High-grade B-cell lymphoma (HGBL), not otherwise specified (NOS), is a recently introduced diagnostic category for aggressive B-cell lymphomas. It includes tumors with Burkitt-like or blastoid morphology that do not have double-hit cytogenetics and that cannot be classified as other well-defined lymphoma subtypes. HBCLs, NOS, are rare and heterogeneous; most have germinal center B-cell phenotype, and up to 45% carry a single-hit MYC rearrangement, but otherwise, they have no unifying immunophenotypic or cytogenetic characteristics. Recent analyses using gene expression profiling (GEP) revealed that up to 15% of tumors currently classified as diffuse large B-cell lymphoma display an HGBL-like GEP signature, indicating a potential to significantly expand the HGBL category using more objective molecular criteria. Optimal treatment of HGBL, NOS, is poorly defined because of its rarity and inconsistent diagnostic patterns. A minority of patients have early-stage disease, which can be managed with standard R-CHOP–based approaches with or without radiation therapy. For advanced-stage HGBL, NOS, which often presents with aggressive disseminated disease, high lactate dehydrogenase, and involvement of extranodal organs (including the central nervous system [CNS]), intensified Burkitt lymphoma–like regimens with CNS prophylaxis may be appropriate. However, many patients diagnosed at age >60 years are not eligible for intensive immunochemotherapy. An improved GEP- and/or genomic-based pathologic classification that could facilitate HGBL-specific trials is needed to improve outcomes for all patients. In this review, we discuss the current clinicopathologic concept of HGBL, NOS, and existing data on its prognosis and treatment and delineate potential future taxonomy enrichments based on emerging molecular diagnostics.

The classification of aggressive B-cell lymphomas has evolved, driven by the delineation of subtypes with unique clinical, pathologic, and molecular features. Accurate classification is important to clinicians, who translate it into refinement of therapeutic approaches. For decades, pathologists have recognized that some B-cell neoplasms morphologically resemble Burkitt lymphoma (BL), while displaying phenotypic or genotypic characteristics inconsistent with this diagnosis. Clinically, such cases behave more aggressively than diffuse large B-cell lymphoma (DLBCL), which led to their provisional designation as high-grade B-cell lymphoma (HGBL), Burkitt-like, in the 1996 Revised European-American Lymphoma Classification.1 The differentiation of HGBL from BL or DLBCL remained problematic, with disagreement between experts in half of cases.2 

In 2008, the World Health Organization (WHO) reclassified these high-grade tumors as “B-cell lymphoma, unclassifiable, with features intermediate between DLBCL and BL3 (BCLU). The BCLU description still emphasized morphology, despite discovery of Burkitt-like gene expression profiles (GEPs) and MYC rearrangements (MYC-Rs).4-6 BCLU further included cases with blastoid morphology, a gray zone between lymphoblastic lymphoma and DLBCL. Interestingly, pediatric hematologists have tailored chemotherapy in aggressive B-cell lymphomas to the clinical presentation (eg, stage, lactate dehydrogenase [LDH] level, and bone marrow or central nervous system [CNS] involvement) rather than specific histology, whereas adult oncologists have strictly separated BL and DLBCL, variably pooling HGBL with either group.7 

Confronted with the poor prognosis of lymphomas carrying concurrent MYC-R and BCL2 (and/or BCL6) rearrangement, the 2016 WHO classification separated these double (DHLs)/triple-hit lymphomas as a formal HGBL category.8-11 Consequently, the BCLU subtype was broken apart, and non-DHL tumors were designated as HGBL, not otherwise specified (NOS).12 This entity is again defined by morphologic criteria. The WHO advocated that HGBL, NOS, be diagnosed sparingly and “only when the pathologist is truly unable to confidently classify the case as DLBCL or BL.”12 (p340) The resulting diagnostic variability hinders translational and clinical research, with a paucity of prognostic or treatment data for these highly aggressive cancers.

In this review, we discuss HGBL, NOS, in the context of emerging molecular and clinical insights. We examine how the definition of HGBL could be reformulated using cytogenetic and molecular criteria, as was first done with DHL, with the goal of establishing specific therapeutic approaches. Hematologists are challenged by treatment decisions for aggressive, rapidly progressing lymphomas, and trials that have used intensified therapy beyond R-CHOP (rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone) for clinically high-risk DLBCL have been unsuccessful in improving survival.13-17 Recognition of molecular HGBL may offer a chance to better identify the most aggressive B-cell lymphomas and to delineate optimal therapy through dedicated clinical trials.

According to the 2016 WHO classification, HGBL, NOS, includes B-cell tumors with either BL-like or blastoid morphology that do not meet criteria for other well-defined categories (Figure 1).12 Adequate classification requires evaluation by cytomorphology, immunohistochemistry, and fluorescence in situ hybridization (FISH). DHL should be classified as HGBL with concurrent MYC-R and BCL2 and/or BCL6 rearrangement. Morphologically clear-cut DLBCL with a single-hit MYC-R should be classified as DLBCL, NOS, regardless of the proliferative fraction or other molecular and cytogenetic features.12,18,19 Historically worse prognosis in DLBCL with MYC-R was largely driven by DHL,9,10,20 and the Lunenburg Lymphoma Biomarker Consortium study of 2383 DLBCLs showed no significant difference in outcomes between DLBCL with or without a single-hit MYC-R.21 Extra copies of MYC have been variably associated with worse prognosis in DLBCL/BCLU.22-24 Extra copies of MYC, coexpression of MYC and BCL2 by immunohistochemistry (dual-expressor lymphoma [DEL]), and alterations in TP53 or other genes are not currently considered in the WHO definition of HGBL, NOS.

Figure 1.

HGBL, NOS and other aggressive B-cell lymphomas. Note that the size of the circles is not to scale, as the precise incidence of some entities is unknown; shaded areas indicate the proportion of cases with MYC rearrangement. Professional illustration by Patrick Lane, ScEYEnce Studios.

Figure 1.

HGBL, NOS and other aggressive B-cell lymphomas. Note that the size of the circles is not to scale, as the precise incidence of some entities is unknown; shaded areas indicate the proportion of cases with MYC rearrangement. Professional illustration by Patrick Lane, ScEYEnce Studios.

Close modal

In a pathologic study of 32 HGBL, NOS, cases from the German High Grade Lymphoma Study Group, CD10 was expressed in 65%, BCL6 in 91%, and BCL2 in 65%, and MYC-R was present in 13%.25,MYC-R may be accompanied by BCL2 amplifications.18,26 An inverse pattern with MYC amplification and BCL2-IGH translocation, or amplification of both oncogenes, can also be encountered.27 Among 50 HGBL, NOS, cases from the University of Pittsburgh (Burkitt-like, n = 41; blastoid, n = 9), 8% showed MYC-R, 32% MYC extra copies, and 60% no MYC alterations.28 In a recent Chinese series of 41 cases, 76% expressed CD10, 83% BCL6, 63% MUM1/IRF4, and 71% MYC.29 

The Lymphoma/Leukemia Molecular Profiling Project study provided the first molecular characterization of HGBL, NOS.30 Notably, 53% of 64 centrally reviewed cases were reclassified as DLBCL or BL, confirming the poor reproducibility of the WHO criteria. There were no significant differences in cytogenetic, GEP, or genomic features between confirmed HGBL, NOS, cases and cases reclassified as DLBCL. Among confirmed HGBLs, 57% had germinal center B-cell (GCB) origin by GEP, but 25% were activated B-cell tumors. MYC-R was present in 46%, BCL2 in 10%, and BCL6 in 12%; the most common mutations involved KMT2D (43%) and TP53 (30%). Mutation-based subtypes and GEP signatures were variable, suggesting that HGBL, NOS, contains several biologic entities, including some activated B-cell tumors with MYD88, CD79B, or TBL1XR1 mutations.

Burkitt-like variant

The Burkitt-like variant of HGBL, NOS, corresponds directly to the historical category of BCLU. It is a GCB (CD10+ BCL6+) lymphoma characterized by Burkitt-like morphology (diffuse infiltrate of intermediate-sized cells, tingible body macrophages, and generally high proliferation rate; Figure 2) but often more pleomorphic appearance and sometimes lower Ki-67 staining (50% to 90%). Other cases are morphologically indistinguishable from BL, yet may show expression of BCL2, lack CD10 expression, or have multiple cytogenetic abnormalities or other characteristics unusual for BL. Of note, 5% to 10% of true BLs may not show MYC-R, so tumors with otherwise classical morphology and immunophenotype should be designated as BL.12,31

Figure 2.

Burkitt-like variant of HGBL, NOS. (A) High magnification of hematoxylin and eosin stain shows a starry-sky pattern with a diffuse infiltrate of medium to large cells with finely dispersed chromatin, several small nucleoli, and frequent mitoses. (B) Malignant B cells express the germinal center markers CD10 and BCL6 but are largely negative for BCL2. (C) Ki-67 proliferation index was ∼100%, and neoplastic cells showed high expression of MYC but were CD43; other negative markers included BCL1, TdT, MUM1, and EBER by in situ hybridization. (D) FISH shows no evidence of MYC-R or BCL2-IGH rearrangement (FISH probes: Vysis LSI IGH/MYC/CEP 8 Tri-Color Dual Fusion Probe, Vysis LSI MYC-Dual Color Break-Apart Probe, and Vysis LSI IGH/BCL2 Dual Color Dual Fusion Probe; Abbott Molecular). (E) Cerebrospinal fluid cytology shows medium to large cells with round to irregular nuclear contours, variably prominent nucleoli, and moderate amount of cytoplasm. (F) The patient, a 43-year-old HIV man, presented with extensive abdominal adenopathy and infiltration of the CNS, gastrointestinal tract, bilateral kidneys, and adrenal glands on computed tomography scan; he attained a complete response (CR) to the R-CODOX-M/IVAC (rituximab plus cyclophosphamide, doxorubicin, vincristine, and high-dose methotrexate alternating with ifosfamide, cytarabine, and etoposide) regimen with intrathecal therapy and remains in remission 1 year later. Original magnification: ×600 (A), ×400 (B-C), and ×1000 (E).

Figure 2.

Burkitt-like variant of HGBL, NOS. (A) High magnification of hematoxylin and eosin stain shows a starry-sky pattern with a diffuse infiltrate of medium to large cells with finely dispersed chromatin, several small nucleoli, and frequent mitoses. (B) Malignant B cells express the germinal center markers CD10 and BCL6 but are largely negative for BCL2. (C) Ki-67 proliferation index was ∼100%, and neoplastic cells showed high expression of MYC but were CD43; other negative markers included BCL1, TdT, MUM1, and EBER by in situ hybridization. (D) FISH shows no evidence of MYC-R or BCL2-IGH rearrangement (FISH probes: Vysis LSI IGH/MYC/CEP 8 Tri-Color Dual Fusion Probe, Vysis LSI MYC-Dual Color Break-Apart Probe, and Vysis LSI IGH/BCL2 Dual Color Dual Fusion Probe; Abbott Molecular). (E) Cerebrospinal fluid cytology shows medium to large cells with round to irregular nuclear contours, variably prominent nucleoli, and moderate amount of cytoplasm. (F) The patient, a 43-year-old HIV man, presented with extensive abdominal adenopathy and infiltration of the CNS, gastrointestinal tract, bilateral kidneys, and adrenal glands on computed tomography scan; he attained a complete response (CR) to the R-CODOX-M/IVAC (rituximab plus cyclophosphamide, doxorubicin, vincristine, and high-dose methotrexate alternating with ifosfamide, cytarabine, and etoposide) regimen with intrathecal therapy and remains in remission 1 year later. Original magnification: ×600 (A), ×400 (B-C), and ×1000 (E).

Close modal

Blastoid variant

The blastoid variant of HGBL, NOS, is rare. It has intermediate-sized cells that show blastoid morphology (medium nuclei with inconspicuous nucleoli and finely dispersed chromatin) while retaining the mature (TdT CD20+) immunophenotype (Figure 3). Blastoid HGBL, NOS, must be distinguished from B-lymphoblastic lymphoma (expressing TdT or CD34) and blastoid mantle cell lymphoma (defined by expression of cyclin D1, SOX11, and/or CCND1-IGH rearrangement). Otherwise, this entity remains poorly characterized. CD10, BCL6, and BCL2 are frequently, but not unvaryingly, expressed, whereas MYC or BCL2 rearrangement is uncommon (excluding DHL).28,32 Some cases represent transformation from follicular lymphoma or other indolent B-cell lymphomas.33 In the largest study of 24 cases (including blastoid DHL), 85% had GCB phenotype, 30% lacked CD10, 14% expressed CD5, and 3 of 4 tested cases stained for IRF4/MUM1.32 Furthermore, 89% had complex karyotype.

Figure 3.

Blastoid variant of HGBL, NOS. (A) Hematoxylin and eosin stain shows an infiltrate of medium blastoid cells with finely dispersed chromatin and inconspicuous nucleoli; touch preparation (inset) showed neoplastic cells with mostly rounded nuclei, inconspicuous nucleoli, and scant cytoplasm; the cells expressed CD19, CD20, PAX5, CD10, BCL6, and Ki-67 (>95%), but not MYC (<10%), cyclin D1, TdT, or BCL2. Additionally, stains from MUM1/IRF4, CD15, CD30, CD23, and PD-L1 were negative. (B) FISH showed normal pattern for MYC, BCL2, and IGH, but 3 copies of BCL6 (Vysis LSI IGH/BCL2 Dual Color Dual Fusion Probe, Vysis LSI BCL6 [ABR] Dual Color Break Apart Rearrangement Probe, and Vysis LSI MYC-Dual Color Break-Apart Probe; Abbott Molecular). (C) The patient, a 33-year-old woman, presented with a large mediastinal mass histologically and immunophenotypically inconsistent with B-lymphoblastic or primary mediastinal B-cell lymphoma; she attained a CR with 6 cycles of DA-EPOCH-R (etoposide, prednisone, vincristine, cyclophosphamide, doxorubicin, and rituximab) and remains in remission 3 years after therapy. Original magnification: ×600 (A).

Figure 3.

Blastoid variant of HGBL, NOS. (A) Hematoxylin and eosin stain shows an infiltrate of medium blastoid cells with finely dispersed chromatin and inconspicuous nucleoli; touch preparation (inset) showed neoplastic cells with mostly rounded nuclei, inconspicuous nucleoli, and scant cytoplasm; the cells expressed CD19, CD20, PAX5, CD10, BCL6, and Ki-67 (>95%), but not MYC (<10%), cyclin D1, TdT, or BCL2. Additionally, stains from MUM1/IRF4, CD15, CD30, CD23, and PD-L1 were negative. (B) FISH showed normal pattern for MYC, BCL2, and IGH, but 3 copies of BCL6 (Vysis LSI IGH/BCL2 Dual Color Dual Fusion Probe, Vysis LSI BCL6 [ABR] Dual Color Break Apart Rearrangement Probe, and Vysis LSI MYC-Dual Color Break-Apart Probe; Abbott Molecular). (C) The patient, a 33-year-old woman, presented with a large mediastinal mass histologically and immunophenotypically inconsistent with B-lymphoblastic or primary mediastinal B-cell lymphoma; she attained a CR with 6 cycles of DA-EPOCH-R (etoposide, prednisone, vincristine, cyclophosphamide, doxorubicin, and rituximab) and remains in remission 3 years after therapy. Original magnification: ×600 (A).

Close modal

Apart from BL and B-lymphoblastic and blastoid mantle cell lymphomas, another entity to be differentiated from HGBL, NOS, is Burkitt-like lymphoma with 11q aberration (BLL-11q), a provisional WHO subtype morphologically indistinguishable from BL.34-36 BLL-11q is predominantly nodal, lacks MYC-R, and occurs mostly in children or younger adults or as a posttransplantation lymphoproliferative disorder.36,37 It is difficult to identify in practice, because its defining cytogenetic lesion (amplification of 11q23.2-q23.3 with telomeric deletion of 11q24.1-qter) is missed by standard karyotyping. It may be suggested by presence of extra copies of ATM (11q22.3) or KMT2A (11q23.3), but cytogenomic arrays or specialized FISH probes are necessary for confirmation. Approximately 5% to 20% of HGBLs, NOS, can be reclassified as BLL-11q.38 11q abnormalities are also found in straightforward BL or other HGBL.39 BLL-11q notably lacks the distinctive BL mutations in ID3, TCF3, MYC, CCND3, or TP53, so it may be more related to HGBL than to BL.34,35,40

Recent analyses using GEP have advanced our understanding of HGBL, revealing that some tumors currently diagnosed as DLBCL have striking biologic similarities to HGBL. Emerging data confirm that these molecular high-grade (MHG) lymphomas have worse prognosis with R-CHOP therapy and might benefit from a refined classification. In 2006, GEP showed that 10% of DLBCLs and most BCLUs express a molecular BL signature characterized by overexpression of MYC targets, with decreased expression of major histocompatibility complex (MHC) genes and NF-κB signaling.4,5 Molecular BL carried more copy number alterations, 17p deletions, and MYC amplifications than classic BL.41,42 Nevertheless, most DLBCLs with a single-hit MYC-R were not molecular BL.

In 2019, this concept was expanded, uncovering that 10% to 25% of DLBCL tumors exhibit distinct GEP signatures similar to high-grade lymphomas (BL or DHL).43,44 Sha et al43 applied a GEP classifier to 928 DLBCLs from the phase 3 REMoDL-B trial (R-CHOP ± bortezomib), identifying 9% with the MHG signature (Table 1). MHG lymphomas were of GCB origin and 49% carried MYC-Rs, but only 36% were DHLs. MHG lymphomas showed low expression of MHC-II genes and downregulation of immune response pathways and were enriched in mutations in MYC, BCL2, DDX3X, TP53, and KMT2D.43,45 Three-year progression-free survival (PFS) was only 37% after R-CHOP, significantly worse than for either GCB or non-GCB DLBCL. MHG lymphomas carried poor prognosis regardless of MYC-R or DHL status. Their unfavorable outcomes after R-CHOP (5-year overall survival [OS], 44%) was confirmed in a UK population-based study.46 

Table 1.

Characteristics of molecular HGBL in studies using GEP for patients diagnosed as having DLBCL

Characteristic(REMoDL-B)43 N = 928(BCCA)44 N = 157 (GCB only)
Study cohort Phase 3 clinical trial Observational cohort 
Molecular HGBL, n (%) 83 (9) 42 (27) 
Median age, y 65 62 
Stage III/IV, n (%) 63 (76) 23 (56) 
LDH Median, 561 units/L 58% elevated 
Extranodal involvement, n (%) 53 (64) NR; 7% >1 site 
High IPI, n (%) 25 (30) 7 (17) 
GCB by GEP, % 90 100 (by definition) 
Cytogenetics, n (%)
MYC-R
 Double/triple hit 
35 (49)
26 (36) 
27 (64)
22 (52) 
Immunohistochemistry, n (%) 
 MYC+
 BCL2+
 MYC+/BCL2+ 
20 (71)
22 (79)
17 (61) 
30 (75)
36 (88)
25 (63) 
PFS or TTP, % 37 at 3 y (R-CHOP)
vs 78 in GCB DLBCL
vs 64 in ABC DLBCL 
57 at 5 y
vs 81 in GCB DLBCL
vs 51 in ABC DLBCL 
OS, % 57 at 3 y (all patients)
vs 88 in GCB DLBCL
vs 81 in ABC DLBCL 
60 at 5 y
vs 81 in GCB DLBCL
vs 56 in ABC DLBCL 
GEP signature Trained on BL vs DLBCL
↑ MYC, TCF3 targets
↓ MHC-II
↓ immune response
↓ inflammation 
Trained on DHL vs DLBCL
↑ MYC, E2F targets
↓ MHC-I and MHC-II
↑ oxidative phosphorylation
↓ TNFα, NF-κB, IL-6/JAK/STAT3 
Mutations MYC, BCL2, TP53, EZH2, DDX3X, KMT2D MYC, BCL2, TP53, EZH2, DDX3X, KMT2D, CREBBP 
Characteristic(REMoDL-B)43 N = 928(BCCA)44 N = 157 (GCB only)
Study cohort Phase 3 clinical trial Observational cohort 
Molecular HGBL, n (%) 83 (9) 42 (27) 
Median age, y 65 62 
Stage III/IV, n (%) 63 (76) 23 (56) 
LDH Median, 561 units/L 58% elevated 
Extranodal involvement, n (%) 53 (64) NR; 7% >1 site 
High IPI, n (%) 25 (30) 7 (17) 
GCB by GEP, % 90 100 (by definition) 
Cytogenetics, n (%)
MYC-R
 Double/triple hit 
35 (49)
26 (36) 
27 (64)
22 (52) 
Immunohistochemistry, n (%) 
 MYC+
 BCL2+
 MYC+/BCL2+ 
20 (71)
22 (79)
17 (61) 
30 (75)
36 (88)
25 (63) 
PFS or TTP, % 37 at 3 y (R-CHOP)
vs 78 in GCB DLBCL
vs 64 in ABC DLBCL 
57 at 5 y
vs 81 in GCB DLBCL
vs 51 in ABC DLBCL 
OS, % 57 at 3 y (all patients)
vs 88 in GCB DLBCL
vs 81 in ABC DLBCL 
60 at 5 y
vs 81 in GCB DLBCL
vs 56 in ABC DLBCL 
GEP signature Trained on BL vs DLBCL
↑ MYC, TCF3 targets
↓ MHC-II
↓ immune response
↓ inflammation 
Trained on DHL vs DLBCL
↑ MYC, E2F targets
↓ MHC-I and MHC-II
↑ oxidative phosphorylation
↓ TNFα, NF-κB, IL-6/JAK/STAT3 
Mutations MYC, BCL2, TP53, EZH2, DDX3X, KMT2D MYC, BCL2, TP53, EZH2, DDX3X, KMT2D, CREBBP 

ABC, activated B cell; BCCA, British Columbia Cancer Agency; IL-6, interleukin-6; IPI, International prognostic index; NR, not reported; TNFα, tumor necrosis factor α; TTP, time to progression.

Ennishi et al44 studied 157 GCB DLBCL tumors, identifying 42 (27%) with a DHL-like GEP signature, although 50% were not cytogenetically DHLs (Table 1). These tumors universally expressed CD10, 64% had MYC-R, and 63% were DELs. The DHL-like lymphomas showed fewer tumor-infiltrating T cells, lower expression of MHC-I genes, and frequent point mutations in MYC, BCL2, DDX3X, TP53, and KMT2D. Importantly, compared with other GCB DLBCLs, 5-year PFS after R-CHOP was worse in DHL-like lymphomas regardless of cytogenetic DHL status. Whole-exome sequencing of non-DHL lymphomas with DHL-like GEP showed that 30% carried cryptic MYC and BCL2 translocations.47 In the Lymphoma/Leukemia Molecular Profiling Project study, the DHL-like GEP signature was present in 54% of HGBL, NOS, cases and in 39% of tumors reclassified as DLBCL.30 

These seminal discoveries suggest that a well-defined subgroup of GCB tumors currently classified as DLBCL are in fact closer to HGBL from both molecular and clinical perspectives. Furthermore, molecular HGBL includes both DHL and non-DHL cases that are prognostically equivalent. If formally recognized in future taxonomy, the molecularly defined HGBL population would become large enough for subtype-specific clinical trials. As described, it requires GEP for identification, which is not available in current practice and would be challenging to implement for rapidly progressing tumors. However, recent sequencing data suggest a potential for genomic-based classification. Wright et al48 reported that the signatures described by Sha et al43 and Ennishi et al44 largely correspond to a subset of genomic EZB tumors enriched in MYC, TP53, DDX3X, GNA13, and FOXO1 mutations. The Haematological Malignancy Research Network study confirmed that subdividing the EZB/C3 cluster according to presence of MYC mutations identifies GCB tumors (EZB-MYC) with a particularly poor prognosis.49,MYC mutations with MYC overexpression also defined the highest-risk GCB subtype in the study by Reddy et al.50 

Other genetically defined forms of HGBL have been proposed and might further expand the molecular HGBL category. One subtype includes tumors with concurrent MYC-R and TP53 alteration (deletion or mutation).26,51TP53 alterations can serve as an alternative second hit to produce high-grade disease with complex karyotype and dismal outcomes.51-53 In 1 series, HGBL, NOS, morphology and poor survival were associated with MYC-R in combination with either TP53 mutation or p53 overexpression.53 In another analysis, the high-grade GEP signature carried poor prognosis only with concurrent TP53 abnormalities.54 A second category includes aggressive activated B-cell DLBCLs with MYD88, CD79B, and TBL1XR1 mutations and CDKN2A/B deletions (subset of MCD/C5 tumors) characterized by immune evasion and propensity for extranodal and CNS involvement.48,55-57 Approximately 9% of HGBLs, NOS, are identified as MCD by the LymphGen classifier, but further research is needed to characterize this subgroup.30 

Clinical characteristics and outcomes

Clinicopathologic series have described HGBL, NOS, as a disease of mostly older adults (median age, ∼70 years), equally frequent in men and women, and often with adverse risk factors (high LDH, high IPI, extranodal involvement, and CNS invasion). In the published BCLU/HGBL series (all <45 cases, often including DHL), 55% to 68% of patients had stage III/IV disease, 60% to 100% high LDH, and 40% to 55% IPI ≥3, and one-third >1 involved extranodal site.29,58-62 Reported 2-year PFS rates ranged from 23% to 69%, and OS rates from 30% to 77%. There are no reliable data on prognostic factors specific to HGBL, NOS, although prognosis appears better with low IPI (0-2).29,58,62

First-line therapy

So far, prospective clinical trials have pooled HGBL, NOS, together with other aggressive B-cell lymphomas. Therefore, clinical practice relies on extrapolations and subset analyses (Table 2). Recently, 2 single-arm studies (using DA-EPOCH-R and R-CHOP plus lenalidomide) have specifically enrolled patients with MYC-R tumors, including some with HGBL, NOS.63,64 Additionally, the Nordic Lymphoma Group NLG-LBC-06 trial used DA-EPOCH-R therapy for aggressive B-cell lymphomas with MYC-R, TP53 deletion or p53 overexpression, DEL, or DHL status.65 However, considering the lack of control groups in these trials, lack of prognostic significance of MYC-R in DLBCL, and inconsistent results from retrospective series of HGBL/BCLU, the significance of a single-hit MYC-R for treatment selection in HGBL, NOS, remains uncertain.59,66 Correlative studies are needed to delineate the prognostic and predictive value of molecular markers, including MYC translocations, amplifications, and hotspot mutations, TP53 or CDKN2A/B alterations, complex karyotype, and GEP signatures. The morphologic and molecular heterogeneities of HGBL, NOS, are likely also reflected in its responsiveness to therapy.

Table 2.

Clinical trial outcomes of adults with HGBL, NOS

StudyTreatmentStudy population*HGBL, NOS, nPFS, % (y)OS, % (y)
Cook et al, 61  R-CHOP ± ASCT DLBCL or HGBL 31 69 (2) 77 (2) 
Persky et al, 71  R-CHOP ± IFRT/IbT Localized DLBL or HGBL 22 87 (5) 89 (5) 
Mead et al, 76  CODOX-M/IVAC BL or DLBCL with Ki-67 >95% NR (DLBCL, 57) 55 (2) 59 (2) 
Corazzelli et al, 77  R-CODOX-M/IVAC BL or HGBL 15 65 (4) NR 
McMillan et al, 82  R-CODOX-M/IVAC DLBCL with IPI 3-5 NR 68 (2) 76 (2) 
Dunleavy et al, 63  DA-EPOCH-R DLBCL with MYC-R 10 71 (4) 77 (4) 
Hoelzer et al, 85  GMALL-B-ALL/NHL2002 BL, HGBL, Burkitt leukemia NR 75 (5) 80 (5) 
Thomas et al, 91  R-hyperCVAD/MA BL, HGBL, B-ALL 16 80 (3) 89 (3) 
Rizzieri et al, 90  CALGB 10002 BL or HGBL 25 64 (2) 64 (2) 
StudyTreatmentStudy population*HGBL, NOS, nPFS, % (y)OS, % (y)
Cook et al, 61  R-CHOP ± ASCT DLBCL or HGBL 31 69 (2) 77 (2) 
Persky et al, 71  R-CHOP ± IFRT/IbT Localized DLBL or HGBL 22 87 (5) 89 (5) 
Mead et al, 76  CODOX-M/IVAC BL or DLBCL with Ki-67 >95% NR (DLBCL, 57) 55 (2) 59 (2) 
Corazzelli et al, 77  R-CODOX-M/IVAC BL or HGBL 15 65 (4) NR 
McMillan et al, 82  R-CODOX-M/IVAC DLBCL with IPI 3-5 NR 68 (2) 76 (2) 
Dunleavy et al, 63  DA-EPOCH-R DLBCL with MYC-R 10 71 (4) 77 (4) 
Hoelzer et al, 85  GMALL-B-ALL/NHL2002 BL, HGBL, Burkitt leukemia NR 75 (5) 80 (5) 
Thomas et al, 91  R-hyperCVAD/MA BL, HGBL, B-ALL 16 80 (3) 89 (3) 
Rizzieri et al, 90  CALGB 10002 BL or HGBL 25 64 (2) 64 (2) 

ASCT, autologous stem cell transplantation; B-ALL, B-cell acute lymphoblastic leukemia; CALGB, Cancer and Leukemia Group B; GMALL, German Multicenter Study Group for Adult Acute Lymphoblastic Leukemia; IFRT/IbT, involved-field radiation therapy followed by ibritumomab tiuxetan consolidation; NR, not reported; R-hyperCVAD/MA, rituximab plus fractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone alternating with high-dose methotrexate and cytarabine.

*

In this table, the term HGBL encompasses Burkitt-like lymphoma, BCLU, DHL, and HGBL, NOS.

Outcomes of the HGBL subset, unless stated otherwise.

Outcomes in the entire study population (results reportedly did not significantly differ for HGBL).

Is R-CHOP adequate for HGBL, NOS?

The main dilemma in HGBL, NOS, is whether DLBCL-like therapy is sufficient, or whether intensified regimens used for BL/DHL are necessary. This area lacks prospective, histology-specific data. Outcomes of HGBCL, NOS, are nearly impossible to disentangle from historical series that variably combined BCLU, DHL, DEL, and DLBCL with MYC-R or even MYC amplification. In the prerituximab era, outcomes of Burkitt-like lymphoma or BCLU in trials seemed similar to those of DLBCL, but MYC-R was associated with poor survival.61,67,68 Retrospective studies do not paint a consistent picture and are subject to bias in patient and treatment selection. One series of 33 pediatric and adult patients with BL or Burkitt-like lymphomas noted that adults without MYC-R who received low-intensity regimens had significantly worse survival compared with those treated with BL protocols because of MYC-R. This difference was absent among children uniformly treated with intensive chemotherapy.66 In contrast, in a series of 52 patients with BCLU treated with R-CHOP or R-hyperCVAD/MA, prognosis was significantly worse for patients with MYC-rearranged tumors who received R-CHOP, but for those without MYC-R, there was no difference in prognosis between the regimens.59 Perry et al58 reported 39 BCLU cases (68% of which would be now classified as HGBL, NOS, and 32% as DHL by FISH) treated mostly with R-CHOP. Only 43% of patients attained a CR, and in most, the disease relapsed soon after completion of chemotherapy (including 35% in the CNS). Median OS was 9 months, with only 30% OS at 5 years and no difference by DHL or MYC-R status. In the aforementioned Chinese case series of HGBL, NOS, survival was significantly worse after R-CHOP compared with more intensive regimens.29 These collective data suggest that, as in the case of DHL, R-CHOP is likely suboptimal for many patients with advanced-stage HGBL, NOS, and we favor intensified regimens, although we acknowledge the uncertainty (Figure 4).

Figure 4.

First-line therapy for HGBL, NOS. Recommendations are based on available experience in aggressive lymphomas and the authors’ preferences, because histology-specific data are sparse; areas of priority research are italicized. HDMTX, high-dose methotrexate; iPET, interim positron emission tomography; IT, intrathecal; PR, partial response; RT, radiation therapy.

Figure 4.

First-line therapy for HGBL, NOS. Recommendations are based on available experience in aggressive lymphomas and the authors’ preferences, because histology-specific data are sparse; areas of priority research are italicized. HDMTX, high-dose methotrexate; iPET, interim positron emission tomography; IT, intrathecal; PR, partial response; RT, radiation therapy.

Close modal

In contrast, for the infrequent cases of early-stage HGBL, NOS, outcomes with R-CHOP are similar to those in DLBCL. One retrospective series of early-stage MYC-rearranged DLBCL/HGBL (50% DHL) found no difference in survival between R-CHOP and intensified regimens.69 Seventeen HGBL, NOS, or Burkitt-like tumors were included in the FLYER trial, which enrolled patients with aggressive B-cell lymphomas with no IPI risk factors and tumors <7.5 cm.70 PFS at 3 years was 96% with 4 cycles of R-CHOP plus 2 doses of rituximab, and no patient with HGBL relapsed. In the S1001 trial, 17% of 132 enrolled patients had HGBL, NOS.71 In this phase 2 trial, patients underwent positron emission tomography after 3 cycles of R-CHOP, and those with a CR (89%) received 1 additional R-CHOP cycle, whereas others received consolidative radiation therapy and radioimmunotherapy. Aggregate 5-year PFS was 87%. Recently, the phase 3 LYSA LNH 09-1B trial reported noninferiority of 4 vs 6 R-CHOP cycles in early-stage aggressive B-cell lymphoma with age-adjusted IPI of 0, also guided by interim positron emission tomography response, and including 16 HGBL cases.72 On the basis of these data, the FLYER, S1001, and LYSA strategies can be considered for early-stage HGBL, NOS, without additional risk factors.

For older or unfit patients with HGBL, NOS, we favor dose-attenuated R-mini-CHOP (rituximab plus reduced-dose CHOP) as a standard approach, guided by geriatric assessment, but recommend clinical trials investigating incorporation of novel targeted approaches whenever available (eg, R-mini-CHOP with azacitidine; registered at www.clinicaltrials.gov as #NCT04799275).73,74

Intensified therapy for HGBL, NOS

R-CODOX-M/IVAC

The dose-intense R-CODOX-M/IVAC regimen was pioneered by Magrath et al75 for younger patients with BL or lymphoblastic lymphoma.31,76 R-CODOX-M/IVAC has been applied in BCLU,77,78 HIV-associated BL,79-81 and high-risk DLBCL.82 In the LY-10 trial, 57 patients with aggressive DLBCL/HGBL achieved 55% PFS and 59% OS at 2 years after CODOX-M/IVAC without rituximab.76 In a recent phase 2 trial using modified CODOX-M/IVAC plus 8 doses of rituximab for DLBCL/HGBL, 2-year PFS was 68%, including patients with baseline CNS involvement (Table 1).82 Toxicities were significant, with grade 4 neutropenia in 88%, thrombocytopenia in 61%, grade ≥3 infections in 71%, and treatment-related mortality of 4% (concentrated among patients who were age >50 years and had poor performance status). Real-world data in BL indicate treatment-related mortality of 5%, mainly resulting from sepsis.31 R-CODOX-M/IVAC has been further modified to ameliorate its toxicity, and we recommend the adjusted schedule, with 6 doses of rituximab.78,82,83 Absent randomized data, R-CODOX-M/IVAC may be preferable for patients age <50 to 60 years and with CNS involvement.

DA-EPOCH-R

DA-EPOCH-R is an intermediate-intensity regimen for high-grade lymphomas including BL and DHL.84 In standard DLBCL, it did not demonstrate an advantage over R-CHOP, while significantly increasing rates of febrile neutropenia (35%) and grade ≥3 peripheral neuropathy (19%).13 Nevertheless, in a phase 2 trial focused on lymphomas with MYC-R, 4-year event-free survival after DA-EPOCH-R in HGBL (HGBL, NOS, n = 10; DHL, n = 24) was 71% (Table 2).63 The study enrolled patients up to age 80 years, indicating that DA-EPOCH-R is an important option for older patients with HGBL, NOS, because administration of other intensified regimens at age >60 years is unsafe. However, even this regimen may be too aggressive for patients age >80 years (with or without comorbidities), because in this group the risk/benefit ratio of full-dose chemotherapy becomes uncertain.

Prephase

The use of prephase therapy to improve a patient’s performance status before the start of definitive chemotherapy has not been specifically studied in HGBL. However, prephase therapy with corticosteroid with or without vincristine and/or fractionated cyclophosphamide is commonly used in BL.81,85,86 In DLBCL, the use of corticosteroid prephase at least 1 week before the start of chemotherapy improved performance status and reduced the number of treatment-associated deaths.87 

CNS prophylaxis

Unlike in BL, which shows predominantly leptomeningeal CNS involvement, parenchymal CNS relapses are more common in HGBL and DLBCL.57,60,88 Systemic prophylaxis using high-dose methotrexate is included in R-CODOX-M/IVAC and can be incorporated into R-CHOP. For HGBL, we favor early intercalated administration when feasible.89 In the phase 2 DA-EPOCH-R trial, CNS prophylaxis comprised 8 administrations of intrathecal methotrexate on days 1 and 5 during cycles 3 through 6.63 Data from BL indicate that this schedule may not be rigorously followed, even in academic centers, thus compromising CNS-related outcomes.88 The phase 3 trial included 4 intrathecal injections delivered during cycles 3 to 6 of DA-EPOCH-R.13 Although the efficacy of intrathecal prophylaxis against parenchymal CNS recurrence is uncertain, in HGBL we favor earlier administration (whenever possible, during a staging lumbar puncture, and regardless of clinical risk factors), with additional doses delivered during cycles 2 through 4. Any benefit of high-dose methotrexate consolidation after DA-EPOCH-R is uncertain, so when a compelling indication for systemic CNS prophylaxis exists, we favor R-CODOX-M/IVAC, which assures early delivery.

Other dose-intense regimens have been studied in BL and HGBL/BCLU with comparable outcomes, but they do not offer conceptual advantages over R-CODOX-M/IVAC.85,90,91 The R-hyperCVAD/MA regimen is included in the National Comprehensive Cancer Network guidelines, but it requires prolonged therapy (8 cycles) and may be associated with a higher treatment-related mortality.31 

Autologous transplantation for consolidation

Consolidation using high-dose chemotherapy and ASCT in first remission is a tempting approach in high-grade lymphomas, which have poor prognosis after chemotherapy alone. However, despite 4 randomized trials, it has not resulted in improved OS, even though some trials showed a PFS advantage in DLBCL with intermediate or high IPI.14-17 These trials did not focus on HGBL, diluting any potential benefit. A large retrospective analysis of 159 DHLs did not show a benefit of consolidative ASCT in first CR.92 Acknowledging the uncertainty related to data extrapolation, at present there is no evidence that ASCT improves outcomes for patients with HGBL, NOS, who attain CR with chemotherapy.

Salvage chemotherapy and transplantation

Data on treatment or outcomes of recurrent HGBL, NOS, are scant. Traditional approaches, modeled after DLBCL, attempt to reinduce remission and consolidate with ASCT. In the randomized CORAL trial, MYC-R was noted in a minority of patients and was notably associated with 4-year PFS of only 18%; outcomes were even worse (2-year OS, 0% to 10%) in observational studies.93,94 Among 267 patients with aggressive B-cell lymphomas (44% HGBL/BCLU) relapsing after intensive immunochemotherapy, 43% responded to salvage chemotherapy, 23% had a CR, and 15% proceeded to ASCT, achieving 3-year PFS of 42%; PFS for all patients was 14%.95 In a retrospective analysis of relapsed DHL, survival seemed better with salvage allogeneic stem cell transplantation (4-year PFS, 40%; OS, 50%).96 With regard to recently approved options for relapsed/refractory DLBCL, no confirmed cases of HGBL were reported in the registration trial of polatuzumab vedotin97; 1 patient with DHL responded to selinexor98; 4 of 7 patients with MYC-rearranged lymphomas (including 2 DHLs) responded to tafasitamab plus lenalidomide,99 and 22% of those with DHL responded to loncastuximab tesirine.100 

T cell–directed immunotherapy

A handful of patients with HGBL, NOS, have participated in trials of CD19-directed chimeric antigen receptor T-cell therapy, with promising results. Nineteen patients with confirmed DHL enrolled in the JULIET trial (tisagenlecleucel) and showed overall response rate of 50%, similar to DLBCL (52%).101 In ZUMA-1 (axicabtagene ciloleucel), 7 patients had HGBL, including 2 with HGBL, NOS; 1 achieved a CR, and 1 a partial response.102 The TRANSCEND-NHL-001 trial of lisocabtagene maraleucel included 36 patients with DHL whose response rate was similar to those with DLBCL (76% and 68%, respectively).103 In 1 study describing real-world outcomes of axicabtagene ciloleucel, 12-month PFS was 39% in HGBL, and OS was 69%, neither significantly different from those in DLBCL.104 A second real-world study identified 17 patients with HGBL, NOS, of whom 15 (88%) responded to chimeric antigen receptor T cells.105 

These responses to T cell–directed immunotherapy are encouraging in HGBL, which is characterized by downregulation of MHC genes and immune response pathways, low expression of PD-L1 on malignant B cells, and low macrophage content in the microenvironment.43,44,106-108 Efficacy of other forms of immunotherapy in HGBL remains to be determined. Anecdotal responses to blinatumomab (n = 1),109 CD20/CD3-bispecific antibody mosunetuzumab (n = 2),110 and anti-CD47 antibody Hu5F9-G4 (n = 1) have been reported.111 

HGBL, NOS, as outlined in the 2016 WHO classification, is a poorly defined and highly heterogeneous entity. Delineation of aggressive high-grade lymphomas that require treatment other than R-CHOP remains a critical priority for research. This goal will likely require replacing the cytomorphologic definition of HGBL, NOS, with more objective GEP or genomic criteria that can account for the biologic similarity between DHL (as identified by FISH) and molecular HGBL, which encompasses >50% of HGBLs, NOS, and ∼15% of DLBCLs, NOS. Successful translation of this knowledge depends on deployment of rapid, readily available, and affordable molecular diagnostics in clinical practice. Examples of studies that can pave the way for future treatment of HGBL include the Alliance A051701 phase 2/3 trial of chemotherapy with or without venetoclax in DEL/DHL (registered at www.clinicaltrials.gov as #NCT03984448) and the ZUMA-12 trial of axicabtagene ciloleucel for patients with DHL or DLBCL who do not achieve early metabolic CR with chemotherapy (registered at www.clinicaltrials.gov as #NCT03761056). Genomic analyses will be important to delineate benefits of these treatments for HGBL. Apart from immunotherapeutic approaches, other potential strategies include epigenetic modifiers, phosphoinositide 3-kinase ihibitors,42,112 bromodomain inhibitors,113 novel immunomodulatory agents, and antibody-drug conjugates.100 Application of these therapies will need to be refined in the context of tumor biology, considering the highly proliferative nature of HGBL and the molecular mechanisms that drive it. After treating unclassifiable tumors as oddities, with diagnoses of exclusion, the prospect of improving outcomes via refined molecular diagnostics and targeted therapeutic approaches for these aggressive B-cell lymphomas is potentially achievable.

Contribution: A.J.O., H.K., and A.M.E. wrote the manuscript and approved the final version of the manuscript.

Conflict-of-interest disclosure: A.J.O. reports research funding from Genentech, Adaptive Biotechnologies, Acrotech Biopharma, Celldex Therapeutics, TG Therapeutics, and Precision BioSciences. A.M.E. reports research funding from ORIEN, Leukemia & Lymphoma Society, National Institutes of Health, and National Cancer Institute; has served on advisory boards (research related) for Seattle Genetics, MorphoSys, Karyopharm, Novartis, Pharmayclics, AbbVie, and Epizyme; and has served as consultant (educational related) for Research to Practice, Curio, Cota, Patient Power, and OncLive. H.K. reports no competing financial interests.

Correspondence: Andrew M. Evens, Rutgers Cancer Institute of New Jersey, 195 Little Albany St, New Brunswick, NJ 08901; e-mail: andrew.evens@rutgers.edu.

1.
Harris
NL
,
Jaffe
ES
,
Stein
H
, et al
.
A revised European-American classification of lymphoid neoplasms: a proposal from the International Lymphoma Study Group
.
Blood.
1994
;
84
(
5
):
1361
-
1392
.
2.
A clinical evaluation of the International Lymphoma Study Group classification of non-Hodgkin’s lymphoma. The Non-Hodgkin’s Lymphoma Classification Project
.
Blood.
1997
;
89
(
11
):
3909
-
3918
.
3.
Swerdlow
SH
,
Campo
E
,
Harris
NL
, et al
.
WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues
, 4th ed.
Lyon, France
:
IARC Press
;
2008
.
4.
Dave
SS
,
Fu
K
,
Wright
GW
, et al;
Lymphoma/Leukemia Molecular Profiling Project
.
Molecular diagnosis of Burkitt’s lymphoma
.
N Engl J Med.
2006
;
354
(
23
):
2431
-
2442
.
5.
Hummel
M
,
Bentink
S
,
Berger
H
, et al;
Molecular Mechanisms in Malignant Lymphomas Network Project of the Deutsche Krebshilfe
.
A biologic definition of Burkitt’s lymphoma from transcriptional and genomic profiling
.
N Engl J Med.
2006
;
354
(
23
):
2419
-
2430
.
6.
Klapper
W
,
Stoecklein
H
,
Zeynalova
S
, et al;
German High-Grade Non-Hodgkin’s Lymphoma Study Group
.
Structural aberrations affecting the MYC locus indicate a poor prognosis independent of clinical risk factors in diffuse large B-cell lymphomas treated within randomized trials of the German High-Grade Non-Hodgkin’s Lymphoma Study Group (DSHNHL)
.
Leukemia.
2008
;
22
(
12
):
2226
-
2229
.
7.
Dunleavy
K
,
Gross
TG
.
Management of aggressive B-cell NHLs in the AYA population: an adult vs pediatric perspective
.
Blood.
2018
;
132
(
4
):
369
-
375
.
8.
Johnson
NA
,
Savage
KJ
,
Ludkovski
O
, et al
.
Lymphomas with concurrent BCL2 and MYC translocations: the critical factors associated with survival
.
Blood.
2009
;
114
(
11
):
2273
-
2279
.
9.
Savage
KJ
,
Johnson
NA
,
Ben-Neriah
S
, et al
.
MYC gene rearrangements are associated with a poor prognosis in diffuse large B-cell lymphoma patients treated with R-CHOP chemotherapy
.
Blood.
2009
;
114
(
17
):
3533
-
3537
.
10.
Barrans
S
,
Crouch
S
,
Smith
A
, et al
.
Rearrangement of MYC is associated with poor prognosis in patients with diffuse large B-cell lymphoma treated in the era of rituximab
.
J Clin Oncol.
2010
;
28
(
20
):
3360
-
3365
.
11.
Petrich
AM
,
Gandhi
M
,
Jovanovic
B
, et al
.
Impact of induction regimen and stem cell transplantation on outcomes in double-hit lymphoma: a multicenter retrospective analysis
.
Blood.
2014
;
124
(
15
):
2354
-
2361
.
12.
Swerdlow
SH
,
Campo
E
,
Harris
NL
, et al
.
WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues
, revised 4th ed.
Lyon, France
:
IARC Press
;
2017
.
13.
Bartlett
NL
,
Wilson
WH
,
Jung
SH
, et al
.
Dose-adjusted EPOCH-R compared with R-CHOP as frontline therapy for diffuse large B-cell lymphoma: clinical outcomes of the phase III intergroup trial Alliance/CALGB 50303
.
J Clin Oncol.
2019
;
37
(
21
):
1790
-
1799
.
14.
Stiff
PJ
,
Unger
JM
,
Cook
JR
, et al
.
Autologous transplantation as consolidation for aggressive non-Hodgkin’s lymphoma
.
N Engl J Med.
2013
;
369
(
18
):
1681
-
1690
.
15.
Cortelazzo
S
,
Tarella
C
,
Gianni
AM
, et al
.
Randomized trial comparing R-CHOP versus high-dose sequential chemotherapy in high-risk patients with diffuse large B-cell lymphomas
.
J Clin Oncol.
2016
;
34
(
33
):
4015
-
4022
.
16.
Chiappella
A
,
Martelli
M
,
Angelucci
E
, et al
.
Rituximab-dose-dense chemotherapy with or without high-dose chemotherapy plus autologous stem-cell transplantation in high-risk diffuse large B-cell lymphoma (DLCL04): final results of a multicentre, open-label, randomised, controlled, phase 3 study
.
Lancet Oncol.
2017
;
18
(
8
):
1076
-
1088
.
17.
Frontzek
F
,
Ziepert
M
,
Nickelsen
M
, et al
.
Rituximab plus high-dose chemotherapy (MegaCHOEP) or conventional chemotherapy (CHOEP-14) in young, high-risk patients with aggressive B-cell lymphoma: 10-year follow-up of a randomised, open-label, phase 3 trial
.
Lancet Haematol.
2021
;
8
(
4
):
e267
-
e277
.
18.
Ok
CY
,
Medeiros
LJ
.
High-grade B-cell lymphoma: a term re-purposed in the revised WHO classification
.
Pathology.
2020
;
52
(
1
):
68
-
77
.
19.
Ott
G
.
Aggressive B-cell lymphomas in the update of the 4th edition of the World Health Organization classification of haematopoietic and lymphatic tissues: refinements of the classification, new entities and genetic findings
.
Br J Haematol.
2017
;
178
(
6
):
871
-
887
.
20.
Horn
H
,
Ziepert
M
,
Becher
C
, et al;
German High-Grade Non-Hodgkin Lymphoma Study Group
.
MYC status in concert with BCL2 and BCL6 expression predicts outcome in diffuse large B-cell lymphoma
.
Blood.
2013
;
121
(
12
):
2253
-
2263
.
21.
Rosenwald
A
,
Bens
S
,
Advani
R
, et al
.
Prognostic significance of MYC rearrangement and translocation partner in diffuse large B-cell lymphoma: a study by the Lunenburg Lymphoma Biomarker Consortium
.
J Clin Oncol.
2019
;
37
(
35
):
3359
-
3368
.
22.
Landsburg
DJ
,
Falkiewicz
MK
,
Petrich
AM
, et al
.
Sole rearrangement but not amplification of MYC is associated with a poor prognosis in patients with diffuse large B cell lymphoma and B cell lymphoma unclassifiable
.
Br J Haematol.
2016
;
175
(
4
):
631
-
640
.
23.
Sermer
D
,
Bobillo
S
,
Dogan
A
, et al
.
Extra copies of MYC, BCL2, and BCL6 and outcome in patients with diffuse large B-cell lymphoma
.
Blood Adv.
2020
;
4
(
14
):
3382
-
3390
.
24.
Schieppati
F
,
Balzarini
P
,
Fisogni
S
, et al
.
An increase in MYC copy number has a progressive negative prognostic impact in patients with diffuse large B-cell and high-grade lymphoma, who may benefit from intensified treatment regimens
.
Haematologica.
2020
;
105
(
5
):
1369
-
1378
.
25.
Hüttl
KS
,
Staiger
AM
,
Richter
J
, et al
.
The “Burkitt-like” immunophenotype and genotype is rarely encountered in diffuse large B cell lymphoma and high-grade B cell lymphoma, NOS
.
Virchows Arch.
2021
;
479
(
3
):
575
-
583
.
26.
Li
S
,
Lin
P
,
Medeiros
LJ
.
Advances in pathological understanding of high-grade B cell lymphomas
.
Expert Rev Hematol.
2018
;
11
(
8
):
637
-
648
.
27.
Li
S
,
Seegmiller
AC
,
Lin
P
, et al
.
B-cell lymphomas with concurrent MYC and BCL2 abnormalities other than translocations behave similarly to MYC/BCL2 double-hit lymphomas
.
Mod Pathol.
2015
;
28
(
2
):
208
-
217
.
28.
Moore
EM
,
Aggarwal
N
,
Surti
U
,
Swerdlow
SH
.
Further exploration of the complexities of large B-cell lymphomas with MYC abnormalities and the importance of a blastoid morphology
.
Am J Surg Pathol.
2017
;
41
(
9
):
1155
-
1166
.
29.
Li
J
,
Liu
X
,
Yao
Z
,
Zhang
M
.
High-grade B-cell lymphomas, not otherwise specified: a study of 41 cases
.
Cancer Manag Res.
2020
;
12
:
1903
-
1912
.
30.
Collinge
BJ
,
Hilton
LK
,
Wong
J
, et al
.
Characterization of the genetic landscape of gigh-grade B-cell Lymphoma, NOS – an LLMPP project
.
Hematol Oncol.
2021
;
39
(
suppl 2
):
abstract 101
.
31.
Evens
AM
,
Danilov
A
,
Jagadeesh
D
, et al
.
Burkitt lymphoma in the modern era: real-world outcomes and prognostication across 30 US cancer centers
.
Blood.
2021
;
137
(
3
):
374
-
386
.
32.
Kanagal-Shamanna
R
,
Medeiros
LJ
,
Lu
G
, et al
.
High-grade B cell lymphoma, unclassifiable, with blastoid features: an unusual morphological subgroup associated frequently with BCL2 and/or MYC gene rearrangements and a poor prognosis
.
Histopathology.
2012
;
61
(
5
):
945
-
954
.
33.
Chiu
A
,
Frizzera
G
,
Mathew
S
, et al
.
Diffuse blastoid B-cell lymphoma: a histologically aggressive variant of t(14;18)-negative follicular lymphoma
.
Mod Pathol.
2009
;
22
(
11
):
1507
-
1517
.
34.
Wagener
R
,
Seufert
J
,
Raimondi
F
, et al
.
The mutational landscape of Burkitt-like lymphoma with 11q aberration is distinct from that of Burkitt lymphoma
.
Blood.
2019
;
133
(
9
):
962
-
966
.
35.
Gonzalez-Farre
B
,
Ramis-Zaldivar
JE
,
Salmeron-Villalobos
J
, et al
.
Burkitt-like lymphoma with 11q aberration: a germinal center-derived lymphoma genetically unrelated to Burkitt lymphoma
.
Haematologica.
2019
;
104
(
9
):
1822
-
1829
.
36.
Salaverria
I
,
Martin-Guerrero
I
,
Wagener
R
, et al;
Berlin-Frankfurt-Münster Non-Hodgkin Lymphoma Group
.
A recurrent 11q aberration pattern characterizes a subset of MYC-negative high-grade B-cell lymphomas resembling Burkitt lymphoma
.
Blood.
2014
;
123
(
8
):
1187
-
1198
.
37.
Ferreiro
JF
,
Morscio
J
,
Dierickx
D
, et al
.
Post-transplant molecularly defined Burkitt lymphomas are frequently MYC-negative and characterized by the 11q-gain/loss pattern
.
Haematologica.
2015
;
100
(
7
):
e275
-
e279
.
38.
Horn
H
,
Kalmbach
S
,
Wagener
R
, et al
.
A diagnostic approach to the identification of Burkitt-like lymphoma with 11q aberration in aggressive B-cell lymphomas
.
Am J Surg Pathol.
2021
;
45
(
3
):
356
-
364
.
39.
Grygalewicz
B
,
Woroniecka
R
,
Rymkiewicz
G
, et al
.
The 11q-gain/loss aberration occurs recurrently in MYC-negative Burkitt-like lymphoma with 11q aberration, as well as MYC-positive Burkitt lymphoma and MYC-positive high-grade B-cell lymphoma, NOS
.
Am J Clin Pathol.
2017
;
149
(
1
):
17
-
28
.
40.
Zayac
AS
,
Olszewski
AJ
.
Burkitt lymphoma: bridging the gap between advances in molecular biology and therapy
.
Leuk Lymphoma.
2020
;
61
(
8
):
1784
-
1796
.
41.
Salaverria
I
,
Zettl
A
,
Beà
S
, et al;
Leukemia and Lymphoma Molecular Profiling Project (LLMPP)
.
Chromosomal alterations detected by comparative genomic hybridization in subgroups of gene expression-defined Burkitt’s lymphoma
.
Haematologica.
2008
;
93
(
9
):
1327
-
1334
.
42.
Bouska
A
,
Bi
C
,
Lone
W
, et al
.
Adult high-grade B-cell lymphoma with Burkitt lymphoma signature: genomic features and potential therapeutic targets
.
Blood.
2017
;
130
(
16
):
1819
-
1831
.
43.
Sha
C
,
Barrans
S
,
Cucco
F
, et al
.
Molecular high-grade B-cell lymphoma: defining a poor-risk group that requires different approaches to therapy [published correction appears in J Clin Oncol. 2019;37(12):1035]
.
J Clin Oncol.
2019
;
37
(
3
):
202
-
212
.
44.
Ennishi
D
,
Jiang
A
,
Boyle
M
, et al
.
Double-hit gene expression signature defines a distinct subgroup of germinal center B-cell-like diffuse large B-cell lymphoma
.
J Clin Oncol.
2019
;
37
(
3
):
190
-
201
.
45.
Cucco
F
,
Barrans
S
,
Sha
C
, et al
.
Distinct genetic changes reveal evolutionary history and heterogeneous molecular grade of DLBCL with MYC/BCL2 double-hit
.
Leukemia.
2020
;
34
(
5
):
1329
-
1341
.
46.
Lacy
SE
,
Barrans
SL
,
Beer
PA
, et al
.
Targeted sequencing in DLBCL, molecular subtypes, and outcomes: a Haematological Malignancy Research Network report
.
Blood.
2020
;
135
(
20
):
1759
-
1771
.
47.
Hilton
LK
,
Tang
J
,
Ben-Neriah
S
, et al
.
The double-hit signature identifies double-hit diffuse large B-cell lymphoma with genetic events cryptic to FISH [published correction appears in Blood. 2020;135(17):1507]
.
Blood.
2019
;
134
(
18
):
1528
-
1532
.
48.
Wright
GW
,
Huang
DW
,
Phelan
JD
, et al
.
A probabilistic classification tool for genetic subtypes of diffuse large B cell lymphoma with therapeutic implications
.
Cancer Cell.
2020
;
37
(
4
):
551
-
568.e14
.
49.
Runge
HFP
,
Lacy
S
,
Barrans
S
, et al
.
Application of the LymphGen classification tool to 928 clinically and genetically-characterised cases of diffuse large B cell lymphoma (DLBCL)
.
Br J Haematol.
2021
;
192
(
1
):
216
-
220
.
50.
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
.
51.
Wang
XJ
,
Bueso-Ramos
CE
,
Tang
G
, et al;
L Jeffrey Medeiros
.
P53 expression correlates with poorer survival and augments the negative prognostic effect of MYC rearrangement, expression or concurrent MYC/BCL2 expression in diffuse large B-cell lymphoma
.
Mod Pathol.
2017
;
30
(
2
):
194
-
203
.
52.
Karube
K
,
Enjuanes
A
,
Dlouhy
I
, et al
.
Integrating genomic alterations in diffuse large B-cell lymphoma identifies new relevant pathways and potential therapeutic targets
.
Leukemia.
2018
;
32
(
3
):
675
-
684
.
53.
Deng
M
,
Xu-Monette
ZY
,
Pham
LV
, et al
.
Aggressive B-cell lymphoma with MYC/TP53 dual alterations displays distinct clinicopathobiological features and response to novel targeted agents
.
Mol Cancer Res.
2021
;
19
(
2
):
249
-
260
.
54.
Song
JY
,
Perry
AM
,
Herrera
AF
, et al
.
Double-hit signature with TP53 abnormalities predicts poor survival in patients with germinal center type diffuse large B-cell lymphoma treated with R-CHOP
.
Clin Cancer Res.
2021
;
27
(
6
):
1671
-
1680
.
55.
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
;
105
(
36
):
13520
-
13525
.
56.
Ollila
TA
,
Kurt
H
,
Waroich
J
, et al
.
Genomic subtypes may predict the risk of central nervous system recurrence in diffuse large B-cell lymphoma
.
Blood.
2021
;
137
(
8
):
1120
-
1124
.
57.
Klanova
M
,
Sehn
LH
,
Bence-Bruckler
I
, et al
.
Integration of cell of origin into the clinical CNS International Prognostic Index improves CNS relapse prediction in DLBCL
.
Blood.
2019
;
133
(
9
):
919
-
926
.
58.
Perry
AM
,
Crockett
D
,
Dave
BJ
, et al
.
B-cell lymphoma, unclassifiable, with features intermediate between diffuse large B-cell lymphoma and Burkitt lymphoma: study of 39 cases
.
Br J Haematol.
2013
;
162
(
1
):
40
-
49
.
59.
Lin
P
,
Dickason
TJ
,
Fayad
LE
, et al
.
Prognostic value of MYC rearrangement in cases of B-cell lymphoma, unclassifiable, with features intermediate between diffuse large B-cell lymphoma and Burkitt lymphoma
.
Cancer.
2012
;
118
(
6
):
1566
-
1573
.
60.
Decker
DP
,
Egan
PC
,
Zayac
AS
,
Treaba
DO
,
Olszewski
AJ
.
Treatment strategies and risk of central nervous system recurrence in high-grade B-cell and Burkitt lymphoma
.
Leuk Lymphoma.
2020
;
61
(
1
):
198
-
201
.
61.
Cook
JR
,
Goldman
B
,
Tubbs
RR
, et al
.
Clinical significance of MYC expression and/or “high-grade” morphology in non-Burkitt, diffuse aggressive B-cell lymphomas: a SWOG S9704 correlative study
.
Am J Surg Pathol.
2014
;
38
(
4
):
494
-
501
.
62.
Miyamoto
K
,
Kobayashi
Y
,
Maeshima
AM
, et al
.
Clinicopathological prognostic factors of 24 patients with B-cell lymphoma, unclassifiable, with features intermediate between diffuse large B-cell lymphoma and Burkitt lymphoma
.
Int J Hematol.
2016
;
103
(
6
):
693
-
702
.
63.
Dunleavy
K
,
Fanale
MA
,
Abramson
JS
, et al
.
Dose-adjusted EPOCH-R (etoposide, prednisone, vincristine, cyclophosphamide, doxorubicin, and rituximab) in untreated aggressive diffuse large B-cell lymphoma with MYC rearrangement: a prospective, multicentre, single-arm phase 2 study
.
Lancet Haematol.
2018
;
5
(
12
):
e609
-
e617
.
64.
Chamuleau
MED
,
Burggraaff
CN
,
Nijland
M
, et al
.
Treatment of patients with MYC rearrangement positive large B-cell lymphoma with R-CHOP plus lenalidomide: results of a multicenter HOVON phase II trial
.
Haematologica.
2020
;
105
(
12
):
2805
-
2812
.
65.
Leppä
S
,
Jørgensen
J
,
Karjalainen-Lindsberg
M-L
, et al
.
Biomarker-driven treatment strategy in high risk diffuse large b-cell lymphoma: results of a nordic phase 2 study
.
Hematol Oncol.
2021
;
39
(
suppl 2
):
abstract 027
.
66.
Sevilla
DW
,
Gong
JZ
,
Goodman
BK
, et al
.
Clinicopathologic findings in high-grade B-cell lymphomas with typical Burkitt morphologic features but lacking the MYC translocation
.
Am J Clin Pathol.
2007
;
128
(
6
):
981
-
991
.
67.
Braziel
RM
,
Arber
DA
,
Slovak
ML
, et al
.
The Burkitt-like lymphomas: a Southwest Oncology Group study delineating phenotypic, genotypic, and clinical features
.
Blood.
2001
;
97
(
12
):
3713
-
3720
.
68.
Akasaka
T
,
Akasaka
H
,
Ueda
C
, et al
.
Molecular and clinical features of non-Burkitt’s, diffuse large-cell lymphoma of B-cell type associated with the c-MYC/immunoglobulin heavy-chain fusion gene
.
J Clin Oncol.
2000
;
18
(
3
):
510
-
518
.
69.
Torka
P
,
Kothari
SK
,
Sundaram
S
, et al
.
Outcomes of patients with limited-stage aggressive large B-cell lymphoma with high-risk cytogenetics
.
Blood Adv.
2020
;
4
(
2
):
253
-
262
.
70.
Poeschel
V
,
Held
G
,
Ziepert
M
, et al;
German Lymphoma Alliance
.
Four versus six cycles of CHOP chemotherapy in combination with six applications of rituximab in patients with aggressive B-cell lymphoma with favourable prognosis (FLYER): a randomised, phase 3, non-inferiority trial
.
Lancet.
2019
;
394
(
10216
):
2271
-
2281
.
71.
Persky
DO
,
Li
H
,
Stephens
DM
, et al
.
Positron emission tomography-directed therapy for patients with limited-stage diffuse large B-cell lymphoma: results of Intergroup National Clinical Trials Network Study S1001 [published correction appears in J Clin Oncol. 2020;38(29):3459]
.
J Clin Oncol.
2020
;
38
(
26
):
3003
-
3011
.
72.
Bologna
S
,
Vander Borght
T
,
Briere
J
, et al
.
Early positron emission tomography response-adapted treatment in localized diffuse large B-cell lymphoma (aaIPI=0): results of the phase 3 LYSA LNH 09-1B trial
.
Hematol Oncol.
2021
;
39
(
suppl 2
):
abstract 005
.
73.
Peyrade
F
,
Jardin
F
,
Thieblemont
C
, et al;
Groupe d’Etude des Lymphomes de l’Adulte (GELA) investigators
.
Attenuated immunochemotherapy regimen (R-miniCHOP) in elderly patients older than 80 years with diffuse large B-cell lymphoma: a multicentre, single-arm, phase 2 trial
.
Lancet Oncol.
2011
;
12
(
5
):
460
-
468
.
74.
Di
M
,
Huntington
SF
,
Olszewski
AJ
.
Challenges and opportunities in the management of diffuse large B-cell lymphoma in older patients
.
Oncologist.
2021
;
26
(
2
):
120
-
132
.
75.
Magrath
I
,
Adde
M
,
Shad
A
, et al
.
Adults and children with small non-cleaved-cell lymphoma have a similar excellent outcome when treated with the same chemotherapy regimen
.
J Clin Oncol.
1996
;
14
(
3
):
925
-
934
.
76.
Mead
GM
,
Barrans
SL
,
Qian
W
, et al;
Australasian Leukaemia and Lymphoma Group
.
A prospective clinicopathologic study of dose-modified CODOX-M/IVAC in patients with sporadic Burkitt lymphoma defined using cytogenetic and immunophenotypic criteria (MRC/NCRI LY10 trial)
.
Blood.
2008
;
112
(
6
):
2248
-
2260
.
77.
Corazzelli
G
,
Frigeri
F
,
Russo
F
, et al
.
RD-CODOX-M/IVAC with rituximab and intrathecal liposomal cytarabine in adult Burkitt lymphoma and ‘unclassifiable’ highly aggressive B-cell lymphoma
.
Br J Haematol.
2012
;
156
(
2
):
234
-
244
.
78.
Lacasce
A
,
Howard
O
,
Lib
S
, et al
.
Modified magrath regimens for adults with Burkitt and Burkitt-like lymphomas: preserved efficacy with decreased toxicity
.
Leuk Lymphoma.
2004
;
45
(
4
):
761
-
767
.
79.
Alwan
F
,
He
A
,
Montoto
S
, et al
.
Adding rituximab to CODOX-M/IVAC chemotherapy in the treatment of HIV-associated Burkitt lymphoma is safe when used with concurrent combination antiretroviral therapy
.
AIDS.
2015
;
29
(
8
):
903
-
910
.
80.
Noy
A
,
Lee
JY
,
Cesarman
E
, et al;
AIDS Malignancy Consortium
.
AMC 048: modified CODOX-M/IVAC-rituximab is safe and effective for HIV-associated Burkitt lymphoma
.
Blood.
2015
;
126
(
2
):
160
-
166
.
81.
Evens
AM
,
Carson
KR
,
Kolesar
J
, et al
.
A multicenter phase II study incorporating high-dose rituximab and liposomal doxorubicin into the CODOX-M/IVAC regimen for untreated Burkitt’s lymphoma
.
Ann Oncol.
2013
;
24
(
12
):
3076
-
3081
.
82.
McMillan
AK
,
Phillips
EH
,
Kirkwood
AA
, et al
.
Favourable outcomes for high-risk diffuse large B-cell lymphoma (IPI 3-5) treated with front-line R-CODOX-M/R-IVAC chemotherapy: results of a phase 2 UK NCRI trial
.
Ann Oncol.
2020
;
31
(
9
):
1251
-
1259
.
83.
Jacobson
C
,
LaCasce
A
.
How I treat Burkitt lymphoma in adults
.
Blood.
2014
;
124
(
19
):
2913
-
2920
.
84.
Roschewski
M
,
Dunleavy
K
,
Abramson
JS
, et al
.
Multicenter study of risk-adapted therapy with dose-adjusted EPOCH-R in adults with untreated Burkitt lymphoma
.
J Clin Oncol.
2020
;
38
(
22
):
2519
-
2529
.
85.
Hoelzer
D
,
Walewski
J
,
Döhner
H
, et al;
German Multicenter Study Group for Adult Acute Lymphoblastic Leukemia
.
Improved outcome of adult Burkitt lymphoma/leukemia with rituximab and chemotherapy: report of a large prospective multicenter trial
.
Blood.
2014
;
124
(
26
):
3870
-
3879
.
86.
Ribrag
V
,
Koscielny
S
,
Bosq
J
, et al
.
Rituximab and dose-dense chemotherapy for adults with Burkitt’s lymphoma: a randomised, controlled, open-label, phase 3 trial
.
Lancet.
2016
;
387
(
10036
):
2402
-
2411
.
87.
Pfreundschuh
M
.
How I treat elderly patients with diffuse large B-cell lymphoma
.
Blood.
2010
;
116
(
24
):
5103
-
5110
.
88.
Zayac
AS
,
Evens
AM
,
Danilov
A
, et al
.
Outcomes of Burkitt lymphoma with central nervous system involvement: evidence from a large multicenter cohort study
.
Haematologica.
2021
;
106
(
7
):
1932
-
1942
.
89.
Abramson
JS
,
Hellmann
M
,
Barnes
JA
, et al
.
Intravenous methotrexate as central nervous system (CNS) prophylaxis is associated with a low risk of CNS recurrence in high-risk patients with diffuse large B-cell lymphoma
.
Cancer.
2010
;
116
(
18
):
4283
-
4290
.
90.
Rizzieri
DA
,
Johnson
JL
,
Byrd
JC
, et al;
Alliance for Clinical Trials In Oncology (ACTION)
.
Improved efficacy using rituximab and brief duration, high intensity chemotherapy with filgrastim support for Burkitt or aggressive lymphomas: cancer and Leukemia Group B study 10 002
.
Br J Haematol.
2014
;
165
(
1
):
102
-
111
.
91.
Thomas
DA
,
Faderl
S
,
O’Brien
S
, et al
.
Chemoimmunotherapy with hyper-CVAD plus rituximab for the treatment of adult Burkitt and Burkitt-type lymphoma or acute lymphoblastic leukemia
.
Cancer.
2006
;
106
(
7
):
1569
-
1580
.
92.
Landsburg
DJ
,
Falkiewicz
MK
,
Maly
J
, et al
.
Outcomes of patients with double-hit lymphoma who achieve first complete remission
.
J Clin Oncol.
2017
;
35
(
20
):
2260
-
2267
.
93.
Cuccuini
W
,
Briere
J
,
Mounier
N
, et al
.
MYC+ diffuse large B-cell lymphoma is not salvaged by classical R-ICE or R-DHAP followed by BEAM plus autologous stem cell transplantation
.
Blood.
2012
;
119
(
20
):
4619
-
4624
.
94.
Epperla
N
,
Maddocks
KJ
,
Salhab
M
, et al
.
C-MYC-positive relapsed and refractory, diffuse large B-cell lymphoma: Impact of additional “hits” and outcomes with subsequent therapy [published correction appears in Cancer. 2018;124(4):867]
.
Cancer.
2017
;
123
(
22
):
4411
-
4418
.
95.
Ayers
EC
,
Li
S
,
Medeiros
LJ
, et al
.
Outcomes in patients with aggressive B-cell non-Hodgkin lymphoma after intensive frontline treatment failure
.
Cancer.
2020
;
126
(
2
):
293
-
303
.
96.
Herrera
AF
,
Rodig
SJ
,
Song
JY
, et al
.
Outcomes after allogeneic stem cell transplantation in patients with double-hit and double-expressor lymphoma
.
Biol Blood Marrow Transplant.
2018
;
24
(
3
):
514
-
520
.
97.
Sehn
LH
,
Herrera
AF
,
Flowers
CR
, et al
.
Polatuzumab vedotin in relapsed or refractory diffuse large B-cell lymphoma
.
J Clin Oncol.
2020
;
38
(
2
):
155
-
165
.
98.
Kalakonda
N
,
Maerevoet
M
,
Cavallo
F
, et al
.
Selinexor in patients with relapsed or refractory diffuse large B-cell lymphoma (SADAL): a single-arm, multinational, multicentre, open-label, phase 2 trial
.
Lancet Haematol.
2020
;
7
(
7
):
e511
-
e522
.
99.
Salles
G
,
Duell
J
,
González Barca
E
, et al
.
Tafasitamab plus lenalidomide in relapsed or refractory diffuse large B-cell lymphoma (L-MIND): a multicentre, prospective, single-arm, phase 2 study
.
Lancet Oncol.
2020
;
21
(
7
):
978
-
988
.
100.
Hamadani
M
,
Radford
J
,
Carlo-Stella
C
, et al
.
Final results of a phase 1 study of loncastuximab tesirine in relapsed/refractory B-cell non-Hodgkin lymphoma
.
Blood.
2021
;
137
(
19
):
2634
-
2645
.
101.
Schuster
SJ
,
Bishop
MR
,
Tam
CS
, et al;
JULIET Investigators
.
Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma
.
N Engl J Med.
2019
;
380
(
1
):
45
-
56
.
102.
Locke
FL
,
Ghobadi
A
,
Jacobson
CA
, et al
.
Long-term safety and activity of axicabtagene ciloleucel in refractory large B-cell lymphoma (ZUMA-1): a single-arm, multicentre, phase 1-2 trial
.
Lancet Oncol.
2019
;
20
(
1
):
31
-
42
.
103.
Abramson
JS
,
Palomba
ML
,
Gordon
LI
, et al
.
Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): a multicentre seamless design study
.
Lancet.
2020
;
396
(
10254
):
839
-
852
.
104.
Nastoupil
LJ
,
Jain
MD
,
Feng
L
, et al
.
Standard-of-care axicabtagene ciloleucel for relapsed or refractory large B-cell lymphoma: results from the US Lymphoma CAR T Consortium
.
J Clin Oncol.
2020
;
38
(
27
):
3119
-
3128
.
105.
Jacobson
CA
,
Hunter
BD
,
Redd
R
, et al
.
Axicabtagene ciloleucel in the non-trial setting: outcomes and correlates of response, resistance, and toxicity
.
J Clin Oncol.
2020
;
38
(
27
):
3095
-
3106
.
106.
Breinholt
MF
,
Oliveira
DVNP
,
Klausen
TW
, et al
.
High-grade B-cell lymphomas with MYC and BCL2 translocations lack tumor-associated macrophages and PD-L1 expression: a possible noninflamed subgroup
.
Hematol Oncol.
2021
;
39
(
3
):
284
-
292
.
107.
Elbaek
MV
,
Pedersen
,
Breinholt
MF
, et al
.
PD-L1 expression is low in large B-cell lymphoma with MYC or double-hit translocation
.
Hematol Oncol.
2019
;
37
(
4
):
375
-
382
.
108.
Landsburg
DJ
,
Koike
A
,
Nasta
SD
, et al
.
Patterns of immune checkpoint protein expression in MYC-overexpressing aggressive B-cell non-Hodgkin lymphomas
.
Cancer Immunol Immunother.
2021
;
70
(
3
):
869
-
874
.
109.
Viardot
A
,
Goebeler
ME
,
Hess
G
, et al
.
Phase 2 study of the bispecific T-cell engager (BiTE) antibody blinatumomab in relapsed/refractory diffuse large B-cell lymphoma
.
Blood.
2016
;
127
(
11
):
1410
-
1416
.
110.
Olszewski
AJ
,
Avigdor
A
,
Babu
S
, et al
.
Single-agent mosunetuzumab is a promising safe and efficacious chemotherapy-free regimen for elderly/unfit patients with previously untreated diffuse large B-cell lymphoma
.
Blood.
2020
;
136
(
suppl 1
):
43
-
45
.
111.
Advani
R
,
Flinn
I
,
Popplewell
L
, et al
.
CD47 blockade by Hu5F9-G4 and rituximab in non-Hodgkin’s lymphoma
.
N Engl J Med.
2018
;
379
(
18
):
1711
-
1721
.
112.
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
.
113.
Trabucco
SE
,
Gerstein
RM
,
Evens
AM
, et al
.
Inhibition of bromodomain proteins for the treatment of human diffuse large B-cell lymphoma
.
Clin Cancer Res.
2015
;
21
(
1
):
113
-
122
.
Sign in via your Institution