Skip to main content
Cold Spring Harbor Perspectives in Medicine logoLink to Cold Spring Harbor Perspectives in Medicine
. 2020 Nov;10(11):a035444. doi: 10.1101/cshperspect.a035444

Impact of Genetics on Mature Lymphoid Leukemias and Lymphomas

Nathanael G Bailey 1, Kojo SJ Elenitoba-Johnson 2
PMCID: PMC7605231  PMID: 31932467

Abstract

Recurrent genetic aberrations have long been recognized in mature lymphoid leukemias and lymphomas. As conventional karyotypic and molecular cloning techniques evolved in the 1970s and 1980s, multiple cytogenetic aberrations were identified in lymphomas, often balanced translocations that juxtaposed oncogenes to the immunoglobulin (IG) or T-cell receptor (TR) loci, leading to dysregulation. However, genetic characterization and classification of lymphoma by conventional cytogenetic methods is limited by the infrequent occurrence of recurrent karyotypic abnormalities in many lymphoma subtypes and by the frequent difficulty in growing clinical lymphoma specimens in culture to obtain informative karyotypes. As higher-resolution genomic techniques developed, such as array comparative genomic hybridization and fluorescence in situ hybridization, many recurrent copy number changes were identified in lymphomas, and copy number assessment of interphase cells became part of routine clinical practice for a subset of diseases. Platforms to globally examine mRNA expression led to major insights into the biology of several lymphomas, although these techniques have not gained widespread application in routine clinical settings. With the advent of next-generation sequencing (NGS) techniques in the early 2000s, numerous insights into the genetic landscape of lymphomas were obtained. In contrast to the myeloid malignancies, most common lymphomas exhibit an at least somewhat mutationally complex genome, with few single driver mutations in the majority of patients. However, many recurrently mutated pathways have been identified across lymphoma subtypes, informing targeted therapeutic approaches that are beginning to make meaningful changes in the treatment of lymphoma. In addition to the ability to identify possible therapeutic targets, NGS techniques are highly amenable to the tracking of residual lymphoma following therapy, because of the presence of unique genetic “fingerprints” in lymphoma cells due to V(D)-J recombination at the antigen receptor loci. This review will provide an overview of the impact of novel genetic technologies on lymphoma classification, biology, and therapy.

OVERVIEW OF GENETIC EVENTS IN SELECTED LYMPHOMAS

Lymphoid neoplasms are clonal proliferations of B cells, T cells, or natural killer (NK) cells that may manifest clinically as lymphadenopathy, extranodal tumors, leukemia, or any combination of the three. B-cell lymphomas comprise more than 30 World Health Organization (WHO)-defined entities and account for the majority of all lymphomas (Swerdlow et al. 2017). Different types of B-cell lymphoma are thought to originate from different stages in normal B-cell development (Fig. 1). Certain genetic events are shared in different lymphomas with similar cells of origin (e.g., mutations of chromatin modifiers in germinal center–derived B-cell lymphomas, B-cell receptor [BCR] pathway activation in postgerminal center B-cell lymphomas). T-cell lymphomas are much rarer than B-cell lymphomas, but they are frequently aggressive. Like B-cell lymphomas, some T-cell lymphomas exhibit phenotypic characteristics of physiologic T-cell subsets (Fig. 2). A comprehensive account of lymphoma genetics is outside of the scope of this review; this section will give an overview of genetic events in selected B-cell and T-cell lymphoma entities.

Figure 1.

Figure 1.

Simplified schematic of B-cell development and the lymphomas thought to originate from specific B-cell developmental stages. Representative frequent and/or clinically relevant genetic alterations are also noted. (MCL) Mantle cell lymphoma, (BL) Burkitt lymphoma, (HGBL) high-grade B-cell lymphoma, (DLBCL) diffuse large B-cell lymphoma, (GCB) germinal center B-cell, (ABC) activated B-cell, (FL) follicular lymphoma, (CLL/SLL) chronic lymphocytic leukemia/small lymphocytic lymphoma, (HCL) hairy cell leukemia, (HCL-v) hairy cell leukemia-variant, (SMZL) splenic marginal zone lymphoma, (LPL) lymphoplasmacytic lymphoma.

Figure 2.

Figure 2.

Simplified schematic of CD4+ T-cell subsets and selected corresponding lymphoma subtypes. Because of T-cell plasticity, the association of some of these entities with the subset is somewhat tenuous. Representative frequent and/or clinically relevant genetic alterations are also noted. (ALCL) Anaplastic large cell lymphoma, (MF/SS) mycosis fungoides/Sézary syndrome, (ATLL) adult T-cell leukemia/lymphoma, (AITL) angioimmunoblastic T-cell lymphoma, (T-PLL) T-cell prolymphocytic leukemia, (Th1) T helper type 1, (Th2) T helper type 2, (Treg) regulatory T cell, (Tfh) follicular helper T cell, (Tcm) central memory T cell.

Chronic Lymphocytic Leukemia/Small Lymphocytic Lymphoma

Chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL) is the most common lymphoid leukemia. CLL/SLL is diagnosed based on characteristic morphologic, clinical, and immunophenotypic characteristics (Rawstron et al. 2018), but it has become clear that the underlying genetics of CLL/SLL are prognostically and therapeutically critical. CLL/SLL may be subdivided into two primary genetic subgroups that each constitute about one-half of cases: those cases that exhibit an unmutated immunoglobulin heavy chain (IGH), and those that harbor a somatic hypermutation of IGH (Damle et al. 1999; Hamblin et al. 1999). These differences reflect whether the founding neoplastic lymphocyte progressed through a germinal center reaction, eliciting somatic hypermutation through the effect of activation-induced cytidine deaminase (AICDA), or whether the tumor arose either from an antigen-naive lymphocyte or one that encountered antigen through a germinal center–independent mechanism. This genomic distinction has major clinical implications, as patients with unmutated CLL/SLL have worse outcomes than do those with mutated CLL/SLL (Byrd et al. 2006).

CLL/SLL commonly exhibits recurrent structural chromosomal alterations that are amenable to clinical detection by fluorescence in situ hybridization (FISH) and microarray platforms (Döhner et al. 2000). Deletions of 13q are the most common structural alteration in CLL/SLL, occurring in ∼55% of patients. 13q deletions encompass MIR15A and MIR16-1, microRNAs that are important regulators of the antiapoptotic proteins BCL2 and MCL1 (Calin et al. 2008). When present in isolation, patients with 13q deletions have a relatively favorable prognosis. Trisomy 12 is relatively common, although its identification does not lead to specific clinical intervention. It is often associated with atypical phenotypic and/or morphologic characteristics, and so its identification may be diagnostically reassuring in a somewhat unusual case. Deletions of 11q23 (targeting ATM and surrounding genes) and 17p (targeting TP53) are associated with adverse prognosis in CLL/SLL. TP53 loss is especially adverse, and its identification has historically been used to direct younger patients to transplant preparation and older patients to agents such as alemtuzumab, as 17p-deleted CLL/SLL patients exhibited especially poor responses to conventional chemoimmunotherapeutic regimens (Gribben 2010; Brown 2018). A new generation of targeted agents such as inhibitors of Bruton tyrosine kinase (BTK), phosphoinositide 3-kinase (PI3K)-δ, and B-cell leukemia/lymphoma 2 (BCL2) have clinical efficacy against this aggressive subset.

Next-generation sequencing (NGS) has allowed identification of numerous recurrently mutated genes in CLL/SLL; although similar to many other lymphomas, there is a long tail of mutated genes, and no single gene is mutated in more than one-half of cases (Puente et al. 2011; Wang et al. 2011; Quesada et al. 2012; Landau et al. 2015). Among the more commonly mutated genes are SF3B1 (in ∼20%), ATM (frequently associated with 11q deletion), TP53 (frequently associated with 17p deletion), and NOTCH1 (associated with unmutated IGHV). Identification of TP53 mutations is critical, as they impart a poor prognosis, as does 17p loss, and ∼30%–40% of patients with TP53 mutation do not have 17p losses that are identified by routine FISH testing (Malcikova et al. 2018).

All patients with CLL/SLL should be screened for mutations in TP53 given its prognostic and therapeutic significance, and FISH studies to identify recurrent copy number changes (such as 17p deletion) are necessary at diagnosis and progression. The mutational status of the CLL should be tested when a patient requires treatment, as fit patients with mutated CLL and without 17p del/TP53 mutation may exhibit excellent responses to FCR chemotherapy, although alternate approaches are needed for other patients (Eichhorst et al. 2016; Fischer et al. 2016). Ibrutinib has recently been approved as frontline therapy for CLL/SLL in all genetic types; however, ibrutinib resistance can arise in a subset of patients. Some relapses following ibrutinib therapy are mediated by acquired mutations in BTK, the target of ibrutinib, or PLCG2, which allows for BTK-independent BCR activation (Woyach et al. 2014). Screening for these mutations may be considered for patients with CLL who lose response to ibrutinib.

Mantle Cell Lymphoma

Mantle cell lymphoma (MCL) is a relatively uncommon B-cell lymphoma that, similar to CLL/SLL, typically exhibits expression of the T-cell-associated antigen CD5. Although most often a morphologically indolent-appearing lymphoma, MCL generally exhibits a progressive disease course and is incurable with conventional chemoimmunotherapeutic approaches. MCL is characterized by translocations between chromosomes 11 and 14, causing deregulation of the cyclin D1 (CCND1) gene by the IGH enhancer region. The identification of this rearrangement in most cases of MCL contributed to its recognition and acceptance as a distinct lymphoma entity (Banks et al. 1992).

Copy number changes of 13q14, 11q23, and 17p are relatively frequent events in mantle cell lymphoma, targeting regions similar to those lost in CLL/SLL (Beà et al. 1999; Bentz et al. 2000). However, mantle cell lymphomas more frequently manifest more complex karyotypes than are seen in CLL/SLL, although there is no clear clinical indication to routinely test for these copy number changes in MCL. NGS studies have identified recurrent mutations in CCND1, ATM, KMT2D (MLL2), and TP53 (Beà et al. 2013; Zhang et al. 2014). TP53 mutations, in particular, have been associated with poorer outcomes in MCL (Eskelund et al. 2017).

Although CCND1 translocations are the genetic hallmark of MCL, lymphomas with morphologic and immunophenotypic characteristics of MCL but without detectable CCND1 translocation or protein expression have been recognized (Yatabe et al. 2000). These so-called CCND1 MCL have a gene expression profile similar to that seen in conventional MCL and consistently overexpress SOX11 (Fu et al. 2005; Mozos et al. 2009). SOX11 is a member of the SOX (sex determining region Y-related HMG-box) proteins and is expressed in ∼90% of MCLs, including in early “in situ” disease and in cyclin D1-negative cases (Dictor et al. 2009; Carvajal-Cuenca et al. 2012). It is now recognized that 50% of the cyclin D1-negative cases harbor CCND2 translocations resulting in juxtaposition with the immunoglobulin genes (Salaverria et al. 2013; Kurita et al. 2016; Martín-Garcia et al. 2018). Additionally, CCND3 translocations to the immunoglobulin light chain enhancers have also recently been recognized in cyclin D1-negative mantle cell lymphomas (Martín-Garcia et al. 2018).

Although most cases of MCL have a progressive disease course, some cases with a predominantly extranodal disease distribution have been recognized with more indolent clinical behavior. These cases have CCND1 translocations, tend not to express SOX11, and commonly have hypermutated IGH (as opposed to most MCL with unmutated IGH) (Fernàndez et al. 2010). This disease has been codified in the WHO classification as “leukemic non-nodal MCL.” Although generally indolent, some patients with leukemic non-nodal MCL exhibit disease progression, often associated with acquisition of 17p/TP53 alterations (Carvajal-Cuenca et al. 2012).

Hairy Cell Leukemia/Splenic Lymphomas

Hairy cell leukemia (HCL) is a rare and clinically indolent mature B-cell leukemia/lymphoma that preferentially involves the spleen, blood, and bone marrow. As opposed to most mature B-cell leukemias that typically present with lymphocytosis, HCL typically manifests with cytopenias, with relatively few circulating neoplastic lymphocytes. HCL cells have characteristic morphologic features and immunophenotypic characteristics (Shao et al. 2013). Until the development of NGS techniques, the genetic basis of HCL was unclear, as few recurrent structural alterations were present in the HCL genome (Forconi et al. 2008). However, in 2011, activating BRAF Val600Glu mutations were identified in nearly all cases of HCL (Tiacci et al. 2011). BRAF mutations are exceptionally uncommon in other B-cell lymphomas, suggesting this is a relatively specific diagnostic marker for HCL (Tiacci et al. 2011; Arcaini et al. 2012). Rare BRAF-negative HCL cases often exhibit preferential usage of IGHV4-34 and frequently have downstream mutations in MAP2K1 (Waterfall et al. 2014). BRAF-mutated HCL may also exhibit loss of wild-type BRAF through deletions, along with infrequent mutations of KMT2C and CDKN1B (Durham et al. 2017).

MAP2K1 mutations are also present in ∼30% of HCL variant (HCL-v) cases (Waterfall et al. 2014), a neoplasm with morphologic and immunophenotypic features similar to but distinct from classic HCL, and with poorer responses to single-agent purine analogs. HCL-v cases additionally exhibit recurrent mutations in CCND3, U2AF1, ARID1A, and TP53 (Waterfall et al. 2014; Durham et al. 2017). The finding of activated MAPK pathways in both HCL and HCL-v unify the pathobiology of these lymphoid leukemias to some degree and point to therapeutic targets in these diseases. Although HCL is relatively well managed by purine analogs, this is not the case for HCL-v. BRAF inhibition has been used successfully in HCL patients in the refractory/relapsed setting (Tiacci et al. 2015), and MEK inhibition has been anecdotally successful in HCL-v (Andritsos et al. 2018).

Splenic Marginal Zone Lymphoma

Splenic marginal zone lymphoma (SMZL) is a low-grade B-cell lymphoma that, like HCL and HCL-v, preferentially involves the blood, bone marrow, and spleen. SMZLs exhibit common karyotypic alterations, with frequent gains of 3 and 3q, gains of 12q, and deletions of 7q and 6q (Salido et al. 2010). NOTCH2 and KLF2 are the most frequently mutated genes in SMZL (Kiel et al. 2012; Rossi et al. 2012; Clipson et al. 2015). Both proteins are important regulators of normal marginal zone formation, and they are preferentially mutated in SMZL versus other low-grade B-cell lymphomas. NOTCH2 and TP53 mutations are associated with decreased treatment-free survival and overall survival, respectively, in SMZL (Parry et al. 2015; Campos-Martín et al. 2017).

Lymphoplasmacytic Lymphoma

Lymphoplasmacytic lymphoma (LPL) is an indolent B-cell lymphoma typically associated with marrow involvement, some degree of plasmacytic differentiation, and IgM monoclonal gammopathy. Waldenström macroglobulinemia (WM) is defined by the presence of an IgM paraprotein, with >10% marrow involvement by LPL. WM/LPL tumors frequently exhibit loss of 6q, deleting TNFAIP3 (Braggio et al. 2009). NGS studies identified MYD88 Leu265Pro mutations in ∼90% of patients with LPL (Treon et al. 2012). MYD88 Leu265Pro mutations are relatively uncommon in other low-grade B-cell lymphomas, although they do rarely occur in SMZL and CLL. Both activating mutations in MYD88 and deletions of TNFAIP3 lead to activation of the NF-κΒ signaling pathway. Patients with MYD88-mutated LPL appear very sensitive to BTK inhibition with ibrutinib, but efficacy is diminished in patients with concurrent mutations of CXCR4 and in the rare MYD88-wild type cases (Treon et al. 2015; Dimopoulos et al. 2017).

Follicular Lymphoma

Follicular lymphoma (FL) is the most common indolent B-cell lymphoma in western countries, accounting for ∼20% of all lymphomas worldwide (The Non-Hodgkin's Lymphoma Classification Project 1997). It is composed of germinal center–derived B cells and frequently exhibits a follicular growth pattern. Its genetic hallmark is the translocation of chromosomes 14 and 18, causing the antiapoptotic gene BCL2 to be up-regulated by the IGH enhancer, similar to the CCND1 rearrangement seen in MCL (Fukuhara et al. 1979; Tsujimoto et al. 1984). IGH/BCL2 is by itself insufficient to cause lymphoma (Roulland et al. 2006) and requires the presence of additional genetic aberrations. In addition to the t(14;18), FL harbors frequent losses of 1p, 6q, 10q, and 17p, with gains of 1q, 6q, 7, 8q, 12q, 17q, and 18 (Cheung et al. 2009). FL contains several highly recurrent mutations in addition to IGH/BCL2 fusion. Mutations in chromatin modifiers are particularly frequent, with KMT2D, CREBBP, EZH2, and EP300 being common mutational targets (Morin et al. 2010, 2011; Pasqualucci et al. 2011). Mutations of chromatin modifiers are present in >90% of FLs, and >70% have mutations in two or more chromatin-modifying genes (Green et al. 2015). In addition to its frequent deletion at chromosome 1p in FL, TNFRSF14 is commonly mutationally inactivated, and TNFRSF14 alterations are present in more than one-half of FL cases (Cheung et al. 2010; Launay et al. 2012). Loss of TNFRSF14 leads to B-cell proliferation through increased BCR signaling and leads to changes in the FL microenvironment that facilitate tumor growth (Boice et al. 2016).

Although FL is an indolent lymphoma, it is generally incurable with conventional immunochemotherapeutic approaches, and transformation to an aggressive B-cell lymphoma is a frequent and clinically devastating event. Several recent studies have explored the underlying genetic basis of transformation in follicular lymphoma. Transformed FL (t-FL) appears to arise from an ancestral clone shared with the dominant FL, rather than as a direct progression of the dominant FL clone. t-FL is enriched for alterations of MYC, CDKN2A/B, TP53, PIM1, EBF1, and B2M (Elenitoba-Johnson et al. 1998; Pasqualucci et al. 2014; Okosun et al. 2014).

A modification of the standard FL international prognostic index (FLIPI) has been proposed that incorporates the presence of mutations in seven commonly mutated genes (m7-FLIPI) (Pastore et al. 2015). In this prognostic model, EZH2, ARID1A, and MEF2B mutations were favorable (particularly EZH2), whereas mutations in EP300, FOXO1, CREBBP, and CARD11 were unfavorable (particularly EP300 and FOXO1). The m7-FLIPI model improved on standard FLIPI risk stratification in FL patients by identifying patients with high-risk FLIPI scores but with favorable outcomes, largely associated with EZH2 mutation.

Pediatric-type FL (PTFL) is an uncommon and highly indolent lymphoma of germinal center B cells that typically occurs in adolescent males. In contrast to conventional FL, PTFL is less genomically complex, with significantly fewer aberrations and without translocations of BCL2 or BCL6. Like FL, PTFL exhibits frequent losses of 1p/TNFRSF14 mutations, but mutations of chromatin modifiers are virtually absent, whereas MAP2K1 mutations are relatively frequent (Louissaint et al. 2016; Schmidt et al. 2016).

Diffuse Large B-Cell Lymphoma

Diffuse large B-cell lymphoma (DLBCL) is the most common lymphoma worldwide (The Non-Hodgkin's Lymphoma Classification Project 1997). The term DLBCL encompasses a genetically diverse group of neoplasms with the common feature of a diffuse proliferation of large, transformed B cells. The heterogeneity of DLBCL was recognized in lymphoma classification systems for decades, but subclassification based on morphologic features is not reproducible among experts and does not clearly associate with outcomes (Harris et al. 1994). In 2000, a landmark study demonstrated that DLBCL could be subdivided into clinically relevant categories based on gene expression as assessed by cDNA microarray analysis (Alizadeh et al. 2000). One large group of DLBCLs exhibited a gene expression profile resembling that of germinal center B cells (GCBs), whereas another exhibited a profile similar to that of activated peripheral blood B cells (ABCs). Some cases remained unclassifiable by this approach. Patients with GCB-DLBCL had better outcomes than patients with ABC-DLBCL when treated with CHOP (Rosenwald et al. 2002), and the survival differences persisted with the addition of rituximab to the CHOP regimen (Lenz et al. 2008). Assessment of gene expression signatures has not historically been clinically feasible, and so surrogate immunohistochemical algorithms were developed and implemented clinically (Hans et al. 2004). Newer gene expression technologies, however, hold the promise of robust distinction between ABC- and GCB-DLBCL in routinely processed formalin-fixed, paraffin-embedded material (Scott et al. 2014).

The GCB/ABC paradigm has informed subsequent genetic exploration of DLBCL. Both ABC and GCB subsets exhibit relatively frequent translocations of BCL6 and mutations of KMT2D, CREBBP, EP300, B2M, TP53, and MEF2B; GCB DLBCL exhibit frequent IGH/BCL2 translocation, mutations of EZH2, and MYC translocations; and ABC DLBCL often have alterations of TNFAIP3, CDKN2A/B, MYD88, PRDM1, CD79A/B, CARD11, and frequently exhibit amplification (rather than translocation) of BCL2 (for review, see Pasqualucci and Dalla-Favera 2015). Many of the mutations identified in ABC-DLBCL lead to chronic active BCR signaling and NF-κB activation, pointing to targetable pathways in this chemoresistant disease (Davis et al. 2010; Ngo et al. 2011).

Recent studies have proposed classifications of DLBCL based on their mutational signatures (Chapuy et al. 2018; Schmitz et al. 2018). Although the studies exhibited some differences in their findings, several common themes emerge, emphasizing that the biologic and clinical heterogeneity of DLBCL cannot be captured with a trichotomous ABC/GCB/other gene expression classification. In both studies, two subgroups were recognized in ABC-DLBCL: One had translocations of BCL6 and mutations of genes in the NOTCH2 pathway, suggesting a possible shared cell of origin with MZL, as splenic MZLs frequently harbor NOTCH2 pathway mutations (Kiel et al. 2012; Rossi et al. 2012) and transformed gastric extranodal MZLs frequently have rearrangements of BCL6 (Chen et al. 2006; Flossbach et al. 2011).This group was associated with relatively favorable survival in both studies and was also enriched in cases with an unclassifiable gene expression profile (neither clearly ABC or GCB). The other ABC cohort consisted of lymphomas with MYD88 Leu265Pro mutations and mutations of CD79B. This group had an unfavorable prognosis and was enriched for lymphomas with extranodal involvement, concordant with results of previous studies that have identified frequent mutations of MYD88 in primary extranodal lymphomas such as primary CNS lymphoma, testicular DLBCL, and primary cutaneous DLBCL, leg-type (Pham-Ledard et al. 2014; Chapuy et al. 2016). In both studies, a group of GCB-DLBCL characterized by BCL2 mutation/rearrangement and mutation of EZH2 was identified with worse survival than other GCB-DLBCLs. The remaining GCB cases were further characterized by Chapuy and colleagues as commonly having mutations in four linker and four core histone genes, along with CD83, CD58, CD70, RHOA, GNA13, SGK1, CARD11, NFKBIE, NFKBIA, BRAF, and STAT3, without mutations in chromatin modifiers. Chapuy and colleagues also recognized a TP53-mutated cohort with multiple structural chromosomal abnormalities, and Schmitz et al. recognized a small cohort of NOTCH1-mutated ABC DBLCL with adverse outcomes.

These efforts point to the biologic complexity of a category that is still classified primarily by its morphologic appearance and that is still treated homogeneously at diagnosis. Although frontline immunochemotherapy for DLBCL has not changed significantly since the introduction of rituximab in the early 2000s, novel agents are now routinely used in the in the relapsed/refractory setting. Particularly favorable responses have been seen in relapsed/refractory ABC DLBCL to agents such as ibrutinib and lenalidomide (Wilson et al. 2015; Czuczman et al. 2017). The specific mutations present in an individual case of DLBCL may better predict response to targeted therapies than the overall gene expression profile, as responses to ibrutinib in ABC DLBCL appear enriched in MYD88/CD79B comutated DLBCL cases (Wilson et al. 2015). The genetic heterogeneity that has been uncovered in DLBCL should inform clinical trials going forward so that possible responses in particular genomic subgroups are not lost.

Burkitt Lymphoma

Burkitt lymphoma (BL) is a highly aggressive mature B-cell neoplasm that arises in different epidemiologic scenarios. Endemic BL is highly associated with EBV infection and occurs in areas with endemic malaria; sporadic BL occurs worldwide and is less frequently associated with EBV; and immunodeficiency-associated BL occurs in patients with immunodeficiency, most commonly HIV infection (Swerdlow et al. 2017). BL is highly aggressive, but curable with intensive chemoimmunotherapy, and its distinction from DLBCL is paramount in adults.

BL is the prototypical example of a lymphoma with a recognized genetic abnormality: extra material on chromosome 14 was first recognized in BL in 1972 (Manolov and Manolova 1972). This material was soon characterized as originating from chromosome 8 (Zech et al. 1976), and the IGH/MYC rearrangement was described soon after (Dalla-Favera et al. 1982; Taub et al. 1982). All epidemiologic types of BL are highly associated with translocations of MYC to immunoglobulin loci, with only rare MYC-fusion-negative cases that may have MYC deregulation by alternate mechanisms (Hummel et al. 2006; Leucci et al. 2008). MYC translocations are not specific to BL, however, as they are present in a minor subset of DBLCL and in many non-BL high-grade B-cell lymphomas (discussed below). Therefore, accurate diagnosis requires integration of genetic, morphologic, and phenotypic findings. Molecular studies may help distinguish between BL and DLBCL, as aside from MYC translocations, BL exhibits few structural alterations, in contrast to DLBCL (Hummel et al. 2006). Sporadic cases of BL exhibit high-frequency mutations of TCF3 or its negative regulator ID3, and oncogenic mutations of CCND3 are common (Schmitz et al. 2012). EBV-associated endemic BL, in contrast, exhibits less frequent mutations of TCF3, ID3, and CCND3, with more frequent mutations of ARID1A, RHOA, and CCNF (Schmitz et al. 2012; Abate et al. 2015). Many genes commonly mutated in DLBCL are only rarely mutated in BL, such as EZH2, SGK1, BCL2, CD79B, and MYD88.

High-Grade B-Cell Lymphomas with MYC and BCL2 and/or BCL6 Rearrangements

High-grade B-cell lymphoma (HGBL) with MYC and BCL2 and/or BCL6 rearrangements is a category introduced in the 2016 revision of the WHO Classification that encompasses cases that previously would have been classified as “B-cell lymphoma, unclassifiable, with features intermediate between DLBCL and BL.” As the name suggests, this category is intended to encompass all mature, aggressive-appearing lymphomas that harbor translocations of MYC, along with a translocation of one or both of BCL2 and BCL6. These lymphomas are commonly referred to as “double-hit” lymphomas (DHLs) and have been associated with very poor outcomes when treated with conventional DLBCL therapy (Johnson et al. 2009; Li et al. 2012). They may have Burkitt-like, blastoid, or DLBCL-like morphology. The introduction of this category and its clinical implications has mandated FISH testing for these fusions in most, if not all, cases of large B-cell lymphoma, as strategies that FISH only cases with a GCB phenotype and MYC/BCL2 protein expression miss a large percentage of rearranged cases (Scott et al. 2018). Selection of FISH probes impacts sensitivity of MYC rearrangement detection as breaks may occur over several megabases. The most comprehensive methods would rely on application of both break-apart and dual-fusion FISH probes (May et al. 2010; Muñoz-Mármol et al. 2013), although this approach is resource-intensive. Several groups have reported that the MYC translocation partner may be prognostically significant, and that lymphomas with nonimmunoglobulin (non-IG) MYC translocation partners have better outcomes than those with IG/MYC fusions (Johnson et al. 2009; Pedersen et al. 2014; Copie-Bergman et al. 2015; Chong et al. 2018), further complicating possible FISH analysis. The most appropriate regimen for DHL is not clear, but patients benefit from regimens more intensive than standard R-CHOP therapy (Friedberg 2017).

Primary Mediastinal (Thymic) Large B-Cell Lymphoma and Classic Hodgkin Lymphoma

Primary mediastinal (thymic) large B-cell lymphoma (PMBL) is an uncommon genetically distinct subtype of large B-cell lymphoma that occurs in the mediastinum, often in young females. Although it exhibits more robust B-cell antigen expression, PMBL exhibits some morphologic, immunophenotypic, and genetic similarity to classic Hodgkin lymphoma (CHL), which also commonly occurs in the mediastinum in younger patients. Early gene expression profiling studies identified PMBL as distinct from DLBCL more generally, and with shared gene expression features with CHL (Rosenwald et al. 2003; Savage et al. 2003). Later mutational studies have confirmed additional shared features with CHL, including 9p24.1 amplifications involving JAK2, CD274 (PDL1) and PDCD1LG2 (PDL2) (Green et al. 2010); CIITA fusions (Steidl et al. 2011); SOCS1 inactivation (Melzner et al. 2005; Weniger et al. 2006); and STAT6 mutations (Ritz et al. 2009; Tiacci et al. 2018). Recent studies have identified IL4R mutations as another activator of JAK-STAT signaling in PMBL (Viganò et al. 2018a), and these mutations are also present in some cases of DLBCL (Viganò et al. 2018b). Nonmediastinal PMBL have been described, yet they are difficult to confidently diagnose (Yuan et al. 2015). As PMBL is often treated more aggressively than most DLBCLs (Dunleavy et al. 2013), recognition of nonmediastinal PMBL could have clinical implications. Mutational analysis or newer gene expression techniques may assist with this rare diagnosis and could be helpful to support the impression of PMBL when diagnosing a large B-cell lymphoma in the mediastinum (Chong et al. 2018).

Genetic studies of CHL have been historically limited by the relative paucity of the malignant Hodgkin Reed–Sternberg (HRS) cells in most biopsy specimens. However, novel techniques have allowed single-cell interrogation of HRS cells, revealing a mutational profile with significant overlap with PMBL, as discussed above. Copy number alterations of the PDL1/PDL2 locus are particularly common in CHL, occurring in nearly every lymphoma in one series (Roemer et al. 2016). Amplifications (as opposed to polysomy or low-level copy gain) of PDL1/PDL2 were associated with decreased progression-free survival and advanced-stage disease in this study and are a likely mechanism for the encouraging clinical responses to immune checkpoint blockade seen in CHL (Ansell et al. 2015). Although relatively few cases of EBV+ CHL have been comprehensively genetically studied, they appear to have a lower mutational burden than EBV CHL and contain fewer copy number alterations (Montgomery et al. 2016; Tiacci et al. 2018).

T-Cell Lymphomas

Anaplastic Large Cell Lymphoma

Anaplastic large cell lymphoma (ALCL) is a CD30+ mature T-cell lymphoma that is divided into two primary groups on the basis of its genetic abnormalities: ALCL, ALK-positive (ALK+ ALCL) and ALCL, ALK-negative (ALK ALCL). ALK+ ALCL is characterized by the fusion of the tyrosine kinase ALK to other genes, most commonly NPM1 (Le Beau et al. 1989; Rimokh et al. 1989; Morris et al. 1994). This leads to aberrant expression of the ALK protein, easily identifiable by immunohistochemical staining (Pittaluga et al. 1997; Pulford et al. 1997; Falini et al. 1998). ALK expression activates multiple signaling pathways, including JAK-STAT, PI3K-AKT, mTOR, MAPK, and others (for review, see Hallberg and Palmer 2013). Small-molecule inhibitors of ALK, such as crizotinib, have demonstrated clinical efficacy in ALK+ ALCL (Mossé et al. 2017).

ALK ALCL, by definition, lacks ALK rearrangement or overexpression. In comparison with ALK+ ALCL, ALK ALCL tend to exhibit a somewhat more complex genome, with a higher frequency of chromosomal gains and losses (Boi et al. 2013). ALK ALCL exhibits a heterogeneous clinical course, and emerging evidence demonstrates that outcomes are heavily dependent on the underlying lymphoma genetics. DUSP22 rearrangements are present in ∼30% of ALK ALCLs, including cutaneous forms of ALCL (Feldman et al. 2009, 2011; Parrilla Castellar et al. 2014). More rarely, TP63 rearrangements occur in ALK ALCL (Vasmatzis et al. 2012; Parrilla Castellar et al. 2014), and together these translocations identify clinically relevant ALK ALCL subgroups. DUSP22-rearranged systemic ALK ALCL have a favorable prognosis similar to that seen in ALK+ ALCL, whereas TP63-rearranged cases do poorly with conventional chemotherapy (Parrilla Castellar et al. 2014; Pedersen et al. 2017). Other recurrent kinase fusions, such as those involving TYK2, occur with less frequency in ALK ALCL (Velusamy et al. 2014; Crescenzo et al. 2015) but activate JAK-STAT signaling, whereas mutations of JAK1 and/or STAT3 are present in ∼20% of cases (Crescenzo et al. 2015). Although the introduction of brentuximab vedotin (an anti-CD30 antibody-drug conjugate) has led to improved therapies in ALK+ and ALK ALCL (Horwitz et al. 2019), the identification of mutationally activated pathways provides alternate drug targets for therapeutic exploration.

T-Cell Prolymphocytic Leukemia

T-cell prolymphocytic leukemia (T-PLL) is a rare mature T-cell leukemia that typically presents with marked lymphocytosis and is associated with poor survival. T-PLL is commonly associated with inv(14) (Malcikova et al. 2018), which juxtaposes the TRA/D locus with the oncogenes TCL1A and TCL1B (Brito-Babapulle et al. 1987; Russo et al. 1989; Pekarsky et al. 1999). This translocation is analogous to the IG translocations commonly seen in B-cell lymphomas, causing the aberrant expression of TCL1A/B owing to its localization near the TRA/D enhancer, leading to increased AKT1 signaling (Pekarsky et al. 2000). Additional common structural events in T-PLL include loss of the ATM gene at 11q23 and trisomy 8. In addition to these large-scale genomic events, point mutations in the JAK-STAT pathway are common (Kiel et al. 2014). Recurrent mutations of JAK1, JAK3, and STAT5B are present in ∼8%, ∼30%, and ∼36% of T-PLL cases, respectively. Inactivating mutations and deletions of EZH2 are additionally present in about one-half of cases (Kiel et al. 2014). Alemtuzumab, an anti-CD52 antibody, has been the mainstay of therapy for T-PLL, but this regimen is not curative and can be associated with significant complications (Dearden 2012). Alternate therapeutic approaches are therefore needed. Although JAK-STAT kinase inhibitors are clinically available, it does not appear that response to JAK-STAT inhibition is readily predictable by mutational status in ex vivo studies (Andersson et al. 2018).

Mycosis Fungoides/Sézary Syndrome

Mycosis fungoides (MF) is the most common cutaneous T-cell lymphoma (CTCL), and it typically exhibits a fairly indolent disease course. However, MF may progress to involve the blood, exhibiting clinical and pathologic overlap with Sézary syndrome (SS), which strictly defined is a de novo aggressive and rare process characterized by erythroderma, lymphadenopathy, and circulating lymphoma cells. MF harbors recurrent mutations of PLCG1 (Kiel et al. 2015), leading to activation of pathways downstream of the T-cell receptor (Vaqué et al. 2014). TP53 mutations occur in MF, more commonly in tumor-stage patients than at earlier stages (McGregor et al. 1999). MF exhibits a relatively simple genome (da Silva Almeida et al. 2015), whereas SS exhibits a highly complex genome, typically with numerous copy number changes and structural rearrangements (Choi et al. 2015; da Silva Almeida et al. 2015; Kiel et al. 2015; Wang et al. 2015b). Common mutational/deletional targets in SS include numerous genes involved in epigenetic regulation (ARID1A, ZEB1, DMNT3A, TET2, CREBBP, and KMT2D), components of several signaling pathways (CARD11, CCR4, PLCG1, BRAF, MAKP1, JAK1, JAK3, STAT3, and STAT5B) and tumor suppressors such as CDKN2A and TP53. CCR4 is a therapeutic target in CTCL, as mogamulizumab, an anti-CCR4 antibody, has recently been Food and Drug Administration (FDA)-approved in the relapsed/refractory setting (Kim et al. 2018). It is not clear that CCR4 mutational status impacts mogamulizumab response in CTCL, although CCR4 mutations sensitize to mogamulizumab therapy in the HTLV1-associated adult T-cell leukemia/lymphoma (Sakamoto et al. 2018).

Nodal T Follicular Helper T-Cell Lymphomas

A large group of nodal peripheral T-cell lymphomas exhibit phenotypic characteristics of normal follicular helper T-cells (TFHs). Angioimmunoblastic T-cell lymphomas (AITLs) and follicular T-cell lymphoma (FTCL) are the two main nodal TFH lymphoma subtypes recognized by the WHO classification (Swerdlow et al. 2017). Recent NGS studies have identified recurrent mutations in epigenetic regulators in both AITL and FTCL, contributing to the recognition that these lymphomas exhibit overlapping biologic features, in addition to a largely shared immunophenotype. Both AITL and FTCL exhibit common mutations of TET2, DNMT3A, and RHOA (Lemonnier et al. 2012; Odejide et al. 2014; Palomero et al. 2014; Sakata-Yanagimoto et al. 2014). IDH2 mutations, in contrast, seem to be restricted to AITL, present in ∼20%–30% of cases (Cairns et al. 2012; Odejide et al. 2014; Wang et al. 2015a). The IDH2 mutations in AITL seem to always target the arginine residue at codon 172, rather than the Arg140 mutations that predominate in acute myeloid leukemia (Marcucci et al. 2010), and in contrast with acute myeloid leukemia, IDH2 and TET2 mutations are not mutually exclusive in AITL (Lemonnier et al. 2012). Arg172 mutations lead to greater 2-HG accumulation than do Arg140 mutations (Ward et al. 2013), and IDH mutations identify a subset of AITL patients with increased H3K27me3 and hypermethylation of gene promotor regions compared to IDH-wild type cases (Wang et al. 2015a). Demethylating agents have demonstrated some clinical efficacy against the lymphoma in AITL patients with concurrent myeloid neoplasms, suggesting that epigenetic modification may be a promising therapeutic approach in AITL (Lemonnier et al. 2018).

Impact of Molecular and Genomic Techniques on Lymphoma Diagnosis and Monitoring

Immunoglobulin/T-Cell Receptor Sequencing

During normal development, lymphocytes undergo somatic V(D)-J recombination of their immunoglobulin (IG) or T-cell receptor (TR) genes. This process generates a genetic “fingerprint” for each unique lymphocyte, as variable numbers of nontemplated nucleotides are inserted at the junctions of the V, D, and J genes, leading to the normal IG and TR repertoire necessary for normal antigen recognition. As lymphomas arise from a single progenitor cell, they exhibit a common, dominant V(D)-J rearrangement, as opposed to a multitude of rearrangements in reactive lymphoproliferations. This phenomenon has long been exploited in the diagnosis of lymphoma, and the detection of monoclonality through assessment of IG or TR diversity through Southern blotting or polymerase chain reaction (PCR) techniques has been a routine ancillary diagnostic technique in hematopathology for decades (van Dongen and Wolvers-Tettero 1991; van Dongen et al. 2003). Routine PCR techniques for monoclonality assessment retain advantages in turnaround time, cost, and ease of analysis. However, using typical capillary electropherograms, the amplified PCR products are separated solely on the basis of size, and each peak in the electropherogram represents the integrated signal of potentially many clones. Although this is not a problem in most cases, this phenomenon can make it difficult to always compare clones between specimens with great confidence and may lead to false-positive calls. Next-generation sequencing (NGS) techniques, on the other hand, generate the sequence itself of each individual V(D)-J read, and reads with shared sequences can be summed to give accurate quantification of each clone present in a specimen.

NGS techniques therefore allow measurement not only of whether a clone is present, but routine determination of hypermutation status and gene family usage, clinically relevant information in common diseases such as CLL/SLL (Byrd et al. 2006; Ghia et al. 2008). NGS techniques allow straightforward and definitive comparison between lymphomas in an individual patient to assess for clonal relatedness, a particularly important determination in the setting of transformation (Rossi et al. 2011). Perhaps the most powerful application of NGS for lymphoid clonality assessment is in the disease monitoring setting. Although minimal residual disease (MRD) testing can be successfully performed with conventional PCR (van der Velden et al. 2007), patient-specific primers are necessary to gain appropriate sensitivity, which is laborious and difficult to implement in most clinical laboratories. As NGS outputs the sequence of each individual read obtained in a specimen, MRD-level detection of lymphoma/leukemia-specific reads can be easily achieved with common primers as long as a sufficiently large number of reads are obtained and the sequence of the diagnostic IG or TR rearrangement is known. This technique has been used for successful disease monitoring in mature lymphomas, acute leukemias, and plasma cell myelomas (Wu et al. 2012; Kurtz et al. 2015; Roschewski et al. 2015; Scherer et al. 2016; Rossi et al. 2017; Perrot et al. 2018; Wood et al. 2018), as the only requirement is the presence of either an IG or TR rearrangement in the neoplastic cells. NGS techniques have recently been FDA-approved in the B-ALL and myeloma MRD setting, and the performance characteristics of the assays compare favorably to that of multiparameter flow cytometry in these diseases.

Clinical Genomics Testing in Lymphoma

Genetic testing in lymphoma has long been performed routinely in the clinical laboratory through comprehensive, but low-resolution, methods such as metaphase karyotyping or through targeted interrogation of specific loci such as through FISH studies. More recently, testing for individual genetic mutations has become common in some circumstances, such as testing for BRAF mutations in suspected HCL or MYD88 mutations in suspected LPL. The recent introduction of high-throughput genomic platforms into the clinical laboratory has made routine extensive genomic characterization of lymphomas feasible, and examples of clinically relevant testing are listed in Table 1.

Table 1.

Examples of genetic biomarkers and possible therapeutic implication in lymphoma entities

Disease Genetic biomarker Therapeutic implication
CLL TP53 mutation/deletion Ibrutinib therapy indication
CLL BTK or PLCG2 mutation Ibrutinib resistance
LPL MYD88 mutation Reduced ibrutinib response if MYD88 is not mutated
LPL CXCR4 mutation Reduced ibrutinib response if CXCR4 is mutated
HCL BRAF mutation Vemurafenib response
HGBL MYC and BCL2 and/or BCL6 rearrangements R-CHOP is suboptimal therapy
DLBCL MYD88/CD79B comutation Increased response to ibrutinib
FL/DLBCL EZH2 mutation EZH2 inhibitors in clinical trials
AITL IDH2 mutation IDH2 inhibitors in clinical trials
ALCL, ALK+ ALK rearrangement May respond to crizotinib
ALCL, ALK DUSP22 rearrangement May be candidate for less intensive therapy than other ALCL, ALK

(CLL) Chronic lymphocytic leukemia, (LPL) lymphoplasmacytic lymphoma, (HCL) hairy cell leukemia, (HGBL) high-grade B-cell lymphoma, (DLBCL) diffuse large B-cell lymphoma, (FL) follicular lymphoma, (AITL) angioimmunoblastic T-cell lymphoma, (ALCL) anaplastic large cell lymphoma, (ALK+) ALK receptor tyrosine kinase positive, (ALK) ALK receptor tyrosine kinase negative.

As discussed previously, recent studies have shown that the traditional, gene expression–based classification of DLBCL (the most common lymphoma type) into ABC and GCB subtypes is overly simplistic, with prognostically important subsets only identifiable by broad genomic profiling (Chapuy et al. 2018; Schmitz et al. 2018). Emerging data suggest that at least some of these genetic fingerprints predict for response to targeted therapies such as ibrutinib (Wilson et al. 2015), but it is likely that other factors in addition to mutational activation impact therapeutic responses, particularly when targeting pathways that are generally important in lymphoid survival (Phelan et al. 2018). As many mutations are present at relatively low frequency in most lymphoma subtypes, broad testing panels would be necessary to identify rare but potentially targetable lesions, such the infrequent but recurrent mutations of BRAF in nodal marginal zone lymphomas (Pillonel et al. 2018). Although examples of genetically directed, targeted therapy remain relatively few in lymphoma, ever-increasing numbers of novel agents are being evaluated in clinical trials, particularly in the setting of relapsed/refractory disease, and comprehensive genetic profiling of lymphomas should be considered in such settings both to identify targets and to identify the determinants of response to novel therapies.

CONCLUSION

NGS technologies have led to an exponential increase in our understanding of the pathogenesis of mature lymphoid leukemias and lymphomas. Although relatively few lymphomas had been associated with karyotypically recognizable abnormalities such as chromosomal translocations, recurrent mutations are now known in virtually every lymphoma subtype. With some exceptions, such as the very high incidence of BRAF mutations in HCL, most lymphoma types are not associated with a single highly recurrently mutated gene or recurrent translocation. Rather, lymphomas exhibit mutational complexity, with a long tail of mutations that are present in relatively few patients. However, within this genetic heterogeneity new subgroups have been recognized, possibly with divergent responses to novel therapeutic agents. Our greater understanding of mutational processes in lymphoma holds the promise of better and less toxic therapies for patients with these devastating diseases.

Footnotes

Editors: Michael G. Kharas, Ross L. Levine, and Ari M. Melnick

Additional Perspectives on Leukemia and Lymphoma: Molecular and Therapeutic Insights available at www.perspectivesinmedicine.org

REFERENCES

  1. Abate F, Ambrosio MR, Mundo L, Laginestra MA, Fuligni F, Rossi M, Zairis S, Gazaneo S, De Falco G, Lazzi S, et al. 2015. Distinct viral and mutational spectrum of endemic Burkitt lymphoma. PLoS Pathog 11: e1005158 10.1371/journal.ppat.1005158 [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Alizadeh AA, Eisen MB, Davis RE, Ma C, Lossos IS, Rosenwald A, Boldrick JC, Sabet H, Yu X, Powell JI, et al. 2000. Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature 403: 503–511. 10.1038/35000501 [DOI] [PubMed] [Google Scholar]
  3. Andersson EI, Pützer S, Yadav B, Dufva O, Khan S, He L, Sellner L, Schrader A, Crispatzu G, Oleś M, et al. 2018. Discovery of novel drug sensitivities in T-PLL by high-throughput ex vivo drug testing and mutation profiling. Leukemia 32: 774–787. 10.1038/leu.2017.252 [DOI] [PubMed] [Google Scholar]
  4. Andritsos LA, Grieselhuber NR, Anghelina M, Rogers KA, Roychowdhury S, Reeser JW, Timmers CD, Freud AG, Blachly JS, Lucas DM, et al. 2018. Trametinib for the treatment of IGHV4-34, MAP2K1-mutant variant hairy cell leukemia. Leuk Lymphoma 59: 1008–1011. 10.1080/10428194.2017.1365853 [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Ansell SM, Lesokhin AM, Borrello I, Halwani A, Scott EC, Gutierrez M, Schuster SJ, Millenson MM, Cattry D, Freeman GJ, et al. 2015. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin's lymphoma. New Engl J Med 372: 311–319. 10.1056/NEJMoa1411087 [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Arcaini L, Zibellini S, Boveri E, Riboni R, Rattotti S, Varettoni M, Guerrera ML, Lucioni M, Tenore A, Merli M, et al. 2012. The BRAF V600E mutation in hairy cell leukemia and other mature B-cell neoplasms. Blood 119: 188–191. 10.1182/blood-2011-08-368209 [DOI] [PubMed] [Google Scholar]
  7. Banks PM, Chan J, Cleary ML, Delsol G, De Wolf-Peeters C, Gatter K, Grogan TM, Harris NL, Isaacson PG, Jaffe ES, et al. 1992. Mantle cell lymphoma. A proposal for unification of morphologic, immunologic, and molecular data. Am J Surg Pathol 16: 637–640. 10.1097/00000478-199207000-00001 [DOI] [PubMed] [Google Scholar]
  8. Beà S, Ribas M, Hernández JM, Bosch F, Pinyol M, Hernández L, García JL, Flores T, González M, López-Guillermo A, et al. 1999. Increased number of chromosomal imbalances and high-level DNA amplifications in mantle cell lymphoma are associated with blastoid variants. Blood 93: 4365–4374. [PubMed] [Google Scholar]
  9. Beà S, Valdés-Mas R, Navarro A, Salaverria I, Martín-Garcia D, Jares P, Giné E, Pinyol M, Royo C, Nadeu F, et al. 2013. Landscape of somatic mutations and clonal evolution in mantle cell lymphoma. Proc Natl Acad Sci 110: 18250–18255. 10.1073/pnas.1314608110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Bentz M, Plesch A, Bullinger L, Stilgenbauer S, Ott G, Müller-Hermelink HK, Baudis M, Barth TF, Möller P, Lichter P, et al. 2000. t(11;14)-positive mantle cell lymphomas exhibit complex karyotypes and share similarities with B-cell chronic lymphocytic leukemia. Genes Chromosomes Cancer 27: 285–294. 10.1002/(SICI)1098-2264(200003)27:33.3.CO;2-D [DOI] [PubMed] [Google Scholar]
  11. Boi M, Rinaldi A, Kwee I, Bonetti P, Todaro M, Tabbò F, Piva R, Rancoita PM, Matolcsy A, Timar B, et al. 2013. PRDM1/BLIMP1 is commonly inactivated in anaplastic large T-cell lymphoma. Blood 122: 2683–2693. 10.1182/blood-2013-04-497933 [DOI] [PubMed] [Google Scholar]
  12. Boice M, Salloum D, Mourcin F, Sanghvi V, Amin R, Oricchio E, Jiang M, Mottok A, Denis-Lagache N, Ciriello G, et al. 2016. Loss of the HVEM tumor suppressor in lymphoma and restoration by modified CAR-T cells. Cell 167: 405–418.e13. 10.1016/j.cell.2016.08.032 [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Braggio E, Keats JJ, Leleu X, Van Wier S, Jimenez-Zepeda VH, Valdez R, Schop RF, Price-Troska T, Henderson K, Sacco A, et al. 2009. Identification of copy number abnormalities and inactivating mutations in two negative regulators of nuclear factor-κB signaling pathways in Waldenström's macroglobulinemia. Cancer Res 69: 3579–3588. 10.1158/0008-5472.CAN-08-3701 [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Brito-Babapulle V, Pomfret M, Matutes E, Catovsky D. 1987. Cytogenetic studies on prolymphocytic leukemia. II: T cell prolymphocytic leukemia. Blood 70: 926–931. 10.1182/blood.V70.4.926.926 [DOI] [PubMed] [Google Scholar]
  15. Brown JR. 2018. How I treat CLL patients with ibrutinib. Blood 131: 379–386. 10.1182/blood-2017-08-764712 [DOI] [PubMed] [Google Scholar]
  16. Byrd JC, Gribben JG, Peterson BL, Grever MR, Lozanski G, Lucas DM, Lampson B, Larson RA, Caligiuri MA, Heerema NA. 2006. Select high-risk genetic features predict earlier progression following chemoimmunotherapy with fludarabine and rituximab in chronic lymphocytic leukemia: Justification for risk-adapted therapy. J Clin Oncol 24: 437–443. 10.1200/JCO.2005.03.1021 [DOI] [PubMed] [Google Scholar]
  17. Cairns RA, Iqbal J, Lemonnier F, Kucuk C, de Leval L, Jais JP, Parrens M, Martin A, Xerri L, Brousset P, et al. 2012. IDH2 mutations are frequent in angioimmunoblastic T-cell lymphoma. Blood 119: 1901–1903. 10.1182/blood-2011-11-391748 [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Calin GA, Cimmino A, Fabbri M, Ferracin M, Wojcik SE, Shimizu M, Taccioli C, Zanesi N, Garzon R, Aqeilan RI, et al. 2008. miR-15a and miR-16-1 cluster functions in human leukemia. Proc Natl Acad Sci 105: 5166–5171. 10.1073/pnas.0800121105 [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Campos-Martín Y, Martínez N, Martínez-López A, Cereceda L, Casado F, Algara P, Oscier D, Menarguez FJ, García JF, Piris MA, et al. 2017. Clinical and diagnostic relevance of NOTCH2- and KLF2-mutations in splenic marginal zone lymphoma. Haematologica 102: e310–e312. 10.3324/haematol.2016.161711 [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Carvajal-Cuenca A, Sua LF, Silva NM, Pittaluga S, Royo C, Song JY, Sargent RL, Espinet B, Climent F, Jacobs SA, et al. 2012. In situ mantle cell lymphoma: Clinical implications of an incidental finding with indolent clinical behavior. Haematologic 97: 270–278. 10.3324/haematol.2011.052621 [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Chapuy B, Roemer MGM, Stewart C, Tan Y, Abo RP, Zhang L, Dunford AJ, Meredith DM, Thorner AR, Jordanova ES, et al. 2016. Targetable genetic features of primary testicular and primary central nervous system lymphomas. Blood 127: 869–881. 10.1182/blood-2015-10-673236 [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Chapuy B, Stewart C, Dunford AJ, Kim J, Kamburov A, Redd RA, Lawrence MS, Roemer MGM, Li AJ, Ziepert M, et al. 2018. Molecular subtypes of diffuse large B cell lymphoma are associated with distinct pathogenic mechanisms and outcomes. Nat Med 24: 679–690. 10.1038/s41591-018-0016-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Chen YW, Hu XT, Liang AC, Au WY, So CC, Wong ML, Shen L, Tao Q, Chu KM, Kwong YL, et al. 2006. High BCL6 expression predicts better prognosis, independent of BCL6 translocation status, translocation partner, or BCL6-deregulating mutations, in gastric lymphoma. Blood 108: 2373–2383. 10.1182/blood-2006-05-022517 [DOI] [PubMed] [Google Scholar]
  24. Cheung KJJ, Shah SP, Steidl C, Johnson N, Relander T, Telenius A, Lai B, Murphy KP, Lam W, Al-Tourah AJ, et al. 2009. Genome-wide profiling of follicular lymphoma by array comparative genomic hybridization reveals prognostically significant DNA copy number imbalances. Blood 113: 137–148. 10.1182/blood-2008-02-140616 [DOI] [PubMed] [Google Scholar]
  25. Cheung KJJ, Johnson NA, Affleck JG, Severson T, Steidl C, Ben-Neriah S, Schein J, Morin RD, Moore R, Shah SP, et al. 2010. Acquired TNFRSF14 mutations in follicular lymphoma are associated with worse prognosis. Cancer Res 70: 9166–9174. 10.1158/0008-5472.CAN-10-2460 [DOI] [PubMed] [Google Scholar]
  26. Choi J, Goh G, Walradt T, Hong BS, Bunick CG, Chen K, Bjornson RD, Maman Y, Wang T, Tordoff J, et al. 2015. Genomic landscape of cutaneous T cell lymphoma. Nat Genet 47: 1011–1019. 10.1038/ng.3356 [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Chong LC, Ben-Neriah S, Slack GW, Freeman C, Ennishi D, Mottok A, Collinge B, Abrisqueta P, Farinha P, Boyle M, et al. 2018. High-resolution architecture and partner genes of MYC rearrangements in lymphoma with DLBCL morphology. Blood Adv 2: 2755–2765. 10.1182/bloodadvances.2018023572 [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Clipson A, Wang M, de Leval L, Ashton-Key M, Wotherspoon A, Vassiliou G, Bolli N, Grove C, Moody S, Escudero-Ibarz L, et al. 2015. KLF2 mutation is the most frequent somatic change in splenic marginal zone lymphoma and identifies a subset with distinct genotype. Leukemia 29: 1177–1185. 10.1038/leu.2014.330 [DOI] [PubMed] [Google Scholar]
  29. Copie-Bergman C, Cuillière-Dartigues P, Baia M, Briere J, Delarue R, Canioni D, Salles G, Parrens M, Belhadj K, Fabiani B, et al. 2015. MYC-IG rearrangements are negative predictors of survival in DLBCL patients treated with immunochemotherapy: A GELA/LYSA study. Blood 126: 2466–2474. 10.1182/blood-2015-05-647602 [DOI] [PubMed] [Google Scholar]
  30. Crescenzo R, Abate F, Lasorsa E, Tabbo F, Gaudiano M, Chiesa N, Di Giacomo F, Spaccarotella E, Barbarossa L, Ercole E, et al. 2015. Convergent mutations and kinase fusions lead to oncogenic STAT3 activation in anaplastic large cell lymphoma. Cancer Cell 27: 516–532. 10.1016/j.ccell.2015.03.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Czuczman MS, Trněný M, Davies A, Rule S, Linton KM, Wagner-Johnston N, Gascoyne RD, Slack GW, Brousset P, Eberhard DA, et al. 2017. A phase 2/3 multicenter, randomized, open-label study to compare the efficacy and safety of lenalidomide versus investigator's choice in patients with relapsed or refractory diffuse large B-cell lymphoma. Clin Cancer Res 23: 4127–4137. 10.1158/1078-0432.CCR-16-2818 [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Dalla-Favera R, Bregni M, Erikson J, Patterson D, Gallo RC, Croce CM. 1982. Human c-myc onc gene is located on the region of chromosome 8 that is translocated in Burkitt lymphoma cells. Proc Natl Acad Sci 79: 7824–7827. 10.1073/pnas.79.24.7824 [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Damle RN, Wasil T, Fais F, Ghiotto F, Valetto A, Allen SL, Buchbinder A, Budman D, Dittmar K, Kolitz J, et al. 1999. Ig V gene mutation status and CD38 expression as novel prognostic indicators in chronic lymphocytic leukemia. Blood 94: 1840–1847. 10.1182/blood.V94.6.1840 [DOI] [PubMed] [Google Scholar]
  34. da Silva Almeida AC, Abate F, Khiabanian H, Martinez-Escala E, Guitart J, Tensen CP, Vermeer MH, Rabadan R, Ferrando A, Palomero T. 2015. The mutational landscape of cutaneous T cell lymphoma and Sézary syndrome. Nat Genet 47: 1465–1470. 10.1038/ng.3442 [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Davis RE, Ngo VN, Lenz G, Tolar P, Young RM, Romesser PB, Kohlhammer H, Lamy L, Zhao H, Yang Y, et al. 2010. Chronic active B-cell-receptor signalling in diffuse large B-cell lymphoma. Nature 463: 88–92. 10.1038/nature08638 [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Dearden C. 2012. How I treat prolymphocytic leukemia. Blood 120: 538–551. 10.1182/blood-2012-01-380139 [DOI] [PubMed] [Google Scholar]
  37. Dictor M, Ek S, Sundberg M, Warenholt J, György C, Sernbo S, Gustavsson E, Abu-Alsoud W, Wadström T, Borrebaeck C. 2009. Strong lymphoid nuclear expression of SOX11 transcription factor defines lymphoblastic neoplasms, mantle cell lymphoma and Burkitt's lymphoma. Haematologica 94: 1563–1568. 10.3324/haematol.2009.008474 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Dimopoulos MA, Trotman J, Tedeschi A, Matous JV, Macdonald D, Tam C, Tournilhac O, Ma S, Oriol A, Heffner LT, et al. 2017. Ibrutinib for patients with rituximab-refractory Waldenström's macroglobulinaemia (iNNOVATE): An open-label substudy of an international, multicentre, phase 3 trial. Lancet Oncol 18: 241–250. 10.1016/S1470-2045(16)30632-5 [DOI] [PubMed] [Google Scholar]
  39. Döhner H, Stilgenbauer S, Benner A, Leupolt E, Kröber A, Bullinger L, Döhner K, Bentz M, Lichter P. 2000. Genomic aberrations and survival in chronic lymphocytic leukemia. N Engl J Med 343: 1910–1916. 10.1056/NEJM200012283432602 [DOI] [PubMed] [Google Scholar]
  40. Dunleavy K, Pittaluga S, Maeda LS, Advani R, Chen CC, Hessler J, Steinberg SM, Grant C, Wright G, Varma G, et al. 2013. Dose-adjusted EPOCH-rituximab therapy in primary mediastinal B-cell lymphoma. N Engl J Med 368: 1408–1416. 10.1056/NEJMoa1214561 [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Durham BH, Getta B, Dietrich S, Taylor J, Won H, Bogenberger JM, Scott S, Kim E, Chung YR, Chung SS, et al. 2017. Genomic analysis of hairy cell leukemia identifies novel recurrent genetic alterations. Blood 130: 1644–1648. 10.1182/blood-2017-01-765107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Eichhorst B, Fink AM, Bahlo J, Busch R, Kovacs G, Maurer C, Lange E, Köppler H, Kiehl M, Sökler M, et al. 2016. First-line chemoimmunotherapy with bendamustine and rituximab versus fludarabine, cyclophosphamide, and rituximab in patients with advanced chronic lymphocytic leukaemia (CLL10): An international, open-label, randomised, phase 3, non-inferiority trial. Lancet Oncol 17: 928–942. 10.1016/S1470-2045(16)30051-1 [DOI] [PubMed] [Google Scholar]
  43. Elenitoba-Johnson KSJ, Gascoyne RD, Lim MS, Chhanabai M, Jaffe ES, Raffeld M. 1998. Homozygous deletions at chromosome 9p21 involving p16 and p15 are associated with histologic progression in follicle center lymphoma. Blood 91: 4677–4685. 10.1182/blood.V91.12.4677 [DOI] [PubMed] [Google Scholar]
  44. Eskelund CW, Dahl C, Hansen JW, Westman M, Kolstad A, Pedersen LB, Montano-Almendras CP, Husby S, Freiburhaus C, Ek S, et al. 2017. TP53 mutations identify younger mantle cell lymphoma patients who do not benefit from intensive chemoimmunotherapy. Blood 130: 1903–1910. 10.1182/blood-2017-04-779736 [DOI] [PubMed] [Google Scholar]
  45. Falini B, Bigerna B, Fizzotti M, Pulford K, Pileri SA, Delsol G, Carbone A, Paulli M, Magrini U, Menestrina F, et al. 1998. ALK expression defines a distinct group of T/null lymphomas (“ALK lymphomas”) with a wide morphological spectrum. Am J Pathol 153: 875–886. 10.1016/S0002-9440(10)65629-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Feldman AL, Law M, Remstein ED, Macon WR, Erickson LA, Grogg KL, Kurtin PJ, Dogan A. 2009. Recurrent translocations involving the IRF4 oncogene locus in peripheral T-cell lymphomas. Leukemia 23: 574–580. 10.1038/leu.2008.320 [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Feldman AL, Dogan A, Smith DI, Law ME, Ansell SM, Johnson SH, Procher JC, Ozsan N, Wieben ED, Eckloff BW, et al. 2011. Discovery of recurrent t(6;7)(p25.3;q32.3) translocations in ALK-negative anaplastic large cell lymphomas by massively parallel genomic sequencing. Blood 117: 915–919. 10.1182/blood-2010-08-303305 [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Fernàndez V, Salamero O, Espinet B, Solé F, Royo C, Navarro A, Camacho F, Beà S, Hartmann E, Amador V, et al. 2010. Genomic and gene expression profiling defines indolent forms of mantle cell lymphoma. Cancer Res 70: 1408–1418. 10.1158/0008-5472.CAN-09-3419 [DOI] [PubMed] [Google Scholar]
  49. Fischer K, Bahlo J, Fink AM, Goede V, Herling CD, Cramer P, Langerbeins P, von Tresckow J, Engelke A, Maurer C, et al. 2016. Long-term remissions after FCR chemoimmunotherapy in previously untreated patients with CLL: Updated results of the CLL8 trial. Blood 127: 208–215. 10.1182/blood-2015-06-651125 [DOI] [PubMed] [Google Scholar]
  50. Flossbach L, Antoneag E, Buck M, Siebert R, Mattfeldt T, Möller P, Barth TF. 2011. BCL6 gene rearrangement and protein expression are associated with large cell presentation of extranodal marginal zone B-cell lymphoma of mucosa-associated lymphoid tissue. Int J Cancer 129: 70–77. 10.1002/ijc.25663 [DOI] [PubMed] [Google Scholar]
  51. Forconi F, Poretti G, Kwee I, Sozzi E, Rossi D, Rancoita PMV, Capello D, Rinaldi A, Zucca E, Raspadori D, et al. 2008. High density genome-wide DNA profiling reveals a remarkably stable profile in hairy cell leukaemia. Br J Haematol 141: 622–630. 10.1111/j.1365-2141.2008.07106.x [DOI] [PubMed] [Google Scholar]
  52. Friedberg JW. 2017. How I treat double-hit lymphoma. Blood 130: 590–596. 10.1182/blood-2017-04-737320 [DOI] [PubMed] [Google Scholar]
  53. Fu K, Weisenburger DD, Greiner TC, Dave S, Wright G, Rosenwald A, Chiorazzi M, Iqbal J, Gesk S, Siebert R, et al. 2005. Cyclin D1-negative mantle cell lymphoma: A clinicopathologic study based on gene expression profiling. Blood 106: 4315–4321. 10.1182/blood-2005-04-1753 [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Fukuhara S, Rowley JD, Variakojis D, Golomb HM. 1979. Chromosome abnormalities in poorly differentiated lymphocytic lymphoma. Cancer Res 39: 3119. [PubMed] [Google Scholar]
  55. Ghia EM, Jain S, Widhopf GF II, Rassenti LZ, Keating MJ, Wierda WG, Gribben JG, Brown JR, Rai KR, Byrd JC, et al. 2008. Use of IGHV3-21 in chronic lymphocytic leukemia is associated with high-risk disease and reflects antigen-driven, post-germinal center leukemogenic selection. Blood 111: 5101–5108. 10.1182/blood-2007-12-130229 [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Green MR, Monti S, Rodig SJ, Juszczynski P, Currie T, O'Donnell E, Chapuy B, Takeyama K, Neuberg D, Golub TR, et al. 2010. 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 116: 3268–3277. 10.1182/blood-2010-05-282780 [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Green MR, Kihira S, Liu CL, Nair RV, Salari R, Gentles AJ, Irish J, Stehr H, Vicente-Dueñas C, Romero-Camarero I, et al. 2015. Mutations in early follicular lymphoma progenitors are associated with suppressed antigen presentation. Proc Natl Acad Sci 112: E1116–E1125. 10.1073/pnas.1501199112 [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Gribben JG. 2010. How I treat CLL up front. Blood 115: 187–197. 10.1182/blood-2009-08-207126 [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Hallberg B, Palmer RH. 2013. Mechanistic insight into ALK receptor tyrosine kinase in human cancer biology. Nat Rev Cancer 13: 685–700. 10.1038/nrc3580 [DOI] [PubMed] [Google Scholar]
  60. Hamblin TJ, Davis Z, Gardiner A, Oscier DG, Stevenson FK. 1999. Unmutated Ig VH genes are associated with a more aggressive form of chronic lymphocytic leukemia. Blood 94: 1848–1854. 10.1182/blood.V94.6.1848 [DOI] [PubMed] [Google Scholar]
  61. Hans CP, Weisenburger DD, Greiner TC, Gascoyne RD, Delabie J, Ott G, Müller-Hermelink HK, Campo E, Braziel RM, Jaffe ES, et al. 2004. Confirmation of the molecular classification of diffuse large B-cell lymphoma by immunohistochemistry using a tissue microarray. Blood 103: 275–282. 10.1182/blood-2003-05-1545 [DOI] [PubMed] [Google Scholar]
  62. Harris NL, Jaffe ES, Stein H, Banks PM, Chan JK, Cleary ML, Delsol G, De Wolf-Peeters C, Falini B, Gatter KC, et al. 1994. A revised European–American classification of lymphoid neoplasms: A proposal from the International Lymphoma Study Group [see comments]. Blood 84: 1361–1392. 10.1182/blood.V84.5.1361.1361 [DOI] [PubMed] [Google Scholar]
  63. Horwitz S, O'Connor OA, Pro B, Illidge T, Fanale M, Advani R, Bartlett NL, Christensen JH, Morschhauser F, Domingo-Domenech E, et al. 2019. Brentuximab vedotin with chemotherapy for CD30-positive peripheral T-cell lymphoma (ECHELON-2): A global, double-blind, randomised, phase 3 trial. Lancet 393: 229–240. 10.1016/S0140-6736(18)32984-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Hummel M, Bentink S, Berger H, Klapper W, Wessendorf S, Barth TFE, Bernd HW, Cogliatti SB, Dierlamm J, Feller AC, et al. 2006. A biologic definition of Burkitt's lymphoma from transcriptional and genomic profiling. New Engl J Med 354: 2419–2430. 10.1056/NEJMoa055351 [DOI] [PubMed] [Google Scholar]
  65. Johnson NA, Savage KJ, Ludkovski O, Ben-Neriah S, Woods R, Steidl C, Dyer MJ, Siebert R, Kuruvilla J, Klasa R, et al. 2009. Lymphomas with concurrent BCL2 and MYC translocations: The critical factors associated with survival. Blood 114: 2273–2279. 10.1182/blood-2009-03-212191 [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Kiel MJ, Velusamy T, Betz BL, Zhao L, Weigelin HG, Chiang MY, Huebner-Chan DR, Bailey NG, Yang DT, Bhagat G, et al. 2012. Whole-genome sequencing identifies recurrent somatic NOTCH2 mutations in splenic marginal zone lymphoma. J Exp Med 209: 1553–1565. 10.1084/jem.20120910 [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Kiel MJ, Velusamy T, Rolland D, Sahasrabuddhe AA, Chung F, Bailey NG, Schrader A, Li B, Li JZ, Ozel AB, et al. 2014. Integrated genomic sequencing reveals mutational landscape of T-cell prolymphocytic leukemia. Blood 124: 1460–1472. 10.1182/blood-2014-03-559542 [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Kiel MJ, Sahasrabuddhe AA, Rolland DCM, Velusamy T, Chung F, Schaller M, Bailey NG, Betz BL, Miranda RN, Porcu P, et al. 2015. Genomic analyses reveal recurrent mutations in epigenetic modifiers and the JAK–STAT pathway in Sézary syndrome. Nat Commun 6: 8470 10.1038/ncomms9470 [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Kim YH, Bagot M, Pinter-Brown L, Rook AH, Porcu P, Horwitz SM, Whittaker S, Tokura Y, Vermeer M, Zinzani PL, et al. 2018. Mogamulizumab versus vorinostat in previously treated cutaneous T-cell lymphoma (MAVORIC): An international, open-label, randomised, controlled phase 3 trial. Lancet Oncol 19: 1192–1204. 10.1016/S1470-2045(18)30379-6 [DOI] [PubMed] [Google Scholar]
  70. Kurita D, Takeuchi K, Kobayashi S, Hojo A, Uchino Y, Sakagami M, Ohtake S, Takahashi H, Miura K, Iriyama N, et al. 2016. A cyclin D1-negative mantle cell lymphoma with an IGL-CCND2 translocation that relapsed with blastoid morphology and aggressive clinical behavior. Virchows Arch 469: 471–476. 10.1007/s00428-016-1995-9 [DOI] [PubMed] [Google Scholar]
  71. Kurtz DM, Green MR, Bratman SV, Scherer F, Liu CL, Kunder CA, Takahashi K, Glover C, Keane C, Kihira S, et al. 2015. Noninvasive monitoring of diffuse large B-cell lymphoma by immunoglobulin high-throughput sequencing. Blood 125: 3679–3687. 10.1182/blood-2015-03-635169 [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Landau DA, Tausch E, Taylor-Weiner AN, Stewart C, Reiter JG, Bahlo J, Kluth S, Bozic I, Lawrence M, Böttcher S, et al. 2015. Mutations driving CLL and their evolution in progression and relapse. Nature 526: 525–530. 10.1038/nature15395 [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Launay E, Pangault C, Bertrand P, Jardin F, Lamy T, Tilly H, Tarte K, Bastard C, Fest T. 2012. High rate of TNFRSF14 gene alterations related to 1p36 region in de novo follicular lymphoma and impact on prognosis. Leukemia 26: 559–562. 10.1038/leu.2011.266 [DOI] [PubMed] [Google Scholar]
  74. Le Beau MM, Bitter MA, Larson RA, Doane LA, Ellis ED, Franklin WA, Rubin CM, Kadin ME, Vardiman JW. 1989. The t(2;5)(p23;q35): A recurring chromosomal abnormality in Ki-1-positive anaplastic large cell lymphoma. Leukemia 3: 866–870. [PubMed] [Google Scholar]
  75. Lemonnier F, Couronné L, Parrens M, Jaïs JP, Travert M, Lamant L, Tournillac O, Rousset T, Fabiani B, Cairns RA, et al. 2012. Recurrent TET2 mutations in peripheral T-cell lymphomas correlate with TFH-like features and adverse clinical parameters. Blood 120: 1466–1469. 10.1182/blood-2012-02-408542 [DOI] [PubMed] [Google Scholar]
  76. Lemonnier F, Dupuis J, Sujobert P, Tournillhac O, Cheminant M, Sarkozy C, Pelletier L, Marçais A, Robe C, Fataccioli V, et al. 2018. Treatment with 5-azacytidine induces a sustained response in patients with angioimmunoblastic T-cell lymphoma. Blood 132: 2305–2309. 10.1182/blood-2018-04-840538 [DOI] [PubMed] [Google Scholar]
  77. Lenz G, Wright G, Dave SS, Xiao W, Powell J, Zhao H, Xu W, Tan B, Goldschmidt N, Iqbal J, et al. 2008. Stromal gene signatures in large-B-cell lymphomas. New Engl J Med 359: 2313–2323. 10.1056/NEJMoa0802885 [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Leucci E, Cocco M, Onnis A, De Falco G, van Cleef P, Bellan C, van Rijk A, Nyagol J, Byakika B, Lazzi S, et al. 2008. MYC translocation-negative classical Burkitt lymphoma cases: An alternative pathogenetic mechanism involving miRNA deregulation. J Pathol 216: 440–450. 10.1002/path.2410 [DOI] [PubMed] [Google Scholar]
  79. Li S, Lin P, Fayad LE, Lennon PA, Miranda RN, Yin CC, Lin E, Medeiros LJ. 2012. 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 25: 145–156. 10.1038/modpathol.2011.147 [DOI] [PubMed] [Google Scholar]
  80. Louissaint A, Schafernak KT, Geyer JT, Kovach AE, Ghandi M, Gratzinger D, Roth CG, Paxton CN, Kim S, Namgyal C, et al. 2016. Pediatric-type nodal follicular lymphoma: A biologically distinct lymphoma with frequent MAPK pathway mutations. Blood 128: 1093–1100. 10.1182/blood-2015-12-682591 [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Malcikova J, Tausch E, Rossi D, Sutton LA, Soussi T, Zenz T, Kater AP, Niemann CU, Gonzalez D, Davi F, et al. 2018. ERIC recommendations for TP53 mutation analysis in chronic lymphocytic leukemia—Update on methodological approaches and results interpretation. Leukemia 32: 1070–1080. 10.1038/s41375-017-0007-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Manolov G, Manolova Y. 1972. Marker band in one chromosome 14 from Burkitt lymphomas. Nature 237: 33–34. 10.1038/237033a0 [DOI] [PubMed] [Google Scholar]
  83. Marcucci G, Maharry K, Wu YZ, Radmacher MD, Mrózek K, Margeson D, Holland KB, Whitman SP, Becker H, Schwind S, et al. 2010. IDH1 and IDH2 gene mutations identify novel molecular subsets within de novo cytogenetically normal acute myeloid leukemia: A Cancer and Leukemia Group B Study. J Clin Oncol 28: 2348–2355. 10.1200/JCO.2009.27.3730 [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Martín-Garcia D, Navarro A, Valdés-Mas R, Clot G, Gutiérrez-Abril J, Prieto M, Ribera-Cortada I, Woroniecka R, Rymkiewicz G, Bens S, et al. 2018. CCND2 and CCND3 hijack immunoglobulin light chain enhancers in cyclin D1-negative mantle cell lymphoma. Blood 133: 940–951. 10.1182/blood-2018-07-862151 [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. May PC, Foot N, Dunn R, Geoghegan H, Neat MJ. 2010. Detection of cryptic and variant IGH-MYC rearrangements in high-grade non-Hodgkin's lymphoma by fluorescence in situ hybridization: Implications for cytogenetic testing. Cancer Genet Cytogenet 198: 71–75. 10.1016/j.cancergencyto.2009.12.010 [DOI] [PubMed] [Google Scholar]
  86. McGregor JM, Crook T, Fraser-Andrews EA, Rozycka M, Crossland S, Brooks L, Whittaker SJ. 1999. Spectrum of p53 gene mutations suggests a possible role for ultraviolet radiation in the pathogenesis of advanced cutaneous lymphomas. J Invest Dermatol 112: 317–321. 10.1046/j.1523-1747.1999.00507.x [DOI] [PubMed] [Google Scholar]
  87. Melzner I, Bucur AJ, Brüderlein S, Dorsch K, Hasel C, Barth TF, Leithäuser F, Möller P. 2005. Biallelic mutation of SOCS-1 impairs JAK2 degradation and sustains phospho-JAK2 action in the MedB-1 mediastinal lymphoma line. Blood 105: 2535–2542. 10.1182/blood-2004-09-3701 [DOI] [PubMed] [Google Scholar]
  88. Montgomery ND, Coward WB IV, Johnson S, Yuan J, Gulley ML, Mathews SP, Kaiser-Rogers K, Rao KW, Sanger WG, Sanmann JN, et al. 2016. Karyotypic abnormalities associated with Epstein–Barr virus status in classical Hodgkin lymphoma. Cancer Genet 209: 408–416. 10.1016/j.cancergen.2016.08.006 [DOI] [PubMed] [Google Scholar]
  89. Morin RD, Johnson NA, Severson TM, Mungall AJ, An J, Goya R, Paul JE, Boyle M, Woolcock BW, Kuchenbauer F, et al. 2010. Somatic mutations altering EZH2 (Tyr641) in follicular and diffuse large B-cell lymphomas of germinal-center origin. Nat Genet 42: 181–185. 10.1038/ng.518 [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Morin RD, Mendez-Lago M, Mungall AJ, Goya R, Mungall KL, Corbett RD, Johnson NA, Severson TM, Chiu R, Field M, et al. 2011. Frequent mutation of histone-modifying genes in non-Hodgkin lymphoma. Nature 476: 298–303. 10.1038/nature10351 [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Morris SW, Kirstein MN, Valentine MB, Dittmer KG, Shapiro DN, Saltman DL, Look AT. 1994. Fusion of a kinase gene, ALK, to a nucleolar protein gene, NPM, in non-Hodgkin's lymphoma. Science 263: 1281–1284. 10.1126/science.8122112 [DOI] [PubMed] [Google Scholar]
  92. Mossé YP, Voss SD, Lim MS, Rolland D, Minard CG, Fox E, Adamson P, Wilner K, Blaney SM, Weigel BJ. 2017. Targeting ALK with Crizotinib in pediatric anaplastic large cell lymphoma and inflammatory myofibroblastic tumor: A Children's Oncology Group Study. J Clin Oncol 35: 3215–3221. 10.1200/JCO.2017.73.4830 [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Mozos A, Royo C, Hartmann E, De Jong D, Baró C, Valera A, Fu K, Weisenburger DD, Delabie J, Chuang SS, et al. 2009. SOX11 expression is highly specific for mantle cell lymphoma and identifies the cyclin D1-negative subtype. Haematologica 94: 1555 10.3324/haematol.2009.010264 [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Muñoz-Mármol AM, Sanz C, Tapia G, Marginet R, Ariza A, Mate JL. 2013. MYC status determination in aggressive B-cell lymphoma: The impact of FISH probe selection. Histopathology 63: 418–424. 10.1111/his.12178 [DOI] [PubMed] [Google Scholar]
  95. Ngo VN, Young RM, Schmitz R, Jhavar S, Xiao W, Lim KH, Kohlhammer H, Xu W, Yang Y, Zhao H, et al. 2011. Oncogenically active MYD88 mutations in human lymphoma. Nature 470: 115–119. 10.1038/nature09671 [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Odejide O, Weigert O, Lane AA, Toscano D, Lunning MA, Kopp N, Kim S, van Bodegom D, Bolla S, Schatz JH, et al. 2014. A targeted mutational landscape of angioimmunoblastic T-cell lymphoma. Blood 123: 1293–1296. 10.1182/blood-2013-10-531509 [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Okosun J, Bodor C, Wang J, Araf S, Yang CY, Pan C, Boller S, Cittaro D, Bozek M, Iqbal S, et al. 2014. Integrated genomic analysis identifies recurrent mutations and evolution patterns driving the initiation and progression of follicular lymphoma. Nat Genet 46: 176–181. 10.1038/ng.2856 [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Palomero T, Couronné L, Khiabanian H, Kim MY, Ambesi-Impiombato A, Perez-Garcia A, Carpenter Z, Abate F, Allegretta M, Jaydu JE, et al. 2014. Recurrent mutations in epigenetic regulators, RHOA and FYN kinase in peripheral T cell lymphomas. Nat Genet 46: 166–170. 10.1038/ng.2873 [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Parrilla Castellar ER, Jaffe ES, Said JW, Swerdlow SH, Ketterling RP, Knudson RA, Sidhu JS, Hsi ED, Karikehalli S, Jiang L, et al. 2014. ALK-negative anaplastic large cell lymphoma is a genetically heterogeneous disease with widely disparate clinical outcomes. Blood 124: 1473–1480. 10.1182/blood-2014-04-571091 [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Parry M, Rose-Zerilli MJJ, Ljungström V, Gibson J, Wang J, Walewska R, Parker H, Parker A, Davis Z, Gardiner A, et al. 2015. Genetics and prognostication in splenic marginal zone lymphoma: Revelations from deep sequencing. Clin Cancer Res 21: 4174–4183. 10.1158/1078-0432.CCR-14-2759 [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Pasqualucci L, Dalla-Favera R. 2015. The genetic landscape of diffuse large B-cell lymphoma. Semin Hematol 52: 67–76. 10.1053/j.seminhematol.2015.01.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Pasqualucci L, Dominguez-Sola D, Chiarenza A, Fabbri G, Grunn A, Trifonov V, Kasper LH, Lerach S, Tang H, Ma J, et al. 2011. Inactivating mutations of acetyltransferase genes in B-cell lymphoma. Nature 471: 189–195. 10.1038/nature09730 [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Pasqualucci L, Khiabanian H, Fangazio M, Vasishtha M, Messina M, Holmes AB, et al. 2014. Genetics of follicular lymphoma transformation. Cell Rep 6: 130–140. 10.1016/j.celrep.2013.12.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Pastore A, Jurinovic V, Kridel R, Hoster E, Staiger AM, Szczepanowski M, Pott C, Kopp N, Murakami M, Horn H, et al. 2015. Integration of gene mutations in risk prognostication for patients receiving first-line immunochemotherapy for follicular lymphoma: A retrospective analysis of a prospective clinical trial and validation in a population-based registry. Lancet Oncol 16: 1111–1122. 10.1016/S1470-2045(15)00169-2 [DOI] [PubMed] [Google Scholar]
  105. Pedersen MØ, Gang AO, Poulsen TS, Knudsen H, Lauritzen AF, Nielsen SL, Klausen TW, Nørgaard P. 2014. MYC translocation partner gene determines survival of patients with large B-cell lymphoma with MYC- or double-hit MYC/BCL2 translocations. Eur J Haematol 92: 42–48. 10.1111/ejh.12212 [DOI] [PubMed] [Google Scholar]
  106. Pedersen MB, Hamilton-Dutoit SJ, Bendix K, Ketterling RP, Bedroske PP, Luoma IM, Sattler CA, Boddicker RL, Bennani NN, Nørgaard P, et al. 2017. DUSP22 and TP63 rearrangements predict outcome of ALK-negative anaplastic large cell lymphoma: A Danish cohort study. Blood 130: 554–557. 10.1182/blood-2016-12-755496 [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Pekarsky Y, Hallas C, Isobe M, Russo G, Croce CM. 1999. Abnormalities at 14q32.1 in T cell malignancies involve two oncogenes. Proc Natl Acad Sci 96: 2949–2951. 10.1073/pnas.96.6.2949 [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Pekarsky Y, Koval A, Hallas C, Bichi R, Tresini M, Malstrom S, Russo G, Tsichlis P, Croce CM. 2000. Tcl1 enhances Akt kinase activity and mediates its nuclear translocation. Proc Natl Acad Sci 97: 3028–3033. 10.1073/pnas.97.7.3028 [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Perrot A, Lauwers-Cances V, Corre J, Robillard N, Hulin C, Chretien ML, Dejoie T, Maheo S, Stoppa AM, Pegourie B, et al. 2018. Minimal residual disease negativity using deep sequencing is a major prognostic factor in multiple myeloma. Blood 132: 2456–2464. 10.1182/blood-2018-06-858613 [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Pham-Ledard A, Beylot-Barry M, Barbe C, Leduc M, Petrella T, Vergier B, Martinez F, Cappellen D, Merlio JP, Grange F. 2014. High frequency and clinical prognostic value of MYD88 L265P mutation in primary cutaneous diffuse large B-cell lymphoma, Leg-type. JAMA Dermatol 150: 1173–1179. 10.1001/jamadermatol.2014.821 [DOI] [PubMed] [Google Scholar]
  111. Phelan JD, Young RM, Webster DE, Roulland S, Wright GW, Kasbekar M, Shaffer AL, Ceribelli M, Wang JQ, Schmitz R, et al. 2018. A multiprotein supercomplex controlling oncogenic signalling in lymphoma. Nature 560: 387–391. 10.1038/s41586-018-0290-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Pillonel V, Juskevicius D, Ng CKY, Bodmer A, Zettl A, Jucker D, Dirnhofer S, Tzankov A. 2018. High-throughput sequencing of nodal marginal zone lymphomas identifies recurrent BRAF mutations. Leukemia 32: 2412–2426. 10.1038/s41375-018-0082-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Pittaluga S, Wlodarska I, Pulford K, Campo E, Morris SW, Van den Berghe H, De Wolf-Petters C. 1997. The monoclonal antibody ALK1 identifies a distinct morphological subtype of anaplastic large cell lymphoma associated with 2p23/ALK rearrangements. Am J Pathol 151: 343–351. [PMC free article] [PubMed] [Google Scholar]
  114. Puente XS, Pinyol M, Quesada V, Conde L, Ordonez GR, Villamor N, Escaramis G, Jares P, Beà S, González-Díaz M, et al. 2011. Whole-genome sequencing identifies recurrent mutations in chronic lymphocytic leukaemia. Nature 475: 101–105. 10.1038/nature10113 [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Pulford K, Lamant L, Morris SW, Butler LH, Wood KM, Stroud D, Delsol G, Mason DY. 1997. Detection of anaplastic lymphoma kinase (ALK) and nucleolar protein nucleophosmin (NPM)-ALK proteins in normal and neoplastic cells with the monoclonal antibody ALK1. Blood 89: 1394–1404. 10.1182/blood.V89.4.1394 [DOI] [PubMed] [Google Scholar]
  116. Quesada V, Conde L, Villamor N, Ordóñez GR, Jares P, Bassaganyas L, Ramsay AJ, Beà S, Pinyol M, Martínez-Trillos A, et al. 2012. Exome sequencing identifies recurrent mutations of the splicing factor SF3B1 gene in chronic lymphocytic leukemia. Nat Genet 44: 47–52. 10.1038/ng.1032 [DOI] [PubMed] [Google Scholar]
  117. Rawstron AC, Kreuzer KA, Soosapilla A, Spacek M, Stehlikova O, Gambell P, McIver-Brown N, Villamor N, Psarra K, Arroz M, et al. 2018. Reproducible diagnosis of chronic lymphocytic leukemia by flow cytometry: An European Research Initiative on CLL (ERIC) & European Society for Clinical Cell Analysis (ESCCA) Harmonisation project. Cytometry B Clin Cytom 94: 121–128. 10.1002/cyto.b.21595 [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Rimokh R, Magaud JP, Berger F, Samarut J, Coiffier B, Germain D, Mason DY. 1989. A translocation involving a specific breakpoint (q35) on chromosome 5 is characteristic of anaplastic large cell lymphoma (‘Ki-1 lymphoma’). Br J Haematol 71: 31–36. 10.1111/j.1365-2141.1989.tb06270.x [DOI] [PubMed] [Google Scholar]
  119. Ritz O, Guiter C, Castellano F, Dorsch K, Melzner J, Jais JP, Dubois G, Gaullard P, Moeller P, Leroy K. 2009. Recurrent mutations of the STAT6 DNA binding domain in primary mediastinal B-cell lymphoma. Blood 114: 1236–1242. 10.1182/blood-2009-03-209759 [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Roemer MGM, Advani RH, Ligon AH, Natkunam Y, Redd RA, Homer H, Connelly CF, Sun HH, Daadi SE, Freeman GJ, et al. 2016. PD-L1 and PD-L2 genetic alterations define classical Hodgkin lymphoma and predict outcome. J Clin Oncol 34: 2690–2697. 10.1200/JCO.2016.66.4482 [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Roschewski M, Dunleavy K, Pittaluga S, Moorhead M, Pepin F, Kong K, Shovlin M, Jaffe ES, Staudt LM, Lai C, et al. 2015. Circulating tumour DNA and CT monitoring in patients with untreated diffuse large B-cell lymphoma: A correlative biomarker study. Lancet Oncol 16: 541–549. 10.1016/S1470-2045(15)70106-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Rosenwald A, Wright G, Chan WC, Connors JM, Campo E, Fisher RI, Gascoyne RD, Muller-Hermlink HK, Smeland EB, Giltnane JM, et al. 2002. The use of molecular profiling to predict survival after chemotherapy for diffuse large-B-cell lymphoma. New Engl J Med 346: 1937–1947. 10.1056/NEJMoa012914 [DOI] [PubMed] [Google Scholar]
  123. Rosenwald A, Wright G, Leroy K, Yu X, Gaulard P, Gascoyne RD, Chan WC, Zhao T, Haioun C, Greiner TC, et al. 2003. 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 198: 851–862. 10.1084/jem.20031074 [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Rossi D, Spina V, Deambrogi C, Rasi S, Laurenti L, Stamatopoulos K, Arcaini L, Lucioni M, Rocque GB, Xu-Monette ZY, et al. 2011. The genetics of Richter syndrome reveals disease heterogeneity and predicts survival after transformation. Blood 117: 3391–3401. 10.1182/blood-2010-09-302174 [DOI] [PubMed] [Google Scholar]
  125. Rossi D, Trifonov V, Fangazio M, Bruscaggin A, Rasi S, Spina V, Monti S, Vaisitti T, Arruga F, Famà R, et al. 2012. The coding genome of splenic marginal zone lymphoma: Activation of NOTCH2 and other pathways regulating marginal zone development. J Exp Med 209: 1537–1551. 10.1084/jem.20120904 [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Rossi D, Diop F, Spaccarotella E, Monti S, Zanni M, Rasi S, Deambrogi C, Spina V, Bruscaggin A, Favini C, et al. 2017. Diffuse large B-cell lymphoma genotyping on the liquid biopsy. Blood 129: 1947–1957. 10.1182/blood-2016-05-719641 [DOI] [PubMed] [Google Scholar]
  127. Roulland S, Navarro JM, Grenot P, Milili M, Agopian J, Montpellier B, Gauduchon P, Lebailly P, Schiff C, Nadel B. 2006. Follicular lymphoma-like B cells in healthy individuals: A novel intermediate step in early lymphomagenesis. J Exp Med 203: 2425–2431. 10.1084/jem.20061292 [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Russo G, Isobe M, Gatti R, Finan J, Batuman O, Huebner K, Nowell PC, Croce CM. 1989. Molecular analysis of a t(14;14) translocation in leukemic T-cells of an ataxia telangiectasia patient. Proc Natl Acad Sci 86: 602–606. 10.1073/pnas.86.2.602 [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Sakamoto Y, Ishida T, Masaki A, Murase T, Yonekura K, Tashiro Y, Tokunaga M, Utsunomiya A, Ito A, Kusumoto S, et al. 2018. CCR4 mutations associated with superior outcome of adult T-cell leukemia/lymphoma under mogamulizumab treatment. Blood 132: 758–761. 10.1182/blood-2018-02-835991 [DOI] [PubMed] [Google Scholar]
  130. Sakata-Yanagimoto M, Enami T, Yoshida K, Shiraishi Y, Ishii R, Miyake Y, Muto H, Tsuyama N, Sato-Otsubo A, Okuno Y, et al. 2014. Somatic RHOA mutation in angioimmunoblastic T cell lymphoma. Nat Genet 46: 171–175. 10.1038/ng.2872 [DOI] [PubMed] [Google Scholar]
  131. Salaverria I, Royo C, Carvajal-Cuenca A, Clot G, Navarro A, Valera A, Song JY, Woroniecka R, Rymkiewicz G, Klapper W, et al. 2013. CCND2 rearrangements are the most frequent genetic events in cyclin D1 mantle cell lymphoma. Blood 121: 1394–1402. 10.1182/blood-2012-08-452284 [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Salido M, Baró C, Oscier D, Stamatopoulos K, Dierlamm J, Matutes E, Traverse-Glehen A, Berger F, Felman P, Thieblemont C, et al. 2010. Cytogenetic aberrations and their prognostic value in a series of 330 splenic marginal zone B-cell lymphomas: A multicenter study of the Splenic B-Cell Lymphoma Group. Blood 116: 1479–1488. 10.1182/blood-2010-02-267476 [DOI] [PubMed] [Google Scholar]
  133. Savage KJ, Monti S, Kutok JL, Cattoretti G, Neuberg D, De Leval L, Kurtin P, Dal Cin P, Ladd C, Feuerhake F, et al. 2003. 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 102: 3871–3879. 10.1182/blood-2003-06-1841 [DOI] [PubMed] [Google Scholar]
  134. Scherer F, Kurtz DM, Newman AM, Stehr H, Craig AF, Esfahani MS, Lovejoy AF, Chabon JJ, Klass DM, Liu CL, et al. 2016. Distinct biological subtypes and patterns of genome evolution in lymphoma revealed by circulating tumor DNA. Sci Transl Med 8: 364ra155 10.1126/scitranslmed.aai8545 [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Schmidt J, Gong S, Marafioti T, Mankel B, Gonzalez-Farre B, Balagué O, Mozos A, Cabecadas J, van der Walt J, Hoehn D, et al. 2016. Genome-wide analysis of pediatric-type follicular lymphoma reveals low genetic complexity and recurrent alterations of TNFRSF14 gene. Blood 128: 1101–1111. 10.1182/blood-2016-03-703819 [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Schmitz R, Young RM, Ceribelli M, Jhavar S, Xiao W, Zhang M, Wright G, Shaffer AL, Hodson DJ, Buras E, et al. 2012. Burkitt lymphoma pathogenesis and therapeutic targets from structural and functional genomics. Nature 490: 116–120. 10.1038/nature11378 [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Schmitz R, Wright GW, Huang DW, Johnson CA, Phelan JD, Wang JQ, Roulland S, Kasbekar M, Young RM, Shaffer AL, et al. 2018. Genetics and pathogenesis of diffuse large B-cell lymphoma. N Engl J Med 378: 1396–1407. 10.1056/NEJMoa1801445 [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Scott DW, Wright GW, Williams PM, Lih CJ, Walsh W, Jaffe ES, Rosenwald A, Campo E, Chan WC, Connors JM, et al. 2014. Determining cell-of-origin subtypes of diffuse large B-cell lymphoma using gene expression in formalin-fixed paraffin-embedded tissue. Blood 123: 1214–1217. 10.1182/blood-2013-11-536433 [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Scott DW, King RL, Staiger AM, Ben-Neriah S, Jiang A, Horn H, Mottok A, Farinha P, Slack GW, Ennishi D, et al. 2018. High-grade B-cell lymphoma with MYC and BCL2 and/or BCL6 rearrangements with diffuse large B-cell lymphoma morphology. Blood 131: 2060–2064. 10.1182/blood-2017-12-820605 [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Shao H, Calvo KR, Grönborg M, Tembhare PR, Kreitman RJ, Stetler-Stevenson M, Yuan CM. 2013. Distinguishing hairy cell leukemia variant from hairy cell leukemia: Development and validation of diagnostic criteria. Leukemia Res 37: 401–409. 10.1016/j.leukres.2012.11.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Steidl C, Shah SP, Woolcock BW, Rui L, Kawahara M, Farinha P, Johnson NA, Zhao Y, Telenius A, Neriah SB, et al. 2011. MHC class II transactivator CIITA is a recurrent gene fusion partner in lymphoid cancers. Nature 471: 377–381. 10.1038/nature09754 [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Swerdlow SH, Campo E, Harris NL, Pileri SA, Jaffe ES, Stein H, et al. 2017. WHO classification of tumours of haematopoietic and lymphoid tissues. WHO Press, Geneva. [Google Scholar]
  143. Taub R, Kirsch I, Morton C, Lenoir G, Swan D, Tronick S, Aaronson S, Leder P. 1982. Translocation of the c-myc gene into the immunoglobulin heavy chain locus in human Burkitt lymphoma and murine plasmacytoma cells. Proc Natl Acad Sci 79: 7837–7841. 10.1073/pnas.79.24.7837 [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. The Non-Hodgkin's Lymphoma Classification Project [No authors listed]. 1997. A clinical evaluation of the International Lymphoma Study Group classification of non-Hodgkin's lymphoma. Blood 89: 3909–3918. 10.1182/blood.V89.11.3909 [DOI] [PubMed] [Google Scholar]
  145. Tiacci E, Trifonov V, Schiavoni G, Holmes A, Kern W, Martelli MP, Pucciarini A, Bigerna B, Pacini R, Wells VA, et al. 2011. BRAF mutations in hairy-cell leukemia. New Engl J Med 364: 2305–2315. 10.1056/NEJMoa1014209 [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Tiacci E, Park JH, De Carolis L, Chung SS, Broccoli A, Scott S, Zaja F, Devlin S, Pulsoni A, Chung YR, et al. 2015. Targeting mutant BRAF in relapsed or refractory hairy-cell leukemia. New Engl J Med 373: 1733–1747. 10.1056/NEJMoa1506583 [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Tiacci E, Ladewig E, Schiavoni G, Penson A, Fortini E, Pettirossi V, Wang Y, Rosseto A, Venanzi A, Vlasevska S, et al. 2018. Pervasive mutations of JAK-STAT pathway genes in classical Hodgkin lymphoma. Blood 131: 2454–2465. 10.1182/blood-2017-11-814913 [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Treon SP, Xu L, Yang G, Zhou Y, Liu X, Cao Y, Sheehy P, Manning RJ, Patterson CJ, Tripsas C, et al. 2012. MYD88 L265P somatic mutation in Waldenström's macroglobulinemia. New Engl J Med 367: 826–833. 10.1056/NEJMoa1200710 [DOI] [PubMed] [Google Scholar]
  149. Treon SP, Tripsas CK, Meid K, Warren D, Varma G, Green R, Argyropoulos KV, Yang G, Cao Y, Xu L, et al. 2015. Ibrutinib in previously treated Waldenström's macroglobulinemia. New Engl J Med 372: 1430–1440. 10.1056/NEJMoa1501548 [DOI] [PubMed] [Google Scholar]
  150. Tsujimoto Y, Finger LR, Yunis J, Nowell PC, Croce CM. 1984. Cloning of the chromosome breakpoint of neoplastic B cells with the t(14;18) chromosome translocation. Science 226: 1097–1099. 10.1126/science.6093263 [DOI] [PubMed] [Google Scholar]
  151. van der Velden VH, Cazzaniga G, Schrauder A, Hancock J, Bader P, Panzer-Grumayer ER, Flohr T, Sutton R, Cave H, Madsen HO, et al. 2007. Analysis of minimal residual disease by Ig/TCR gene rearrangements: Guidelines for interpretation of real-time quantitative PCR data. Leukemia 21: 604–611. 10.1038/sj.leu.2404586 [DOI] [PubMed] [Google Scholar]
  152. van Dongen JJ, Wolvers-Tettero IL. 1991. Analysis of immunoglobulin and T cell receptor genes. Part II: Possibilities and limitations in the diagnosis and management of lymphoproliferative diseases and related disorders. Clin Chim Acta 198: 93–174. 10.1016/0009-8981(91)90247-A [DOI] [PubMed] [Google Scholar]
  153. van Dongen JJ, Langerak AW, Bruggemann M, Evans PA, Hummel M, Lavender FL, Delabesse E, Davi F, Schuuring E, García-Sanz R, et al. 2003. Design and standardization of PCR primers and protocols for detection of clonal immunoglobulin and T-cell receptor gene recombinations in suspect lymphoproliferations: Report of the BIOMED-2 Concerted Action BMH4-CT98-3936. Leukemia 17: 2257–2317. 10.1038/sj.leu.2403202 [DOI] [PubMed] [Google Scholar]
  154. Vaqué JP, Gómez-López G, Monsálvez V, Varela I, Martínez N, Pérez C, Domínguez O, Graña O, Rodríguez-Peralto JL, Rodríguez-Pinilla SM, et al. 2014. PLCG1 mutations in cutaneous T-cell lymphomas. Blood 123: 2034–2043. 10.1182/blood-2013-05-504308 [DOI] [PubMed] [Google Scholar]
  155. Vasmatzis G, Johnson SH, Knudson RA, Ketterling RP, Braggio E, Fonseca R, Viswanatha DS, Law ME, Kip NS, Ozsan N, et al. 2012. Genome-wide analysis reveals recurrent structural abnormalities of TP63 and other p53-related genes in peripheral T-cell lymphomas. Blood 120: 2280–2289. 10.1182/blood-2012-03-419937 [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Velusamy T, Kiel MJ, Sahasrabuddhe AA, Rolland D, Dixon CA, Bailey NG, Betz BL, Brown NA, Hristov AC, Wilcox RA, et al. 2014. A novel recurrent NPM1-TYK2 gene fusion in cutaneous CD30-positive lymphoproliferative disorders. Blood 124: 3768–3771. 10.1182/blood-2014-07-588434 [DOI] [PubMed] [Google Scholar]
  157. Viganò E, Gunawardana J, Mottok A, Van Tol T, Mak K, Chan FC, Chong L, Chavez E, Woolcock B, Takata K, et al. 2018a. Somatic IL4R mutations in primary mediastinal large B-cell lymphoma lead to constitutive JAK-STAT signaling activation. Blood 131: 2036–2046. 10.1182/blood-2017-09-808907 [DOI] [PubMed] [Google Scholar]
  158. Viganò E, Duns G, Ennishi D, Gascoyne RD, Morin RD, Scott DW, Steidl C. 2018b. Recurrent IL4R somatic mutations in diffuse large B-cell lymphoma lead to an altered gene expression profile and changes in tumor microenvironment composition. Blood 132: 669 10.1182/blood-2018-99-110473 [DOI] [Google Scholar]
  159. Wang L, Lawrence MS, Wan Y, Stojanov P, Sougnez C, Stevenson K, Werner L, Sivachenko A, DeLuca DS, Zhang L, et al. 2011. SF3B1 and other novel cancer genes in chronic lymphocytic leukemia. N Engl J Med 365: 2497–2506. 10.1056/NEJMoa1109016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Wang C, McKeithan TW, Gong Q, Zhang W, Bouska A, Rosenwald A, Gascoyne RD, Wu X, Wang J, Muhammad Z, et al. 2015a. IDH2R172 mutations define a unique subgroup of patients with angioimmunoblastic T-cell lymphoma. Blood 126: 1741–1752. 10.1182/blood-2015-05-644591 [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Wang L, Ni X, Covington KR, Yang BY, Shiu J, Zhang X, Xi L, Meng Q, Langridge T, Drummond J, et al. 2015b. Genomic profiling of Sézary syndrome identifies alterations of key T cell signaling and differentiation genes. Nat Genet 47: 1426–1434. 10.1038/ng.3444 [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Ward PS, Lu C, Cross JR, Abdel-Wahab O, Levine RL, Schwartz GK, Thompson CB. 2013. The potential for isocitrate dehydrogenase mutations to produce 2-hydroxyglutarate depends on allele specificity and subcellular compartmentalization. J Biol Chem 288: 3804–3815. 10.1074/jbc.M112.435495 [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Waterfall JJ, Arons E, Walker RL, Pineda M, Roth L, Killian JK, Abaan OD, Davis SR, Kreitman RJ, Meltzer PS. 2014. High prevalence of MAP2K1 mutations in variant and IGHV4-34–expressing hairy-cell leukemias. Nat Genet 46: 8–10. 10.1038/ng.2828 [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Weniger MA, Melzner I, Menz CK, Wegener S, Bucur AJ, Dorsch K, Mattfeldt T, Barth TF, Möller P. 2006. Mutations of the tumor suppressor gene SOCS-1 in classical Hodgkin lymphoma are frequent and associated with nuclear phospho-STAT5 accumulation. Oncogene 25: 2679–2684. 10.1038/sj.onc.1209151 [DOI] [PubMed] [Google Scholar]
  165. Wilson WH, Young RM, Schmitz R, Yang Y, Pittaluga S, Wright G, Lih CJ, Williams PM, Shaffer AL, Gerecitano J, et al. 2015. Targeting B cell receptor signaling with ibrutinib in diffuse large B cell lymphoma. Nat Med 21: 922–926. 10.1038/nm.3884 [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Wood B, Wu D, Crossley B, Dai Y, Williamson D, Gawad C, Borowitz MJ, Devidas M, Maloney KW, Larsen E, et al. 2018. Measurable residual disease detection by high-throughput sequencing improves risk stratification for pediatric B-ALL. Blood 131: 1350–1359. 10.1182/blood-2017-09-806521 [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Woyach JA, Furman RR, Liu TM, Ozer HG, Zapatka M, Ruppert AS, Xue L, Li DH, Steggerda SM, Versele M, et al. 2014. Resistance mechanisms for the Bruton's tyrosine kinase inhibitor ibrutinib. New Engl J Med 370: 2286–2294. 10.1056/NEJMoa1400029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Wu D, Sherwood A, Fromm JR, Winter SS, Dunsmore KP, Loh ML, Greisman HA, Sabath DE, Wood BL, Robins H. 2012. High-throughput sequencing detects minimal residual disease in acute T lymphoblastic leukemia. Sci Transl Med 4: 134ra63. [DOI] [PubMed] [Google Scholar]
  169. Yatabe Y, Suzuki R, Tobinai K, Matsuno Y, Ichinohasama R, Okamoto M, Yamagauchi M, Tamaru J, Uike N, Hashimoto Y, et al. 2000. Significance of cyclin D1 overexpression for the diagnosis of mantle cell lymphoma: A clinicopathologic comparison of cyclin D1-positive MCL and cyclin D1-negative MCL-like B-cell lymphoma. Blood 95: 2253–2261. [PubMed] [Google Scholar]
  170. Yuan J, Wright G, Rosenwald A, Steidl C, Gascoyne RD, Connors JM, Mottok A, Weisenburger DD, Greiner TC, Fu K, et al. 2015. Identification of primary mediastinal large B-cell lymphoma at nonmediastinal sites by gene expression profiling. Am J Surg Pathol 39: 1322–1330. 10.1097/PAS.0000000000000473 [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. Zech L, Haglund U, Nilsson K, Klein G. 1976. Characteristic chromosomal abnormalities in biopsies and lymphoid-cell lines from patients with Burkitt and non-Burkitt lymphomas. Int J Cancer 17: 47–56. 10.1002/ijc.2910170108 [DOI] [PubMed] [Google Scholar]
  172. Zhang J, Jima D, Moffitt AB, Liu Q, Czader M, Hsi ED, Fedoriw Y, Dunphy CH, Richards KL, Gill JI, et al. 2014. The genomic landscape of mantle cell lymphoma is related to the epigenetically determined chromatin state of normal B cells. Blood 123: 2988–2996. 10.1182/blood-2013-07-517177 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Cold Spring Harbor Perspectives in Medicine are provided here courtesy of Cold Spring Harbor Laboratory Press

RESOURCES