E, hematoxylin and eosin; LP, lymphocyte-predominant; NLP, nodular lymphocyte predominant Hodgkin lymphoma/nodular lymphocyte predominant B-cell lymphoma; THRLBL, T-cell/histiocytes-rich large B-cell lymphoma.

THRLBL is considered an aggressive B-cell lymphoma with sparse large tumor cells scattered in a background of abundant T-cells and/or histiocytes. It most frequently affects middle-aged or older male patients, whereas NLP often affects relatively younger patients, including children. Patients with THRLBL typically present with advanced disease with B symptoms, bulky lymphadenopathy, and/or hepatosplenomegaly. Systemic involvement is much more common than in NLP and frequently involves the spleen, liver, bone marrow, and bone, in addition to lymph nodes.

Tumor cells in THRLBL carry clonal IG gene rearrangement and high levels of somatic mutations. The genomic profile is similar to that of NLP, further supporting a close relationship.48,49

Histologically, tumor cells in THRLBL are evenly dispersed in a background of diffuse T-cells with a variable number of histiocytes. The tumor cells may exhibit variations in size or pleomorphism, and lobated nuclei can be present in some cases (Fig. 3a). A nodular growth pattern is not appreciated on H&E-stained sections. Focal residual nodules, if present, confirm NLP Fan pattern E and argue against de novo THRLBL. Other subtle differences may be helpful. In THRLBL, the background usually contains more histiocytes and fine fibrosis, whereas fibrosis is less often encountered in lymph nodes involved by NLP. Necrosis is also more commonly found in THRLBL.

The immunophenotype of the tumor cells in THRLBL is similar to that of NLP (Fig. 3b). They are negative for CD15, CD30, and EBER in most cases. By flow cytometry, the phenotype of the neoplastic cells in THRLBL and NLP is nearly identical, limiting the utility of flow cytometry for differential diagnosis.50,51 Nevertheless, in THRLBL, the background lymphocytes are more often CD8+ T-cells with an absence of small B-cells (Fig. 3b–c).52 These T-cells show strong activation by 3D measurement.53 PD-1+ T-cell rosettes surrounding tumor cells are generally absent. FDC meshworks are absent as well. In contrast, LP cells in NLP Fan pattern E may still show vague clusters, best highlighted by CD20 and Oct-2 (Fig. 3d–e). In addition, a few residual scattered small B-cells, traces of FDC meshworks, and T-cell rosettes all favor the diagnosis of NLP Fan pattern E (Fig. 3e–f).

In some cases, a clear distinction between THRLBL and NLP Fan pattern E may not be possible. However, if NLP is observed in another site or at the time of recurrence, the diagnosis of NLP may be confirmed.

Morphologic and IHC features of the two entities are summarized in Table 1.

Table 1

Differential diagnosis of NLP: Summary of histopathologic and IHC features

THRLBLNLPLRCHL
Neoplastic cellsMorphologic featuresSparse, widely dispersed, variable degrees of pleomorphism. May resemble centroblasts or immunoblastsSparse, multilobated nuclei, thin nuclear membranes, basophilic nucleoli. Tend to form loose clusters. May be intrafollicular or interfollicular.Sparse; may resemble lacunar cells or classic HRS cells. Most common in the marginal zone surrounding regressed follicles.
CD30−/+Negative to weakly +Strongly +
CD15−/rare ++/−
EBERRarely EBV+EBV + (15–20%)
EMAvariable−/+
Pan-B cell markers++−/weak and variable +
MUM-1−/+−/++, strong
Oct-2+, strong+, strong−/+
Pax5++ (may be weak)+, weak
OthersN/APositive for MEF2B;
may be positive for STAT6 in rare cases
Often positive for Fascin, Gata-3, and STAT6
Non-neoplastic BackgroundMorphologic featuresDiffuse lymphocytic background with increased histiocytes and fine fibrosisNodular or diffuse lymphocytic background. Variable histiocytes.Mainly nodular pattern. Regressed follicles present. Rare to absence of eosinophils, neutrophils and plasma cells. Diffuse pattern less common
CD20Absence of small B-cellsBackground B-cells nearly always presentIntact follicles
CD4/CD8Often CD8> CD4Mainly CD4+Mainly CD4+
T-cell (PD-1+) rosettesAbsentPresentSometimes seen
CD21/CD23AbsentExpanded FDC meshworksCompact tight intact FDC meshworks

NLP vs. LRCHL

LRCHL is a subtype of CHL with scattered HRS cells in a background rich in small lymphocytes and usually lacking other inflammatory cells, such as plasma cells and eosinophils. It is less common than NLP and usually affects older patients compared to NLP and other CHL subtypes, showing a male predominance. Patients with LRCHL show an indolent clinical course, and B symptoms are rare. The disease mainly affects peripheral lymph nodes. Mediastinal involvement is less common than in nodular sclerosis CHL but can be present, while mediastinal disease is rare in NLP. The prognosis of LRCHL is somewhat better than other CHL subtypes.12

Histologically, the most common form of LRCHL exhibits a nodular growth pattern (Fig. 4a–f).1,54 The follicles contain regressed germinal centers and intact mantle zones. CD21 or CD23 highlights small, dense, compact, and intact FDC meshworks associated with the eccentrically located regressed germinal centers (Fig. 4b). The HRS cells are best observed in the marginal zone, surrounding the regressed follicles. This pattern contrasts with what is seen in NLP, where the LP cells are scattered within the altered follicles (Fig. 4g–l). LRCHL with a diffuse pattern is rare and probably closer to the mixed cellularity subtype of CHL.

The histologic and immunohistochemical features of LRCHL vs. EBV+ NLP.
Fig. 4  The histologic and immunohistochemical features of LRCHL vs. EBV+ NLP.

(a–f) Representative images of LRCHL, EBV+. (a) H&E stain section of LRCHL involving a lymph node (original magnification 30×); the insets show HRS cells at higher power (original magnification 40× and 80×). (b) CD21 shows the regressed lymphoid follicles and tight FDC meshworks (original magnification 2×); the inset shows eccentrically located compact FDC meshworks (arrows), which are associated with regressed germinal centers (original magnification 20×). (c) CD30 highlights the HRS cells with strong and homogeneous staining mainly in the interfollicular and marginal zone areas; the eccentrically located regressed germinal center is highlighted by arrows (original magnification 15×); the insets show HRS cells at higher power (original magnification 40×). (d) MUM-1 is positive in HRS cells with strong staining (red arrows), outside the small regressed germinal center (black arrows) (original magnification 10×); the inset shows HRS cells at higher power (original magnification 40×). (e) Dim staining for Pax5 in HRS cells (red arrows) (original magnification 40×). (f) HRS cells positive for EBER (original magnification 4×, inset 40×). (g–l): Representative images of EBV+ NLP Fan pattern A. (g) CD20 shows downregulated CD20 expression with variable staining in LP cells (original magnification 10×, inset 40×). (h) Pax5 expression is downregulated in LP cells (red arrows) (original magnification 10×, inset 40×). (i) CD30 highlights clusters of LP cells with weak and variable staining (original magnification 10×, inset 40×). (j) EBER highlights clusters of LP cells (original magnification 4×, inset 40×). (k) Oct-2 highlights LP cells with strong staining, in contrast to weaker staining in background small B-cells (original magnification 20×). The inset (CD3) shows T-cell rosettes around LP cells (original magnification 30×). (l) CD21 shows expanded and fragmented FDC meshworks with irregular contours (original magnification 2×). EBER, EBV-encoded small RNA; EBV, Epstein-Barr virus; FDC, follicular dendritic cells; H&E, hematoxylin and eosin; HRS, Hodgkin/Reed-Sternberg; LP, lymphocyte-predominant; LRCHL, lymphocyte-rich classic Hodgkin lymphoma; MUM-1, multiple myeloma 1/interferon regulatory factor 4; NLP, Nodular lymphocyte predominant Hodgkin lymphoma/nodular lymphocyte predominant B-cell lymphoma; Oct-2, Octamer-binding protein 2; Pax5, paired box protein 5.

The HRS cells in LRCHL may resemble “lacunar” type HRS cells with ample cytoplasm and sometimes multilobated nuclei (Fig. 4a). They can also resemble LP cells. The HRS cells exhibit the typical immunophenotype of CHL with strong staining for CD30, CD15 (subset), MUM-1, and dim expression of Pax5 (Fig. 4c–e). CD20 and CD79a are most often negative but can be variably expressed. Occasionally, HRS cells in this subtype can express variable B-cell transcription factors such as Oct-2, Bob-1, and Bcl-6. EBER and latent membrane protein 1 (LMP1) can be positive or negative (Fig. 4f). It should be noted that LP cells in NLP can be rarely positive for CD30 or CD15.21,55–57 However, if positive, CD30 staining tends to be weak in LP cells (Fig. 4i). EBV-positive NLP is another rare finding (Fig. 4j).14,15 In reported cases, Oct-2 usually remains strongly expressed. (Fig. 4k)

Other markers reported in the differential diagnosis of LRCHL and NLP include fascin, GATA-binding protein 3 (Gata-3), and STAT6. These antigens are more likely to be positive in the HRS cells of CHL than in LP cells.58–61 The specificity of STAT6 expression in HRS cells is controversial.61,62 In addition, HRS cells also show strong staining with MUM-1, generally negative in LP cells. While epithelial membrane antigen has been reported to be positive in LP cells, the incidence of truly positive cases is low. A recent study showed that light chain restriction can be demonstrated by ultrasensitive in situ hybridization for kappa and lambda in LP cells but not in HRS cells.63 PD-1+/CD57+ T-cell rosettes around HRS cells can be present but are usually less common than in NLP (Fig. 3k, inset).

Specific studies focusing on the genomic features of HRS cells in LRCHL have not been published. In other work, the failure of HRS cells to express IG transcripts has been partially attributed to a deficiency in the transcription factors Oct-2 and Bob-1. It is of interest that these transcription factors are more often positive in LRCHL.55 Whether this impacts Ig expression in the neoplastic cells is unknown.

The genomic profile of HRS cells differs from that of LP cells in NLP and may provide clues for their distinction if available. HRS cells show alterations in both the NF-kappa B and JAK/STAT pathways.64–66 These include mutations of SOCS1.67–70 The other frequent genetic abnormalities include gains of 2p, 9p, 16p, 17q, 19q, and 20q, losses of 6q, 11q, and 13q, as well as aberrations of 9p24.1 leading to amplification of JAK2 and increased expression of PD-1 ligands.71,72 Inactivating B2M mutations, as well as driver mutations in BCL7A, GNA13, and PTPN1, are also detected in CHL.73,74

Morphologic and IHC features of the two entities are summarized in Table 1.

Conclusions

NLP is an uncommon form of lymphoma. In recent years, there has been a greater appreciation of its distinctive clinical and biological features, with emphasis on major differences from CHL. This information has led to changes in the clinical management of NLP. For these reasons, an accurate diagnosis is of major importance to the practicing pathologist. The lymph node biopsy should always be examined with knowledge of the clinical history. Key diagnostic features include the nature of the neoplastic cells, as well as the characteristic microenvironment. In addition, ancillary techniques, including immunohistochemistry, can lead to the correct interpretation in most cases.

Declarations

Acknowledgement

None.

Funding

This work was supported by the Intramural Research Program of the Center for Cancer Research, National Cancer Institute (ZIA SC000550).

Conflict of interest

The authors declare no competing financial interest.

Authors’ contributions

Manuscript writing and editing (YD, ESJ). Both authors have made a significant contribution to this study and have approved the final manuscript.

References

  1. Anagnostopoulos I, Hansmann ML, Franssila K, Harris M, Harris NL, Jaffe ES, et al. European Task Force on Lymphoma project on lymphocyte predominance Hodgkin disease: histologic and immunohistologic analysis of submitted cases reveals 2 types of Hodgkin disease with a nodular growth pattern and abundant lymphocytes. Blood 2000;96(5):1889-1899 View Article PubMed/NCBI
  2. Rets AV, Gottesman SR. Nodular Lymphocyte Predominant Hodgkin Lymphoma versus T-Cell/Histiocyte-Rich Large B-Cell Lymphoma: A Diagnostic Challenge. Case Rep Pathol 2014;2014:956217 View Article PubMed/NCBI
  3. Pinkus GS, Said JW. Hodgkin’s disease, lymphocyte predominance type, nodular—further evidence for a B cell derivation. L & H variants of Reed-Sternberg cells express L26, a pan B cell marker. Am J Pathol 1988;133(2):211-217 View Article PubMed/NCBI
  4. Marafioti T, Hummel M, Anagnostopoulos I, Foss HD, Falini B, Delsol G, et al. Origin of nodular lymphocyte-predominant Hodgkin’s disease from a clonal expansion of highly mutated germinal-center B cells. N Engl J Med 1997;337(7):453-458 View Article PubMed/NCBI
  5. Stein H, Swerdlow SH, Gascoyne RD, Poppema S, Jaffe ES, Pileri SA. WHO Classification of Tumorus of Haematopoietic And Lymphoid Tissues. Revised 4th ed. Lyon, France: IARC; 2017, 431-434 View Article PubMed/NCBI
  6. Feldman AL, Poppema S. Hematopathology. 2nd ed. Philidelphia, PA: Elsevier; 2017, 507-524 View Article PubMed/NCBI
  7. Mason DY, Banks PM, Chan J, Cleary ML, Delsol G, de Wolf Peeters C, et al. Nodular lymphocyte predominance Hodgkin’s disease. A distinct clinicopathological entity. Am J Surg Pathol 1994;18(5):526-530 View Article PubMed/NCBI
  8. Campo E, Jaffe ES, Cook JR, Quintanilla-Martinez L, Swerdlow SH, Anderson KC, et al. The International Consensus Classification of Mature Lymphoid Neoplasms: a report from the Clinical Advisory Committee. Blood 2022;140(11):1229-1253 View Article PubMed/NCBI
  9. Laurent C, Do C, Gourraud PA, de Paiva GR, Valmary S, Brousset P. Prevalence of Common Non-Hodgkin Lymphomas and Subtypes of Hodgkin Lymphoma by Nodal Site of Involvement: A Systematic Retrospective Review of 938 Cases. Medicine (Baltimore) 2015;94(25):e987 View Article PubMed/NCBI
  10. Eichenauer DA, Engert A. How I treat nodular lymphocyte-predominant Hodgkin lymphoma. Blood 2020;136(26):2987-2993 View Article PubMed/NCBI
  11. Miettinen M, Franssila KO, Saxen E. Hodgkin’s disease, lymphocytic predominance nodular. Increased risk for subsequent non-Hodgkin’s lymphomas. Cancer 1983;51(12):2293-2300 View Article PubMed/NCBI
  12. Diehl V, Sextro M, Franklin J, Hansmann ML, Harris N, Jaffe E, et al. Clinical presentation, course, and prognostic factors in lymphocyte-predominant Hodgkin’s disease and lymphocyte-rich classical Hodgkin’s disease: report from the European Task Force on Lymphoma Project on Lymphocyte-Predominant Hodgkin’s Disease. J Clin Oncol 1999;17(3):776-783 View Article PubMed/NCBI
  13. Al-Mansour M, Connors JM, Gascoyne RD, Skinnider B, Savage KJ. Transformation to aggressive lymphoma in nodular lymphocyte-predominant Hodgkin’s lymphoma. J Clin Oncol 2010;28(5):793-799 View Article PubMed/NCBI
  14. Huppmann AR, Nicolae A, Slack GW, Pittaluga S, Davies-Hill T, Ferry JA, 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(3):316-324 View Article PubMed/NCBI
  15. Wang S, Medeiros LJ, Xu-Monette ZY, Zhang S, O’Malley DP, Orazi A, et al. Epstein-Barr virus-positive nodular lymphocyte predominant Hodgkin lymphoma. Ann Diagn Pathol 2014;18(4):203-209 View Article PubMed/NCBI
  16. Prakash S, Fountaine T, Raffeld M, Jaffe ES, Pittaluga S. IgD positive L&H cells identify a unique subset of nodular lymphocyte predominant Hodgkin lymphoma. Am J Surg Pathol 2006;30(5):585-592 View Article PubMed/NCBI
  17. Ali N, Moussa E, Khorshed E, Zaghloul MS, Elnashar A, Abdalla A. Variant histology of pediatric nodular lymphocyte-predominant Hodgkin lymphoma with IgD and CD30 expression. Pediatr Blood Cancer 2023;70(11):e30647 View Article PubMed/NCBI
  18. Senchenko MA, Konovalov DM. [IgD expression in various immunoarchitectural patterns of nodular lymphocyte predominant Hodgkin lymphoma in children]. Arkh Patol 2024;86(1):21-26 View Article PubMed/NCBI
  19. Thurner L, Hartmann S, Fadle N, Regitz E, Kemele M, Kim YJ, et al. Lymphocyte predominant cells detect Moraxella catarrhalis-derived antigens in nodular lymphocyte-predominant Hodgkin lymphoma. Nat Commun 2020;11(1):2465 View Article PubMed/NCBI
  20. Thurner L, Fadle N, Regitz E, Roth S, Cetin O, Kos IA, et al. B-cell receptor reactivity against Rothia mucilaginosa in nodular lymphocyte-predominant Hodgkin lymphoma. Haematologica 2023;108(12):3347-3358 View Article PubMed/NCBI
  21. Fan Z, Natkunam Y, Bair E, Tibshirani R, Warnke RA. Characterization of variant patterns of nodular lymphocyte predominant hodgkin lymphoma with immunohistologic and clinical correlation. Am J Surg Pathol 2003;27(10):1346-1356 View Article PubMed/NCBI
  22. Hartmann S, Eichenauer DA, Plutschow A, Mottok A, Bob R, Koch K, et al. The prognostic impact of variant histology in nodular lymphocyte-predominant Hodgkin lymphoma: a report from the German Hodgkin Study Group (GHSG). Blood 2013;122(26):4246-4252 View Article PubMed/NCBI
  23. Hicks J, Flaitz C. Progressive transformation of germinal centers: review of histopathologic and clinical features. Int J Pediatr Otorhinolaryngol 2002;65(3):195-202 View Article PubMed/NCBI
  24. Sadanand A, Aljudi A, Bergsagel DJ, Keller FG. A single-center retrospective review of pediatric cases of progressive transformation of germinal centers. Pediatr Blood Cancer 2023;70(6):e30283 View Article PubMed/NCBI
  25. Moore EM, Swerdlow SH, Gibson SE. J chain and myocyte enhancer factor 2B are useful in differentiating classical Hodgkin lymphoma from nodular lymphocyte predominant Hodgkin lymphoma and primary mediastinal large B-cell lymphoma. Hum Pathol 2017;68:47-53 View Article PubMed/NCBI
  26. Krenacs D, Borbenyi Z, Bedekovics J, Mehes G, Bagdi E, Krenacs L. Pattern of MEF2B expression in lymphoid tissues and in malignant lymphomas. Virchows Arch 2015;467(3):345-355 View Article PubMed/NCBI
  27. Torabi A, Fromm JR, Naresh KN. MEF2B is the ideal immunohistochemical marker to highlight neoplastic LP cells in nodular lymphocyte-predominant Hodgkin lymphoma. EJHaem 2023;4(2):517-519 View Article PubMed/NCBI
  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(7):747-755 View Article PubMed/NCBI
  29. Hummel M, Anagnostopoulos I, Dallenbach F, Korbjuhn P, Dimmler C, Stein H. EBV infection patterns in Hodgkin’s disease and normal lymphoid tissue: expression and cellular localization of EBV gene products. Br J Haematol 1992;82(4):689-694 View Article PubMed/NCBI
  30. Murray PG, Young LS, Rowe M, Crocker J. Immunohistochemical demonstration of the Epstein-Barr virus-encoded latent membrane protein in paraffin sections of Hodgkin’s disease. J Pathol 1992;166(1):1-5 View Article PubMed/NCBI
  31. Menke JR, Spinner MA, Natkunam Y, Warnke RA, Advani RH, Gratzinger DA. CD20-Negative Nodular Lymphocyte-Predominant Hodgkin Lymphoma: A 20-Year Consecutive Case Series From a Tertiary Cancer Center. Arch Pathol Lab Med 2021;145(6):753-758 View Article PubMed/NCBI
  32. Bein J, Thurner L, Hansmann ML, Hartmann S. Lymphocyte predominant cells of nodular lymphocyte predominant Hodgkin lymphoma interact with rosetting T cells in an immunological synapse. Am J Hematol 2020;95(12):1495-1502 View Article PubMed/NCBI
  33. Hartmann S, Scharf S, Steiner Y, Loth AG, Donnadieu E, Flinner N, et al. Landscape of 4D Cell Interaction in Hodgkin and Non-Hodgkin Lymphomas. Cancers (Basel) 2021;13(20):5208 View Article PubMed/NCBI
  34. Churchill HR, Roncador G, Warnke RA, Natkunam Y. Programmed death 1 expression in variant immunoarchitectural patterns of nodular lymphocyte predominant Hodgkin lymphoma: comparison with CD57 and lymphomas in the differential diagnosis. Hum Pathol 2010;41(12):1726-1734 View Article PubMed/NCBI
  35. Nam-Cha SH, Roncador G, Sanchez-Verde L, Montes-Moreno S, Acevedo A, Dominguez-Franjo P, et al. PD-1, a follicular T-cell marker useful for recognizing nodular lymphocyte-predominant Hodgkin lymphoma. Am J Surg Pathol 2008;32(8):1252-1257 View Article PubMed/NCBI
  36. Atayar C, Kok K, Kluiver J, Bosga A, van den Berg E, van der Vlies P, et al. BCL6 alternative breakpoint region break and homozygous deletion of 17q24 in the nodular lymphocyte predominance type of Hodgkin’s lymphoma-derived cell line DEV. Hum Pathol 2006;37(6):675-683 View Article PubMed/NCBI
  37. Renne C, Martin-Subero JI, Hansmann ML, Siebert R. Molecular cytogenetic analyses of immunoglobulin loci in nodular lymphocyte predominant Hodgkin’s lymphoma reveal a recurrent IGH-BCL6 juxtaposition. J Mol Diagn 2005;7(3):352-356 View Article PubMed/NCBI
  38. Wlodarska I, Stul M, De Wolf-Peeters C, Hagemeijer A. Heterogeneity of BCL6 rearrangements in nodular lymphocyte predominant Hodgkin’s lymphoma. Haematologica 2004;89(8):965-972 View Article PubMed/NCBI
  39. Liso A, Capello D, Marafioti T, Tiacci E, Cerri M, Distler V, et al. Aberrant somatic hypermutation in tumor cells of nodular-lymphocyte-predominant and classic Hodgkin lymphoma. Blood 2006;108(3):1013-1020 View Article PubMed/NCBI
  40. Hartmann S, Schuhmacher B, Rausch T, Fuller L, Doring C, Weniger M, et al. Highly recurrent mutations of SGK1, DUSP2 and JUNB in nodular lymphocyte predominant Hodgkin lymphoma. Leukemia 2016;30(4):844-853 View Article PubMed/NCBI
  41. Schuhmacher B, Bein J, Rausch T, Benes V, Tousseyn T, Vornanen M, et al. JUNB, DUSP2, SGK1, SOCS1 and CREBBP are frequently mutated in T-cell/histiocyte-rich large B-cell lymphoma. Haematologica 2019;104(2):330-337 View Article PubMed/NCBI
  42. Schumacher MA, Schmitz R, Brune V, Tiacci E, Doring C, Hansmann ML, et al. Mutations in the genes coding for the NF-kappaB regulating factors IkappaBalpha and A20 are uncommon in nodular lymphocyte-predominant Hodgkin’s lymphoma. Haematologica 2010;95(1):153-157 View Article PubMed/NCBI
  43. Hartmann S, Eichenauer DA. Nodular lymphocyte predominant Hodgkin lymphoma: pathology, clinical course and relation to T-cell/histiocyte rich large B-cell lymphoma. Pathology 2020;52(1):142-153 View Article PubMed/NCBI
  44. Jardin F. NFkB Pathway and Hodgkin Lymphoma. Biomedicines 2022;10(9):2153 View Article PubMed/NCBI
  45. Straus DJ. Progress in understanding the biology of nodular lymphocyte-predominant Hodgkin lymphoma. Haematologica 2021;106(10):2538 View Article PubMed/NCBI
  46. Lorenzi L, Tabellini G, Vermi W, Moratto D, Porta F, Notarangelo LD, et al. Occurrence of nodular lymphocyte-predominant hodgkin lymphoma in hermansky-pudlak type 2 syndrome is associated to natural killer and natural killer T cell defects. PLoS One 2013;8(11):e80131 View Article PubMed/NCBI
  47. Straus SE, Jaffe ES, Puck JM, Dale JK, Elkon KB, Rosen-Wolff A, et al. The development of lymphomas in families with autoimmune lymphoproliferative syndrome with germline Fas mutations and defective lymphocyte apoptosis. Blood 2001;98(1):194-200 View Article PubMed/NCBI
  48. Brune V, Tiacci E, Pfeil I, Doring C, Eckerle S, van Noesel CJ, et al. Origin and pathogenesis of nodular lymphocyte-predominant Hodgkin lymphoma as revealed by global gene expression analysis. J Exp Med 2008;205(10):2251-2268 View Article PubMed/NCBI
  49. Hartmann S, Doring C, Jakobus C, Rengstl B, Newrzela S, Tousseyn T, et al. Nodular lymphocyte predominant hodgkin lymphoma and T cell/histiocyte rich large B cell lymphoma—endpoints of a spectrum of one disease?. PLoS One 2013;8(11):e78812 View Article PubMed/NCBI
  50. Ally F, Gajzer D, Fromm JR. A Review of the Flow Cytometric Findings in Classic Hodgkin Lymphoma, Nodular Lymphocyte Predominant Hodgkin Lymphoma and T Cell/Histiocyte-Rich Large B Cell Lymphoma. Clin Lab Med 2023;43(3):427-444 View Article PubMed/NCBI
  51. Glynn E, Fromm JR. Immunophenotypic Characterization and Purification of Neoplastic Cells from Lymph Nodes Involved by T-Cell/Histiocyte-rich Large B-cell Lymphoma by Flow Cytometry and Flow Cytometric Cell Sorting. Cytometry B Clin Cytom 2020;98(1):88-98 View Article PubMed/NCBI
  52. Boudova L, Torlakovic E, Delabie J, Reimer P, Pfistner B, Wiedenmann S, et al. Nodular lymphocyte-predominant Hodgkin lymphoma with nodules resembling T-cell/histiocyte-rich B-cell lymphoma: differential diagnosis between nodular lymphocyte-predominant Hodgkin lymphoma and T-cell/histiocyte-rich B-cell lymphoma. Blood 2003;102(10):3753-3758 View Article PubMed/NCBI
  53. Sadeghi Shoreh Deli A, Scharf S, Steiner Y, Bein J, Hansmann ML, Hartmann S. 3D analyses reveal T cells with activated nuclear features in T-cell/histiocyte-rich large B-cell lymphoma. Mod Pathol 2022;35(10):1431-1438 View Article PubMed/NCBI
  54. Ashton-Key M, Thorpe PA, Allen JP, Isaacson PG. Follicular Hodgkin’s disease. Am J Surg Pathol 1995;19(11):1294-1299 View Article PubMed/NCBI
  55. Nam-Cha SH, Montes-Moreno S, Salcedo MT, Sanjuan J, Garcia JF, Piris MA. Lymphocyte-rich classical Hodgkin’s lymphoma: distinctive tumor and microenvironment markers. Mod Pathol 2009;22(8):1006-1015 View Article PubMed/NCBI
  56. Quintanilla-Martinez L, de Jong D, de Mascarel A, Hsi ED, Kluin P, Natkunam Y, et al. Gray zones around diffuse large B cell lymphoma. Conclusions based on the workshop of the XIV meeting of the European Association for Hematopathology and the Society of Hematopathology in Bordeaux, France. J Hematop 2009;2(4):211-236 View Article PubMed/NCBI
  57. Venkataraman G, Raffeld M, Pittaluga S, Jaffe ES. CD15-expressing nodular lymphocyte-predominant Hodgkin lymphoma. Histopathology 2011;58(5):803-805 View Article PubMed/NCBI
  58. Bhargava P, Pantanowitz L, Pinkus GS, Pinkus JL, Paessler ME, Roullet M, et al. Utility of fascin and JunB in distinguishing nodular lymphocyte predominant from classical lymphocyte-rich Hodgkin lymphoma. Appl Immunohistochem Mol Morphol 2010;18(1):16-23 View Article PubMed/NCBI
  59. Kezlarian B, Alhyari M, Venkataraman G, Karner K, Inamdar KV, Menon MP. GATA3 Immunohistochemical Staining in Hodgkin Lymphoma: Diagnostic Utility in Differentiating Classic Hodgkin Lymphoma From Nodular Lymphocyte Predominant Hodgkin Lymphoma and Other Mimicking Entities. Appl Immunohistochem Mol Morphol 2019;27(3):180-184 View Article PubMed/NCBI
  60. Salama ME, Rajan Mariappan M, Inamdar K, Tripp SR, Perkins SL. The value of CD23 expression as an additional marker in distinguishing mediastinal (thymic) large B-cell lymphoma from Hodgkin lymphoma. Int J Surg Pathol 2010;18(2):121-128 View Article PubMed/NCBI
  61. Van Slambrouck C, Huh J, Suh C, Song JY, Menon MP, Sohani AR, et al. Diagnostic utility of STAT6(YE361) expression in classical Hodgkin lymphoma and related entities. Mod Pathol 2020;33(5):834-845 View Article PubMed/NCBI
  62. Hartmann S, Eichenauer DA, Plutschow A, Mottok A, Bob R, Koch K, et al. Histopathological features and their prognostic impact in nodular lymphocyte-predominant Hodgkin lymphoma—a matched pair analysis from the German Hodgkin Study Group (GHSG). Br J Haematol 2014;167(2):238-242 View Article PubMed/NCBI
  63. Kaseb H, Wang Z, Cook JR. Ultrasensitive RNA In Situ Hybridization for Kappa and Lambda Light Chains Assists in the Differential Diagnosis of Nodular Lymphocyte-predominant Hodgkin Lymphoma. Am J Surg Pathol 2022;46(8):1078-1083 View Article PubMed/NCBI
  64. Hinz M, Lemke P, Anagnostopoulos I, Hacker C, Krappmann D, Mathas S, et al. Nuclear factor kappaB-dependent gene expression profiling of Hodgkin’s disease tumor cells, pathogenetic significance, and link to constitutive signal transducer and activator of transcription 5a activity. J Exp Med 2002;196(5):605-617 View Article PubMed/NCBI
  65. Hinz M, Loser P, Mathas S, Krappmann D, Dorken B, Scheidereit C. Constitutive NF-kappaB maintains high expression of a characteristic gene network, including CD40, CD86, and a set of antiapoptotic genes in Hodgkin/Reed-Sternberg cells. Blood 2001;97(9):2798-2807 View Article PubMed/NCBI
  66. Hopken UE, Foss HD, Meyer D, Hinz M, Leder K, Stein H, et al. Up-regulation of the chemokine receptor CCR7 in classical but not in lymphocyte-predominant Hodgkin disease correlates with distinct dissemination of neoplastic cells in lymphoid organs. Blood 2002;99(4):1109-1116 View Article PubMed/NCBI
  67. Joos S, Granzow M, Holtgreve-Grez H, Siebert R, Harder L, Martin-Subero JI, et al. Hodgkin’s lymphoma cell lines are characterized by frequent aberrations on chromosomes 2p and 9p including REL and JAK2. Int J Cancer 2003;103(4):489-495 View Article PubMed/NCBI
  68. Melzner I, Weniger MA, Menz CK, Moller P. Absence of the JAK2 V617F activating mutation in classical Hodgkin lymphoma and primary mediastinal B-cell lymphoma. Leukemia 2006;20(1):157-158 View Article PubMed/NCBI
  69. Skinnider BF, Elia AJ, Gascoyne RD, Patterson B, Trumper L, Kapp U, et al. Signal transducer and activator of transcription 6 is frequently activated in Hodgkin and Reed-Sternberg cells of Hodgkin lymphoma. Blood 2002;99(2):618-626 View Article PubMed/NCBI
  70. Weniger MA, Melzner I, Menz CK, Wegener S, Bucur AJ, Dorsch K, et al. Mutations of the tumor suppressor gene SOCS-1 in classical Hodgkin lymphoma are frequent and associated with nuclear phospho-STAT5 accumulation. Oncogene 2006;25(18):2679-2684 View Article PubMed/NCBI
  71. Green MR, Monti S, Rodig SJ, Juszczynski P, Currie T, O’Donnell E, 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(17):3268-3277 View Article PubMed/NCBI
  72. Roemer MG, Advani RH, Ligon AH, Natkunam Y, Redd RA, Homer H, et al. PD-L1 and PD-L2 Genetic Alterations Define Classical Hodgkin Lymphoma and Predict Outcome. J Clin Oncol 2016;34(23):2690-2697 View Article PubMed/NCBI
  73. Reichel J, Chadburn A, Rubinstein PG, Giulino-Roth L, Tam W, Liu Y, et al. Flow sorting and exome sequencing reveal the oncogenome of primary Hodgkin and Reed-Sternberg cells. Blood 2015;125(7):1061-1072 View Article PubMed/NCBI
  74. Maura F, Ziccheddu B, Xiang JZ, Bhinder B, Rosiene J, Abascal F, et al. Molecular Evolution of Classic Hodgkin Lymphoma Revealed Through Whole-Genome Sequencing of Hodgkin and Reed Sternberg Cells. Blood Cancer Discov 2023;4(3):208-227 View Article PubMed/NCBI