The diffuse aggressive large B-cell lymphomas are a heterogeneous group of B-cell malignancies. Although many are readily recognized due to characteristic clinical and pathologic features, several problematic areas still exist in diagnosis of these lymphomas due to a variety of reasons that include imprecise or difficult-to-apply diagnostic criteria, gaps in our understanding of lymphoma biology, and limitations in technologies available in the clinical laboratory compared to the research laboratory. This may result in some degree of confusion in the pathology report, particularly if the issues are not clearly explained, leading to frustration or misinterpretation on the part of the reader. In this review, I will discuss the pathologic features of a subset of the WHO 2008 classification diffuse aggressive large B-cell lymphomas, focusing on areas in which difficulties exist in diagnosis and/or biomarker marker assessment. A deeper understanding of the issues and areas of uncertainty due to limitations in our knowledge about the biology of these diseases should lead to better communication between pathologists and clinicians.

Learning Objectives
  • Recognize clinical and pathologic features of primary mediastinal large B-cell lymphomas and mediastinal gray zone lymphomas

  • Be familiar with the concept of EBV-positive B-cell lymphomas in non-immunocompromised patients

  • Understand the concepts of cell of origin in diffuse large B-cell lymphoma and differentiate between the double-hit and double-expresser lymphomas

The diffuse aggressive large B-cell lymphomas are a heterogeneous group of B-cell malignancies. Recognized entities in the WHO 2008 classification are shown in Table 1.1  Distinctive entities with characteristic and specific pathologic and/or clinical features have been recognized, and include entities, such as plasmablastic lymphoma, primary effusion lymphoma, lymphomatoid granulomatosis, and intravascular lymphoma. Application of relatively unambiguous diagnostic criteria in combination with clinical presentations typically allow specific diagnoses. However, several problematic areas exist due to a variety of reasons that include imprecise or difficult-to-apply diagnostic criteria, gaps in our understanding of lymphoma biology, and limitations in technologies available in the clinical laboratory compared to the research laboratory (in which entities or biomarkers were defined). This may result in some degree of confusion in the pathology report, particularly if the issues are not clearly explained. In this review, I will discuss the pathologic features of a subset of the WHO 2008 classification diffuse aggressive large B-cell lymphomas, focusing on areas in which difficulties exist in diagnosis and/or biomarker marker assessment. In particular, I will focus on primary mediastinal large B-cell lymphoma, B-cell lymphoma, unclassifiable with features intermediate between diffuse large B-cell lymphoma and Hodgkin lymphoma, EBV+ diffuse large B-cell lymphoma of the elderly, diffuse large B-cell lymphoma not otherwise specified (DLBCL, nos), and B-cell lymphoma, unclassifiable, with features intermediate between diffuse large B-cell lymphoma and Burkitt lymphoma.

Table 1.

DLBCL subtypes of entities (WHO 2008)

DLBCL subtypes of entities (WHO 2008)
DLBCL subtypes of entities (WHO 2008)

PMBL is an uncommon lymphoma, accounting for 2.4% of NHLs.2  It typically occurs in young adults (median age 32 years) with a female predominance and presents as a bulky mediastinal mass that can invade local structures including lung, pleura, and pericardium. Spread to regional lymph nodes including cervical and supraclavicular nodes may be seen. Thus, most patients (75%) present with stage I or II disease. Because of the mass effect, patients may suffer from superior vena cava syndrome. Other common features include presence of pleural effusion and B-symptoms. Although spread to distant sites may occur, involvement of the bone marrow is very uncommon.3-5  The putative cell of origin is the thymic B-cell and gene expression profiling studies have demonstrated a PMBL gene signature, supporting the consideration of this lymphoma as a distinct entity.6,7 

Histologically, the pattern is diffuse but this lymphoma may have a varied appearance. The lymphoma is composed of medium-to-large sized cells with round to lobulated nuclei, some of which may resemble Hodgkin cells. The cells often have moderate to abundant amounts of pale cytoplasm. There is frequently a delicate, compartmentalizing fibrosis. The immunophenotype is that of a mature B-cell expressing PAX5, CD19, CD20, and CD79a. CD10, BCL6, and MUM1 are variably expressed and CD21 is absent. BCL2 is often expressed (80%).8-10  CD23, a marker expressed on normal thymic B cells, is expressed in up to 70% of PMBL cases.11  CD30 is also commonly expressed but is generally weak and heterogeneous in pattern.12  Although OCT2, BOB.1 and PU.1 are consistently expressed, PMBL cases often lack detectable immunoglobulin by flow cytometry.10  Unfortunately none of these markers are specific for PMBL and may be expressed in DLBCL, nos. MAL, a gene involved in lipid raft organization, apical transport in epithelial cells, and T-cell signaling is also overexpressed in PMBL and immunohistochemical staining shows expression in 54%-70% of cases, whereas <5% of diffuse large B-cell lymphomas express MAL protein.13-15  EBV is only rarely, if ever, present in PMBL.16 BCL2, BCL6, and MYC rearrangements are not features of PMBL.17-19 

Gene expression profiling studies not only demonstrated that PMBL was distinct from DLBCL nos but also that the profile and deregulated pathways showed similarities to classical Hodgkin lymphoma (cHL).7,17  This has implications both in diagnosis and overlap cases to be discussed shortly.

Genetic studies have demonstrated recurrent structural genetic changes to include gains of chromosome 9p24 in up to 79% of cases (JAK2, PDL1, and PDL2), 2p in ∼50% of cases (REL and BCL11A), Xp11.4-21 and Xq24-26 in ∼30%.20-22  Other studies have also implicated copy number alterations or mutations in JMJD2C, SOCS1, BCL11A, STAT6, TNFAIP3, MYC, and TP53, or epigenetic changes (promoter hypermethylation) in CDKN2A.18,23-29  These alterations can be generally organized into specific pathways related NFκB activation, JAK/STAT signaling, transcriptional regulation, cell cycle/TP53 pathway, and histone modification.30  More recently, recurrent translocations involving CIITA, a master regulator of HLA class II expression regulation, have been discovered in 38% of PMBL cases, and help explain decreased HLA class II expression seen in PMBL.31  Thus, avoiding immune surveillance may be a theme in PMBL. Indeed PDL1 and PDL2 have been identified as partner genes in CIITA translocations, the end result of which is to overexpress these PD1 receptor ligands under the control of the CIITA promoter.30,31  This is not the only mechanism for increased expression PDL1 and PDL2 because the above mentioned gains in chromosome 9p have been shown to result in increased expression of these molecules.23,32,33 

Although much as been learned about the pathobiology of PMBL, application to everyday diagnostic practice has lagged. Assessment for gene expression signatures or rearrangements of CIITA, PDL1, and PDL2 have not made their way to the clinical laboratory due to factors including technological limitations or lack of commercially available reagents. Furthermore, although rearrangements in CIITA, PDL1, and PDL2 appear to be relatively specific for PMBL, sensitivity is modest. Thus, a major diagnostic dilemma faced by pathologists is distinguishing PMBL from DLBCL, nos presenting with a mediastinal mass. Pathologists still rely on classic PMBL pathologic findings of a diffuse infiltrate of large, often multilobulated, cells with variable amounts of pale cytoplasm, set in a fine compartmentalizing fibrotic stroma. Immunophenotypic clues favoring PMBL include variable expression of CD30 and CD23 and these markers, in association with other mature B-cell markers, and exclusion of other specific types of diffuse aggressive B-cell lymphomas (such as Burkitt lymphoma, lymphoblastic lymphoma, or transformed follicular lymphoma), and classical Hodgkin lymphoma (to be discussed in the next section) should allow a specific and definitive diagnosis in many cases when considering the constellation of clinical and pathologic features. To further complicate the issue, cases of diffuse large B-cell lymphoma have been identified that have the gene expression signature of PMBL despite the fact that there is no detectable mediastinal mass.34  Four of 6 such cases had pathologic features of PMBL such as fine sclerosis or variable expression of CD30 and CD23. However, one was felt to histologically be a DLBCL, whereas the other case had gray zone features between cHL and PMBL.34  Thus, one may now consider the possibility of a “non-mediastinal PMBL.”

Newer markers are now being examined and may further enhance one's ability to specifically and positively identify PMBL from other diffuse aggressive large B-cell lymphoma, notably DLBCL, nos presenting with a mediastinal mass. Among the proposed useful markers are expression of MAL, PDL2, and CD200, and coexpression of cytoplasmic TRAF1 and nuclear cREL.14,35-37  Given the current lack of suitable commercially available MAL and PDL2 antibodies and difficulties in cREL staining (as well as reported modest diagnostic sensitivity of 53% for cREL and TRAF1 coexpression), CD200 may be more promising but requires verification in other centers. Of note, we are currently evaluating the performance of a commercially available MAL antibody for use in identifying PMBL. Due to the lack of highly sensitive and specific phenotypic markers, pathology reports may reflect a degree of diagnostic uncertainty in some cases of suspected PMBL. Close communication between pathologists and clinicians may be required to agree on the most appropriate diagnosis.

Because PMBL and cHL share biologic features at the molecular level, it stands to reason that some degree of diagnostic overlap might exist in which pathologic features are intermediate between the two entities. Indeed cases with sequential NHL and cHL, composite lymphomas, and cases with features of both a DLBCL and cHL have long been recognized.38-40  The WHO 2008 classification recognized the existence of cases that might have pathologic features intermediate between these two entities, commonly referred to as mediastinal gray zone lymphoma (MGZL).1  These cases, occur in younger patients with a median age of 20-40 years and a male predominance. Initial descriptions recognized 2 groups of cases.41  The first are those in which the initial impression is that of PMBL. However, occasional RS-like cells or nodules may be present. Additionally, phenotypic features may be atypical such as weak/absent CD20, expression of CD15, and weak expression of B-cell transcription factors such as PAX5, OCT2, and BOB.1, which are reminiscent of Reed–Sternberg cells. Alternatively, other cases resemble nodular sclerosis Hodgkin lymphoma but have features, such as strong CD20 expression, expression of CD79a and/or CD45, lack of the characteristic mixed inflammatory background, lack of necrosis, and have increased large mononuclear Hodgkin-like cells that are more in keeping with PMBL.41,42  Thus, there is a histologic spectrum of cytologic appearances in the same case, which can also vary in different areas of the tumor, and mixed immunophenotypic signals.

This theme of overlap also extends to molecular features since common genetic abnormalities such as gains in chromosome 2p, 9p, 16p, and 8q are found in both cHL and PMBL. Interestingly abnormalities in these same loci have been found in MGZLs.43  FISH studies showed gains or amplifications in 2p16.1 (BCL11/REL) in 33% of cases, gains of 8q24 (MYC) in 27%, and gains or amplifications of 9p24.1 (JAK2/PDL2). CIITA probes showed gain or amplification in 10% and translocation in 27% of cases.43  Of note, the frequency of translocation is intermediate between the 38% described for PMBL and 15% for cHL.31  Given the fact that metachronous cases of PMBL and cHL have occurred, suggesting some degree of “reprogramming” of the cells, one could surmise that epigenetic mechanisms might be relevant in this process. Methylation profiling of PMBL, cHL, and MGZL cases has shown distinct profiles for each type and a 22-gene methylation class prediction model was developed. Interestingly, composite PMBL/cHL cases (even when microdissected and individual components analyzed separately) maintained the profile of MGZL rather than splitting into PMBL and cHL components.44  In aggregate, these data appear to provide evidence and rationale for the relatedness of these lymphomas and support for identifying MGZL as a separable entity within the group.

As can now be understood, pathologists and clinicians are faced with the issue of imprecise diagnostic criteria for this overlap entity. In practice, pathologists should reserve the diagnosis of MGZL for cases that truly send mixed/hybrid signals; namely, morphology suggested one entity (PMBL or cHL) but phenotypic data more in keeping with the other, ideally supported by more than one aberrant marker. For example, cHL morphology with strong expression of CD20 and robust coexpression of B-cell associated transcription factors OCT-2 and BOB.1 or cases with PMBL morphology with expression of CD15 and downregulated OCT-2 or BOB.1. Likewise, minor morphologic variation, such as the occasional RS-like cell in typical PMBL, should not be the sole reason for designating a case as MGZL. Clearly stating the data supporting the intermediate features in the report will help the clinician understand the reasoning for arriving at the diagnosis. Recognizing that composite cases can occur, MGZL designation should not be used for such composite lymphoma cases.42  Of note, the methylation profiling data might suggest that these composite cases, from a clinical perspective, could be managed as a MGZL case. Finally, one should avoid a definitive diagnosis of MGZL in a needle biopsy given the inherent morphologic spectrum of these cases and need to exclude composite lymphomas in this setting.

EBV-positive DLBCL of the elderly as defined in the WHO 2008 occurs in adults over the age of 50 who have no known immunodeficiency or other specific EBV+ lymphoproliferative disorder, such as lymphomatoid granulomatosis or EBV+ DLBCL associated with chronic inflammation. It is believed to be related to deterioration of the immune system that may occur in elderly patients (immune senescence). It comprises 8%-10% of DLBCL in Asian countries but is likely lower (3%) in the United States.45-47  Most patients (80%) present with lymphadenopathy. Extranodal involvement may occur and often involves skin, lung, tonsil, and stomach. Elevated LDH (58%) and B symptoms (49%) are common.48  This lymphoma is aggressive and has a poor prognosis compared to EBV-negative tumors and prognostic factors include presence of B symptoms and age >70 years.46 

Pathologically, cases show architectural effacement by an atypical large cell infiltrate. The cells often show a morphologic heterogeneity, with a polymorphous infiltrate demonstrating varying degrees of plasmacytic differentiation. Large centroblasts, immunoblasts, and multilobated Reed–Sternberg-like cells can be seen. Necrosis is also a characteristic feature. Immunophenotyping shows expression of CD20 and other pan-B cell markers such as CD79a. A non-germinal center B-cell phenotype with MUM1 expression is often seen and reflects the plasmacytic maturation.45,46  By definition EBV (best assessed by EBER staining) is positive in the vast majority of cells. CD30 is expressed in a high proportion of cases (75%).46  RS-like cells may express CD15 and show decreased CD20 expression, but usually retain expression of PAX5 and OCT2, unlike true RS-cells in which OCT2 is downregulated. Thus, classical Hodgkin lymphoma may become a consideration. Features that favor an age-related EBV-positive lymphoproliferative disorders/lymphoma include extranodal involvement, lack of the typical mixed inflammatory background of Hodgkin lymphoma, and the variation in cell morphology expressing CD20, CD30, and EBER.

Other EBV+ lymphoproliferative disorders (expanding the spectrum of disease)

The spectrum of EBV-positive lymphoproliferative disorders has been expanded to include reactive and neoplastic processes, described are reactive lymphoid hyperplasia, polymorphic extranodal and nodal LPDs, and DLBCL. Polymorphic extranodal LPDs were divided into isolated lesions and lesions that fit descriptions of EBV+ mucocutaneous ulcer.49  This latter lesion is a relatively recently described entity in which an EBV+ superficial ulcerating lesion involving gastrointestinal tract, skin, and oropharyngeal mucosa occurs. It is composed of a polymorphous infiltrate with a spectrum of cells to include atypical large B-blasts and Reed–Sternberg-like cells. Despite the cytologically atypical appearance, prognosis is excellent with conservative therapy.49,50  Although the polymorphic extranodal LPDs had a good prognosis (5 year disease-specific survival of 93%), the nodal polymorphic LPDs and DLBCL cases had poor outcomes (5 year disease-specific survival of 57% and 25%).49  Distinguishing features for DLBCL include lack of the full spectrum of plasmacytic differentiation, less variation in cell size, and marked pleomorphism compared with nodal polymorphic LPDs.

Immunoglobulin heavy chain gene rearrangements are seen more frequently in EBV-positive polymorphic nodal lymphoproliferative disorders and DLBCL categories (56%-63%) compared to reactive hyperplasia and extranodal lymphoproliferative disorders (16%-33%). Interestingly T-cell receptor gamma gene rearrangements were detected in 27%-50% of reactive hyperplasia and extranodal lymphoproliferative disorders and 15%-24% of polymorphic nodal lymphoproliferative disorders and DLBCL cases.49  The presence of T-cell clones is felt to reflect the limited T-cell repertoire in the immunesenescent setting.51-54 

It has been recognized that the somewhat arbitrary age limitation of 50 years may not be appropriate. A series of 46 cases was reported in patients <45 years of age. The median age was 23 years with a male–female ratio of 3.6. All patients presented with lymphadenopathy and only 11% had extranodal disease. Interestingly the histopathology was most commonly that of T-cell histiocyte-rich large B-cell lymphoma (78%), mediastinal gray zone lymphoma (15%), and DLBCL (7%). Tumor cells often expressed CD30, as well as PDL1 and IDO, the latter markers suggesting a local immunosuppressive or immune tolerant microenvironment.55,56 

This entity has altered practice and increased the necessary work-up in large B-cell lymphomas. Very little data exists on how frequently EBV is assessed by pathologists in cases of diffuse aggressive B-cell lymphoma. However, given the prognostic implications, assessment should be routinely performed. Fortunately, there are known histopathologic and phenotype clues that can focus efforts at EBV detection. These include a polymorphous infiltrate with plasmacytic differentiation, presence of Hodgkin-like cells, necrosis, and non-germinal center B-cell phenotype. Understanding the spectrum of EBV-positive LPDs is important so that hyperplastic processes and extranodal polymorphic lesions, particularly EBV-positive mucocutaneous ulcer are not overinterpreted as overt lymphoma.49  On the other hand, understanding that EBV-positive DLBCL in the older adult population and polymorphic LPDs in lymph nodes that resemble polymorphic post-transplant lymphomas are aggressive B-cell lymphomas will prompt consideration of appropriate conventional immunochemotherapy and, perhaps in the future, strategies to improve immunity toward EBV. Recognizing advances in our understanding of EBV-driven B-cell LPDs may affect nomenclature in the anticipated WHO update. Cases of EBV+ DLBCL of the elderly may be designated as EBV+ DLBCL nos, without reference to age of the patient.

The diagnosis of DLBCL, nos is generally straight forward, but currently there is a degree of uncertainty with regard to the types of studies for risk assessment that should be done and what techniques to use. Pathologist's tasks are twofold when making a diagnosis of DLBCL, nos. The first is to exclude other large B-cell lymphoma entities and subtypes of DLBCL (Table 1), whereas the second is to perform biomarker assessment that might yield important prognostic or risk assessment information. The pathology report should contain enough relevant information for both these purposes and we will concern ourselves with these two issues.

Other forms of diffuse aggressive B-cell lymphomas, such as PMBL, intravascular lymphoma, DLBCL associated with chronic inflammation, lymphomatoid granulomatosis, ALK+ large B-cell lymphoma, oral plasmablastic lymphoma, and HHV8-related lymphomas (primary effusion lymphoma and lymphomas occurring in the setting of multicentric Castleman disease) are usually distinguishable from DLBCL nos by a constellation of characteristic clinical and pathologic features. Because of the great importance of clinical setting on these diagnoses, pathologists must actively attempt to understand the clinical setting and clinicians should make every attempt to inform their pathologists about relevant clinical information, such as location/distribution of disease, HIV status, or other potential causes of immunocompromise. In addition, pathologists must use sufficient ancillary studies to exclude these subtypes and other forms of lymphoma. Discretion of course can be used, with certain markers done secondarily when other primary markers suggest a particular lymphoma type is more likely to be in the differential diagnosis based on the clinical, histologic, and phenotypic features. For example, ALK does not need to be routinely assessed unless the proper morphology (immunoblastic) and plasmablastic phenotype are present. Likewise, other entities that may be confused with DLBCL, nos should be excluded, such as pleomorphic blastoid mantle cell lymphoma. Table 2 shows an example diagnostic panel and conditions in which markers might be secondarily assessed. Use of such a panel with described results in the body of the report should allow the pathologist to convey the differential diagnostic considerations of the case and the supporting evidence for the final diagnosis.

Table 2.

Example diffuse aggressive B-cell lymphoma marker panel

Example diffuse aggressive B-cell lymphoma marker panel
Example diffuse aggressive B-cell lymphoma marker panel

Plasmablastic phenotype: CD20−/CD138+/PAX5−/MUM1+.

COO indicates cell of origin; PEL, primary effusion lymphoma; LBCL, large B-cell lymphoma; LPD, lymphoproliferative disorder; DH, “double-hit”; HRS, Hodgkin and Reed–Sternberg; PMBL, primary mediastinal large B-cell lymphoma; and FDC, follicular dendritic cell.

One can see that the intensity of the work-up, even for something as seemingly straight forward as DLBCL, nos, is substantial and far greater than required in years past. Fortunately, the availability of these tests (whether done on-site or through reference laboratory services) in formalin-fixed paraffin-embedded tissues (FFPET) now enables any pathologist to have these studies performed. Unfortunately, we are still limited by the amount and quality of tissues provided by the biopsy. With the proliferation of guided needle core biopsies, the amount of tissue available for diagnosis is drastically decreased. In instances where architectural assessment is important, necrosis is present, or tissues are partially involved (common scenarios include mediastinal tumors and assessment of gray zone lymphomas, question of underlying follicular lymphoma, DLBCL versus grade 3 follicular lymphoma, EBV+ LPDs including mononucleosis), excisional biopsies may be required. However, many studies have suggested that needle core biopsy is a reasonable first option that can result in an actionable diagnosis in a majority of cases. The degree of acceptable result varies from study to study depending on whether an actionable result was defined as the outcome or whether definitive WHO subtype was required. Variability also derives from whether cases were limited to those in which lymphoma was present or a wider variety of diagnoses including reactive conditions were allowed. However, general themes are that close cooperation with hematopathologists, incorporation of ancillary testing, such as flow cytometry, immunohistochemistry, and molecular studies, as well as larger size and greater number of cores, optimize diagnostic yield.57-62  Fine-needle aspirate cytology should not be used for primary diagnosis of lymphoma.63 

Biology and biomarkers in DLBCL, nos

Over the past 15 years, our understanding of the biology of DLBCL nos has improved and we now recognize the biologic heterogeneity of DLBCL nos that underlies and helps explain some of the clinical heterogeneity in terms of response and outcomes to modern therapies. Landmark gene expression profiling studies have defined several signatures that relate DLBCL nos to normal cell counterparts (cell of origin; COO).64-67  In particular, cases can be related to that of a germinal center B-cell (GCB) or an in vitro activated B-cell (ABC). In addition to elucidating some of the biological heterogeneity of DLBCL, nos, this dichotomization also has clinical relevance with the ABC type having a worse prognosis than GCB type, independent of known clinical risk factors such as the International Prognostic Index (IPI). These data have been confirmed in patients treated with rituximab-CHOP therapy.67  Other signatures were also defined with clinical relevance. A favorable stromal signature (stromal signature 1) was identified that reflects extracellular matrix deposition and tumor infiltrating macrophages. An unfavorable stromal signature (stromal signature 2) was also identified that reflects angiogenesis and high blood vessel density.67 

In addition to these important expression signatures, genetic abnormalities have been discovered. For example, numerical abnormalities, such as trisomy 3 and deletion of CDKN2A, are associated with poor prognosis.68  In fact, these abnormalities track with the COO and further support the concept that different subtypes of DLBCL arise in different molecular genetic backgrounds, reflecting their biologic distinctiveness that is also manifest in clinical differences in outcome.68  Mutations in specific pathways that are associated with cell of origin have also been discovered. For example, mutations in the B-cell receptor signaling pathway leading to NFκB activation are associated with ABC type of DLBCL, nos and mutation in the EZH2 involved in histone modification is seen in GCB DLBCLs. Both pathways are potential targets of therapy.

High throughput sequencing studies have also begun to define the mutational landscape of DLBCL, nos.69-72  These studies have demonstrated sets of commonly mutated genes and suggested a number of important gene ontogeny categories. In one large study of 73 primary DLBLC cases 34 of which had paired normal DNA, 322 recurrently mutated candidate genes were identified. The genes could be organized into pathways and >50% of the genes could be placed into one of 12 gene ontologies. The most common processes were signal transduction (including genes in the JAK-STAT, ubiquitin, WNT, NFkB, NOTCH, and PI3 kinase pathways) and chromatin modification (including SETD2, MLL3, ARID1A, and MEF2B). The degree of non-overlap in gene lists found amongst studies likely reflects the high degree of biologic heterogeneity, with a long tail of uncommon but recurrently mutated genes.72 Table 3 shows selected commonly mutated genes and associated pathways from multiple next-generation-sequencing studies.

Table 3.

Selected recurrently mutated genes in DLBCL, nos

Selected recurrently mutated genes in DLBCL, nos
Selected recurrently mutated genes in DLBCL, nos

Despite the explosion of biologic data from broad scale gene expression profiling, array-based DNA copy number, and high throughput sequencing technologies, we have been challenged to reduce these discoveries to practice for reasons that include requirements for fresh frozen tissue, requirement for highly specialized equipment, potential long assay turnaround times, and cost. Clearly, for FFPET, immunohistochemistry (IHC) has great appeal for broad applicability and cost. However, issues related to standardization and pre-analytic variability that may affect antigen preservation and interobserver variability have been obstacles. Improvements in staining technology, advances in antibody technology, and experience in other tumor systems, such as breast cancer, with regard to standardization of tissue fixation, interpretive guidelines, and proficiency testing suggest that IHC biomarker assessment can be done, and but faces significant obstacles.73-77 

In particular, there has been great interest in determining COO (GCB vs non-GCB) and IHC algorithms, generally using 3-5 markers have been developed.78-81  As can be seen from Figure 1, the algorithms differ in terms of approach, markers used, and cutoffs for each marker. Published concordance rates range from 86%-93%. Other performance characteristics such as sensitivity, specificity, and positive predictive value for GCB and ABC types also vary but range from 81%-99% (Table 4).78-81  The COO not only has prognostic value but is currently being used as an integral biomarker for clinical trials enriching for ABC types of DLBCL. Therefore, accurate determination and reporting of this data is becoming critical. In addition, based on the strength of the biological data and prognostic importance, it is likely the upcoming WHO Classification update will include COO determination as a subtype of DLBCL nos rather than a variant, requiring determination in daily practice.82 

Figure 1.

Algorithms for COO determination.

Figure 1.

Algorithms for COO determination.

Close modal
Table 4.

Cell of origin immunostaining algorithms and published performance

Cell of origin immunostaining algorithms and published performance
Cell of origin immunostaining algorithms and published performance

NA indicates not available; *, modified algorithms.

Advances in molecular technology have the potential to offer a molecular-based alternative to immunophenotypic surrogates. Experience has shown that whole genome expression profiling is not required to perform COO determination and limited gene sets have been identified. For example, Wright et al reported a 14 gene classifier capable of determining COO.66  This classifier has been reduced to practice using a single-tube multiplex capillary electrophoresis-based real-time QPCR assay and is currently available as a test in our clinical laboratories.83  A 20 gene classifier has also been reported and is being actively developed as a commercial test.84  Of note, such molecular assays may also be used to calibrate and validate IHC-based assays to maximize availability, minimize turnaround times, and decrease cost if adequate performance can be maintained. Indeed we have validated our IHC algorithms using such an approach with sensitivities and specificities for GCB and ABC subtypes in the 90% range (Tables 4 and 5).

Table 5.

Performance of IHC for ABC identification134 

Performance of IHC for ABC identification134
Performance of IHC for ABC identification134

N = 63 with ICEPlex molecular classifier.

Can we use IHC in the routine laboratory for COO prediction? Given that molecular classifiers operate on a 90% probability threshold for determining molecular classification, IHC performance is arguably appropriate for use in the clinical setting, provided laboratories verify the IHC performance characteristics against gene-expression-based assays in their laboratories. In addition, the trend toward small biopsies or confounding issues such as presence of necrosis and inclusion of non-tumor tissues in biopsies may limit application of some molecular assays, thus IHC-based alternatives/back-ups may be required. With several possible IHC algorithms and now molecular test alternatives to choose from, pathology reports should be clear regarding which method is used and pathologists should adhere to the specified cutoffs if a particular IHC algorithm is used. Ideally, these should be reported in synoptic format for clarity. Reporting templates for biomarker data in clinical reports have prepared for this type of data and are published by the College of American Pathologists to serve as a guide for pathologists.85 

In the search for biomarkers that assist in risk stratification, recent interest has turned to expression of MYC and BCL2 (double expressors; DEs). It is known that cases of DLBCL with MYC and BCL2 translocations have an extremely poor prognosis.86-88  However, other mechanisms besides translocations can result in overexpression of these proteins.68,89  With the development of a monoclonal antibody suitable for FFPET, 2 groups studied expression of BCL2 and MYC by immunohistochemistry.90,91  Green et al studied a series of 193 cases of DLBCL treated with R-CHOP and, using median expression values, determined cutoffs of ≥70% for BCL2 and ≥40% for MYC identified a high-risk patient population, which was then validated in a separate cohort of 116 patients.90  Johnson et al evaluated a training set of 167 DLBCL patients treated with RCHOP and determined cutoffs of ≥50% for BCL2 and ≥40% for MYC. This was validated in a separate set of 140 cases.91  Both series noted a strong association between DEs and COO classification (ABC type or non-GCB phenotype).90,91  Furthermore, the DE status was an independent predictor of PFS and OS when IPI and COO were included in statistical models. The adverse effect of DE held when genetic double-hit (DH) lymphomas were excluded.91 

In a follow-up large multicenter study, Hu et al92  confirmed this effect by evaluating 40% and 70% cutoffs for MYC and BCL2 proteins, respectively, where cutoffs were arrived at through receiver-operator-curve analysis. The adverse impact on outcome was confirmed as well as the association with ABC type, independence from IPI, and was also shown once DH lymphomas were excluded. Of note, due to the high association with COO, ABC type was no longer associated with poor prognosis once DE status was included.92 

Several other studies have been published examining DE status, each confirming the poor prognostic impact of DE status. However, cutoffs for BCL2 varied,90,91,93  and in 1 study treatment was not homogeneous.94  Thus, the preponderance of evidence shows the importance of DE status for risk stratification in DLBCL, nos; however, it should be mentioned that not all studies have shown the poor prognostic effects perhaps due to differences in study population.95  Notwithstanding, there are still some unsettled issues related to precise cutoffs to use in practice. Reproducibility has been shown to be good (91%) for BCL2.91  It also appears good for MYC, because one pathologist scored MYC in the training set of Johnson et al, and different pathologists scored the validation set.91  There is also general agreement on antibody clones for these markers because the Y69 clone for MYC and the 124 clone for BCL2 were used in all studies cited. We have recently validated these immunostains in our cohort of 69 RCHOP-treated DLBCL, nos patients, with the purpose of evaluating our technical platforms and the cutoffs of ≥40% for MYC and ≥70% for BCL2. Using automated immunostainers commercially available IVD kits for MYC (clone Y69) and BCL2 (clones 124 and SP66), we confirmed that ≥40%/≥70% cutoffs for MYC and BCL2, respectively, identified patients with poor OS, independent of the IPI. Furthermore, we found that ≥40%/≥50% also performed similarly and that clones SP66 and 124 also performed similarly.96  Thus, for laboratories wishing to implement this type of testing, these reagents appear suitable for use in this indication. Because a 50% cutoff for BCL2 may be easier to estimate, we suggest cutoffs of ≥40% for MYC and ≥50% for BCL2 be adopted, although those wishing to use 70% for BCL2 will be on firm footing. As with the COO, pathology reporting should be clear in terms of cutoffs used.

The microenvironment

Although much effort has been directed at understanding the biologic feature intrinsic to the tumor cells, features of related to immune response and microenvironment have been also been defined. As noted previously, gene profiling efforts identified stromal signatures that were of prognostic importance.67  Other gene expression studies have similarly found that microenvironmental components are differentially expressed and provide prognostic information.97-99  Attempts to reduce these findings to practice using immunohistochemical markers for gene products such as fibronectin and SPARC, microvessel density, or infiltrating T cells, Tregs, and macrophages have been successful in demonstrating feasibility, but have yet to be extensively reproduced to the same extent that COO or MYC/BCL2 expression have been.100-105  Interest is mounting in manipulating the immune microenvironment and reconstituting an anti-tumor response through PD1 blockade and a promising phase II trial has been completed in DLBCL.106  Further work, including whether assessment of PDL1 as a predictive biomarker is of value, is warranted.

BCLu

BCLu is an imprecisely defined entity in the 2008 WHO classification and was meant to recognize the overlap and gray zone between DLBCL and BL.1  Such cases were demonstrated in GEP studies, in which expression profiles intermediate between signatures of molecular BL (mBL) and DLBCL were seen.107,108  Histologic features vary. Some cases have high-grade features, such as starry sky and intermediately sized cells, but demonstrate excessive pleomorphism that exclude BL. Others may have morphologic features that closely resemble BL; however, phenotype (such as expression of BCL2) or genotype (concurrent MYC and BCL2 or BCL6 translocations, or MYC translocation with complex karyotype) make a case unacceptable for BL. Still others may have morphology of DLBCL but an intermediate GEP with or without MYC translocation.1,107,108  Too make matters worse, some case of mBL as defined by GEP, have genetic or morphologic feature that are atypical for BL, and include some cases with MYC and BCL2 translocations (“double-hit” cases) or complex genetic backgrounds with MYC translocations that would not be acceptable for BL. As a group, patients tend to be older and have a poor prognosis.109 Table 6 has the current 2008 WHO features of BCLu, with comparison to DLBCL and BL.

Table 6.

WHO 2008 Features of BCLu1 

WHO 2008 Features of BCLu1
WHO 2008 Features of BCLu1

One can immediately see the problem in daily practice, because GEP and detailed genetic analysis by array-based and/or karyotyping is needed to recapitulate studies done to recognize BCLu. As a consequence, pathologists primary entry point for BCLu is appropriate morphology, ie, a case with “high grade” morphology (Table 6), which is then assessed for other supporting phenotypic or molecular features. An operational guideline would be to reserve this designation in a pathology report for cases that histologically closely resemble BL but in which the immunophenotype (such as lack of CD10, expression of strong BCL2) or genotype (complex karyotype, presence of BCL2 or BCL6 rearrangement along with MYC rearrangement, MYC rearrangement with non-IG partner gene) are unacceptable for BL. Some cases may have mixed morphology in which some intermediately sized Burkitt-type cells as well as large cells are present. As a corollary, cases with typical DLBCL, nos morphology, whether or not a MYC rearrangement is present, should not be placed into the BCLu designation. Also implied is that cases with appropriate morphology must be studied for genetic features that could be determined from routine karyotyping. Because karyotyping is not common practice, extensive FISH testing for MYC, BCL2, and BCL6 rearrangements becomes imperative (see below). How frequently does “high grade morphology” occur in diffuse aggressive B-cell lymphoma? In a review of diffuse aggressive B-cell lymphomas from a phase 3 trial of 260 patients, 12% were deemed to have BCLu morphology.110 

Double-hit lymphoma

The term “double-hit” (DH) lymphoma, recently reviewed at the 2014 American Society of Hematology meeting education sessions, has been used variably in the literature and typically refers to cases with MYC translocation in combination of either BCL2 or BCL6 translocations.111  However, some studies have included CCND1 translocations or advocate inclusion of cases with numerical abnormalities in MYC or BCL2.87,112,113  Cases with CCND1 and MYC translocation are best considered as forms of mantle cell lymphoma (blastoid variants) and there is not consensus whether numerical abnormalities in MYC and BCL2 should be included. More data is needed to accept such cases in formal classification systems. DH lymphomas are thus not a specific entity in the WHO 2008. They represent approximately 2%-14% of DLBCL and 32%-78% of BCLu.111,113-116  Variation can be explained by different patient populations studied as well as differences in methods used to identify cases. The histopathology of DH or triple-hit (TH; rare cases with concurrent MYC, BCL2, and BCL6 rearrangements) lymphomas cases is variable. In select series where inclusion was largely based on molecular features, DLBCL nos and BCLu histology is reported in 32%-72% (median 62%) and 2%-62% (median 28%), respectively, with a small proportion (<15%) being follicular lymphoma or transformed from low grade lymphoma.86,88,112,113,117,118  Coming at the issue from the starting point of the histopathology, 2%-12% of DLBL are DH, whereas 32%-78% of BCLu are DH.111  Morphology may matter with some studies suggesting DLBCL, nos morphology has a better overall survival compared with non-DLBCL or BCLu morphology.86,88  The immunophenotype of DH lymphomas is heavily weighted to a germinal center B-cell phenotype with approximately 80%-90% of cases expressing CD10 and BCL6 with MUM1/IRF4 expressed in <20% of cases.86-88,119-122  In MYC/BCL6 DH cases, CD10 is less likely to be expressed compared with MYC/BCL2 DH cases.119  DH and the rare TH lymphomas generally have an aggressive clinical course with many studies showing mean or median survivals on the order of 1 year or less.86,88,114,117,118,123  Thus alternate treatments are needed.

Because routine karyotyping is seldom performed in NHLs, most cases are defined by fluorescent in situ hybridization (FISH) studies. FISH studies are most often used and depending on probes used, cases may not be recognized. In general, break-apart probes will be the most sensitive method to detect translocations; however, partner genes are not identified. Specific fusion probes are required to positively identify the fusion partners. For example, break apart probes for MYC will identify the great majority of translocated cases, but ∼10% of MYC translocated cases may be missed in break-apart only probes are used.123  However, IGH-MYC, IGK-MYC, and IGL-MYC probes must be used to determine whether the IG genes are involved in the translocation. Unfortunately, the latter 2 probe sets are not commercially available. If MYC rearrangement is present but IGH-MYC dual fusion probes are negative, break-apart IGK or IGL may be used to infer that these genes are involved; however, it is not definitive because it is possible that other genes may be involved in fusion with IGK or IGL.

Does the partner gene for MYC matter? Some studies,86,124,125  but not all,112  suggest that only IG-MYC fusions are associated with poor prognosis in DH lymphomas, whereas MYC fusion with non-IG partner genes is not associated with aggressive behavior.86,124,125  However, it should be remembered that most studies that have defined the very poor prognosis of DH lymphoma used break apart MYC probes to help identify cases.88,113,114,117,126-128  To further confound the issue of definition of DH, one study has suggested that so-called “atypical DH lymphomas” (those with single translocations of MYC or BCL2 with copy number alterations of the other gene, or copy number alterations of both genes) have similar poor outcome to conventional DH lymphomas.87,129,130  In addition to potential clinical importance associated with MYC partner genes, there may be histologic associations as well. A non-IG partner gene was associated with DLBCL morphology.86  The importance of MYC partner gene remains to be resolved.

In interest of full disclosure, not all studies have shown the importance of recognizing DH lymphomas. For example, Aukema et al in a series of non-uniformly treated patients found no difference in outcome between patients with SH and DH lymphomas.112  However, cases with a mBL gene expression signature were excluded and treatment protocols were predominantly in the pre-rituximab era. Tzankhov et al reported that in a multivariate analysis of DLBCL patients, MYC-R and high level of MYC protein expression were the most important factors along with IPI group and R-CHOP-like treatment in predicting outcome. Thus DH status lost its importance once these factors were known.123 

In view of these data, it is not surprising that current laboratory practice is quite variable. Many laboratories test for MYC with break-apart probes. A subset may follow-up a positive result with IGH-MYC dual fusion probes. Very few laboratories if any, routinely attempt IGK and IGL FISH as secondary assays in the event that a MYC translocation is present while an IGH-MYC dual fusion probe set is negative. In an attempt to minimize cost, some laboratories have adopted screening using IHC for MYC. With the development of suitable MYC antibodies, one may enrich for translocated cases by only FISH testing cases that have MYC protein expression above a certain threshold, often 40%-50%.131,132  In a small series of 56 DLBCL, a threshold of 50% nuclear staining for MYC detected all cases with MYC translocation.132  However, results in other larger series has not been as good. With cutoffs ranging from 30% to 50%, the percentage of MYC-R cases below the cutoff ranged from 18% to 41% with lower cutoffs generally being associated with lower percentage of MYC-R DLBCL cases.90,93,95,123,128  Indeed, Horn et al argue against using a combined IHC/FISH algorithm in favor of FISH due to the potential for missing cases screened by IHC, but do concede that a 20% threshold could be considered (with a sensitivity of 92.3% for picking up MYC-R cases).95  One point should be made clear, Ki67 proliferative index should not be used as a screen because many cases of MYC translocated cases will have a low proliferative fraction. For example 42% of MYC translocated cases were shown to have a Ki67 proliferation index <80%.133  Practices seeking to maximize detection of DH lymphomas will likely adopt FISH testing in all diffuse aggressive B-cell lymphomas using MYC break-apart and IGH-MYC fusion probes along with BCL2 and BCL6 break apart probes. Those looking to use balance cost and sensitivity may adopt an IHC screen. However, individual laboratories should understand the performance characteristics of the MYC IHC stain and chosen cutoff for this purpose. Routine FISH testing will likely become obligatory as the much anticipated update to the current WHO classification may call for a high grade B-cell lymphoma, not otherwise specified (HGBCL, nos) parsed into 2 subgroups. The first is HGBCL,nos with MYC and BCL2 or BCL6 rearrangement (with specified morphology of either DLBCL, nos or BCLu). The second is HGBCL, nos without DH genetic features but with BCLu or other “high-grade” histologic features.82 

The diagnosis, characterization, and reporting of diffuse aggressive large B-cell lymphomas has grown more complex. Molecular profiling has defined new entities, helped us recognize pathologic gray zones, revealed clinically relevant biologic subtypes, and helped identify prognostic and potentially predictive biomarkers. High-throughput sequencing studies are also providing insight into the mutational landscape that will inform new therapeutic strategies and perhaps provide a new framework with which to understand and classify these lymphomas. Inclusion of current relevant pathologic, immunophenotypic, and genetic features requires close interaction between pathologists and clinicians so that these data are presented in a clear and clinically meaningful way. Incorporation into local and national guidelines that are periodically reviewed and updated will drive the application in all types of practice settings.

Eric D. Hsi, Department of Laboratory Medicine, L30, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195; Phone: 216-444-5230; Fax: 216-445-7253; e-mail: hsie@ccf.org.

1
Swerdlow
 
SH
Harris
 
NL
Jaffe
 
ES
Pileri
 
SA
Stein
 
H
Thiele
 
J
Vardiman
 
JW
WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues
2008
Ed 4
Lyon
IARC
2
The Non-Hodgkin's Lymphoma Classification Project
A clinical evaluation of the International Lymphoma Study Group classification of non-Hodgkin's lymphoma
Blood
1997
, vol. 
89
 
11
(pg. 
3909
-
3918
)
3
Jacobson
 
JO
Aisenberg
 
AC
Lamarre
 
L
, et al. 
Mediastinal large cell lymphoma: an uncommon subset of adult lymphoma curable with combined modality therapy
Cancer
1988
, vol. 
62
 
9
(pg. 
1893
-
1898
)
4
Lamarre
 
L
Jacobson
 
JO
Aisenberg
 
AC
Harris
 
NL
Primary large cell lymphoma of the mediastinum: a histologic and immunophenotypic study of 29 cases
Am J Surg Pathol
1989
, vol. 
13
 
9
pg. 
730739
 
5
Johnson
 
PW
Davies
 
AJ
Primary mediastinal B-cell lymphoma
Hematology Am Soc Hematol Educ Program
2008
pg. 
349358
 
6
Isaacson
 
PG
Norton
 
AJ
Addis
 
BJ
The human thymus contains a novel population of B lymphocytes
Lancet
1987
, vol. 
2
 
8574
(pg. 
1488
-
1491
)
7
Rosenwald
 
A
Wright
 
G
Leroy
 
K
, 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
, vol. 
198
 
6
(pg. 
851
-
862
)
8
Hutchinson
 
CB
Wang
 
E
Primary mediastinal (thymic) large B-cell lymphoma: a short review with brief discussion of mediastinal gray zone lymphoma
Arch Pathol Lab Med
2011
, vol. 
135
 
3
(pg. 
394
-
398
)
9
Pileri
 
SA
Zinzani
 
PL
Gaidano
 
G
, et al. 
Pathobiology of primary mediastinal B-cell lymphoma
Leuk Lymphoma
2003
, vol. 
44
 
Suppl 3
(pg. 
S21
-
S26
)
10
Pileri
 
SA
Gaidano
 
G
Zinzani
 
PL
, et al. 
Primary mediastinal B-cell lymphoma: high frequency of BCL-6 mutations and consistent expression of the transcription factors OCT-2, BOB.1, and PU.1 in the absence of immunoglobulins
Am J Pathol
2003
, vol. 
162
 
1
(pg. 
243
-
253
)
11
Calaminici
 
M
Piper
 
K
Lee
 
AM
Norton
 
AJ
CD23 expression in mediastinal large B-cell lymphomas
Histopathology
2004
, vol. 
45
 
6
(pg. 
619
-
624
)
12
Higgins
 
JP
Warnke
 
RA
CD30 expression is common in mediastinal large B-cell lymphoma
Am J Clin Pathol
1999
, vol. 
112
 
2
(pg. 
241
-
247
)
13
Copie-Bergman
 
C
Gaulard
 
P
Maouche-Chretien
 
L
, et al. 
The MAL gene is expressed in primary mediastinal large B-cell lymphoma
Blood
1999
, vol. 
94
 
10
(pg. 
3567
-
3575
)
14
Copie-Bergman
 
C
Plonquet
 
A
Alonso
 
MA
, et al. 
MAL expression in lymphoid cells: further evidence for MAL as a distinct molecular marker of primary mediastinal large B-cell lymphomas
Mod Pathol
2002
, vol. 
15
 
11
(pg. 
1172
-
1180
)
15
Hsi
 
ED
Sup
 
SJ
Alemany
 
C
, et al. 
MAL is expressed in a subset of Hodgkin lymphoma and identifies a population of patients with poor prognosis
Am J Clin Pathol
2006
, vol. 
125
 
5
pg. 
776782
 
16
Cazals-Hatem
 
D
Lepage
 
E
Brice
 
P
, et al. 
Primary mediastinal large B-cell lymphoma: a clinicopathologic study of 141 cases compared with 916 nonmediastinal large B-cell lymphomas, a GELA (“Groupe d'Etude des Lymphomes de l'Adulte”) study
Am J Surg Pathol
1996
, vol. 
20
 
7
(pg. 
877
-
888
)
17
Savage
 
KJ
Monti
 
S
Kutok
 
JL
, et al. 
The molecular signature of mediastinal large B-cell lymphoma differs from that of other diffuse large B-cell lymphomas and shares features with classical Hodgkin lymphoma
Blood
2003
, vol. 
102
 
12
(pg. 
3871
-
3879
)
18
Scarpa
 
A
Moore
 
PS
Rigaud
 
G
, et al. 
Molecular features of primary mediastinal B-cell lymphoma: involvement of p16INK4A, p53 and c-myc
Br J Haematol
1999
, vol. 
107
 
1
(pg. 
106
-
113
)
19
Tsang
 
P
Cesarman
 
E
Chadburn
 
A
Liu
 
YF
Knowles
 
DM
Molecular characterization of primary mediastinal B cell lymphoma
Am J Pathol
1996
, vol. 
148
 
6
(pg. 
2017
-
2025
)
20
Bentz
 
M
Barth
 
TF
Bruderlein
 
S
, et al. 
Gain of chromosome arm 9p is characteristic of primary mediastinal B-cell lymphoma (MBL): comprehensive molecular cytogenetic analysis and presentation of a novel MBL cell line
Genes Chromosomes Cancer
2001
, vol. 
30
 
4
(pg. 
393
-
401
)
21
Joos
 
S
Otano-Joos
 
MI
Ziegler
 
S
, et al. 
Primary mediastinal (thymic) B-cell lymphoma is characterized by gains of chromosomal material including 9p and amplification of the REL gene
Blood
1996
, vol. 
87
 
4
(pg. 
1571
-
1578
)
22
Wessendorf
 
S
Barth
 
TF
Viardot
 
A
, et al. 
Further delineation of chromosomal consensus regions in primary mediastinal B-cell lymphomas: an analysis of 37 tumor samples using high-resolution genomic profiling (array-CGH)
Leukemia
2007
, vol. 
21
 
12
(pg. 
2463
-
2469
)
23
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
, vol. 
116
 
17
(pg. 
3268
-
3277
)
24
Melzner
 
I
Bucur
 
AJ
Bruderlein
 
S
, et al. 
Biallelic mutation of SOCS-1 impairs JAK2 degradation and sustains phospho-JAK2 action in the MedB-1 mediastinal lymphoma line
Blood
2005
, vol. 
105
 
6
(pg. 
2535
-
2542
)
25
Ritz
 
O
Guiter
 
C
Castellano
 
F
, et al. 
Recurrent mutations of the STAT6 DNA binding domain in primary mediastinal B-cell lymphoma
Blood
2009
, vol. 
114
 
6
(pg. 
1236
-
1242
)
26
Rui
 
L
Emre
 
NC
Kruhlak
 
MJ
, et al. 
Cooperative epigenetic modulation by cancer amplicon genes
Cancer Cell
2010
, vol. 
18
 
6
(pg. 
590
-
605
)
27
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
, vol. 
206
 
5
(pg. 
981
-
989
)
28
Weniger
 
MA
Gesk
 
S
Ehrlich
 
S
, et al. 
Gains of REL in primary mediastinal B-cell lymphoma coincide with nuclear accumulation of REL protein
Genes Chromosomes Cancer
2007
, vol. 
46
 
4
(pg. 
406
-
415
)
29
Weniger
 
MA
Pulford
 
K
Gesk
 
S
, et al. 
Gains of the proto-oncogene BCL11A and nuclear accumulation of BCL11A(XL) protein are frequent in primary mediastinal B-cell lymphoma
Leukemia
2006
, vol. 
20
 
10
(pg. 
1880
-
1882
)
30
Steidl
 
C
Gascoyne
 
RD
The molecular pathogenesis of primary mediastinal large B-cell lymphoma
Blood
2011
, vol. 
118
 
10
(pg. 
2659
-
2669
)
31
Steidl
 
C
Shah
 
SP
Woolcock
 
BW
, et al. 
MHC class II transactivator CIITA is a recurrent gene fusion partner in lymphoid cancers
Nature
2011
, vol. 
471
 
7338
(pg. 
377
-
381
)
32
Twa
 
DD
Steidl
 
C
Structural genomic alterations in primary mediastinal large B-cell lymphoma
Leuk Lymphoma
2015
(pg. 
1
-
12
)
33
Twa
 
DD
Chan
 
FC
Ben-Neriah
 
S
, et al. 
Genomic rearrangements involving programmed death ligands are recurrent in primary mediastinal large B-cell lymphoma
Blood
2014
, vol. 
123
 
13
(pg. 
2062
-
2065
)
34
Yuan
 
J
Wright
 
G
Rosenwald
 
A
, et al. 
Identification of primary mediastinal large B-cell lymphoma at nonmediastinal sites by gene expression profiling
Am J Surg Pathol
2015
, vol. 
39
 
10
pg. 
1322
 
35
Dorfman
 
DM
Shahsafaei
 
A
Alonso
 
MA
Utility of CD200 immunostaining in the diagnosis of primary mediastinal large B cell lymphoma: comparison with MAL, CD23, and other markers
Mod Pathol
2012
, vol. 
25
 
12
(pg. 
1637
-
1643
)
36
Rodig
 
SJ
Savage
 
KJ
LaCasce
 
AS
, et al. 
Expression of TRAF1 and nuclear c-Rel distinguishes primary mediastinal large cell lymphoma from other types of diffuse large B-cell lymphoma
Am J Surg Pathol
2007
, vol. 
31
 
1
(pg. 
106
-
112
)
37
Shi
 
M
Roemer
 
MG
Chapuy
 
B
, et al. 
Expression of programmed cell death 1 ligand 2 (PD-L2) is a distinguishing feature of primary mediastinal (thymic) large B-cell lymphoma and associated with PDCD1LG2 copy gain
Am J Surg Pathol
2014
, vol. 
38
 
12
(pg. 
1715
-
1723
)
38
Jaffe
 
ES
Zarate-Osorno
 
A
Kingma
 
DW
Raffeld
 
M
Medeiros
 
LJ
The interrelationship between Hodgkin's disease and non-Hodgkin's lymphomas
Ann Oncol
1994
, vol. 
5
 
Suppl 1
(pg. 
7
-
11
)
39
Jaffe
 
ES
Zarate-Osorno
 
A
Medeiros
 
LJ
The interrelationship of Hodgkin's disease and non-Hodgkin's lymphomas: lessons learned from composite and sequential malignancies
Semin Diagn Pathol
1992
, vol. 
9
 
4
(pg. 
297
-
303
)
40
Perrone
 
T
Frizzera
 
G
Rosai
 
J
Mediastinal diffuse large-cell lymphoma with sclerosis: a clinicopathologic study of 60 cases
Am J Surg Pathol
1986
, vol. 
10
 
3
(pg. 
176
-
191
)
41
Traverse-Glehen
 
A
Pittaluga
 
S
Gaulard
 
P
, et al. 
Mediastinal gray zone lymphoma: the missing link between classic Hodgkin's lymphoma and mediastinal large B-cell lymphoma
Am J Surg Pathol
2005
, vol. 
29
 
11
(pg. 
1411
-
1421
)
42
Quintanilla-Martinez
 
L
de Jong
 
D
de Mascarel
 
A
, 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
, vol. 
2
 
4
(pg. 
211
-
236
)
43
Eberle
 
FC
Salaverria
 
I
Steidl
 
C
, et al. 
Gray zone lymphoma: chromosomal aberrations with immunophenotypic and clinical correlations
Mod Pathol
2011
, vol. 
24
 
12
(pg. 
1586
-
1597
)
44
Eberle
 
FC
Rodriguez-Canales
 
J
Wei
 
L
, et al. 
Methylation profiling of mediastinal gray zone lymphoma reveals a distinctive signature with elements shared by classical Hodgkin's lymphoma and primary mediastinal large B-cell lymphoma
Haematologica
2011
, vol. 
96
 
4
(pg. 
558
-
566
)
45
Gibson
 
SE
Hsi
 
ED
Epstein-Barr virus-positive B-cell lymphoma of the elderly at a United States tertiary medical center: an uncommon aggressive lymphoma with a nongerminal center B-cell phenotype
Hum Pathol
2009
, vol. 
40
 
5
(pg. 
653
-
661
)
46
Oyama
 
T
Yamamoto
 
K
Asano
 
N
, et al. 
Age-related EBV-associated B-cell lymphoproliferative disorders constitute a distinct clinicopathologic group: a study of 96 patients
Clin Cancer Res
2007
, vol. 
13
 
17
(pg. 
5124
-
5132
)
47
Park
 
S
Lee
 
J
Ko
 
YH
, et al. 
The impact of Epstein-Barr virus status on clinical outcome in diffuse large B-cell lymphoma
Blood
2007
, vol. 
110
 
3
(pg. 
972
-
978
)
48
Shimoyama
 
Y
Yamamoto
 
K
Asano
 
N
Oyama
 
T
Kinoshita
 
T
Nakamura
 
S
Age-related Epstein-Barr virus-associated B-cell lymphoproliferative disorders: special references to lymphomas surrounding this newly recognized clinicopathologic disease
Cancer Sci
2008
, vol. 
99
 
6
(pg. 
1085
-
1091
)
49
Dojcinov
 
SD
Venkataraman
 
G
Pittaluga
 
S
, et al. 
Age-related EBV-associated lymphoproliferative disorders in the Western population: a spectrum of reactive lymphoid hyperplasia and lymphoma
Blood
2011
, vol. 
117
 
18
(pg. 
4726
-
4735
)
50
Dojcinov
 
SD
Venkataraman
 
G
Raffeld
 
M
Pittaluga
 
S
Jaffe
 
ES
EBV positive mucocutaneous ulcer–a study of 26 cases associated with various sources of immunosuppression
Am J Surg Pathol
2010
, vol. 
34
 
3
(pg. 
405
-
417
)
51
Ghia
 
P
Prato
 
G
Stella
 
S
Scielzo
 
C
Geuna
 
M
Caligaris-Cappio
 
F
Age-dependent accumulation of monoclonal CD4+CD8+ double positive T lymphocytes in the peripheral blood of the elderly
Br J Haematol
2007
, vol. 
139
 
5
(pg. 
780
-
790
)
52
Messaoudi
 
I
Warner
 
J
Nikolich-Zugich
 
D
Fischer
 
M
Nikolich-Zugich
 
J
Molecular, cellular, and antigen requirements for development of age-associated T cell clonal expansions in vivo
J Immunol
2006
, vol. 
176
 
1
(pg. 
301
-
308
)
53
Nakahara
 
K
Utsunomiya
 
A
Hanada
 
S
, et al. 
Transient appearance of CD3+CD8+ T lymphocytes with monoclonal gene rearrangement of T-cell receptor beta locus
Br J Haematol
1998
, vol. 
100
 
2
(pg. 
411
-
414
)
54
Pawelec
 
G
Larbi
 
A
Immunity and ageing in man: Annual Review 2006/2007
Exp Gerontol
2008
, vol. 
43
 
1
(pg. 
34
-
38
)
55
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
, vol. 
126
 
7
(pg. 
863
-
872
)
56
Choe
 
JY
Yun
 
JY
Jeon
 
YK
, et al. 
Indoleamine 2,3-dioxygenase (IDO) is frequently expressed in stromal cells of Hodgkin lymphoma and is associated with adverse clinical features: a retrospective cohort study
BMC Cancer
2014
, vol. 
14
 pg. 
335
 
57
Amador-Ortiz
 
C
Chen
 
L
Hassan
 
A
, et al. 
Combined core needle biopsy and fine-needle aspiration with ancillary studies correlate highly with traditional techniques in the diagnosis of nodal-based lymphoma
Am J Clin Pathol
2011
, vol. 
135
 
4
(pg. 
516
-
524
)
58
Burke
 
C
Thomas
 
R
Inglis
 
C
, et al. 
Ultrasound-guided core biopsy in the diagnosis of lymphoma of the head and neck. A 9 year experience
Br J Radiol
2011
, vol. 
84
 
1004
(pg. 
727
-
732
)
59
He
 
Y
Ji
 
X
Xie
 
Y
, et al. 
Clinical application of ultrasound-guided core needle biopsy with multiple punches in the diagnosis of lymphoma
World J Surg Oncol
2015
, vol. 
13
 pg. 
126
 
60
Hu
 
Q
Naushad
 
H
Xie
 
Q
Al-Howaidi
 
I
Wang
 
M
Fu
 
K
Needle-core biopsy in the pathologic diagnosis of malignant lymphoma showing high reproducibility among pathologists
Am J Clin Pathol
2013
, vol. 
140
 
2
(pg. 
238
-
247
)
61
Lachar
 
WA
Shahab
 
I
Saad
 
AJ
Accuracy and cost-effectiveness of core needle biopsy in the evaluation of suspected lymphoma: a study of 101 cases
Arch Pathol Lab Med
2007
, vol. 
131
 
7
(pg. 
1033
-
1039
)
62
Skelton
 
E
Jewison
 
A
Okpaluba
 
C
, et al. 
Image-guided core needle biopsy in the diagnosis of malignant lymphoma
Eur J Surg Oncol
2015
, vol. 
41
 
7
(pg. 
852
-
858
)
63
Hehn
 
ST
Grogan
 
TM
Miller
 
TP
Utility of fine-needle aspiration as a diagnostic technique in lymphoma
J Clin Oncol
2004
, vol. 
22
 
15
(pg. 
3046
-
3052
)
64
Alizadeh
 
AA
Eisen
 
MB
Davis
 
RE
, et al. 
Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling
Nature
2000
, vol. 
403
 
6769
(pg. 
503
-
511
)
65
Rosenwald
 
A
Wright
 
G
Chan
 
WC
, et al. 
The use of molecular profiling to predict survival after chemotherapy for diffuse large-B-cell lymphoma
N Engl J Med
2002
, vol. 
346
 
25
(pg. 
1937
-
1947
)
66
Wright
 
G
Tan
 
B
Rosenwald
 
A
Hurt
 
EH
Wiestner
 
A
Staudt
 
LM
A gene expression-based method to diagnose clinically distinct subgroups of diffuse large B cell lymphoma
Proc Natl Acad Sci U S A
2003
, vol. 
100
 
17
(pg. 
9991
-
9996
)
67
Lenz
 
G
Wright
 
G
Dave
 
SS
, et al. 
Stromal gene signatures in large-B-cell lymphomas
N Engl J Med
2008
, vol. 
359
 
22
(pg. 
2313
-
2323
)
68
Lenz
 
G
Wright
 
GW
Emre
 
NC
, et al. 
Molecular subtypes of diffuse large B-cell lymphoma arise by distinct genetic pathways
Proc Natl Acad Sci U S A
2008
, vol. 
105
 
36
(pg. 
13520
-
13525
)
69
Lohr
 
JG
Stojanov
 
P
Lawrence
 
MS
, et al. 
Discovery and prioritization of somatic mutations in diffuse large B-cell lymphoma (DLBCL) by whole-exome sequencing
Proc Natl Acad Sci U S A
2012
, vol. 
109
 
10
(pg. 
3879
-
3884
)
70
Morin
 
RD
Mungall
 
K
Pleasance
 
E
, et al. 
Mutational and structural analysis of diffuse large B-cell lymphoma using whole-genome sequencing
Blood
2013
, vol. 
122
 
7
(pg. 
1256
-
1265
)
71
Pasqualucci
 
L
Trifonov
 
V
Fabbri
 
G
, et al. 
Analysis of the coding genome of diffuse large B-cell lymphoma
Nat Genet
2011
, vol. 
43
 
9
(pg. 
830
-
837
)
72
Zhang
 
J
Grubor
 
V
Love
 
CL
, et al. 
Genetic heterogeneity of diffuse large B-cell lymphoma
Proc Natl Acad Sci U S A
2013
, vol. 
110
 
4
(pg. 
1398
-
1403
)
73
Cohen
 
DA
Dabbs
 
DJ
Cooper
 
KL
, et al. 
Interobserver agreement among pathologists for semiquantitative hormone receptor scoring in breast carcinoma
Am J Clin Pathol
2012
, vol. 
138
 
6
(pg. 
796
-
802
)
74
Dyhdalo
 
KS
Fitzgibbons
 
PL
Goldsmith
 
JD
Souers
 
RJ
Nakhleh
 
RE
Laboratory compliance with the American Society of Clinical Oncology/College of American Pathologists human epidermal growth factor receptor 2 testing guidelines: a 3-year comparison of validation procedures
Arch Pathol Lab Med
2014
, vol. 
138
 
7
(pg. 
876
-
884
)
75
Perez
 
EA
Cortes
 
J
Gonzalez-Angulo
 
AM
Bartlett
 
JM
HER2 testing: current status and future directions
Cancer Treat Rev
2014
, vol. 
40
 
2
(pg. 
276
-
284
)
76
Wolff
 
AC
Hammond
 
ME
Hicks
 
DG
, et al. 
Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update
J Clin Oncol
2013
, vol. 
31
 
31
(pg. 
3997
-
4013
)
77
Salles
 
G
de Jong
 
D
Xie
 
W
, et al. 
Prognostic significance of immunohistochemical biomarkers in diffuse large B-cell lymphoma: a study from the Lunenburg Lymphoma Biomarker Consortium
Blood
2011
, vol. 
117
 
26
(pg. 
7070
-
7078
)
78
Choi
 
WW
Weisenburger
 
DD
Greiner
 
TC
, et al. 
A new immunostain algorithm classifies diffuse large B-cell lymphoma into molecular subtypes with high accuracy
Clin Cancer Res
2009
, vol. 
15
 
17
(pg. 
5494
-
5502
)
79
Hans
 
CP
Weisenburger
 
DD
Greiner
 
TC
, et al. 
Confirmation of the molecular classification of diffuse large B-cell lymphoma by immunohistochemistry using a tissue microarray
Blood
2004
, vol. 
103
 
1
(pg. 
275
-
282
)
80
Meyer
 
PN
Fu
 
K
Greiner
 
TC
, et al. 
Immunohistochemical methods for predicting cell of origin and survival in patients with diffuse large B-cell lymphoma treated with rituximab
J Clin Oncol
2011
, vol. 
29
 
2
(pg. 
200
-
207
)
81
Visco
 
C
Li
 
Y
Xu-Monette
 
ZY
, et al. 
Comprehensive gene expression profiling and immunohistochemical studies support application of immunophenotypic algorithm for molecular subtype classification in diffuse large B-cell lymphoma: a report from the International DLBCL Rituximab-CHOP Consortium Program Study
Leukemia
2012
, vol. 
26
 
9
(pg. 
2103
-
2113
)
82
Harris
 
NL
“Aggressive” B-cell lymphomas
Society for Hematopathology Companion Meeting, United States and Canadian Academy of Pathology Annual Meeting
2015
Boston, MA
83
Collie
 
AM
Nolling
 
J
Divakar
 
KM
, et al. 
Molecular subtype classification of formalin-fixed, paraffin-embedded diffuse large B-cell lymphoma samples on the ICEPlex(R) system
Br J Haematol
2014
, vol. 
167
 
2
(pg. 
281
-
285
)
84
Scott
 
DW
Wright
 
GW
Williams
 
PM
, et al. 
Determining cell-of-origin subtypes of diffuse large B-cell lymphoma using gene expression in formalin-fixed paraffin-embedded tissue
Blood
2014
, vol. 
123
 
8
(pg. 
1214
-
1217
)
85
Duncavage
 
E
Advani
 
RH
Agosti
 
S
, et al. 
Template for reporting results of biomarker testing of specimens from patients with diffuse large B-cell lymphoma, not otherewise specified (nos)
CAP Cancer Protocols
2014
, vol. 
Vol. 2015
 
Northfield, IL
College of American Pathologists
86
Johnson
 
NA
Savage
 
KJ
Ludkovski
 
O
, et al. 
Lymphomas with concurrent BCL2 and MYC translocations: the critical factors associated with survival
Blood
2009
, vol. 
114
 
11
(pg. 
2273
-
2279
)
87
Li
 
S
Lin
 
P
Fayad
 
LE
, et al. 
B-cell lymphomas with MYC/8q24 rearrangements and IGH@BCL2/t(14;18)(q32;q21): an aggressive disease with heterogeneous histology, germinal center B-cell immunophenotype and poor outcome
Mod Pathol
2012
, vol. 
25
 
1
(pg. 
145
-
156
)
88
Snuderl
 
M
Kolman
 
OK
Chen
 
YB
, et al. 
B-cell lymphomas with concurrent IGH-BCL2 and MYC rearrangements are aggressive neoplasms with clinical and pathologic features distinct from Burkitt lymphoma and diffuse large B-cell lymphoma
Am J Surg Pathol
2010
, vol. 
34
 
3
(pg. 
327
-
340
)
89
Bea
 
S
Zettl
 
A
Wright
 
G
, et al. 
Diffuse large B-cell lymphoma subgroups have distinct genetic profiles that influence tumor biology and improve gene-expression-based survival prediction
Blood
2005
, vol. 
106
 
9
(pg. 
3183
-
3190
)
90
Green
 
TM
Nielsen
 
O
de Stricker
 
K
Xu-Monette
 
ZY
Young
 
KH
Moller
 
MB
High levels of nuclear MYC protein predict the presence of MYC rearrangement in diffuse large B-cell lymphoma
Am J Surg Pathol
2012
, vol. 
36
 
4
(pg. 
612
-
619
)
91
Johnson
 
NA
Slack
 
GW
Savage
 
KJ
, et al. 
Concurrent expression of MYC and BCL2 in diffuse large B-cell lymphoma treated with rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone
J Clin Oncol
2012
, vol. 
30
 
28
(pg. 
3452
-
3459
)
92
Hu
 
S
Xu-Monette
 
ZY
Tzankov
 
A
, et al. 
MYC/BCL2 protein coexpression contributes to the inferior survival of activated B-cell subtype of diffuse large B-cell lymphoma and demonstrates high-risk gene expression signatures: a report from The International DLBCL Rituximab-CHOP Consortium Program
Blood
2013
, vol. 
121
 
20
(pg. 
4021
-
4031
quiz 4250
93
Perry
 
AM
Alvarado-Bernal
 
Y
Laurini
 
JA
, et al. 
MYC and BCL2 protein expression predicts survival in patients with diffuse large B-cell lymphoma treated with rituximab
Br J Haematol
2014
, vol. 
165
 
3
(pg. 
382
-
391
)
94
Yan
 
LX
Liu
 
YH
Luo
 
DL
, et al. 
MYC expression in concert with BCL2 and BCL6 expression predicts outcome in Chinese patients with diffuse large B-cell lymphoma, not otherwise specified
PLoS One
2014
, vol. 
9
 
8
pg. 
e104068
 
95
Horn
 
H
Ziepert
 
M
Becher
 
C
, et al. 
MYC status in concert with BCL2 and BCL6 expression predicts outcome in diffuse large B-cell lymphoma
Blood
2013
, vol. 
121
 
12
(pg. 
2253
-
2263
)
96
Schneider
 
KB
Collie
 
AMB
Lanigan
 
C.P
, et al. 
Dual expression of c-MYC and BCL2 proteins predicts worse outcomes in diffuse large B-cell lymphoma
Leuk Lymphoma
2015
 
in press
97
Monti
 
S
Savage
 
KJ
Kutok
 
JL
, et al. 
Molecular profiling of diffuse large B-cell lymphoma identifies robust subtypes including one characterized by host inflammatory response
Blood
2005
, vol. 
105
 
5
(pg. 
1851
-
1861
)
98
Linderoth
 
J
Eden
 
P
Ehinger
 
M
, et al. 
Genes associated with the tumour microenvironment are differentially expressed in cured versus primary chemotherapy-refractory diffuse large B-cell lymphoma
Br J Haematol
2008
, vol. 
141
 
4
(pg. 
423
-
432
)
99
Alizadeh
 
AA
Gentles
 
AJ
Alencar
 
AJ
, et al. 
Prediction of survival in diffuse large B-cell lymphoma based on the expression of 2 genes reflecting tumor and microenvironment
Blood
2011
, vol. 
118
 
5
(pg. 
1350
-
1358
)
100
Brandt
 
S
Montagna
 
C
Georgis
 
A
, et al. 
The combined expression of the stromal markers fibronectin and SPARC improves the prediction of survival in diffuse large B-cell lymphoma
Exp Hematol Oncol
2013
, vol. 
2
 
1
pg. 
27
 
101
Cardesa-Salzmann
 
TM
Colomo
 
L
Gutierrez
 
G
, et al. 
High microvessel density determines a poor outcome in patients with diffuse large B-cell lymphoma treated with rituximab plus chemotherapy
Haematologica
2011
, vol. 
96
 
7
(pg. 
996
-
1001
)
102
Meyer
 
PN
Fu
 
K
Greiner
 
T
, et al. 
The stromal cell marker SPARC predicts for survival in patients with diffuse large B-cell lymphoma treated with rituximab
Am J Clin Pathol
2011
, vol. 
135
 
1
(pg. 
54
-
61
)
103
Coutinho
 
R
Clear
 
AJ
Mazzola
 
E
, et al. 
Revisiting the immune microenvironment of diffuse large B-cell lymphoma using a tissue microarray and immunohistochemistry: robust semi-automated analysis reveals CD3 and FoxP3 as potential predictors of response to R-CHOP
Haematologica
2015
, vol. 
100
 
3
(pg. 
363
-
369
)
104
Nam
 
SJ
Go
 
H
Paik
 
JH
, et al. 
An increase of M2 macrophages predicts poor prognosis in patients with diffuse large B-cell lymphoma treated with rituximab, cyclophosphamide, doxorubicin, vincristine and prednisone
Leuk Lymphoma
2014
, vol. 
55
 
11
(pg. 
2466
-
2476
)
105
Riihijarvi
 
S
Fiskvik
 
I
Taskinen
 
M
, et al. 
Prognostic influence of macrophages in patients with diffuse large B-cell lymphoma: a correlative study from a Nordic phase II trial
Haematologica
2015
, vol. 
100
 
2
(pg. 
238
-
245
)
106
Armand
 
P
Nagler
 
A
Weller
 
EA
, et al. 
Disabling immune tolerance by programmed death-1 blockade with pidilizumab after autologous hematopoietic stem-cell transplantation for diffuse large B-cell lymphoma: results of an international phase II trial
J Clin Oncol
2013
, vol. 
31
 
33
(pg. 
4199
-
4206
)
107
Dave
 
SS
Fu
 
K
Wright
 
GW
, et al. 
Molecular diagnosis of Burkitt's lymphoma
N Engl J Med
2006
, vol. 
354
 
23
(pg. 
2431
-
2442
)
108
Hummel
 
M
Bentink
 
S
Berger
 
H
, et al. 
A biologic definition of Burkitt's lymphoma from transcriptional and genomic profiling
N Engl J Med
2006
, vol. 
354
 
23
(pg. 
2419
-
2430
)
109
Salaverria
 
I
Zettl
 
A
Bea
 
S
, et al. 
Chromosomal alterations detected by comparative genomic hybridization in subgroups of gene expression-defined Burkitt's lymphoma
Haematologica
2008
, vol. 
93
 
9
(pg. 
1327
-
1334
)
110
Cook
 
JR
Goldman
 
B
Tubbs
 
RR
, et al. 
Clinical significance of MYC expression and/or “high-grade” morphology in non-Burkitt, diffuse aggressive B-cell lymphomas: a SWOG S9704 correlative study
Am J Surg Pathol
2014
, vol. 
38
 
4
(pg. 
494
-
501
)
111
Swerdlow
 
SH
Diagnosis of ‘double hit’ diffuse large B-cell lymphoma and B-cell lymphoma, unclassifiable, with features intermediate between DLBCL and Burkitt lymphoma: when and how, FISH versus IHC
Hematology Am Soc Hematol Educ Program
2014
, vol. 
2014
 
1
(pg. 
90
-
99
)
112
Aukema
 
SM
Siebert
 
R
Schuuring
 
E
, et al. 
Double-hit B-cell lymphomas
Blood
2011
, vol. 
117
 
8
(pg. 
2319
-
2331
)
113
Oki
 
Y
Noorani
 
M
Lin
 
P
, et al. 
Double hit lymphoma: the MD Anderson Cancer Center clinical experience
Br J Haematol
2014
, vol. 
166
 
6
(pg. 
891
-
901
)
114
Akyurek
 
N
Uner
 
A
Benekli
 
M
Barista
 
I
Prognostic significance of MYC, BCL2, and BCL6 rearrangements in patients with diffuse large B-cell lymphoma treated with cyclophosphamide, doxorubicin, vincristine, and prednisone plus rituximab
Cancer
2012
, vol. 
118
 
17
(pg. 
4173
-
4183
)
115
Friedberg
 
JW
Double-hit diffuse large B-cell lymphoma
J Clin Oncol
2012
, vol. 
30
 
28
(pg. 
3439
-
3443
)
116
Quintanilla-Martinez
 
L
IX. Is it only about MYC? How to approach the diagnosis of diffuse large B-cell lymphomas
Hematol Oncol
2015
, vol. 
33
 
Suppl 1
(pg. 
50
-
55
)
117
Cohen
 
JB
Geyer
 
SM
Lozanski
 
G
, et al. 
Complete response to induction therapy in patients with Myc-positive and double-hit non-Hodgkin lymphoma is associated with prolonged progression-free survival
Cancer
2014
, vol. 
120
 
11
(pg. 
1677
-
1685
)
118
Landsburg
 
DJ
Nasta
 
SD
Svoboda
 
J
Morrissette
 
JJ
Schuster
 
SJ
‘Double-Hit’ cytogenetic status may not be predicted by baseline clinicopathological characteristics and is highly associated with overall survival in B cell lymphoma patients
Br J Haematol
2014
, vol. 
166
 
3
(pg. 
369
-
374
)
119
Pillai
 
RK
Sathanoori
 
M
Van Oss
 
SB
Swerdlow
 
SH
Double-hit B-cell lymphomas with BCL6 and MYC translocations are aggressive, frequently extranodal lymphomas distinct from BCL2 double-hit B-cell lymphomas
Am J Surg Pathol
2013
, vol. 
37
 
3
(pg. 
323
-
332
)
120
Le Gouill
 
S
Talmant
 
P
Touzeau
 
C
, et al. 
The clinical presentation and prognosis of diffuse large B-cell lymphoma with t(14;18) and 8q24/c-MYC rearrangement
Haematologica
2007
, vol. 
92
 
10
(pg. 
1335
-
1342
)
121
Niitsu
 
N
Okamoto
 
M
Miura
 
I
Hirano
 
M
Clinical features and prognosis of de novo diffuse large B-cell lymphoma with t(14;18) and 8q24/c-MYC translocations
Leukemia
2009
, vol. 
23
 
4
(pg. 
777
-
783
)
122
Tomita
 
N
Tokunaka
 
M
Nakamura
 
N
, et al. 
Clinicopathological features of lymphoma/leukemia patients carrying both BCL2 and MYC translocations
Haematologica
2009
, vol. 
94
 
7
(pg. 
935
-
943
)
123
Tzankov
 
A
Xu-Monette
 
ZY
Gerhard
 
M
, et al. 
Rearrangements of MYC gene facilitate risk stratification in diffuse large B-cell lymphoma patients treated with rituximab-CHOP
Mod Pathol
2014
, vol. 
27
 
7
(pg. 
958
-
971
)
124
Pedersen
 
MO
Gang
 
AO
Poulsen
 
TS
, et al. 
Double-hit BCL2/MYC translocations in a consecutive cohort of patients with large B-cell lymphoma: a single centre's experience
Eur J Haematol
2012
, vol. 
89
 
1
(pg. 
63
-
71
)
125
Pedersen
 
MO
Gang
 
AO
Poulsen
 
TS
, et al. 
MYC translocation partner gene determines survival of patients with large B-cell lymphoma with MYC- or double-hit MYC/BCL2 translocations
Eur J Haematol
2014
, vol. 
92
 
1
(pg. 
42
-
48
)
126
Kanungo
 
A
Medeiros
 
LJ
Abruzzo
 
LV
Lin
 
P
Lymphoid neoplasms associated with concurrent t(14;18) and 8q24/c-MYC translocation generally have a poor prognosis
Mod Pathol
2006
, vol. 
19
 
1
(pg. 
25
-
33
)
127
Barrans
 
S
Crouch
 
S
Smith
 
A
, et al. 
Rearrangement of MYC is associated with poor prognosis in patients with diffuse large B-cell lymphoma treated in the era of rituximab
J Clin Oncol
2010
, vol. 
28
 
20
(pg. 
3360
-
3365
)
128
Valera
 
A
Lopez-Guillermo
 
A
Cardesa-Salzmann
 
T
, et al. 
MYC protein expression and genetic alterations have prognostic impact in patients with diffuse large B-cell lymphoma treated with immunochemotherapy
Haematologica
2013
, vol. 
98
 
10
(pg. 
1554
-
1562
)
129
Li
 
S
Seegmiller
 
AC
Lin
 
P
, et al. 
B-cell lymphomas with concurrent MYC and BCL2 abnormalities other than translocations behave similarly to MYC/BCL2 double-hit lymphomas
Mod Pathol
2015
, vol. 
28
 
2
(pg. 
208
-
217
)
130
Lin
 
P
Dickason
 
TJ
Fayad
 
LE
, et al. 
Prognostic value of MYC rearrangement in cases of B-cell lymphoma, unclassifiable, with features intermediate between diffuse large B-cell lymphoma and Burkitt lymphoma
Cancer
2012
, vol. 
118
 
6
(pg. 
1566
-
1573
)
131
Ruzinova
 
MB
Caron
 
T
Rodig
 
SJ
Altered subcellular localization of c-Myc protein identifies aggressive B-cell lymphomas harboring a c-MYC translocation
Am J Surg Pathol
2010
, vol. 
34
 
6
(pg. 
882
-
891
)
132
Kluk
 
MJ
Chapuy
 
B
Sinha
 
P
, et al. 
Immunohistochemical detection of MYC-driven diffuse large B-cell lymphomas
PLoS One
2012
, vol. 
7
 
4
pg. 
e33813
 
133
Savage
 
KJ
Johnson
 
NA
Ben-Neriah
 
S
, et al. 
MYC gene rearrangements are associated with a poor prognosis in diffuse large B-cell lymphoma patients treated with R-CHOP chemotherapy
Blood
2009
, vol. 
114
 
17
(pg. 
3533
-
3537
)
134
Collie
 
AMB
Nolling
 
J
Divakar
 
KM
, et al. 
Validation of multiple immunohistochemical algorithms for assigning diffuse large B-cell lymphoma subtypes using a clincal molecular DLBCL subtyping assay
XVII annual meeting of the European Association for Hematopathology
October 17-22, 2014
Istanbul, Turkey

Competing Interests

Conflict-of-interest disclosure: The author has received research funding from Abbvie, Eli Lilly, and Cellerant Therapeutics; and has been affiliated with the Speakers Bureau for Seattle Genetics.

Author notes

Off-label drug use: None disclosed.