Our understanding of the pathogenesis of lymphoid malignancies has been transformed by next-generation sequencing. The studies in this review have used whole-genome, exome, and transcriptome sequencing to identify recurring structural genetic alterations and sequence mutations that target key cellular pathways in acute lymphoblastic leukemia (ALL) and the lymphomas. Although each tumor type is characterized by a unique genomic landscape, several cellular pathways are mutated in multiple tumor types—transcriptional regulation of differentiation, antigen receptor signaling, tyrosine kinase and Ras signaling, and epigenetic modifications—and individual genes are mutated in multiple tumors, notably TCF3, NOTCH1, MYD88, and BRAF. In addition to providing fundamental insights into tumorigenesis, these studies have also identified potential new markers for diagnosis, risk stratification, and therapeutic intervention. Several genetic alterations are intuitively “druggable” with existing agents, for example, kinase-activating lesions in high-risk B-cell ALL, NOTCH1 in both leukemia and lymphoma, and BRAF in hairy cell leukemia. Future sequencing efforts are required to comprehensively define the genetic basis of all lymphoid malignancies, examine the relative roles of germline and somatic variation, dissect the genetic basis of clonal heterogeneity, and chart a course for clinical sequencing and translation to improved therapeutic outcomes.

The lymphoid malignancies represent a diverse range of tumors characterized by variable stages of maturation ranging from pro-B or T cells in acute lymphoblastic leukemia (ALL) to cells representative of the lymph node in the non-Hodgkin lymphomas (NHLs) to mature plasma cells in myeloma and related disorders. These disorders have a diverse range of clinical manifestations, sites of organ involvement, and responsiveness to therapy. Somatic genetic alterations are a hallmark of lymphoid malignancies, and it is now known that each tumor type typically harbors a constellation of genetic alterations, including gross chromosomal alterations (aneuploidy and/or chromosomal rearrangements), structural genetic alterations (deletions and gains of DNA), and sequence mutations. Genomic techniques that have identified these alterations may be grouped into three broad categories: (1) cytogenetic studies that have identified structural chromosomal alterations, coupled with limited candidate gene sequencing or structural profiling studies; (2) first-generation genome-wide profiling using array-based comparative genomic hybridization or single nucleotide polymorphism microarrays and gene expression profiling; and (3) second-generation sequencing studies using whole-genome sequencing (WGS), transcriptome sequencing (messenger mRNA [mRNA-seq]) and/or whole-exome sequencing (WES) to more comprehensively define the genomic landscape of these diseases. For each tumor type discussed below, key genetic alterations identified from cytogenetic and first-generation genomic profiling studies are presented to provide context for the results of next-generation sequencing (NGS) studies summarized in Table 1.

Table 1

Key genetic alterations identified by NGS studies in lymphoid neoplasms

Tumor typeGenetic alteration
T-ALL PHF6, CNOT3, RPL5, RPL10 
ETP ALL Loss-of-function mutations in hematopoietic regulators (GATA3, IKZF1, RUNX1, ETV6
Gain-of-function mutations in Ras, FLT3, IL7R 
Inactivating mutations in epigenetic regulators (EZH2, SUZ12, EED, SETD2, DNMT3A
BCR-ABL1–like ALL Rearrangement of CRLF2 in 50% of cases; concomitant activating JAK mutations in 50% of CRLF2-rearranged cases 
Rearrangement of multiple kinase genes: ABL1, ABL2, EPOR, PDGFRB 
Hypodiploid ALL Ras mutations (NF1, PTPN11, NRAS, KRAS) in near-haploid ALL 
IKZF2 and TP53 mutations in low hypodiploid ALL; TP53 mutations are commonly germline,  
Relapsed ALL CREBBP mutations enriched at relapse 
NT5C2 mutations enriched at relapse 
Familial ALL TP53 mutations in low-hypodiploid ALL; PAX5 p.Gly183Ser in autosomal-dominant ALL 
DLBCL and NHL Lymphoid signaling (CD79B
NF-kB signaling (CARD11, MYD88
Histone modification (CREBBP/EP300, EZH2, MEF2B, MLL2/MLL3
SMZL NOTCH2 mutations 
MCL NOTCH1 mutations, associated with poor outcome 
HL CIITA rearrangements 
PMBCL CIITA rearrangements 
BL TCF3/ID3 mutations in BL and other MYC-rearranged lymphomas 
CCND mutations 
CLL NOTCH1 mutations; associated with disease progression 
mRNA splicing mutations (eg, SF3B1
DNA damage and/or repair mutations (ATM, POT1
Regulation of apoptosis (BIRC3
Innate immunity (MYD88, TLR2
HCL Activating BRAF mutations 
MM Multiple targets of mutation, including NRAS, KRAS, TP53, CCND1, DIS3, BRAF, NF-κB signaling and histone modification 
WM MYD88 p.Leu265Pro in >90% of cases 
Tumor typeGenetic alteration
T-ALL PHF6, CNOT3, RPL5, RPL10 
ETP ALL Loss-of-function mutations in hematopoietic regulators (GATA3, IKZF1, RUNX1, ETV6
Gain-of-function mutations in Ras, FLT3, IL7R 
Inactivating mutations in epigenetic regulators (EZH2, SUZ12, EED, SETD2, DNMT3A
BCR-ABL1–like ALL Rearrangement of CRLF2 in 50% of cases; concomitant activating JAK mutations in 50% of CRLF2-rearranged cases 
Rearrangement of multiple kinase genes: ABL1, ABL2, EPOR, PDGFRB 
Hypodiploid ALL Ras mutations (NF1, PTPN11, NRAS, KRAS) in near-haploid ALL 
IKZF2 and TP53 mutations in low hypodiploid ALL; TP53 mutations are commonly germline,  
Relapsed ALL CREBBP mutations enriched at relapse 
NT5C2 mutations enriched at relapse 
Familial ALL TP53 mutations in low-hypodiploid ALL; PAX5 p.Gly183Ser in autosomal-dominant ALL 
DLBCL and NHL Lymphoid signaling (CD79B
NF-kB signaling (CARD11, MYD88
Histone modification (CREBBP/EP300, EZH2, MEF2B, MLL2/MLL3
SMZL NOTCH2 mutations 
MCL NOTCH1 mutations, associated with poor outcome 
HL CIITA rearrangements 
PMBCL CIITA rearrangements 
BL TCF3/ID3 mutations in BL and other MYC-rearranged lymphomas 
CCND mutations 
CLL NOTCH1 mutations; associated with disease progression 
mRNA splicing mutations (eg, SF3B1
DNA damage and/or repair mutations (ATM, POT1
Regulation of apoptosis (BIRC3
Innate immunity (MYD88, TLR2
HCL Activating BRAF mutations 
MM Multiple targets of mutation, including NRAS, KRAS, TP53, CCND1, DIS3, BRAF, NF-κB signaling and histone modification 
WM MYD88 p.Leu265Pro in >90% of cases 

PMBCL, primary mediastinal B-cell lymphoma.

ALL is a proliferation of B- or T-lymphoid progenitor cells, is the commonest childhood malignancy, and is the most common cause of cancer-related death in children and young adults.1  Chromosomal abnormalities are present in approximately three quarters of childhood ALL cases, including aneuploidy (most commonly, hyperdiploidy with more than 50 chromosomes and, less commonly, hypodiploidy with less than 44 chromosomes) and a range of translocations, including t(12;21)(p13;q22) encoding ETV6-RUNX1 (TEL-AML1), t(1;19)(q23;p13.3) encoding TCF3-PBX1 (E2A-PBX1), t(9;22)(q34;q11.2) encoding BCR-ABL1, rearrangement of MLL, and rearrangement of CRLF2 in B-progenitor ALL, and rearrangement of T-cell antigen receptor loci to transcription factor genes (eg, HOX11, LYL1, TAL1, and MLL) in T-lineage ALL.2,3  The genetic basis of ALL in adolescents and adults with ALL is less well understood, although the frequency of favorable alterations (eg, hyperdiploidy and ETV6-RUNX1) falls and unfavorable alterations (BCR-ABL1 and MLL rearrangement) rise with increasing age.4  Although these alterations are important initiating lesions in leukemogenesis, they are usually insufficient to induce leukemia in experimental models, and extensive microarray and candidate gene sequencing studies have identified a number of additional recurring genetic alterations in leukemogenesis, treatment failure, and relapse.5 

ALL genomes typically harbor fewer structural genetic alterations than many solid tumors. Although it is tempting to assume that mutational load is dependent on patient age, mutation frequency is highly dependent on tumor type. Factors such as exposure to environmental mutagens and mutations in TP53 and other genes encoding mediators of DNA integrity are clearly important but do not explain the striking variation in the type and frequency of mutations between different childhood and adult tumors.6  Infant leukemia harbors few alterations apart from MLL rearrangement, but other early-onset leukemias harbor more mutations (eg, ETV6-RUNX1 and BCR-ABL1), and other childhood solid and brain tumors often have a remarkably high mutation frequency.7  More than 50 recurring regions of DNA copy number alteration have been identified, which are commonly focal deletions limited to one or few genes. These include transcriptional regulators of lymphoid development (PAX5, IKZF1, EBF1, LEF1), tumor suppressors (CDKN2A, CDKN2B, RB1, TP53), lymphoid signaling genes (BTLA, CD200 TOX), transcriptional regulators and coactivators (TBL1XR1, ERG), and regulators of chromatin structure and epigenetic regulators (CTCF, CREBBP).7,8  Several of these alterations are associated with high-risk ALL and an increased risk of treatment failure and relapse, notably deletions of IKZF1 (IKAROS)9  and deletion or sequence mutation of CREBBP.10  Sanger sequencing studies have also identified recurring sequence mutations. In B-lineage ALL, these most commonly affect lymphoid development (PAX5), Ras signaling (NRAS, KRAS, and NF1), cytokine receptor signaling (IL7R, JAK2), and tumor suppression (TP53).11  Similarly, a number of targets of structural genetic alteration and/or sequence mutation have been identified in T-lineage ALL, including activating mutations of NOTCH1,12  deletion and/or mutation of PTEN,13 WT1,14 FBXW7,15  and amplification of MYB.7,16 

The most extensive NGS studies in ALL include the St. Jude Children’s Research Hospital–Washington University Pediatric Cancer Genome Project (PCGP)6  and the Children’s Oncology Group–National Cancer Institute Therapeutically Applicable Research to Generate Effective Treatments (TARGET) initiative (http://ocg.cancer.gov/programs/target). Sequencing of the full spectrum of ALL subtypes is incomplete; however, the results of NGS analysis of several high-risk ALL subtypes have recently been reported. These data have shown that alteration of multiple cellular pathways, including cytokine receptor and Ras signaling, tumor suppression, lymphoid development, and epigenetic regulation, are hallmarks of multiple ALL subtypes.

T-ALL is characterized by an older age of onset than that for B-ALL, male sex preponderance, and inferior outcome in comparison with B-ALL. In the first NGS study of ALL, Ferrando and colleagues examined the male sex preponderance by performing targeted capture and NGS of X chromosome genes. This identified sequence mutations and, less commonly, deletion of PHF6 in 16% and 38% of childhood and adult T-ALL cases, respectively. The PHF6 alterations result in loss of PHF6 expression and are associated with TLX1/3- and TAL1-rearranged ALL.17,18  The role of PHF6 in leukemogenesis is poorly understood, but the loss-of-function alterations suggest that PHF6 is a tumor suppressor.

Early T-cell precursor (ETP) ALL is an aggressive subtype of immature leukemia that accounts for a high proportion of T-ALL treatment failures.19  Various laboratory criteria have been proposed to identify these immature cases, but the original definition proposed by Campana and colleagues is based on immunophenotype: leukemic cells that express T-lineage markers (eg, cytoplasmic CD3) but lack expression of CD1a and CD8, have weak or negative CD5 expression, and exhibit aberrant expression of myeloid and/or stem cell markers. This pattern is reminiscent of the murine ETP,20  the earliest stage of thymic T-cell maturation that retains lineage plasticity.

In the first report of WGS of a lymphoid malignancy, the PCGP performed WGS of tumor and matched nontumor DNA of 12 ETP ALL cases, WES and mRNA-seq in selected cases, and mutation recurrence testing of selected genes in 94 additional ETP and non-ETP T-ALL cases.21  Unexpectedly, no common structural rearrangement or sequence mutation was identified. There was marked diversity in the frequency and nature of genetic alterations, with several cases exhibiting complex multichromosomal structural alterations with the hallmarks of chromothripsis,22  and other cases with no structural alterations whatsoever. However, the majority of cases harbored alterations in three pathways: loss-of-function mutations in genes encoding regulators of hematopoietic development (ETV6, GATA3, IKZF1, RUNX1; see also Della Gatta et al23  and Van Vlierberghe et al24 ); activating mutations in cytokine receptor and Ras signaling (NRAS, KRAS, FLT3, JAK1, JAK3, and IL7R; see also Flex et al,25  Shochat et al,26  Zenatti et al,27  and Neumann et al28 ); and inactivating mutations targeting epigenetic regulators, most commonly components of the polycomb repressor complex 2 that mediates histone 3 lysine 27 (H3K27) trimethylation (EZH2, SUZ12, EED; see also Ntziachristos et al29 ), the histone 3 lysine 36 trimethylase gene SETD2, and the histone acetyltransferase and CREBBP homolog EP300 (p300). Subsequent studies have also identified recurring mutations in the DNA methyltransferase gene DNMT3A in adult ETP ALL.30  Additional new targets of mutation were identified, including DNM2, ECT2L, and RELN, and several genes—and indeed specific somatic mutations—were identified that had previously been reported as germline mutations in inherited developmental disorders, notably those in the zinc finger domain of the hematopoietic transcription factor gene GATA3. Several genes, including RUNX1, ETV6, and IKZF1, were targeted by multiple types of genomic aberration, including sequence mutation, deletion, and translocation, indicating the need for WGS to completely define the spectrum of somatic genomic alterations in hematologic neoplasms. The mutational spectrum of ETP ALL is similar to that observed in myeloid leukemias, and comparison of the transcriptional profile of ETP ALL with those of normal human hematopoietic progenitors showed significant similarity to hematopoietic stem and early myeloid progenitors.21  Thus, ETP ALL likely represents part of a spectrum of immature, stem cell–like leukemias. The potential for new therapeutic approaches, including epigenetic modifiers and agents targeting JAK-STAT signaling, are currently being explored.

Pre-NGS studies identified a number of recurring targets of mutation in typical (non-ETP) T-ALL,3  including deletions and mutations in CDKN2A/CDKN2B (encoding the INK4/ARF family of cell cycle regulators and tumor suppressors), NOTCH1, FBXW7, PTEN, WT1, and MYB, and recurring, if infrequent, rearrangements encoding chimeric fusions that may be amenable to tyrosine kinase inhibitor (TKI) therapy (notably NUP214-ABL131 ). Comprehensive NGS of typical T-ALL has not been reported; however, a recent WES study of more than 60 T-ALL cases identified recurrent mutations in CNOT3, a member of transcriptional regulatory complex, and ribosomal protein genes.32  Since structural genetic alterations and rearrangements are hallmarks of leukemia, these data must be complemented with transcriptome and WGS studies to identify all genomic alterations in T-ALL.

In 2009, two study groups identified a group of childhood B-progenitor ALLs lacking a recurring chromosomal alteration that exhibited a gene expression profile similar to BCR-ABL1–positive ALL, deletion of IKZF1 (also common in BCR-ABL1–positive ALL9 ), and poor outcome.33,34  The similarity of the gene expression profile to BCR-ABL1 positive ALL in these BCR-ABL1–like or Philadelphia-like ALL cases suggested the presence of previously unrecognized genetic alterations that activate tyrosine kinase signaling pathways. BCR-ABL1–like ALL is common, comprising up to 10% to 15% of childhood B-ALL and a higher frequency of ALL in adolescents and young adults (unpublished data), and it is associated with poor outcome.35  Up to half of BCR-ABL1–like ALL cases harbor rearrangement of CRLF2 located at the pseudoautosomal region of Xp22.3/Yp11.3, either as a translocation to the immunoglobulin heavy chain enhancer region at 14q32.33 (IGH-CRLF2), or a focal deletion proximal to CRLF2 resulting in the expression of a P2RY8-CRLF2 fusion transcript.36-38  Both alterations result in overexpression of CRLF2 on the surface of lymphoid blasts that may be detected by immunophenotyping.36,38  Approximately half of CRLF2-rearranged cases harbor concomitant activating mutations of the Janus kinase genes JAK1 and JAK2, most commonly at p.Arg683 in the pseudokinase domain of JAK2 and, less commonly, in the kinase domains of JAK1 and JAK2. The JAK2 p.Val617Phe mutation commonly observed in myeloproliferative neoplasms39  does not occur in ALL, although the corresponding mutation in the pseudokinase domain in JAK1 (p.Val678Phe) is observed.11,40  These alterations result in activation of JAK-STAT signaling that may be amenable to therapy with JAK inhibitors such as ruxolitinib,41  and this is currently being explored as a therapeutic strategy.

Transcriptome sequencing and WGS of 15 BCR-ABL1–like ALL cases, 12 of which lacked CRLF2 rearrangement, identified a strikingly diverse array of genetic alterations activating cytokine receptor and tyrosine signaling.42  These were most commonly rearrangements resulting in chimeric fusion genes deregulating tyrosine kinases (NUP214-ABL1, ETV6-ABL1, RANBP2-ABL1, RCSD1-ABL1, BCR-JAK2, PAX5-JAK2, STRN3-JAK2, and EBF1-PDGFRB) and cytokine receptors (IGH-EPOR). Up to 20% of BCR-ABL1–like cases lack a chimeric fusion on mRNA-seq analysis, and sequence mutations (eg, activating mutations of FLT3 and IL7R) and structural alterations (eg, focal deletions of SH2B3, or LNK, which constrains JAK signaling43 ) that activate signaling have been identified in fusion-negative cases. These diverse genetic alterations activate a limited number of signaling pathways, notably ABL1 and PDGFRB (both of which may be inhibited with the TKIs imatinib and dasatinib) and JAK-STAT signaling. These rearrangements have been shown to activate signaling pathways in model cell lines and in primary leukemic cells, and xenografts of BCR-ABL1–like ALL are highly sensitive to TKIs in vivo. Moreover, a recent report of a child with refractory EBF1-PDGFRB–positive ALL that was exquisitely sensitive to imatinib emphasizes the potential clinical utility of TKI therapy in BCR-ABL1–like ALL.44  Ongoing studies are performing NGS of childhood and adult ALL to comprehensively define the repertoire of kinase-activating alterations in BCR-ABL1–like ALL and to develop clinical trials to direct patients with BCR-ABL1–like ALL to appropriate TKI therapy.

Hypodiploidy with less than 44 chromosomes is observed in up to 3% of ALL cases and is associated with poor prognosis. Two subtypes of hypodiploid ALL have been described according to the severity of aneuploidy: near-haploid cases with 24 to 31 chromosomes and low-hypodiploid cases with 32 to 39 chromosomes.45-47  However, the nature of additional genetic alterations driving leukemogenesis and poor outcome in hypodiploid ALL is unknown. Microarray and NGS analysis of a large cohort of more than 120 hypodiploid ALL samples has demonstrated that near-haploid and low-hypodiploid ALL have distinct transcriptomic signatures and submicroscopic genetic alterations.48  The majority of near-haploid cases harbor mutations activating Ras signaling, most commonly in NF1, and the IKAROS family gene IKZF3 (AIOLOS). In contrast, low-hypodiploid cases have near universal mutation of the tumor suppressor TP53 (p53), with the mutations present in the germline in approximately half the cases and inactivating mutations of a third IKAROS family member IKZF2 (HELIOS). Parallel analysis of primary hypodiploid xenografts (primagrafts) demonstrated activation of Ras-Raf-MEK-ERK and phosphatidylinositol-3-OH kinase (PI3K) signaling in the majority of hypodiploid primagrafts that was sensitive to PI3K and PI3K/mTOR inhibitors, but not MEK inhibitors, suggesting that PI3K inhibition represents a novel therapeutic approach. In addition, the identification of low-hypodiploid ALL as a manifestation of Li-Fraumeni syndrome indicates that all children with low-hypodiploid ALL should be tested for TP53 mutational status.49,50  Recent NGS studies have also identified other germline mutations in familial leukemia, such as a germline PAX5 p.Gly183Ser mutation in two kindreds with autosomal dominant pre-B ALL51 ; thus, there is considerable interest in defining the full range of inherited genetic variants that influence susceptibility to ALL.

Although several subtypes of ALL are associated with a high risk of treatment failure, relapse occurs across the spectrum of ALL subtypes and is associated with very poor outcome. Moreover, it has long been recognized that ALL genomes are not static but exhibit acquisition of chromosomal abnormalities over time.52  Single nucleotide polymorphism microarray profiling studies of matched diagnosis-relapse ALL samples demonstrated that the majority of ALL cases show changes in the patterns of structural genomic alterations from diagnosis to relapse52  and that many relapse-acquired lesions, including those targeting genes associated with high-risk ALL (IKZF1, IKZF2, CDKN2A, and CDKN2B), are present at low levels at diagnosis, suggesting that genetically determined tumor heterogeneity is a key determinant of treatment failure and relapse.53,54  Sequencing of 300 genes in matched diagnosis-relapse samples recapitulated these findings and identified mutations in the transcriptional coactivator and acetyltransferase CREBBP (CREB-binding protein, or CBP) as a relapse-acquired lesion in up to 20% of relapsed ALL samples.10,55 CREBBP mutations are also observed at diagnosis in NHL, particularly diffuse large B-cell lymphoma (DLBCL),56  and they impair histone acetylation.10,56  CREBBP in part mediates the transcriptional response to glucocorticoids, and histone deacetylase inhibitors were effective in killing steroid-resistant ALL cell lines. Two study groups recently identified relapse-acquired mutations in NT5C2 that encode a 5′-nucleotidase enzyme responsible for the inactivation of nucleoside-analog drugs.57,58  Thus, mutations that confer resistance to drugs commonly used to treat ALL represent a key mechanism of treatment failure and resistance.

Genomic profiling studies, including NGS, have shed important light on the genetic basis and molecular heterogeneity of NHL, which comprises a diverse range of entities with distinct pathologic and genetic features, including DLBCL, follicular lymphoma, mantle cell lymphoma (MCL), splenic marginal zone lymphoma (SMZL), chronic lymphocytic leukemia (CLL), and hairy cell leukemia (HCL).59  As with ALL, there is an extensive literature describing microarray-based profiling of transcriptional signatures, structural genetic alterations, and candidate gene sequencing in NHL. These studies have identified important genetic alterations that are beyond the scope of this review,60  but key findings from NGS studies are reviewed below.

DLBCL is the most common form of NHL. Gene expression profiling studies identified at least three subtypes of DLBCL with distinct transcriptional profiles reflective of their cell of origin: activated B-cell, germinal center B-cell, and primary mediastinal B-cell lymphoma, which is thought to be of thymic B-cell origin. Multiple studies have used candidate gene, WGS, WES, and mRNA-seq to identify recurring genetic alterations and rearrangements in DLBCL and to examine their association with clinical features, transcriptomic subtype, and outcome.56,61-68  Notably, mutations affecting B-cell receptor signaling (CD79B) and activating nuclear factor κB (NF-κB) signaling (CARD11 and MYD88) are enriched in activated B-cell DLBCL, and genes encoding histone modifiers (CREBBP, EP300, EZH2, MEF2B, MLL2/3) are more commonly mutated in the germinal center B-cell subtype. The high frequency of mutations in histone-modifying genes reinforces the central role of these alterations in the pathogenesis of a range of lymphoid malignancies but also highlights distinct roles of different alterations in different tumors. For example, the mutations in the histone acetyltransferase and transactivator CREBBP are similar to those observed in ALL but, in contrast to ALL, they are observed in the predominant clone at diagnosis and may act by impairing acetylation of TP53 and BCL6. Similarly, although EZH2 is mutated in both NHL61,63  and ALL,21  the type of EZH2 alteration and predicted functional consequences are different. In DLBCL and follicular lymphoma, the most common alterations are a missense mutation at p.Tyr641, which is a gain-of-function mutation that enhances histone 3 lysine 27 trimethylation and transcriptional repression.69,70  In contrast, the EZH2 alterations in ETP ALL are loss-of-function deletions and sequence mutations that are predicted to have the opposite effect.

These findings have been extended by a recent NGS study that performed WGS of 40 DLBCL samples and 13 cell lines, coupled with transcriptome sequencing and DNA copy number alterations analysis of 96 samples.71  This study expanded the number of significantly mutated genes to 74, more than 40 of which had not been previously reported, including cell surface receptor genes (CD70, CD83), purinergic receptor genes (P2RY8, P2RX5) and G-protein–coupled receptors. In contrast to precursor B-cell neoplasms, recurring chromosomal rearrangements leading to the expression of chimeric fusions are uncommon in DLBCL, with the exception of those involving TP63.72 

NGS studies have also identified recurring targets of mutation associated with other subtypes of NHL, including NOTCH2 mutations in approximately 25% of SMZLs.73  Transcriptome sequencing of MCL has identified NOTCH1 mutations in 12% of cases, most commonly in the C-terminal domain in the recognition sequence for FBXW7.74  This pattern is similar to that observed in CLL, and the NOTCH1 mutations are associated with poor outcome in MCL. Additional recurring targets of mutation in MCL include ATM, CCND1, TP53, and the E3 ligase UBR5.75 

Prior to the advent of NGS, few structural genetic alterations had been described in Hodgkin lymphoma (HL). A remarkable finding of mRNA-seq of HL cell lines and tumor samples was a high frequency of rearrangement of CIITA to a range of fusion partners including the uncharacterized gene BX648577, CD274, CD273, and RALGDS, commonly resulting in expression of chimeric fusion transcripts in HL (15% of cases) and the phenotypically related disorder primary mediastinal B-cell lymphoma (38%), whereas the fusions were rare in DLBCL.76  CIITA is an important transactivator of class II major histocompatibility complex expression. The role of the fusions in lymphomagenesis is incompletely understood, but modulation of HLA expression and escape from immunosurveillance has been postulated.

Burkitt lymphoma (BL) is a highly aggressive B-lymphoid malignancy that, like DLBCL, is thought to arise from the germinal center. Deregulation of MYC by translocation into the immunoglobulin loci enhancer regions, most frequently, the IGH, is a hallmark of BL. BL is observed in multiple contexts, including sporadic BL in developed countries, Epstein-Barr virus–associated BL typically observed in the developing world, and HIV-associated BL. Three study groups recently described the first NGS studies of BL and used a combination of WGS, WES, and/or mRNA-seq,77-79  and in one study, high-throughput RNA interference.79  These studies identified at least 70 recurrently mutated genes in BL, the most common of which were alterations affecting TCF3 activity in 70% of sporadic BL cases, either due to mutations of TCF3 or, more commonly, loss-of-function mutations or deletions of ID3, which encodes a negative regulator of TCF3. ID3 alterations were observed in 35% to 68% of BL, 13% of other MYC-rearranged lymphoma, but not DLBCL. These alterations activated B-cell receptor and PI3K signaling. In addition, 38% of sporadic BLs harbored oncogenic mutations of the CCND3, encoding cyclin D3, which promotes cell cycle progression. These remarkable findings highlight two new avenues for potential therapeutic exploitation in this highly aggressive malignancy and suggest that further sequencing efforts to examine the genetic differences between the different forms of BL will be of considerable interest.

CLL is the most common leukemia, and it is characterized by monoclonal proliferation of CD5+ B cells and variable lymphadenopathy, splenomegaly, cytopenias, and autoimmune phenomena. The clinical course of CLL is variable, and genetic features previously associated with prognosis include the extent of somatic hypermutation of immunoglobulin genes and cytogenetic alterations, including deletions of 11q23, 17p13, and 13q14.80  The presence of or lack of hypermutation of Ig genes may reflect the cell of origin of CLL—naive B cells in cases lacking hypermutation and germinal center B cells in cases with somatic hypermutation. Several recent studies have reported WGS and WES of CLL, which have provided insights into the mutational landscape and clonal heterogeneity of this disease.81-87  Compared with other lymphoid malignancies, few genes are mutated in more than 10% of cases. In contrast, multiple genes are mutated at lower frequency. In approximately one third of cases, no recurring targets of mutation have been identified. Mutations are enriched in several pathways, which are likely driver events, including NOTCH1 signaling (NOTCH1 and the U3 ubiquitin ligase gene FBXW7); mRNA splicing, transport, and processing (DDX3X, SF3B1, SFRS1, U2AF2, and XPO1); DNA damage, repair, and telomere maintenance (ATM, POT1, and TP53); regulation of apoptosis (BIRC3); and innate immunity and inflammation (MAPK1, MYD88, and TLR2). Recurrent structural alterations leading to chimeric fusion genes are uncommon in CLL. The frequency of mutations has varied across studies. This may in part reflect the nature of the cohorts studied (early stage vs advanced disease), the enrichment for cases with unmutated or mutated immunoglobulin genes, and possibly the sequencing approach used, but the most common targets of mutation are NOTCH1, SF3B1, TP53, and MYD88. The identification of mutations targeting the spliceosome machinery is one of the most notable novel findings from these studies. These mutations may result in aberrant splicing, intron retention, and expression of aberrant protein isoforms that may potentially deregulate a diverse range of cellular pathways. Several of these alterations, notably NOTCH1 mutations, are associated with poor survival.83,88  Mutational profiling has been integrated with cytogenetic analysis to refine outcome prediction in CLL, with TP53 and BIRC3 associated with poor survival; NOTCH1, SF3B1, and/or del(11q22) with intermediate risk; trisomy 12 or normal karyotype with low risk; and del (13q14) with very low risk.89  Mutations in SF3B1 and NOTCH1 are commonly identified as subclonal events early in the disease course and may drive the emergence of resistant clones and disease progression.90 

One of the most remarkable findings from recent NGS studies of lymphoid neoplasms is the identification of near universal BRAF mutations in HCL. HCL is characterized by splenomegaly, cytopenias, and the presence of malignant cells with distinctive villous projections (ie, hairy cells) in the marrow and spleen. Although it is often indolent and amenable to nucleoside analog therapy, the molecular pathogenesis has remained elusive. WES of an HCL genome identified a somatic BRAF p.Val600Glu mutation91  that was also identified as a universal phenomenon in a recurrence cohort of 47 cases. BRAF p.Val600Glu is observed in more than half of the malignant melanomas, papillary carcinoma of the thyroid, and Langerhans cell histiocytosis, and infrequently in myeloma, but it is uncommon in other lymphoid neoplasms.92  The presence of this mutation in nearly all cases of HCL has been confirmed by other studies.93  The p.Val600Glu mutation lies in the activation loop of the kinase domain of BRAF and explains the activation of the Raf-MEK-ERK pathway characteristic of HCL. Detection of BRAF p.Val600Glu has rapidly entered the clinical arena, as a diagnostic test for HCL and to monitor response to therapy.94  Moreover, reports of responsiveness of treatment-refractory HCL to the mutant BRAF inhibitor vemurafenib have spurred trials testing BRAF inhibitors in HCL.95  Although comprehensive definition of the genomic landscape of HCL by WGS is awaited, these findings indicate that mutant BRAF is a key driver and rational therapeutic target in this disease.

Multiple myeloma (MM) is characterized by proliferation of malignant plasma cells in the bone marrow, bony destruction, and end organ dysfunction, either directly from plasma cell infiltration or from the effects of the secreted monoclonal Ig (paraprotein). MM is one disorder in a spectrum of plasma cell dyscrasias that include monoclonal gammopathy of uncertain significance (MGUS), smoldering MM, MM and plasma cell leukemia. Chromosomal alterations include hyperdiploidy and translocations, commonly rearrangements of CCDKN1, CCDN3, and WHSC1 into the IGH locus. The first genome-wide mutational data on 38 MM cases studied by WGS or WES found 10 genes to be significant recurring targets of mutation,96  including NRAS, KRAS, TP53, CCND1, and six genes not previously found to be mutated in cancer: FAM46C, DIS3, ALOX12B, HLA-A, and MAGED1. Notably, DIS3 (RRP44; mutated in 11% of cases) encodes an RNA exonuclease, and the mutations identified are predicted to be loss of function, implicating altered RNA processing in MM. In addition, mutations were identified in genes regulating protein folding and translation, including XBP1. Recurring mutations were identified in BRAF, including p.Val600Glu and p.Lys601Asn. Mutations were also observed in genes regulating NF-κB signaling and histone modification (MLL, MLL2, MLL3, KDM6A [UTX], WHSC1, and WHSC1L1), and in sites of somatic mutation in the noncoding region (eg, 5′ and within BCL6). Thus, these results have implicated new pathogenic pathways in MM, but additional analysis and sequencing of MM is required to fully define the mutational landscape of MM for several reasons. MM genomes are structurally complex, and accurate resolution of structural genetic alterations in MM has not yet been comprehensively performed. Moreover, the nature of mutations that define each stage of the plasma cell disorders has not been fully investigated. Consistent with sequencing studies of other leukemias and lymphomas, MM is characterized by substantial clonal heterogeneity that is likely to be an important determinant of disease progression. Accordingly, reports are emerging of mutations associated with aggressive, extramedullary disease, and sequencing of serial cohorts of MGUS, smoldering MM, and MM and plasma cell lymphoma have begun to elucidate the subclonal structure of MM and its relationship to disease evolution and progression.97 

The genetic basis of Waldenström macroglobulinemia (WM), a lymphoplasmacytic lymphoma with secretion of monoclonal IgM, was unknown until recently. Remarkably, WGS of 40 WM samples identified a somatic mutation of MYD88, p.Leu265Pro, in 91% of IgM- and non-IgM–secreting lymphoplasmacytic lymphomas.98  MYD88 mutations were uncommon in MGUS or SMZL—disorders that must be distinguished from WM—and thus mutation detection may be useful at diagnosis. Other recurring mutations included SWI-SNF chromatin remodeling gene ARID1A and the histone gene HIST1H1E, but at a much lower frequency than MYD88. MYD88 is an adaptor molecule that facilitates Toll-like receptor and interleukin-1 receptor signaling through the NF-κB pathway via phosphorylation of IκBα. WM cells harboring MYD88 mutations exhibit biochemical evidence of NF-κB signaling, and because IκBα blockade using proteasome inhibitors is effective in WM, inhibition of this pathway represents a potential therapeutic approach.99 

It is clear that although additional sequencing of expanded cohorts of samples is required to fully define the genomic landscape of each tumor type, existing studies have identified genetic alterations that are immediately attractive for diagnosis, risk stratification, and therapeutic targeting. The most suitable sequencing approach is dependent on the type of alterations, and it should also be highlighted that clinical sequencing is rapidly evolving, both in terms of technological approaches and implementing rapid sequencing and analysis in a regulated environment. It is envisaged that diagnostic approaches will range from conventional molecular diagnostics targeting one or few genetic alterations (eg, BRAF or MYD88 alterations in HCL and WM; reverse transcriptase polymerase chain reaction for recurring fusions identified in high-risk ALL) to panels of targets for disease subtypes with multiple types of mutation, through to genome-wide sequencing approaches (WES, transcriptome sequencing, and/or WGS) for entities driven by multiple types of genetic alteration. Although NGS is likely to rapidly enter the clinic, a note of caution is warranted because, although many recurring alterations are of considerable biologic interest, many are not intuitively actionable or druggable. Moreover, the identification of potential druggable mutations in a novel disease context is increasingly common, and evidence to recommend the use of a targeted agent is often limited or absent (for a more detailed review on clinical sequencing, see Simon and Roychowdhury100 ).

NGS studies reported in the last 3 years have advanced our understanding of the molecular basis of the lymphoid neoplasms by implicating new genes and pathways, refining classification schema, and identifying new targets for therapeutic intervention. As described above, a remarkable finding of these studies is the identification of similarly mutated genes and pathways in different hematopoietic and nonhematopoietic tumors: notably, transcriptional regulation of lymphoid development; lymphoid antigen receptor signaling; cytokine receptor, tyrosine kinase, and Ras signaling; and epigenetic alterations. These findings indicate that perturbation of these core pathways is critical in many tumor types. Conversely, several genes are mutated in multiple tumor types, including BRAF in HCL and myeloma (as well as solid tumors such as melanoma), NOTCH1 in T-ALL and CLL, and MYD88 in multiple types of leukemia. This underscores the importance of these mutations both in pathogenesis and in the deployment of targeted therapies across multiple tumor types.

These studies should be considered works in progress, and several important areas for future work are envisaged. Sequencing of many additional cases of each tumor type using complementary NGS modalities is needed. Most studies have examined relatively small numbers of tumors, and it is clear that the different types of NGS are complementary in defining the full landscape of genetic alterations. Moreover, several types of genetic alterations, including complex sequence mutations and structural rearrangements, are often difficult to robustly identify from the short reads generated by NGS101 ; advances in sequencing and computational approaches will yield additional insights. Most studies have sequenced bulk tumor populations, and these have inferred extensive intratumoral heterogeneity. Since it is now clear that subclonal mutations may be important determinants of disease progression and treatment failure,10,53  sequencing of purified cell populations, including single-cell analysis, will be required to fully dissect this clonal complexity and determine the relationship to disease behavior. Most studies have focused on somatic genetic alterations by directly comparing tumor genomes to matched nontumor genomes. The role of inherited variations in tumorigenesis has not been thoroughly investigated but is clearly important in view of recent studies identifying pathogenic mutations in familial and sporadic tumors48,51,102  and in the literature describing associations between common inherited variants and susceptibility to ALL.103-107  Finally, most studies have focused on cataloging genetic alterations in the protein-coding genome. The role of genetic alterations in the noncoding genome and the relationship between structural, transcriptomic, and epigenetic profiles remains to be fully explored and is particularly important in view of the high frequency of mutations in epigenetic regulators in many lymphoid neoplasms.

The author thanks the many colleagues and collaborators, particularly those from St. Jude Children’s Research Hospital, the PCGP, the Children’s Oncology Group, and the TARGET initiative, who have made key contributions to the studies described and apologizes to those whose work could not be cited for space limitations.

This work was supported by the American Lebanese Syrian Associated Charities of St. Jude Children’s Research Hospital; C.G.M. and is a Pew Scholar in the Biomedical Sciences, and a St. Baldrick’s Foundation Scholar.

Contribution: C.G.M. wrote the manuscript.

Conflict-of-interest disclosure: The author declares no competing financial interests.

Correspondence: Charles G. Mullighan, Department of Pathology, Mail Stop 342, 262 Danny Thomas Place, Memphis TN 38105; e-mail: charles.mullighan@stjude.org.

1
Inaba
 
H
Greaves
 
M
Mullighan
 
CG
Acute lymphoblastic leukaemia.
Lancet
2013
, vol. 
381
 
9881
(pg. 
1943
-
1955
)
2
Harrison
 
CJ
Foroni
 
L
Cytogenetics and molecular genetics of acute lymphoblastic leukemia.
Rev Clin Exp Hematol
2002
, vol. 
6
 
2
(pg. 
91
-
113, discussion 200-202
)
3
Aifantis
 
I
Raetz
 
E
Buonamici
 
S
Molecular pathogenesis of T-cell leukaemia and lymphoma.
Nat Rev Immunol
2008
, vol. 
8
 
5
(pg. 
380
-
390
)
4
Nachman
 
J
Clinical characteristics, biologic features and outcome for young adult patients with acute lymphoblastic leukaemia.
Br J Haematol
2005
, vol. 
130
 
2
(pg. 
166
-
173
)
5
Mullighan
 
CG
Molecular genetics of B-precursor acute lymphoblastic leukemia.
J Clin Invest
2012
, vol. 
122
 
10
(pg. 
3407
-
3415
)
6
Downing
 
JR
Wilson
 
RK
Zhang
 
J
, et al. 
The pediatric cancer genome project.
Nat Genet
2012
, vol. 
44
 
6
(pg. 
619
-
622
)
7
Mullighan
 
CG
Goorha
 
S
Radtke
 
I
, et al. 
Genome-wide analysis of genetic alterations in acute lymphoblastic leukaemia.
Nature
2007
, vol. 
446
 
7137
(pg. 
758
-
764
)
8
Kuiper
 
RP
Schoenmakers
 
EF
van Reijmersdal
 
SV
Hehir-Kwa
 
JY
van Kessel
 
AG
van Leeuwen
 
FN
Hoogerbrugge
 
PM
High-resolution genomic profiling of childhood ALL reveals novel recurrent genetic lesions affecting pathways involved in lymphocyte differentiation and cell cycle progression.
Leukemia
2007
, vol. 
21
 
6
(pg. 
1258
-
1266
)
9
Mullighan
 
CG
Miller
 
CB
Radtke
 
I
, et al. 
BCR-ABL1 lymphoblastic leukaemia is characterized by the deletion of Ikaros.
Nature
2008
, vol. 
453
 
7191
(pg. 
110
-
114
)
10
Mullighan
 
CG
Zhang
 
J
Kasper
 
LH
, et al. 
CREBBP mutations in relapsed acute lymphoblastic leukaemia.
Nature
2011
, vol. 
471
 
7337
(pg. 
235
-
239
)
11
Zhang
 
J
Mullighan
 
CG
Harvey
 
RC
, et al. 
Key pathways are frequently mutated in high-risk childhood acute lymphoblastic leukemia: a report from the Children’s Oncology Group.
Blood
2011
, vol. 
118
 
11
(pg. 
3080
-
3087
)
12
Weng
 
AP
Ferrando
 
AA
Lee
 
W
, et al. 
Activating mutations of NOTCH1 in human T cell acute lymphoblastic leukemia.
Science
2004
, vol. 
306
 
5694
(pg. 
269
-
271
)
13
Gutierrez
 
A
Sanda
 
T
Grebliunaite
 
R
, et al. 
High frequency of PTEN, PI3K, and AKT abnormalities in T-cell acute lymphoblastic leukemia.
Blood
2009
, vol. 
114
 
3
(pg. 
647
-
650
)
14
Tosello
 
V
Mansour
 
MR
Barnes
 
K
, et al. 
WT1 mutations in T-ALL.
Blood
2009
, vol. 
114
 
5
(pg. 
1038
-
1045
)
15
O’Neil
 
J
Grim
 
J
Strack
 
P
, et al. 
FBW7 mutations in leukemic cells mediate NOTCH pathway activation and resistance to gamma-secretase inhibitors.
J Exp Med
2007
, vol. 
204
 
8
(pg. 
1813
-
1824
)
16
Clappier
 
E
Cuccuini
 
W
Kalota
 
A
, et al. 
The C-MYB locus is involved in chromosomal translocation and genomic duplications in human T-cell acute leukemia (T-ALL), the translocation defining a new T-ALL subtype in very young children.
Blood
2007
, vol. 
110
 
4
(pg. 
1251
-
1261
)
17
Van Vlierberghe
 
P
Palomero
 
T
Khiabanian
 
H
, et al. 
PHF6 mutations in T-cell acute lymphoblastic leukemia.
Nat Genet
2010
, vol. 
42
 
4
(pg. 
338
-
342
)
18
Van Vlierberghe
 
P
Patel
 
J
Abdel-Wahab
 
O
, et al. 
PHF6 mutations in adult acute myeloid leukemia.
Leukemia
2011
, vol. 
25
 
1
(pg. 
130
-
134
)
19
Coustan-Smith
 
E
Mullighan
 
CG
Onciu
 
M
, et al. 
Early T-cell precursor leukaemia: a subtype of very high-risk acute lymphoblastic leukaemia.
Lancet Oncol
2009
, vol. 
10
 
2
(pg. 
147
-
156
)
20
Rothenberg
 
EV
Moore
 
JE
Yui
 
MA
Launching the T-cell-lineage developmental programme.
Nat Rev Immunol
2008
, vol. 
8
 
1
(pg. 
9
-
21
)
21
Zhang
 
J
Ding
 
L
Holmfeldt
 
L
, et al. 
The genetic basis of early T-cell precursor acute lymphoblastic leukaemia.
Nature
2012
, vol. 
481
 
7380
(pg. 
157
-
163
)
22
Korbel
 
JO
Campbell
 
PJ
Criteria for inference of chromothripsis in cancer genomes.
Cell
2013
, vol. 
152
 
6
(pg. 
1226
-
1236
)
23
Della Gatta
 
G
Palomero
 
T
Perez-Garcia
 
A
, et al. 
Reverse engineering of TLX oncogenic transcriptional networks identifies RUNX1 as tumor suppressor in T-ALL.
Nat Med
2012
, vol. 
18
 
3
(pg. 
436
-
440
)
24
Van Vlierberghe
 
P
Ambesi-Impiombato
 
A
Perez-Garcia
 
A
, et al. 
ETV6 mutations in early immature human T cell leukemias.
J Exp Med
2011
, vol. 
208
 
13
(pg. 
2571
-
2579
)
25
Flex
 
E
Petrangeli
 
V
Stella
 
L
, et al. 
Somatically acquired JAK1 mutations in adult acute lymphoblastic leukemia.
J Exp Med
2008
, vol. 
205
 
4
(pg. 
751
-
758
)
26
Shochat
 
C
Tal
 
N
Bandapalli
 
OR
, et al. 
Gain-of-function mutations in interleukin-7 receptor-α (IL7R) in childhood acute lymphoblastic leukemias.
J Exp Med
2011
, vol. 
208
 
5
(pg. 
901
-
908
)
27
Zenatti
 
PP
Ribeiro
 
D
Li
 
W
, et al. 
Oncogenic IL7R gain-of-function mutations in childhood T-cell acute lymphoblastic leukemia.
Nat Genet
2011
, vol. 
43
 
10
(pg. 
932
-
939
)
28
Neumann
 
M
Heesch
 
S
Gokbuget
 
N
, et al. 
 
Clinical and molecular characterization of early T-cell precursor leukemia: a high-risk subgroup in adult T-ALL with a high frequency of FLT3 mutations. Blood cancer journal. 2012;2(1):e55
29
Ntziachristos
 
P
Tsirigos
 
A
Van Vlierberghe
 
P
, et al. 
Genetic inactivation of the polycomb repressive complex 2 in T cell acute lymphoblastic leukemia.
Nat Med
2012
, vol. 
18
 
2
(pg. 
298
-
301
)
30
Neumann
 
M
Heesch
 
S
Schlee
 
C
, et al. 
Whole-exome sequencing in adult ETP-ALL reveals a high rate of DNMT3A mutations.
Blood
2013
, vol. 
121
 
23
(pg. 
4749
-
4752
)
31
Graux
 
C
Cools
 
J
Melotte
 
C
, et al. 
Fusion of NUP214 to ABL1 on amplified episomes in T-cell acute lymphoblastic leukemia.
Nat Genet
2004
, vol. 
36
 
10
(pg. 
1084
-
1089
)
32
De Keersmaecker
 
K
Atak
 
ZK
Li
 
N
, et al. 
Exome sequencing identifies mutation in CNOT3 and ribosomal genes RPL5 and RPL10 in T-cell acute lymphoblastic leukemia.
Nat Genet
2013
, vol. 
45
 
2
(pg. 
186
-
190
)
33
Mullighan
 
CG
Su
 
X
Zhang
 
J
, et al. 
Children’s Oncology Group
Deletion of IKZF1 and prognosis in acute lymphoblastic leukemia.
N Engl J Med
2009
, vol. 
360
 
5
(pg. 
470
-
480
)
34
Den Boer
 
ML
van Slegtenhorst
 
M
De Menezes
 
RX
, et al. 
A subtype of childhood acute lymphoblastic leukaemia with poor treatment outcome: a genome-wide classification study.
Lancet Oncol
2009
, vol. 
10
 
2
(pg. 
125
-
134
)
35
Loh
 
ML
Zhang
 
J
Harvey
 
RC
, et al. 
Tyrosine kinome sequencing of pediatric acute lymphoblastic leukemia: a report from the Children’s Oncology Group TARGET Project.
Blood
2013
, vol. 
121
 
3
(pg. 
485
-
488
)
36
Russell
 
LJ
Capasso
 
M
Vater
 
I
, et al. 
Deregulated expression of cytokine receptor gene, CRLF2, is involved in lymphoid transformation in B-cell precursor acute lymphoblastic leukemia.
Blood
2009
, vol. 
114
 
13
(pg. 
2688
-
2698
)
37
Harvey
 
RC
Mullighan
 
CG
Chen
 
IM
, et al. 
Rearrangement of CRLF2 is associated with mutation of JAK kinases, alteration of IKZF1, Hispanic/Latino ethnicity, and a poor outcome in pediatric B-progenitor acute lymphoblastic leukemia.
Blood
2010
, vol. 
115
 
26
(pg. 
5312
-
5321
)
38
Mullighan
 
CG
Collins-Underwood
 
JR
Phillips
 
LA
, et al. 
Rearrangement of CRLF2 in B-progenitor- and Down syndrome-associated acute lymphoblastic leukemia.
Nat Genet
2009
, vol. 
41
 
11
(pg. 
1243
-
1246
)
39
Levine
 
RL
Gilliland
 
DG
Myeloproliferative disorders.
Blood
2008
, vol. 
112
 
6
(pg. 
2190
-
2198
)
40
Mullighan
 
CG
Zhang
 
J
Harvey
 
RC
, et al. 
JAK mutations in high-risk childhood acute lymphoblastic leukemia.
Proc Natl Acad Sci USA
2009
, vol. 
106
 
23
(pg. 
9414
-
9418
)
41
Maude
 
SL
Tasian
 
SK
Vincent
 
T
, et al. 
Targeting JAK1/2 and mTOR in murine xenograft models of Ph-like acute lymphoblastic leukemia.
Blood
2012
, vol. 
120
 
17
(pg. 
3510
-
3518
)
42
Roberts
 
KG
Morin
 
RD
Zhang
 
J
, et al. 
Genetic alterations activating kinase and cytokine receptor signaling in high-risk acute lymphoblastic leukemia.
Cancer Cell
2012
, vol. 
22
 
2
(pg. 
153
-
166
)
43
Bersenev
 
A
Wu
 
C
Balcerek
 
J
, et al. 
Lnk constrains myeloproliferative diseases in mice.
J Clin Invest
2010
, vol. 
120
 
6
(pg. 
2058
-
2069
)
44
Weston
 
BW
Hayden
 
MA
Roberts
 
KG
, et al. 
Tyrosine kinase inhibitor therapy induces remission in a patient with refractory EBF1-PDGFRB-positive acute lymphoblastic leukemia.
J Clin Oncol
2013
, vol. 
31
 
25
(pg. 
e413
-
e416
)
45
Harrison
 
CJ
Moorman
 
AV
Broadfield
 
ZJ
, et al. 
Childhood and Adult Leukaemia Working Parties
Three distinct subgroups of hypodiploidy in acute lymphoblastic leukaemia.
Br J Haematol
2004
, vol. 
125
 
5
(pg. 
552
-
559
)
46
Heerema
 
NA
Nachman
 
JB
Sather
 
HN
, et al. 
Hypodiploidy with less than 45 chromosomes confers adverse risk in childhood acute lymphoblastic leukemia: a report from the children’s cancer group.
Blood
1999
, vol. 
94
 
12
(pg. 
4036
-
4045
)
47
Raimondi
 
SC
Zhou
 
Y
Mathew
 
S
, et al. 
Reassessment of the prognostic significance of hypodiploidy in pediatric patients with acute lymphoblastic leukemia.
Cancer
2003
, vol. 
98
 
12
(pg. 
2715
-
2722
)
48
Holmfeldt
 
L
Wei
 
L
Diaz-Flores
 
E
, et al. 
The genomic landscape of hypodiploid acute lymphoblastic leukemia.
Nat Genet
2013
, vol. 
45
 
3
(pg. 
242
-
252
)
49
Villani
 
A
Tabori
 
U
Schiffman
 
J
, et al. 
Biochemical and imaging surveillance in germline TP53 mutation carriers with Li-Fraumeni syndrome: a prospective observational study.
Lancet Oncol
2011
, vol. 
12
 
6
(pg. 
559
-
567
)
50
Powell
 
BC
Jiang
 
L
Muzny
 
DM
, et al. 
Identification of TP53 as an acute lymphocytic leukemia susceptibility gene through exome sequencing.
Pediatr Blood Cancer
2013
, vol. 
60
 
6
(pg. 
E1
-
E3
)
51
Shah
 
S
Schrader
 
KA
Waanders
 
E
, et al. 
A recurrent germline PAX5 mutation confers susceptibility to pre-B cell acute lymphoblastic leukemia [published online ahead of print September 8, 2013].
Nat Genet
52
Raimondi
 
SC
Pui
 
CH
Head
 
DR
Rivera
 
GK
Behm
 
FG
Cytogenetically different leukemic clones at relapse of childhood acute lymphoblastic leukemia.
Blood
1993
, vol. 
82
 
2
(pg. 
576
-
580
)
53
Mullighan
 
CG
Phillips
 
LA
Su
 
X
Ma
 
J
Miller
 
CB
Shurtleff
 
SA
Downing
 
JR
Genomic analysis of the clonal origins of relapsed acute lymphoblastic leukemia.
Science
2008
, vol. 
322
 
5906
(pg. 
1377
-
1380
)
54
Yang
 
JJ
Bhojwani
 
D
Yang
 
W
, et al. 
Genome-wide copy number profiling reveals molecular evolution from diagnosis to relapse in childhood acute lymphoblastic leukemia.
Blood
2008
, vol. 
112
 
10
(pg. 
4178
-
4183
)
55
Inthal
 
A
Zeitlhofer
 
P
Zeginigg
 
M
, et al. 
CREBBP HAT domain mutations prevail in relapse cases of high hyperdiploid childhood acute lymphoblastic leukemia.
Leukemia
2012
, vol. 
26
 
8
(pg. 
1797
-
1803
)
56
Pasqualucci
 
L
Dominguez-Sola
 
D
Chiarenza
 
A
, et al. 
Inactivating mutations of acetyltransferase genes in B-cell lymphoma.
Nature
2011
, vol. 
471
 
7337
(pg. 
189
-
195
)
57
Meyer
 
JA
Wang
 
J
Hogan
 
LE
, et al. 
Relapse-specific mutations in NT5C2 in childhood acute lymphoblastic leukemia.
Nat Genet
2013
, vol. 
45
 
3
(pg. 
290
-
294
)
58
Tzoneva
 
G
Perez-Garcia
 
A
Carpenter
 
Z
, et al. 
Activating mutations in the NT5C2 nucleotidase gene drive chemotherapy resistance in relapsed ALL.
Nat Med
2013
, vol. 
19
 
3
(pg. 
368
-
371
)
59
Campo
 
E
Swerdlow
 
SH
Harris
 
NL
Pileri
 
S
Stein
 
H
Jaffe
 
ES
The 2008 WHO classification of lymphoid neoplasms and beyond: evolving concepts and practical applications.
Blood
2011
, vol. 
117
 
19
(pg. 
5019
-
5032
)
60
Pasqualucci
 
L
The genetic basis of diffuse large B-cell lymphoma.
Curr Opin Hematol
2013
, vol. 
20
 
4
(pg. 
336
-
344
)
61
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
, vol. 
42
 
2
(pg. 
181
-
185
)
62
Morin
 
RD
Mendez-Lago
 
M
Mungall
 
AJ
, et al. 
Frequent mutation of histone-modifying genes in non-Hodgkin lymphoma.
Nature
2011
, vol. 
476
 
7360
(pg. 
298
-
303
)
63
Pasqualucci
 
L
Trifonov
 
V
Fabbri
 
G
, et al. 
Analysis of the coding genome of diffuse large B-cell lymphoma.
Nat Genet
2011
, vol. 
43
 
9
(pg. 
830
-
837
)
64
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 USA
2012
, vol. 
109
 
10
(pg. 
3879
-
3884
)
65
Zhang
 
J
Grubor
 
V
Love
 
CL
, et al. 
Genetic heterogeneity of diffuse large B-cell lymphoma.
Proc Natl Acad Sci USA
2013
, vol. 
110
 
4
(pg. 
1398
-
1403
)
66
Lenz
 
G
Wright
 
GW
Emre
 
NC
, et al. 
Molecular subtypes of diffuse large B-cell lymphoma arise by distinct genetic pathways.
Proc Natl Acad Sci USA
2008
, vol. 
105
 
36
(pg. 
13520
-
13525
)
67
Davis
 
RE
Ngo
 
VN
Lenz
 
G
, et al. 
Chronic active B-cell-receptor signalling in diffuse large B-cell lymphoma.
Nature
2010
, vol. 
463
 
7277
(pg. 
88
-
92
)
68
Trinh
 
DL
Scott
 
DW
Morin
 
RD
, et al. 
Analysis of FOXO1 mutations in diffuse large B-cell lymphoma.
Blood
2013
, vol. 
121
 
18
(pg. 
3666
-
3674
)
69
Sneeringer
 
CJ
Scott
 
MP
Kuntz
 
KW
Knutson
 
SK
Pollock
 
RM
Richon
 
VM
Copeland
 
RA
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 USA
2010
, vol. 
107
 
49
(pg. 
20980
-
20985
)
70
Yap
 
DB
Chu
 
J
Berg
 
T
, et al. 
Somatic mutations at EZH2 Y641 act dominantly through a mechanism of selectively altered PRC2 catalytic activity, to increase H3K27 trimethylation.
Blood
2011
, vol. 
117
 
8
(pg. 
2451
-
2459
)
71
Morin
 
RD
Mungall
 
K
Pleasance
 
E
, et al. 
Mutational and structural analysis of diffuse large B-cell lymphoma using whole-genome sequencing.
Blood
2013
, vol. 
122
 
7
(pg. 
1256
-
1265
)
72
Scott
 
DW
Mungall
 
KL
Ben-Neriah
 
S
, et al. 
TBL1XR1/TP63: a novel recurrent gene fusion in B-cell non-Hodgkin lymphoma.
Blood
2012
, vol. 
119
 
21
(pg. 
4949
-
4952
)
73
Kiel
 
MJ
Velusamy
 
T
Betz
 
BL
, et al. 
Whole-genome sequencing identifies recurrent somatic NOTCH2 mutations in splenic marginal zone lymphoma.
J Exp Med
2012
, vol. 
209
 
9
(pg. 
1553
-
1565
)
74
Kridel
 
R
Meissner
 
B
Rogic
 
S
, et al. 
Whole transcriptome sequencing reveals recurrent NOTCH1 mutations in mantle cell lymphoma.
Blood
2012
, vol. 
119
 
9
(pg. 
1963
-
1971
)
75
Meissner
 
B
Kridel
 
R
Lim
 
RS
, et al. 
The E3 ubiquitin ligase UBR5 is recurrently mutated in mantle cell lymphoma.
Blood
2013
, vol. 
121
 
16
(pg. 
3161
-
3164
)
76
Steidl
 
C
Shah
 
SP
Woolcock
 
BW
, et al. 
MHC class II transactivator CIITA is a recurrent gene fusion partner in lymphoid cancers.
Nature
2011
, vol. 
471
 
7338
(pg. 
377
-
381
)
77
Love
 
C
Sun
 
Z
Jima
 
D
, et al. 
The genetic landscape of mutations in Burkitt lymphoma.
Nat Genet
2012
, vol. 
44
 
12
(pg. 
1321
-
1325
)
78
Richter
 
J
Schlesner
 
M
Hoffmann
 
S
, et al. 
ICGC MMML-Seq Project
Recurrent mutation of the ID3 gene in Burkitt lymphoma identified by integrated genome, exome and transcriptome sequencing.
Nat Genet
2012
, vol. 
44
 
12
(pg. 
1316
-
1320
)
79
Schmitz
 
R
Young
 
RM
Ceribelli
 
M
, et al. 
Burkitt lymphoma pathogenesis and therapeutic targets from structural and functional genomics.
Nature
2012
, vol. 
490
 
7418
(pg. 
116
-
120
)
80
Martín-Subero
 
JI
López-Otín
 
C
Campo
 
E
Genetic and epigenetic basis of chronic lymphocytic leukemia.
Curr Opin Hematol
2013
, vol. 
20
 
4
(pg. 
362
-
368
)
81
Wang
 
L
Lawrence
 
MS
Wan
 
Y
, et al. 
SF3B1 and other novel cancer genes in chronic lymphocytic leukemia.
N Engl J Med
2011
, vol. 
365
 
26
(pg. 
2497
-
2506
)
82
Puente
 
XS
Pinyol
 
M
Quesada
 
V
, et al. 
Whole-genome sequencing identifies recurrent mutations in chronic lymphocytic leukaemia.
Nature
2011
, vol. 
475
 
7354
(pg. 
101
-
105
)
83
Fabbri
 
G
Rasi
 
S
Rossi
 
D
, et al. 
Analysis of the chronic lymphocytic leukemia coding genome: role of NOTCH1 mutational activation.
J Exp Med
2011
, vol. 
208
 
7
(pg. 
1389
-
1401
)
84
Quesada
 
V
Conde
 
L
Villamor
 
N
, et al. 
Exome sequencing identifies recurrent mutations of the splicing factor SF3B1 gene in chronic lymphocytic leukemia.
Nat Genet
2012
, vol. 
44
 
1
(pg. 
47
-
52
)
85
Quesada
 
V
Ramsay
 
AJ
Rodríguez
 
D
Puente
 
XS
Campo
 
E
López-Otín
 
C
The genomic landscape of chronic lymphocytic leukemia: clinical implications.
BMC Med
2013
, vol. 
11
 pg. 
124
 
86
Ramsay
 
AJ
Quesada
 
V
Foronda
 
M
, et al. 
POT1 mutations cause telomere dysfunction in chronic lymphocytic leukemia.
Nat Genet
2013
, vol. 
45
 
5
(pg. 
526
-
530
)
87
Ramsay
 
AJ
Rodríguez
 
D
Villamor
 
N
, et al. 
Frequent somatic mutations in components of the RNA processing machinery in chronic lymphocytic leukemia.
Leukemia
2013
, vol. 
27
 
7
(pg. 
1600
-
1603
)
88
Rossi
 
D
Rasi
 
S
Fabbri
 
G
, et al. 
Mutations of NOTCH1 are an independent predictor of survival in chronic lymphocytic leukemia.
Blood
2012
, vol. 
119
 
2
(pg. 
521
-
529
)
89
Rossi
 
D
Rasi
 
S
Spina
 
V
, et al. 
Integrated mutational and cytogenetic analysis identifies new prognostic subgroups in chronic lymphocytic leukemia.
Blood
2013
, vol. 
121
 
8
(pg. 
1403
-
1412
)
90
Landau
 
DA
Carter
 
SL
Stojanov
 
P
, et al. 
Evolution and impact of subclonal mutations in chronic lymphocytic leukemia.
Cell
2013
, vol. 
152
 
4
(pg. 
714
-
726
)
91
Tiacci
 
E
Trifonov
 
V
Schiavoni
 
G
, et al. 
BRAF mutations in hairy-cell leukemia.
N Engl J Med
2011
, vol. 
364
 
24
(pg. 
2305
-
2315
)
92
Trifa
 
AP
Popp
 
RA
Cucuianu
 
A
, et al. 
Absence of BRAF V600E mutation in a cohort of 402 patients with various chronic and acute myeloid neoplasms.
Leuk Lymphoma
2012
, vol. 
53
 
12
(pg. 
2496
-
2497
)
93
Arcaini
 
L
Zibellini
 
S
Boveri
 
E
, et al. 
The BRAF V600E mutation in hairy cell leukemia and other mature B-cell neoplasms.
Blood
2012
, vol. 
119
 
1
(pg. 
188
-
191
)
94
Akarca
 
AU
Shende
 
VH
Ramsay
 
AD
, et al. 
BRAF V600E mutation-specific antibody, a sensitive diagnostic marker revealing minimal residual disease in hairy cell leukaemia.
Br J Haematol
2013
, vol. 
162
 
6
(pg. 
848
-
851
)
95
Dietrich
 
S
Glimm
 
H
Andrulis
 
M
von Kalle
 
C
Ho
 
AD
Zenz
 
T
BRAF inhibition in refractory hairy-cell leukemia.
N Engl J Med
2012
, vol. 
366
 
21
(pg. 
2038
-
2040
)
96
Chapman
 
MA
Lawrence
 
MS
Keats
 
JJ
, et al. 
Initial genome sequencing and analysis of multiple myeloma.
Nature
2011
, vol. 
471
 
7339
(pg. 
467
-
472
)
97
Walker
 
BA
Wardell
 
CP
Melchor
 
L
, et al. 
Intraclonal heterogeneity is a critical early event in the development of myeloma and precedes the development of clinical symptoms [published online ahead of print July 2, 2013].
Leukemia
98
Treon
 
SP
Xu
 
L
Yang
 
G
, et al. 
MYD88 L265P somatic mutation in Waldenström’s macroglobulinemia.
N Engl J Med
2012
, vol. 
367
 
9
(pg. 
826
-
833
)
99
Treon
 
SP
Hunter
 
ZR
Matous
 
J
, et al. 
Multicenter clinical trial of bortezomib in relapsed/refractory Waldenstrom’s macroglobulinemia: results of WMCTG Trial 03-248.
Clin Cancer Res
2007
, vol. 
13
 
11
(pg. 
3320
-
3325
)
100
Simon
 
R
Roychowdhury
 
S
Implementing personalized cancer genomics in clinical trials.
Nat Rev Drug Discov
2013
, vol. 
12
 
5
(pg. 
358
-
369
)
101
Wang
 
J
Mullighan
 
CG
Easton
 
J
, et al. 
CREST maps somatic structural variation in cancer genomes with base-pair resolution.
Nat Methods
2011
, vol. 
8
 
8
(pg. 
652
-
654
)
102
Perez-Garcia
 
A
Ambesi-Impiombato
 
A
Hadler
 
M
, et al. 
Genetic loss of SH2B3 in acute lymphoblastic leukemia [published online ahead of print August 1, 2013].
Blood
103
Papaemmanuil
 
E
Hosking
 
FJ
Vijayakrishnan
 
J
, et al. 
Loci on 7p12.2, 10q21.2 and 14q11.2 are associated with risk of childhood acute lymphoblastic leukemia.
Nat Genet
2009
, vol. 
41
 
9
(pg. 
1006
-
1010
)
104
Xu
 
H
Yang
 
W
Perez-Andreu
 
V
, et al. 
Novel susceptibility variants at 10p12.31-12.2 for childhood acute lymphoblastic leukemia in ethnically diverse populations.
J Natl Cancer Inst
2013
, vol. 
105
 
10
(pg. 
733
-
742
)
105
Yang
 
JJ
Cheng
 
C
Devidas
 
M
, et al. 
Ancestry and pharmacogenomics of relapse in acute lymphoblastic leukemia.
Nat Genet
2011
, vol. 
43
 
3
(pg. 
237
-
241
)
106
Yang
 
JJ
Cheng
 
C
Yang
 
W
, et al. 
Genome-wide interrogation of germline genetic variation associated with treatment response in childhood acute lymphoblastic leukemia.
JAMA
2009
, vol. 
301
 
4
(pg. 
393
-
403
)
107
Treviño
 
LR
Yang
 
W
French
 
D
, et al. 
Germline genomic variants associated with childhood acute lymphoblastic leukemia.
Nat Genet
2009
, vol. 
41
 
9
(pg. 
1001
-
1005
)
Sign in via your Institution