Abstract

Comprehensive genetic analysis of tumors with exome or whole-genome sequencing has enabled the identification of the genes that are recurrently mutated in cancer. This has stimulated a series of exciting advances over the past 15 years, guiding us to new molecular biomarkers and therapeutic targets among the common mature B-cell neoplasms. In particular, diffuse large B-cell lymphoma (DLBCL), follicular lymphoma (FL), and Burkitt lymphoma (BL) have each been the subject of considerable attention in this field. Currently, >850 genes have been reported as targets of protein-coding mutations in at least 1 of these entities. To reduce this to a manageable size, we describe a systematic approach to prioritize and categorize these genes, based on the quality and type of supporting data. For each entity, we provide a list of candidate driver genes categorized into Tier 1 (high-confidence genes), Tier 2 (candidate driver genes), or Tier 3 (lowest-confidence genes). Collectively, this reduces the number of high-confidence genes for these 3 lymphomas to a mere 144. This further affirms the substantial overlap between the genes relevant in DLBCL and each of FL and BL. These highly curated and annotated gene lists will continue to be maintained as a resource to the community. These results emphasize the extent of the knowledge gap regarding the role of each of these genes in lymphomagenesis. We offer our perspective on how to accelerate the experimental confirmation of drivers using a variety of model systems, using these lists as a guide for prioritizing genes.

A central goal in cancer genomics is to delineate the genetic and molecular changes that contribute to the onset and progression of cancer. By definition, driver mutations afford a fitness advantage to cells that ultimately give rise to a neoplastic clone. The term “driver” can refer to the individual mutations that contribute to cancer and as shorthand for any gene whose mutation can promote oncogenesis. Driver genes are identified by comparing the mutation profiles of tumors from a cohort of patients with the same diagnosis or pathology.1 Although the computational strategies to accomplish this vary, each seeks to identify the genes with a somatic mutation rate and/or pattern distinct from that expected by chance, often reported as significantly mutated genes. When applied to a single cancer diagnosis such as Burkitt lymphoma (BL), these genes can be considered candidate drivers within that entity, or “BL genes.” Although some driver genes are pervasive, with similar roles in many cancers, others appear mutated in a limited number of entities. For a surprising fraction of these genes, the mechanism by which their mutation contributes to lymphoma initiation or progression is merely speculative. In this review, we summarize the current landscape of knowledge for the established and suspected drivers across 3 major types of B-cell lymphomas: diffuse large B-cell lymphoma (DLBCL),2-7 follicular lymphoma (FL),2,8-11 and BL.12-15 

An unwritten rule in cancer genomics is that each cohort-based study is accompanied by a significantly mutated gene list, typically comprising a mixture of known and novel genes. For any well-studied entity, this paradigm has 2 consequences: an inconsistency in the genes reported in each study, and a perpetually growing collection of drivers. Accordingly, there has been a surge of candidate lymphoma genes since the availability of massively parallel sequencing. The ongoing “discovery” of new cancer genes might not be surprising, considering the estimate that up to 5000 samples may be needed to saturate the drivers for some malignancies.1 Based on this projection, one could reason that the genes unique to any study (orphan genes) merely reflect interpatient heterogeneity,16 and will eventually be confirmed in subsequent studies. We see this explanation as incompatible with the data. Instead, the literature represents a mix of true drivers alongside genes that have no relevance to lymphoma, and without addressing them, such errors continue to be propagated, and the collection of genes continue to grow. This leaves the scientific community with the challenge of how to prioritize these genes and on which to focus efforts for future experiments involving targeted sequencing or functional characterization.

Collectively, the 3 B-cell lymphomas covered by this review have been the subject of dozens of genomic screens. With this level of attention, one might naively assume that we can agree on which genes are the common target of mutations in each. One of our goals with this review was to address the following question: how many (and which) genes are relevant drivers in each? Here, we propose a framework to curate and categorize candidate drivers based on the supporting evidence. We provide annotated lists of candidate drivers for DLBCL, FL, and BL, which are in the supplemental Data and a public version–controlled repository, as a resource to the community that will continue to be actively maintained (https://github.com/morinlab/LLMPP/).

In a perfect world, every genomic survey of the same malignancy would arrive at essentially the same list of driver genes. In lieu of this, a single publication might be designated as the gold standard based on some objective criteria. Experimental design considerations such as the type and volume of data (coverage), source of nucleic acids (frozen/formalin-fixed paraffin-embedded) and the inclusion of matched constitutional DNA to remove germ line variants can affect the accuracy of a gene list.17 Small cohorts, insufficient coverage, and limited gene panels, can all hamper the detection of genes with lower mutation rates or miss genes completely. Inadequate removal of systematic artifacts or germ line variants can increase the rate of false positives. We compared gene lists from 12 DLBCL studies spanning a decade to estimate the accuracy of each result. By comparing the gene list from each study to the rest, the relative number of orphan genes can serve as a proxy for the false-positive rate. A caveat is that more recent gene lists naturally have higher orphan rates because there has been less opportunity for those genes to be subsequently confirmed. We partially mitigated this by comparing smaller sets of studies, grouping them by age (Figure 1A). These comparisons reveal a striking range of discordance in the published gene lists, with some studies having orphan rates of 82% (263 of 319 genes in Zhang et al16) and 55% (73 of 133 genes in Fan et al18).

Figure 1.

Orphan genes and the usual suspects. (A) The shared and orphan genes obtained by intersecting gene lists from the first 6 large DLBCL studies (top) and the remaining 6 studies (bottom). Each set of studies is also intersected with the current Tier 1 DLBCL gene list. The genes in each intersection are also identified by red or blue circles on panel C. (B) The number of DLBCL genes in Tier 1 and the full list is shown at the time of each of the 12 large studies.2,3,5-7,16,18-23 The totals at each time point include genes that were identified previously or in other contemporaneous studies that are not part of this figure. The citations for studies that nominated these additional genes are as follows: FAS,24,CARD11,25,CXCR4,26,PIM1,27,CREBBP/EP300,28,EZH2,29,MYD88,30,BCL2,31,IL4R,32,NFKBIZ,33,MS4A1,34,XPO1,35,BRAF,36,PRDM1,37,TNFAIP3,38,MYC,27 and STAT6.39 (C) For each column below a study in panel B, the Tier 1 DLBCL genes newly introduced by the study are grouped together and indicated in the corresponding color. For all subsequent studies, a gene is colored violet if it was on the significantly mutated gene (SMG) list of that study, which is considered a confirmation of that gene. The total number of confirming studies is indicated next to each gene using a gradient.

Figure 1.

Orphan genes and the usual suspects. (A) The shared and orphan genes obtained by intersecting gene lists from the first 6 large DLBCL studies (top) and the remaining 6 studies (bottom). Each set of studies is also intersected with the current Tier 1 DLBCL gene list. The genes in each intersection are also identified by red or blue circles on panel C. (B) The number of DLBCL genes in Tier 1 and the full list is shown at the time of each of the 12 large studies.2,3,5-7,16,18-23 The totals at each time point include genes that were identified previously or in other contemporaneous studies that are not part of this figure. The citations for studies that nominated these additional genes are as follows: FAS,24,CARD11,25,CXCR4,26,PIM1,27,CREBBP/EP300,28,EZH2,29,MYD88,30,BCL2,31,IL4R,32,NFKBIZ,33,MS4A1,34,XPO1,35,BRAF,36,PRDM1,37,TNFAIP3,38,MYC,27 and STAT6.39 (C) For each column below a study in panel B, the Tier 1 DLBCL genes newly introduced by the study are grouped together and indicated in the corresponding color. For all subsequent studies, a gene is colored violet if it was on the significantly mutated gene (SMG) list of that study, which is considered a confirmation of that gene. The total number of confirming studies is indicated next to each gene using a gradient.

Close modal

If all gene lists are affected by false positives and/or false negatives, how can we determine how many driver genes exist for a given malignancy? The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours (WHO-HAEM5) places the number of DLBCL genes at ∼150,40 which is based on the list proposed by Reddy et al.19 Notably, this lacks important genes that were established as DLBCL genes in earlier studies. These include ZFP36L1 and HIST1H1C,2 and DTX1, the latter having been reported as a driver by the same group.16,41 Additional DLBCL drivers identified in separate studies, such as NOTCH1,3,5,PRKDC,5,20,ITPKB,5,ID3,5,7,HVCN1,6,7 and SIN3A6,7 are also missing from that list.5-7,21 In all cases, the importance of mutations in these genes has been established by observation in a second study or characterization in model systems.42-45 Along with these absences, the list of Reddy et al had the third highest orphan gene rate (31%), implying a significant number of false positives. Some of the more recent studies of large cohorts were more conservative, each reporting <100 genes.5,6 Of the 99 genes nominated by Chapuy et al6 only 58 overlap with the list of Reddy et al. Based on this intersection, we can confidently state that there are at least 58 DLBCL genes, but this is far from complete.

A fair and thorough consolidation effort should somehow consider the gene lists from every study while avoiding propagating errors. The reason for this becomes clear if we instead accept every gene, even if reported by a single study. Exploring how this affects DLBCL as a function of time, the total number of genes grows from 81 at the end of 20112,3,28,30 to nearly 700 a decade later (Figure 1B), with most of these genes originating from a pair of papers from the same group.16,18 Although this strategy yields a comprehensive list, it clutters our list with genes unlikely to be relevant to DLBCL, thus is of limited value without further contextualization. A conservative way to mitigate this issue is to limit our list to the genes reported by a reasonable number of studies. Requiring a consensus across many studies, however, is equally problematic. For example, only 7 genes lie at the intersection of the first 6 DLBCL lists. The result is similar if the more recent group of studies are compared (Figure 1A). A more balanced approach to reduce the perpetuation of false positives without discarding valid candidates is to prioritize the genes reported by at least 2 studies. Based on the 12 studies, this brings us to 153 DLBCL genes, a number close to the current WHO-HAEM5 estimate.

Based on these concepts, we implemented a flexible approach to consolidate, curate, and categorize candidate B-cell lymphoma drivers into a separate gene list for each pathology. We began by identifying all studies that promoted at least 1 gene as recurrently mutated (by protein-coding mutations) in DLBCL, BL, or FL. Starting with the superset of novel genes from all such surveys of a given pathology, we record the initial study mentioning the gene (ie, the “originating study”). Genes are assigned to tiers to reflect our confidence in their role in that entity, with Tier 1 and Tier 2, respectively, representing the high- and moderate-confidence genes. Genes of particularly low confidence can also be assigned to a third tier; however, not all lists contain Tier 3 genes. This process draws from gene lists in all available peer-reviewed studies that sequenced patient samples of the entity and, in some situations, manual review and/or reanalysis of the primary data (supplemental Methods).41,46 Genes are assigned to Tier 2 by default, in which they can remain indefinitely. Promotion to Tier 1 typically results from additional evidence of recurrent mutation demonstrated by a subsequent study. Figure 1C shows the origin of each Tier 1 DLBCL gene and the confirmation of many of these across subsequent studies.

The availability of functional experimental data demonstrating the role of a mutation in lymphoma biology also allows a gene to be promoted to Tier 1. For example, B2M was a Tier 2 DLBCL gene as of its first description in 2011.2 It became eligible for Tier 1 when another group demonstrated their role in modulating recognition by immune cells.47 In rare situations, the originating study may include functional data demonstrating the effect of the mutations in an appropriate model system, in which case the gene is immediately eligible for Tier 1. MYD88, MS4A1, CREBBP, and EP300 in DLBCL are examples of such genes.28,30,34 We do not consider hits from generic pooled CRISPR screens as sufficient evidence to validate a candidate driver because they do not model the effect of the specific mutations observed. In general, determining the adequacy of functional data involves a more nuanced assessment of each study that is discussed hereafter.

Another complication affecting the genetic analysis of these lymphomas is the influence of aberrant somatic hypermutation on the genome, which causes an elevated mutation rate in certain genes.27 Many genes affected by aberrant somatic hypermutation have been assigned to Tier 1 because of the mere recurrence of mutations.48 Additional caution should be used when considering these mutations as drivers without additional evidence of their relevance. Importantly, although these may not all represent drivers, these mutations contribute to the classification of DLBCL,6,48 BL,13 and FL49,50 into genetic subgroups. The Tier 1 list for DLBCL currently contains 126 protein-coding genes, which represents the lower bound for the full set of drivers (supplemental Table 1). The experimental demonstration of a gene or mutation represents a laborious and costly barrier to the growth of Tier 1 genes. We anticipate the promotion of additional Tier 2 genes as more genetic and functional data emerge and, in a subsequent section, discuss how this might be accelerated.

Although Tier 1 genes can be considered bona fide drivers, the genes in Tier 2 should represent only those with a reasonable chance of eventually joining Tier 1. Across the Tier 2 genes, we observed a substantial range in the quantity and quality of supporting data. To reduce the size of Tier 2, genes can be moved to Tier 3 if they meet either of 2 strict criteria. First, a gene is assigned to Tier 3 if a minority of the variants in that gene fail to meet minimal quality thresholds upon manual review of the original data.41 This is based on the rationale that, if stricter criteria were used, the gene would not likely have been nominated in the first place. Secondly, we have reduced confidence in any study whose results cannot be reproduced upon reanalysis of the same data. Accordingly, if a subsequent analysis of the same samples fails to reproduce at least half of the driver mutations reported in the initial study, all Tier 2 genes from that study are assigned to Tier 3. Thus far, this criterion has only been applied to 2 studies. More information is provided in supplemental Table 7 and the supplemental Results.16,18 Collectively, this results in 387 Tier 3 DLBCL genes and 70 Tier 3 BL genes.

Although this practice intends to enrich Tier 2 for genes with potential merit, the contents of the Tier 3 lists should not be entirely dismissed. Although there is minimal evidence that their mutation contributes to lymphomagenesis, some may nonetheless be relevant to lymphoma biology. For example, mutations in SYK and MYB have not been confirmed but these genes respectively have established roles in B-cell receptor signaling and hematopoiesis.19 It is conceivable that the reliance of a few studies on RNA sequencing for detecting mutations could bias their gene list toward genes that are highly expressed in malignant cells.2,14,51 We acknowledge that low mutation incidence alone is not evidence against a gene acting as a driver. TET2 and NOTCH1, respectively mutated in 5% and 7% of DLBCLs, were recognized as drivers in this entity relatively recently, likely escaping detection earlier because of this low incidence. Tier 3 is significantly enriched for genes that have an elevated rate of rare germ line variants in the human population (supplemental Results).52 This suggest that many of the genes in Tier 3 result from inadequate removal of germ line variants.

Importantly, some genes in Tier 3 for 1 entity represent high-confidence genes for another. For example, 11 of Tier 3 BL genes are high-confidence DLBCL genes, including NOTCH1, BRAF, CD79B, ETS1, and BTG2. Each of these were attributed to BL by Love et al or Panea et al.14,53 The existence of DLBCL-associated mutations in BL cohorts could be explained by insufficient pathology review. Consistent with this, a classifier trained on DLBCL and BL genetics assigned a third of the patients from that study to the DLBCL group.54 When cost is a consideration when designing custom sequencing panels, we recommend that Tier 3 genes should be considered low priority and excluded if necessary.

The gene lists for BL, FL, and DLBCL naturally contain significant overlap because many of the same genes are mutated in multiple B-cell neoplasms.55 In particular, it has long been recognized that FL and germinal center B-cell–like DLBCL share many driver mutations.2,20,49 For example, EZH2 hot spot mutations are a feature of FL and DLBCL but are rarely found in any of the other common B-cell lymphomas.54 In contrast, BRAF is commonly mutated across a variety of solid tumors including epithelial cancers,56 whereas these mutations are relatively rare across the mature B-cell neoplasms, with the exception of hairy cell leukemia. Interestingly, there are more documented drivers in DLBCL than any other mature B-cell neoplasm, perhaps reflecting that DLBCL represents an umbrella term comprising at least 5 to 7 subtypes with distinct molecular underpinnings, most sharing some biological features with another B-cell malignancy.57 Accordingly, the number of genes that are unique to DLBCL as a whole appears low.

Despite sharing many drivers with DLBCL, the FL list is substantially shorter, comprising 54 Tier 1 genes and 58 Tier 2 genes. This might partially be a consequence of fewer large-scale studies that have focused on this pathology. In just the past few years, multiple studies focusing on FL have emerged (Figure 2) and we anticipate that additional such studies will allow the more recent Tier 2 genes to be promoted to Tier 1. We also expect additional Tier 1 DLBCL genes to eventually enter the FL list. The separate gene list for FL is justified, in part, for genes that are scantily mutated in DLBCL and unlikely to be attributed to both entities (Figure 3). Currently, this is limited to MAP2K1, ATP6AP1, ATP6V1B2, CTSS, and VMA21. MAP2K1 mutations are limited to pediatric-type FL,58 whereas the others are seen in adult FL. For completeness, we anticipate that researchers studying either FL, BL, or DLBCL may prefer to pool the Tier 1 genes from all lists (supplemental Table 1), which is only slightly longer than the DLBCL list (supplemental Table 2) because of overlap with the others (supplemental Tables 3 and 4).

Figure 2.

The source and status of each B-cell lymphoma gene. (A) Studies that reported >1 SMG in FL, BL, or DLBCL are summarized according to the method used to identify mutations (Sanger sequencing,27,59 RNA sequencing,51 whole-genome sequencing [WGS],7,12,20,49,54,60 exome,5,6,10,18,19,21,22,33,53,61 panel,31,32,62 or a combination thereof2,14,15,29,63). Studies are also separated by the lymphoma type that was sequenced, with those using a combination of FL and DLBCL grouped in the middle section along with FL-centric studies. Additional details for each study are also provided in supplemental Table 5. The height of each vertical bar is proportional to the number of newly identified SMGs reported by the study. A gene was not counted if it had already been reported in that entity at the time of the study, but a gene may appear more than once if it was reported in multiple entities (eg, FL and DLBCL). Representative examples of genes that are in either Tier 2 or 3 are shown in the middle, adjacent to the corresponding citation. The number of genes originating from each study and their assignment to 1 of the 3 tiers is summarized on the right (B).

Figure 2.

The source and status of each B-cell lymphoma gene. (A) Studies that reported >1 SMG in FL, BL, or DLBCL are summarized according to the method used to identify mutations (Sanger sequencing,27,59 RNA sequencing,51 whole-genome sequencing [WGS],7,12,20,49,54,60 exome,5,6,10,18,19,21,22,33,53,61 panel,31,32,62 or a combination thereof2,14,15,29,63). Studies are also separated by the lymphoma type that was sequenced, with those using a combination of FL and DLBCL grouped in the middle section along with FL-centric studies. Additional details for each study are also provided in supplemental Table 5. The height of each vertical bar is proportional to the number of newly identified SMGs reported by the study. A gene was not counted if it had already been reported in that entity at the time of the study, but a gene may appear more than once if it was reported in multiple entities (eg, FL and DLBCL). Representative examples of genes that are in either Tier 2 or 3 are shown in the middle, adjacent to the corresponding citation. The number of genes originating from each study and their assignment to 1 of the 3 tiers is summarized on the right (B).

Close modal
Figure 3.

Overlaps and contrasts between the 3 gene lists. (A) A modified Venn diagram illustrating the intersections between the DLBCL, FL, and BL gene lists with nested circles representing the Tier 1 and Tier 2 genes for each entity. When not all genes in a group can be shown because of space limitations, these lists end in an ellipsis. Tier 3 genes are excluded to simplify the figure. Representative genes in each intersection are shown but re not exhaustive for the larger intersections. Intersections sharing no genes are denoted with the “∅” symbol. (B) A 3-dimensional plot showing the relative enrichment of mutations in each entity relative to the other 2. Each axis represents the log (odds ratio) from a Fisher exact test comparing the 2 entities indicated on that axis. For example, the DLBCL vs BL dimension is based on the relative incidence of mutations in these genes in a large collection of DLBCL and BL tumors. The points represent a selection of representative genes from the Tier 1 intersections from the Venn diagram. Points are colored according to the gene list(s) in which they reside. The 3 arrows indicate the location of points with relative enrichments in each of the entities. Most of the DLBCL and BL genomes used for this analysis were described in our recent study.54 Additional DLBCLs and FLs were originally sequenced as part of the ICGC malignant lymphoma (Germany) MALY-DE project,20 a Genome Canada-funded project in BC Cancer,49,64 or the Burkitt Lymphoma Genome Sequencing Project.12 The DLBCL exomes largely from the study by Schmitz et al,5 Chapuy et al,6 Lohr et al,22 and our previous studies.34,64 

Figure 3.

Overlaps and contrasts between the 3 gene lists. (A) A modified Venn diagram illustrating the intersections between the DLBCL, FL, and BL gene lists with nested circles representing the Tier 1 and Tier 2 genes for each entity. When not all genes in a group can be shown because of space limitations, these lists end in an ellipsis. Tier 3 genes are excluded to simplify the figure. Representative genes in each intersection are shown but re not exhaustive for the larger intersections. Intersections sharing no genes are denoted with the “∅” symbol. (B) A 3-dimensional plot showing the relative enrichment of mutations in each entity relative to the other 2. Each axis represents the log (odds ratio) from a Fisher exact test comparing the 2 entities indicated on that axis. For example, the DLBCL vs BL dimension is based on the relative incidence of mutations in these genes in a large collection of DLBCL and BL tumors. The points represent a selection of representative genes from the Tier 1 intersections from the Venn diagram. Points are colored according to the gene list(s) in which they reside. The 3 arrows indicate the location of points with relative enrichments in each of the entities. Most of the DLBCL and BL genomes used for this analysis were described in our recent study.54 Additional DLBCLs and FLs were originally sequenced as part of the ICGC malignant lymphoma (Germany) MALY-DE project,20 a Genome Canada-funded project in BC Cancer,49,64 or the Burkitt Lymphoma Genome Sequencing Project.12 The DLBCL exomes largely from the study by Schmitz et al,5 Chapuy et al,6 Lohr et al,22 and our previous studies.34,64 

Close modal

Although having been the subject of multiple whole-genome sequencing,12,54 whole-exome sequncing,14,15,53 and targeted sequencing studies,62,65 BL currently has only 34 Tier 1 genes (Figure 2) with another 77 genes residing in Tier 2. Some of the earliest exome-based studies nominated a substantial number of genes but most have since been assigned to Tier 3 (Figure 2; supplemental Table 4).53 As the most recent genome-wide studies have individually contributed a small number of new genes, the Tier 1 BL list may be approaching saturation (supplemental Table 5). This would suggest that BL is more genetically homogeneous than FL or DLBCL. Interestingly, more than a third of the BL genes overlap with DLBCL and FL, including KMT2D, FOXO1, GNA13, TP53, CCND3, ARID1A, and SMARCA4. There is further sharing of driver genes between Tier 1 BL genes and DLBCL that does not extend to FL, such as DDX3X, HNRNPU, SIN3A, and RHOA. Some of these likely represent mutations that are only relevant in conjunction with other common BL drivers. For example, DDX3X mutations protect against the collateral proteotoxic stress experienced by cells harboring IGH::MYC.66 Only 6 of the Tier 1 BL genes are not shared with either the FL or BL Tier 1 lists whereas many more Tier 2 BL genes overlap the Tier 1 DLBCL list (Figure 3).

We noted large disparities in the sampling of Tier 1 genes across panel-based studies. For example, the panel used by Burkhardt et al included only 22 of the Tier 1 BL genes, with notable exclusions such as KMT2D, HNRNPU, and USP7.62 Similarly, Crouch et al sequenced 44 of the 54 Tier 1 FL genes, excluding important genes such as ATP6AP1, ATP6V1B2, and CTSS.50 In designing a general panel for application to any of these 3 entities, we suggest the inclusion of the 144 genes that exist in at least 1 of the Tier 1 lists (supplemental Table 1). Importantly, none of the panel-based studies sequenced all 144 Tier 1 genes.64 The panels that included the largest fraction of these genes were used in Krysiak et al and Russler-Germain et al (supplemental Table 6).31,32 

The recurrent mutation of a gene within a disease entity is suggestive that the altered function of the gene product contributed to initiation or progression of malignancy. However, the strongest evidence of a driver comes from experimental demonstration of a lymphoma-driving impact on cell behavior or fitness. Although the list of drivers may seem manageable, with only 144 Tier 1 genes across BL, DLBCL, and FL, we published studies detailing the function for a mere 58 of these (Figure 4). Functional characterization of drivers is complicated by the fact that most genes have many different recurrent mutations, not all of which can be considered functionally equivalent. Moreover, the consequence of any mutation may involve genetic interactions, which can only be revealed in combination with specific alterations in other genes. Finally, to exert their relevant effect, the effect of some mutations may require modeling within a specific developmental stage of B-cell biology and/or microenvironmental setting. The importance of modeling each mutation in the appropriate biological context is supported by contrasting the mutation patterns in the same gene between entities. For example, the dominant class of mutations in CREBBP is distinct between DLBCL and FL, with the latter predominantly harboring missense rather than truncating mutations.49 Although this observation implies different selective pressures, it remains unclear whether this is because of different microenvironmental pressures or cellular context of the premalignant cells (cell of origin). Biological sex is another variable that must be considered, as illustrated by the distinct mutation patterns seen in DDX3X among male and female patients with BL.67 

Figure 4.

Mutation patterns and effect on all Tier 1 genes. Genes in Tier 1 on any of the FL, DLBCL, or BL lists are shown. Because genes can exist in >1 list and may not be assigned the same tier, the tier for each entity is indicated on the right (Tier 1, green; Tier 2, yellow; Tier 3, amber). The heat map shows the proportion of cases with mutations in that gene is shown above each entity with deeper red indicating a higher incidence. Genes known to be affected by aberrant somatic hypermutation (aSHM) are indicated with a gray rectangle to the right. The existence of functional data is indicated on the far right. The color shows whether mutations have been functionally determined to represent a loss of function (violet), gain of function (red), or neomorph (yellow). The studies demonstrating this result for these genes are as follows: ARID1A,68,ID3,51,TCF3,51,CCND3,51,FOXO1,69,GNA13,59,S1PR2,59,RHOA,59,P2RY8,59,KMT2D,70,71,SMARCA4,72,BCL6,73,BCL7A,74,DDX3X,66,FBXO11,75,TFAP4,76,CREBBP,77,78,EP300,28,EZH2,79,80,IRF8,81,PIM1,82,BTG1,83,84,CDKN2A,85,CD79B,86,DTX1,87,NOTCH1,43,POU2AF1,88,POU2F2,89,B2M, CD58,47,BCL10,90,CARD11,25,FAS,91,RRAGC,60,ATP6V1B2,60,ATP6AP1,60,STAT6,92,TBL1XR1,93,TNFAIP3,94,KLHL6,95,MEF2B,96,SGK1,97,SOCS1,98,MYD88,99 and TMEM30A.100 The citations for these and all remaining genes are included in the supplemental Tables.

Figure 4.

Mutation patterns and effect on all Tier 1 genes. Genes in Tier 1 on any of the FL, DLBCL, or BL lists are shown. Because genes can exist in >1 list and may not be assigned the same tier, the tier for each entity is indicated on the right (Tier 1, green; Tier 2, yellow; Tier 3, amber). The heat map shows the proportion of cases with mutations in that gene is shown above each entity with deeper red indicating a higher incidence. Genes known to be affected by aberrant somatic hypermutation (aSHM) are indicated with a gray rectangle to the right. The existence of functional data is indicated on the far right. The color shows whether mutations have been functionally determined to represent a loss of function (violet), gain of function (red), or neomorph (yellow). The studies demonstrating this result for these genes are as follows: ARID1A,68,ID3,51,TCF3,51,CCND3,51,FOXO1,69,GNA13,59,S1PR2,59,RHOA,59,P2RY8,59,KMT2D,70,71,SMARCA4,72,BCL6,73,BCL7A,74,DDX3X,66,FBXO11,75,TFAP4,76,CREBBP,77,78,EP300,28,EZH2,79,80,IRF8,81,PIM1,82,BTG1,83,84,CDKN2A,85,CD79B,86,DTX1,87,NOTCH1,43,POU2AF1,88,POU2F2,89,B2M, CD58,47,BCL10,90,CARD11,25,FAS,91,RRAGC,60,ATP6V1B2,60,ATP6AP1,60,STAT6,92,TBL1XR1,93,TNFAIP3,94,KLHL6,95,MEF2B,96,SGK1,97,SOCS1,98,MYD88,99 and TMEM30A.100 The citations for these and all remaining genes are included in the supplemental Tables.

Close modal

Understanding the mechanistic contribution of each driver gene to lymphoma biology and deciphering this complex network of cooperating mutations is an essential challenge we must overcome if we are to exploit the full potential of genomic medicine in lymphoma. The evidence required for this functional annotation may come in many forms, typically starting by drawing inferences from the mutation pattern. For example, genes with a preponderance of truncating mutations are presumed to be tumor suppressors whereas those with mutation hot spots are expected to exhibit gain of function. Among the lymphoma genes, counter-examples of each of these have been documented.97 The strongest support, sufficient to validate a driver, is established using an experimental system that allows quantification of a biological phenotype after the introduction of a defined genetic perturbation in an experimentally controlled manner. The technology available for such experiments is constantly evolving, allowing many genetic perturbations to be interrogated in high throughput in multiple different cellular contexts. Scalability is paramount when we consider the goal of modeling hundreds or thousands of mutations in combinations of 2, 3, or 4, the number of permutations to model rapidly becomes very large.

Perhaps the most direct evidence of a lymphoma-promoting function of an individual mutation is provided by using genetically modified mouse models. Intercrossing allows multiple mutant alleles to be combined. These models have the advantage of recapitulating the immune microenvironment, and conditional gene targeting allows introduction as specific stages of B-cell development. Numerous such models have demonstrated the ability of specific genetic combinations to drive transformation of B cells to diseases that mimic human lymphomas.101-105 However, transgenic mouse models are costly, slow to generate and unsuited to high-throughput analysis. Moreover, the requirements for lymphocyte transformation in the mouse do not always faithfully recapitulate the situation in human cells.

An alternative approach has been to use human lymphoma cell lines.106 These are ideally suited to high-throughput screening technologies such as genome-wide CRISPR screening and have proven a workhorse tool for mechanistic investigation of lymphoma functional genomics. However, cell lines do not exist for some lymphoma types. For instance, all cell lines associated with FL come from tumors that have transformed to DLBCL. The absence of lines representing untransformed FL may reflect the essential dependence of this lymphoma type upon the microenvironment. Moreover, cell lines may have been growing for decades during which time they have evolved a complex and skewed repertoire of genetic mutations, that is already perfectly optimized for in vitro growth. This limits the capacity to test the introduction of new mutations on a specific genetic background, but also makes it hard to test the ability of any genetic perturbation to increase the fitness of the cell line. To overcome these issues, we developed a novel coculture system.107,108 By mimicking the survival signals provided by the lymph node microenvironment we were able to support the growth and proliferation of normal, nonmalignant germinal center B cells purified from discarded pediatric tonsillectomy tissue. Although these cells cannot be transduced with conventional vesicular stomatitis virus G pseudotyped virus, the use of a gibbon ape leukemia virus and murine leukemia virus fusion envelope allowed high-efficiency transduction with both lentivirus and retrovirus. This allows for efficient genetic modification of nonmalignant human GC B cells and the potential to overexpress or knock out by CRISPR any desired putative cancer driver gene, in its wild type or mutant form and into a genetically normal background or in any desired genetic combination. Moreover, this system is scalable and suited to high-throughput screening using targeted or whole genome CRISPR libraries, or over expression of wild type or mutant open reading frame libraries. Established lymphoma cell lines are most amenable to dropout screening, which identifies genes essential for survival of malignant B cells in the context of an existing set of somatic mutations.99 In contrast, the use of primary cells allows for the controlled introduction and identification of genetic perturbations that afford a gain of fitness to the cell, providing an ideal system for the assessment of putative lymphoma driver genes. However, this powerful system is not without limitation, most prominently the inability to model interaction with the host immune system. In an attempt to model this microenvironmental interaction several groups have established autologous coculture systems from normal109 or malignant lymphoid tissue110 to establish in vitro organoid models of the germinal center or of B-cell lymphomas. These have not yet been combined with genetic modification but may provide a future strategy to assess the potential of lymphoma driver genes to modify interaction with the host immune system.

The technology for genetic editing and phenotypic interrogation continues to see rapid improvements. CRISPR provides a strategy to mimic loss of function mutations by knockout of the gene, which is routinely done at genome scale. Single-nucleotide variants can be introduced by targeted CRISPR editing to recapitulate specific mutations, but this is relatively inefficient and laborious. In contrast, newer technologies such as base editing now allow near nucleotide-level interrogation of individual nucleotide changes in high throughput in cell lines or primary B cells.111 Improvements in single-cell RNA sequencing now allow hundreds of thousands of cell transcriptomes to be profiled in a single experiment. These can be combined with barcoded perturbations such as CRISPR knockout, base editing, or open-reading-frame libraries to profile the transcriptional impact of many hundreds of genetic perturbations in a single experiment, providing mechanistic insight into the functional impact of each genetic change beyond its impact on competitive fitness.

Each experimental strategy balances advantages and limitations, but together these model systems offer a complementary set of tools to interrogate the function of putative lymphoma genes, at the resolution of individual nucleotide changes. They provide the tools to do this at scale and to screen for synergy between cooperating mutations. As techniques for genetic editing, cell barcoding and phenotypic interrogation continues to advance, we hope to witness an acceleration in our understanding of the role and function of lymphoma drivers.

The authors are grateful to the courageous patients who selflessly donated their valuable samples to further cancer research.

Contribution: R.D.M. and D.J.H. conceived of the study; K.M.C., R.D.M., and D.J.H. wrote the manuscript; and K.D. and R.D.M. created the figures and tables.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Ryan D. Morin, Simon Fraser University, 8888 University Dr, Burnaby, BC V5A 1S6, Canada; email: rdmorin@sfu.ca.

1.
Lawrence
MS
,
Stojanov
P
,
Mermel
CH
, et al
.
Discovery and saturation analysis of cancer genes across 21 tumour types
.
Nature
.
2014
;
505
(
7484
):
495
-
501
.
2.
Morin
RD
,
Mendez-Lago
M
,
Mungall
AJ
, et al
.
Frequent mutation of histone-modifying genes in non-Hodgkin lymphoma
.
Nature
.
2011
;
476
(
7360
):
298
-
303
.
3.
Pasqualucci
L
,
Trifonov
V
,
Fabbri
G
, et al
.
Analysis of the coding genome of diffuse large B-cell lymphoma
.
Nat Genet
.
2011
;
43
(
9
):
830
-
837
.
4.
Chong
LC
,
Ben-Neriah
S
,
Slack
GW
, et al
.
High-resolution architecture and partner genes of MYC rearrangements in lymphoma with DLBCL morphology
.
Blood Adv
.
2018
;
2
(
20
):
2755
-
2765
.
5.
Schmitz
R
,
Wright
GW
,
Huang
DW
, et al
.
Genetics and pathogenesis of diffuse large B-cell lymphoma
.
N Engl J Med
.
2018
;
378
(
15
):
1396
-
1407
.
6.
Chapuy
B
,
Stewart
C
,
Dunford
AJ
, et al
.
Molecular subtypes of diffuse large B cell lymphoma are associated with distinct pathogenic mechanisms and outcomes
.
Nat Med
.
2018
;
24
(
5
):
679
-
690
.
7.
Arthur
SE
,
Jiang
A
,
Grande
BM
, et al
.
Genome-wide discovery of somatic regulatory variants in diffuse large B-cell lymphoma
.
Nat Commun
.
2018
;
9
(
1
):
4001
.
8.
Kridel
R
,
Chan
FC
,
Mottok
A
, et al
.
Histological transformation and progression in follicular lymphoma: a clonal evolution study
.
PLoS Med
.
2016
;
13
(
12
):
e1002197
.
9.
Kalmbach
S
,
Grau
M
,
Zapukhlyak
M
, et al
.
Novel insights into the pathogenesis of follicular lymphoma by molecular profiling of localized and systemic disease forms
.
Leukemia
.
2023
;
37
(
10
):
2058
-
2065
.
10.
Russler-Germain
DA
,
Krysiak
K
,
Ramirez
C
, et al
.
Mutations associated with progression in follicular lymphoma predict inferior outcomes at diagnosis: Alliance A151303
.
Blood Adv
.
2023
;
7
(
18
):
5524
-
5539
.
11.
Green
MR
,
Kihira
S
,
Liu
CL
, et al
.
Mutations in early follicular lymphoma progenitors are associated with suppressed antigen presentation
.
Proc Natl Acad Sci USA
.
2015
;
112
(
10
):
E1116
-
E1125
.
12.
Grande
BM
,
Gerhard
DS
,
Jiang
A
, et al
.
Genome-wide discovery of somatic coding and noncoding mutations in pediatric endemic and sporadic Burkitt lymphoma
.
Blood
.
2019
;
133
(
12
):
1313
-
1324
.
13.
Thomas
N
,
Dreval
K
,
Gerhard
DS
, et al
.
Genetic subgroups inform on pathobiology in adult and pediatric Burkitt lymphoma
.
Blood
.
2023
;
141
(
8
):
904
-
916
.
14.
Panea
R
,
Love
C
,
Shingleton
JR
, et al
.
The whole genome landscape of Burkitt lymphoma subtypes
.
Blood
.
2019
;
134
(
19
):
1598
-
1607
.
15.
Richter
J
,
Schlesner
M
,
Hoffmann
S
, et al
.
Recurrent mutation of the ID3 gene in Burkitt lymphoma identified by integrated genome, exome and transcriptome sequencing
.
Nat Genet
.
2012
;
44
(
12
):
1316
-
1320
.
16.
Zhang
J
,
Grubor
V
,
Love
CL
, et al
.
Genetic heterogeneity of diffuse large B-cell lymphoma
.
Proc Natl Acad Sci U S A
.
2013
;
110
(
4
):
1398
-
1403
.
17.
Dreval
K
,
Boutros
PC
,
Morin
RD
.
Minimum information for reporting a genomics experiment
.
Blood
.
2022
;
140
(
24
):
2549
-
2555
.
18.
Fan
Z
,
Pei
R
,
Sha
K
,
Chen
L
,
Wang
T
,
Lu
Y
.
Comprehensive characterization of driver genes in diffuse large B cell lymphoma
.
Oncol Lett
.
2020
;
20
(
1
):
382
-
390
.
19.
Reddy
A
,
Zhang
J
,
Davis
NS
, et al
.
Genetic and functional drivers of diffuse large B cell lymphoma
.
Cell
.
2017
;
171
(
2
):
481
-
494.e15
.
20.
Hübschmann
D
,
Kleinheinz
K
,
Wagener
R
, et al
.
Mutational mechanisms shaping the coding and noncoding genome of germinal center derived B-cell lymphomas
.
Leukemia
.
2021
;
35
(
7
):
2002
-
2016
.
21.
Albuquerque
MA
,
Grande
BM
,
Ritch
EJ
, et al
.
Enhancing knowledge discovery from cancer genomics data with Galaxy
.
GigaScience
.
2017
;
6
(
5
):
1
-
13
.
22.
Lohr
JG
,
Stojanov
P
,
Lawrence
MS
, et al
.
Discovery and prioritization of somatic mutations in diffuse large B-cell lymphoma (DLBCL) by whole-exome sequencing
.
Proc Natl Acad Sci U S A
.
2012
;
109
(
10
):
3879
-
3884
.
23.
Morin
RD
,
Mungall
K
,
Pleasance
E
, et al
.
Mutational and structural analysis of diffuse large B-cell lymphoma using whole-genome sequencing
.
Blood
.
2013
;
122
(
7
):
1256
-
1265
.
24.
Scholl
V
,
Stefanoff
CG
,
Hassan
R
,
Spector
N
,
Renault
IZ
.
Mutations within the 5′ region of FAS/CD95 gene in nodal diffuse large B-cell lymphoma
.
Leuk Lymphoma
.
2007
;
48
(
5
):
957
-
963
.
25.
Lenz
G
,
Davis
RE
,
Ngo
VN
, et al
.
Oncogenic CARD11 mutations in human diffuse large B cell lymphoma
.
Science
.
2008
;
319
(
5870
):
1676
-
1679
.
26.
Khodabakhshi
AH
,
Morin
RD
,
Fejes
AP
, et al
.
Recurrent targets of aberrant somatic hypermutation in lymphoma
.
Oncotarget
.
2012
;
3
(
11
):
1308
-
1319
.
27.
Pasqualucci
L
,
Neumeister
P
,
Goossens
T
, et al
.
Hypermutation of multiple proto-oncogenes in B-cell diffuse large-cell lymphomas
.
Nature
.
2001
;
412
(
6844
):
341
-
346
.
28.
Pasqualucci
L
,
Dominguez-Sola
D
,
Chiarenza
A
, et al
.
Inactivating mutations of acetyltransferase genes in B-cell lymphoma
.
Nature
.
2011
;
471
(
7337
):
189
-
195
.
29.
Morin
RD
,
Johnson
NA
,
Severson
TM
, et al
.
Somatic mutations altering EZH2 (Tyr641) in follicular and diffuse large B-cell lymphomas of germinal-center origin
.
Nat Genet
.
2010
;
42
(
2
):
181
-
185
.
30.
Ngo
VN
,
Young
RM
,
Schmitz
R
, et al
.
Oncogenically active MYD88 mutations in human lymphoma
.
Nature
.
2011
;
470
(
7332
):
115
-
119
.
31.
Tanaka
S
,
Louie
DC
,
Kant
JA
,
Reed
JC
.
Frequent incidence of somatic mutations in translocated BCL2 oncogenes of non-Hodgkin’s lymphomas
.
Blood
.
1992
;
79
(
1
):
229
-
237
.
32.
Duns
G
,
Viganò
E
,
Ennishi
D
, et al
.
Characterization of DLBCL with a PMBL gene expression signature
.
Blood
.
2021
;
138
(
2
):
136
-
148
.
33.
Morin
RD
,
Assouline
S
,
Alcaide
M
, et al
.
Genetic landscapes of relapsed and refractory diffuse large B-cell lymphomas
.
Clin Cancer Res
.
2016
;
22
(
9
):
2290
-
2300
.
34.
Rushton
CK
,
Arthur
SE
,
Alcaide
M
, et al
.
Genetic and evolutionary patterns of treatment resistance in relapsed B-cell lymphoma
.
Blood Adv
.
2020
;
4
(
13
):
2886
-
2898
.
35.
Mareschal
S
,
Dubois
S
,
Viailly
P-J
, et al
.
Whole exome sequencing of relapsed/refractory patients expands the repertoire of somatic mutations in diffuse large B-cell lymphoma
.
Genes Chromosomes Cancer
.
2016
;
55
(
3
):
251
-
267
.
36.
Tiacci
E
,
Trifonov
V
,
Schiavoni
G
, et al
.
BRAF mutations in hairy-cell leukemia
.
N Engl J Med
.
2011
;
364
(
24
):
2305
-
2315
.
37.
Pasqualucci
L
,
Compagno
M
,
Houldsworth
J
, et al
.
Inactivation of the PRDM1/BLIMP1 gene in diffuse large B cell lymphoma
.
J Exp Med
.
2006
;
203
(
2
):
311
-
317
.
38.
Compagno
M
,
Lim
WK
,
Grunn
A
, et al
.
Mutations of multiple genes cause deregulation of NF-kappaB in diffuse large B-cell lymphoma
.
Nature
.
2009
;
459
(
7247
):
717
-
721
.
39.
Yildiz
M
,
Li
H
,
Bernard
D
, et al
.
Activating STAT6 mutations in follicular lymphoma
.
Blood
.
2015
;
125
(
4
):
668
-
679
.
40.
Alaggio
R
,
Amador
C
,
Anagnostopoulos
I
, et al
.
The 5th edition of the World Health Organization Classification of haematolymphoid tumours: lymphoid neoplasms. [published correction appears in Leukemia. 2023;37(9):1944-1951]
.
Leukemia
.
2022
;
36
(
7
):
1720
-
1748
.
41.
Dreval
K
,
Cruz
M
,
Rushton
C
, et al
.
Revisiting Reddy: a DLBCL do-over
.
bioRxiv
.
Preprint posted online 22 November 2023
.
42.
Arima
H
,
Nishikori
M
,
Otsuka
Y
, et al
.
B cells with aberrant activation of Notch1 signaling promote Treg and Th2 cell-dominant T-cell responses via IL-33
.
Blood Adv
.
2018
;
2
(
18
):
2282
-
2295
.
43.
Ryan
RJH
,
Petrovic
J
,
Rausch
DM
, et al
.
A B cell regulome links notch to downstream oncogenic pathways in small B cell lymphomas
.
Cell Rep
.
2017
;
21
(
3
):
784
-
797
.
44.
Tiacci
E
,
Ladewig
E
,
Schiavoni
G
, et al
.
Pervasive mutations of JAK-STAT pathway genes in classical Hodgkin lymphoma
.
Blood
.
2018
;
131
(
22
):
2454
-
2465
.
45.
Yusufova
N
,
Kloetgen
A
,
Teater
M
, et al
.
Histone H1 loss drives lymphoma by disrupting 3D chromatin architecture
.
Nature
.
2021
;
589
(
7841
):
299
-
305
.
46.
Rushton
CK
,
Dreval
K
,
Morin
RD
.
Concerning data inconsistencies in Burkitt lymphoma genome study
.
Blood
.
2023
;
142
(
10
):
933
-
936
.
47.
Challa-Malladi
M
,
Lieu
YK
,
Califano
O
, et al
.
Combined genetic inactivation of β2-microglobulin and CD58 reveals frequent escape from immune recognition in diffuse large B cell lymphoma
.
Cancer Cell
.
2011
;
20
(
6
):
728
-
740
.
48.
Wright
GW
,
Huang
D
,
Phelan
J
, et al
.
A probabilistic classification tool for genetics subtypes of diffuse large B cell lymphoma with therapeutic implications
.
Cancer Cell
.
2020
;
37
(
4
):
551
-
568.e14
.
49.
Dreval
K
,
Hilton
LK
,
Cruz
M
, et al
.
Genetic subdivisions of follicular lymphoma defined by distinct coding and noncoding mutation patterns
.
Blood
.
2023
;
142
(
6
):
561
-
573
.
50.
Crouch
S
,
Painter
D
,
Barrans
S
, et al
.
Molecular subclusters of follicular lymphoma: a report from the United Kingdom’s Haematological Malignancy Research Network
.
Blood Adv
.
2022
;
6
(
21
):
5716
-
5731
.
51.
Schmitz
R
,
Young
RM
,
Ceribelli
M
, et al
.
Burkitt lymphoma pathogenesis and therapeutic targets from structural and functional genomics
.
Nature
.
2012
;
490
(
7418
):
116
-
120
.
52.
Shyr
C
,
Tarailo-Graovac
M
,
Gottlieb
M
,
Lee
JJY
,
van Karnebeek
C
,
Wasserman
WW
.
FLAGS, frequently mutated genes in public exomes. [published correction appears in BMC Med Genomics. 2017;10(1):69]
.
BMC Med Genomics
.
2014
;
7
:
64
.
53.
Love
C
,
Sun
Z
,
Jima
D
, et al
.
The genetic landscape of mutations in Burkitt lymphoma
.
Nat Genet
.
2012
;
44
(
12
):
1321
-
1325
.
54.
Thomas
N
,
Dreval
K
,
Gerhard
D
, et al
.
Genetic subgroups inform on pathobiology in adult and pediatric Burkitt lymphoma
.
Blood
.
2023
;
141
(
8
):
904
-
916
.
55.
de Leval
L
,
Alizadeh
AA
,
Bergsagel
PL
, et al
.
Genomic profiling for clinical decision making in lymphoid neoplasms
.
Blood
.
2022
;
140
(
21
):
2193
-
2227
.
56.
Bailey
MH
,
Tokheim
C
,
Porta-Pardo
E
, et al
.
Comprehensive characterization of cancer driver genes and mutations
.
Cell
.
2018
;
174
(
4
):
1034
-
1035
.
57.
Morin
RD
,
Arthur
SE
,
Hodson
DJ
.
Molecular profiling in diffuse large B-cell lymphoma: why so many types of subtypes?
.
Br J Haematol
.
2022
;
196
(
4
):
814
-
829
.
58.
Louissaint
A
,
Schafernak
KT
,
Geyer
JT
, et al
.
Pediatric-type nodal follicular lymphoma: a biologically distinct lymphoma with frequent MAPK pathway mutations
.
Blood
.
2016
;
128
(
8
):
1093
-
1100
.
59.
Muppidi
J
,
Schmitz
R
,
Green
JA
, et al
.
Loss of signaling via Gα13 in germinal center B cell-derived lymphoma
.
Nature
.
2014
;
516
(
7530
):
254
-
258
.
60.
Okosun
J
,
Wolfson
RL
,
Wang
J
, et al
.
Recurrent mTORC1-activating RRAGC mutations in follicular lymphoma
.
Nat Genet
.
2016
;
48
(
2
):
183
-
188
.
61.
Krysiak
K
,
Gomez
F
,
White
BS
, et al
.
Recurrent somatic mutations affecting B-cell receptor signaling pathway genes in follicular lymphoma
.
Blood
.
2017
;
129
(
4
):
473
-
483
.
62.
Burkhardt
B
,
Michgehl
U
,
Rohde
J
, et al
.
Clinical relevance of molecular characteristics in Burkitt lymphoma differs according to age
.
Nat Commun
.
2022
;
13
(
1
):
3881
.
63.
Pararajalingam
P
,
Coyle
KM
,
Arthur
SE
, et al
.
Coding and noncoding drivers of mantle cell lymphoma identified through exome and genome sequencing
.
Blood
.
2020
;
136
(
5
):
572
-
584
.
64.
Hilton
LK
,
Ngu
HS
,
Collinge
B
, et al
.
Relapse timing is associated with distinct evolutionary dynamics in diffuse large B-cell lymphoma
.
J Clin Oncol
.
2023
;
41
(
25
):
4164
-
4177
.
65.
Johnston
JM
,
Carroll
WL
.
c-myc hypermutation in Burkitt’s lymphoma
.
Leuk Lymphoma
.
1992
;
8
(
6
):
431
-
439
.
66.
Gong
C
,
Krupka
J
,
Gao
J
, et al
.
Sequential inverse dysregulation of the RNA helicases DDX3X and DDX3Y facilitates MYC-driven lymphomagenesis
.
Mol Cell
.
2021
;
81
(
19
):
4059
-
4075.e11
.
67.
Coyle
KM
,
Hillman
T
,
Cheung
M
, et al
.
Shared and distinct genetic features in human and canine B-cell lymphomas
.
Blood Adv
.
2022
;
6
(
11
):
3404
-
3409
.
68.
Barisic
D
,
Chin
CR
,
Meydan
C
, et al
.
ARID1A orchestrates SWI/SNF-mediated sequential binding of transcription factors with ARID1A loss driving pre-memory B cell fate and lymphomagenesis
.
Cancer Cell
.
2024
;
42
(
4
):
720
-
722
.
69.
Trinh
DL
,
Scott
DW
,
Morin
RD
, et al
.
Analysis of FOXO1 mutations in diffuse large B-cell lymphoma
.
Blood
.
2013
;
121
(
18
):
3666
-
3674
.
70.
Zhang
J
,
Dominguez-Sola
D
,
Hussein
S
, et al
.
Disruption of KMT2D perturbs germinal center B cell development and promotes lymphomagenesis
.
Nat Med
.
2015
;
21
(
10
):
1190
-
1198
.
71.
Ortega-Molina
A
,
Boss
IW
,
Canela
A
, et al
.
The histone lysine methyltransferase KMT2D sustains a gene expression program that represses B cell lymphoma development
.
Nat Med
.
2015
;
21
(
10
):
1199
-
1208
.
72.
Deng
Q
,
Lakra
P
,
Gou
P
, et al
.
SMARCA4 is a haploinsufficient B cell lymphoma tumor suppressor that fine-tunes centrocyte cell fate decisions
.
Cancer Cell
.
2024
;
42
(
4
):
605
-
622.e11
.
73.
Mascle
X
,
Albagli
O
,
Lemercier
C
.
Point mutations in BCL6 DNA-binding domain reveal distinct roles for the six zinc fingers
.
Biochem Biophys Res Commun
.
2003
;
300
(
2
):
391
-
396
.
74.
Baliñas-Gavira
C
,
Rodríguez
MI
,
Andrades
Á
, et al
.
Frequent mutations in the amino-terminal domain of BCL7A impair its tumor suppressor role in DLBCL
.
Leukemia
.
2020
;
34
(
10
):
2722
-
2735
.
75.
Duan
S
,
Cermak
L
,
Pagan
JK
, et al
.
FBXO11 targets BCL6 for degradation and is inactivated in diffuse large B-cell lymphomas
.
Nature
.
2012
;
481
(
7379
):
90
-
93
.
76.
Tonc
E
,
Takeuchi
Y
,
Chou
C
, et al
.
Unexpected suppression of tumorigenesis by c-MYC via TFAP4-dependent restriction of stemness in B lymphocytes
.
Blood
.
2021
;
138
(
24
):
2526
-
2538
.
77.
Jiang
Y
,
Ortega-Molina
A
,
Geng
H
, et al
.
CREBBP inactivation promotes the development of HDAC3-dependent lymphomas
.
Cancer Discov
.
2017
;
7
(
1
):
38
-
53
.
78.
Zhang
J
,
Vlasevska
S
,
Wells
VA
, et al
.
The CREBBP acetyltransferase is a haploinsufficient tumor suppressor in B-cell lymphoma
.
Cancer Discov
.
2017
;
7
(
3
):
322
-
337
.
79.
Sneeringer
CJ
,
Scott
MP
,
Kuntz
KW
, et al
.
Coordinated activities of wild-type plus mutant EZH2 drive tumor-associated hypertrimethylation of lysine 27 on histone H3 (H3K27) in human B-cell lymphomas
.
Proc Natl Acad Sci U S A
.
2010
;
107
(
49
):
20980
-
20985
.
80.
Béguelin
W
,
Popovic
R
,
Teater
M
, et al
.
EZH2 is required for germinal center formation and somatic EZH2 mutations promote lymphoid transformation
.
Cancer Cell
.
2013
;
23
(
5
):
677
-
692
.
81.
Qiu
Z
,
Khalife
J
,
Ethiraj
P
, et al
.
IRF8-mutant B cell lymphoma evades immunity through a CD74-dependent deregulation of antigen processing and presentation in MHCII complexes
.
Sci Adv
.
2024
;
10
(
28
):
eadk2091
.
82.
Kuo
H-P
,
Ezell
SA
,
Hsieh
S
, et al
.
The role of PIM1 in the ibrutinib-resistant ABC subtype of diffuse large B-cell lymphoma
.
Am J Cancer Res
.
2016
;
6
(
11
):
2489
-
2501
.
83.
Almasmoum
HA
,
Airhihen
B
,
Seedhouse
C
,
Winkler
GS
.
Frequent loss of BTG1 activity and impaired interactions with the Caf1 subunit of the Ccr4-Not deadenylase in non-Hodgkin lymphoma
.
Leuk Lymphoma
.
2021
;
62
(
2
):
281
-
290
.
84.
Mlynarczyk
C
,
Teater
M
,
Pae
J
, et al
.
BTG1 mutation yields supercompetitive B cells primed for malignant transformation
.
Science
.
2023
;
379
(
6629
):
eabj7412
.
85.
Kannengiesser
C
,
Brookes
S
,
del Arroyo
AG
, et al
.
Functional, structural, and genetic evaluation of 20 CDKN2A germ line mutations identified in melanoma-prone families or patients
.
Hum Mutat
.
2009
;
30
(
4
):
564
-
574
.
86.
Davis
RE
,
Ngo
VN
,
Lenz
G
, et al
.
Chronic active B-cell-receptor signalling in diffuse large B-cell lymphoma
.
Nature
.
2010
;
463
(
7277
):
88
-
92
.
87.
Meriranta
L
,
Pasanen
A
,
Louhimo
R
, et al
.
Deltex-1 mutations predict poor survival in diffuse large B-cell lymphoma
.
Haematologica
.
2017
;
102
(
5
):
e195
-
e198
.
88.
González-Rincón
J
,
Méndez
M
,
Gómez
S
, et al
.
Unraveling transformation of follicular lymphoma to diffuse large B-cell lymphoma
.
PLoS One
.
2019
;
14
(
2
):
e0212813
.
89.
Hodson
DJ
,
Shaffer
AL
,
Xiao
W
, et al
.
Regulation of normal B-cell differentiation and malignant B-cell survival by OCT2
.
Proc Natl Acad Sci U S A
.
2016
;
113
(
14
):
E2039
-
E2046
.
90.
Xia
M
,
David
L
,
Teater
M
, et al
.
BCL10 mutations define distinct dependencies guiding precision therapy for DLBCL
.
Cancer Discov
.
2022
;
12
(
8
):
1922
-
1941
.
91.
Wang
L
,
Yang
JK
,
Kabaleeswaran
V
, et al
.
The Fas-FADD death domain complex structure reveals the basis of DISC assembly and disease mutations
.
Nat Struct Mol Biol
.
2010
;
17
(
11
):
1324
-
1329
.
92.
Mentz
M
,
Keay
W
,
Strobl
CD
, et al
.
PARP14 is a novel target in STAT6 mutant follicular lymphoma
.
Leukemia
.
2022
;
36
(
9
):
2281
-
2292
.
93.
Venturutti
L
,
Teater
M
,
Zhai
A
, et al
.
TBL1XR1 mutations drive extranodal lymphoma by inducing a pro-tumorigenic memory fate
.
Cell
.
2020
;
182
(
2
):
297
-
316.e27
.
94.
Novak
U
,
Rinaldi
A
,
Kwee
I
, et al
.
The NF-kappaB negative regulator TNFAIP3 (A20) is inactivated by somatic mutations and genomic deletions in marginal zone lymphomas
.
Blood
.
2009
;
113
(
20
):
4918
-
4921
.
95.
Choi
J
,
Lee
K
,
Ingvarsdottir
K
, et al
.
Loss of KLHL6 promotes diffuse large B-cell lymphoma growth and survival by stabilizing the mRNA decay factor roquin2
.
Nat Cell Biol
.
2018
;
20
(
5
):
586
-
596
.
96.
Pon
JR
,
Wong
J
,
Saberi
S
, et al
.
MEF2B mutations in non-Hodgkin lymphoma dysregulate cell migration by decreasing MEF2B target gene activation
.
Nat Commun
.
2015
;
6
(
1
):
7953
.
97.
Gao
J
,
Sidiropoulou
E
,
Walker
I
, et al
.
SGK1 mutations in DLBCL generate hyperstable protein neoisoforms that promote AKT independence
.
Blood
.
2021
;
138
(
11
):
959
-
964
.
98.
Melzner
I
,
Bucur
AJ
,
Brüderlein
S
, et al
.
Biallelic mutation of SOCS-1 impairs JAK2 degradation and sustains phospho-JAK2 action in the MedB-1 mediastinal lymphoma line
.
Blood
.
2005
;
105
(
6
):
2535
-
2542
.
99.
Phelan
JD
,
Young
RM
,
Webster
DE
, et al
.
A multiprotein supercomplex controlling oncogenic signalling in lymphoma
.
Nature
.
2018
;
560
(
7718
):
387
-
391
.
100.
Ennishi
D
,
Healy
S
,
Bashashati
A
, et al
.
TMEM30A loss-of-function mutations drive lymphomagenesis and confer therapeutically exploitable vulnerability in B-cell lymphoma
.
Nat Med
.
2020
;
26
(
4
):
577
-
588
.
101.
Brescia
P
,
Schneider
C
,
Holmes
AB
, et al
.
MEF2B instructs germinal center development and acts as an oncogene in B cell lymphomagenesis
.
Cancer Cell
.
2018
;
34
(
3
):
453
-
465.e9
.
102.
Sander
S
,
Calado
DP
,
Srinivasan
L
, et al
.
Synergy between PI3K signaling and MYC in Burkitt lymphomagenesis
.
Cancer Cell
.
2012
;
22
(
2
):
167
-
179
.
103.
Egle
A
,
Harris
AW
,
Bath
ML
,
O’Reilly
L
,
Cory
S
.
VavP-Bcl2 transgenic mice develop follicular lymphoma preceded by germinal center hyperplasia
.
Blood
.
2004
;
103
(
6
):
2276
-
2283
.
104.
Flümann
R
,
Hansen
J
,
Meinel
J
, et al
.
An inducible Cd79b mutation confers ibrutinib sensitivity in mouse models of Myd88-driven diffuse large B-cell lymphoma
.
Blood Adv
.
2024
;
8
(
5
):
1063
-
1074
.
105.
Dominguez
PM
,
Ghamlouch
H
,
Rosikiewicz
W
, et al
.
TET2 deficiency causes germinal center hyperplasia, impairs plasma cell differentiation, and promotes B-cell lymphomagenesis
.
Cancer Discov
.
2018
;
8
(
12
):
1632
-
1653
.
106.
Hahn
WC
,
Counter
CM
,
Lundberg
AS
,
Beijersbergen
RL
,
Brooks
MW
,
Weinberg
RA
.
Creation of human tumour cells with defined genetic elements
.
Nature
.
1999
;
400
(
6743
):
464
-
468
.
107.
Caeser
R
,
Di Re
M
,
Krupka
JA
, et al
.
Genetic modification of primary human B cells to model high-grade lymphoma
.
Nat Commun
.
2019
;
10
(
1
):
4543
.
108.
Caeser
R
,
Gao
J
,
Di Re
M
,
Gong
C
,
Hodson
DJ
.
Genetic manipulation and immortalized culture of ex vivo primary human germinal center B cells
.
Nat Protoc
.
2021
;
16
(
5
):
2499
-
2519
.
109.
Wagar
LE
,
Salahudeen
A
,
Constantz
CM
, et al
.
Modeling human adaptive immune responses with tonsil organoids
.
Nat Med
.
2021
;
27
(
1
):
125
-
135
.
110.
Kastenschmidt
JM
,
Schroers-Martin
JG
,
Sworder
BJ
, et al
.
A human lymphoma organoid model for evaluating and targeting the follicular lymphoma tumor immune microenvironment
.
Cell Stem Cell
.
2024
;
31
(
3
):
410
-
420.e4
.
111.
Schmidt
R
,
Ward
CC
,
Dajani
R
, et al
.
Base-editing mutagenesis maps alleles to tune human T cell functions
.
Nature
.
2024
;
625
(
7996
):
805
-
812
.

Author notes

The gene lists for diffuse large B-cell lymphoma, follicular lymphoma, and Burkitt lymphoma, and the combined Tier 1 list will continue to be maintained. The latest versions will always be available from the GitHub repository: https://github.com/morinlab/LLMPP/blob/main/resources/curated.

The full-text version of this article contains a data supplement.