Primary mediastinal large B-cell lymphoma (PMBCL) is a separate entity in the World Health Organization’s classification, based on clinicopathologic features and a distinct molecular signature that overlaps with nodular sclerosis classic Hodgkin lymphoma (cHL). Molecular classifiers can distinguish PMBCL from diffuse large B-cell lymphoma (DLBCL) using ribonucleic acid derived from paraffin-embedded tissue and are integral to future studies. However, given that ∼5% of DLBCL can have a molecular PMBCL phenotype in the absence of mediastinal involvement, clinical information remains critical for diagnosis. Studies during the past 10 to 20 years have elucidated the biologic hallmarks of PMBCL that are reminiscent of cHL, including the importance of the JAK-STAT and NF-κB signaling pathways, as well as an immune evasion phenotype through multiple converging genetic aberrations. The outcome of PMBCL has improved in the modern rituximab era; however, whether there is a single standard treatment for all patients and when to integrate radiotherapy remains controversial. Regardless of the frontline therapy, refractory disease can occur in up to 10% of patients and correlates with poor outcome. With emerging data supporting the high efficacy of PD1 inhibitors in PMBCL, studies are underway that integrate them into the up-front setting.

Primary mediastinal (thymic) large B-cell lymphoma (PMBCL) was initially defined as a subtype of diffuse large B-cell lymphoma (DLBCL) in the Revised European American Lymphoma Classification based on unique clinicopathologic features. Subsequent molecular profiling studies highlighted a characteristic gene expression signature with striking similarities to classic Hodgkin lymphoma (cHL),1,2 and established PMBCL as a distinct entity in the World Health Organization (WHO) classification.3 Recurrent somatic gene mutations, copy number alterations, and structural variants, including those in 9p24.1 and 2p16.1,4-6 leading to JAK-STAT and NFKB constitutive pathway activation, are recognized disease hallmarks. In addition, an immune evasion phenotype predominates and is achieved through multiple converging genetic mechanisms, including structural changes at 9p24.1 leading to expression of PDL (CD274 [PDL1] and, in particular, PDCD1LG12 [PDL2]), as well as structural variants of CIITA, leading to reduced expression of major histocompatibility complex class 2 (MHC 2).7-11 Coupled with advances in the understanding of the biology of PMBCL are studies demonstrating improved outcome using rituximab chemoimmunotherapy, but still ongoing is the controversy about the optimal primary therapy and the role of consolidative radiotherapy (RT). Further, similar to cHL, a strong biological rationale for targeting the PD1-PDL pathway has led to studies evaluating PD1 inhibitors in relapsed/refractory (R/R) PMBCL, with excellent efficacy and drug approval in this setting. This review is focused on the overall unique pathogenetic features of PMBCL and provides a detailed analysis of the current treatment landscape.

PMBCL accounts for ∼2% to 4% of all non-Hodgkin lymphomas (NHLs) and 10% of all large B-cell lymphomas. It occurs mainly in young adults with a median age of ∼35 years and has a female predominance (female/male ratio, 2:1). Patients may present with compressive symptoms caused by the anterior mediastinal mass, and thrombosis, including superior vena cava syndrome, may occur. Most patients have stage I or II disease but may have intrathoracic extension into the lung and/or chest wall or pleural/pericardial effusions.12 Extrathoracic disease can also occur in ∼10% of patients, including rare involvement of the kidney and adrenal gland,13-15 but the predominant site of disease must be in the anterior mediastinum. These sites, as well as the central nervous system (CNS), may be involved at relapse.

Pathology and immunophenotype of PMBCL

Pathologically, PMBCL is typified by a diffuse proliferation of large cells, often compartmentalized by fine bands of sclerosis (Figure 1). It can exhibit a wide morphological/cytological spectrum with variable size and shape of the neoplastic cells, and multilobated cells resembling Hodgkin Reed-Sternberg cells may be present. Of note, the diagnosis can be challenging, given the tumor location, and biopsy specimens may be small with crush artifact. With compartmentalizing fibrosis, PMBCL can mimic a carcinoma or thymoma, and adequate sampling is essential for full immunohistochemical analyses. It must also be distinguished from nodular sclerosis cHL and DLBCL with secondary mediastinal involvement. In some cases, core biopsies may not be adequate to distinguish PMBCL from the related entity, mediastinal gray zone lymphoma (MGZL), and obtaining a larger biopsy specimen is advised.

Figure 1.

Primary mediastinal large B-cell lymphoma. Hematoxylin and eosin staining illustrating the clear cytoplasm of the neoplastic cells and the fibrotic bands without annular distribution that compartmentalize the tumor cells. Tumor cells express CD20 and most express CD30, which is weak and heterogeneous. The arrow depicts a multilobulated Hodgkin Reed Sternberg-like cell. Black bars = 50 μm; gray bar = 20 μm.

Figure 1.

Primary mediastinal large B-cell lymphoma. Hematoxylin and eosin staining illustrating the clear cytoplasm of the neoplastic cells and the fibrotic bands without annular distribution that compartmentalize the tumor cells. Tumor cells express CD20 and most express CD30, which is weak and heterogeneous. The arrow depicts a multilobulated Hodgkin Reed Sternberg-like cell. Black bars = 50 μm; gray bar = 20 μm.

Close modal

In addition to the leukocyte common antigen (CD45), the B-cell lineage antigens CD19, CD20, and CD22 are expressed (Figure 1; Table 1). However, unlike other B-cell lymphomas, PMBCL tumors often lack surface or cytoplasmic immunoglobulin, despite expression of the immunoglobulin co-receptor CD79a. CD10 and CD21 are typically negative, whereas most cases have a MUM1/IRF4+ phenotype with variable BCL6 expression.12 CD30 is positive in ∼80% of cases, but is usually weak and heterogeneous (Figure 1; Table 1), and CD15 is absent. Expression of CD200, CD23, and MAL, a lipid raft component, as well as TRAF1 and nuclear cREL, are characteristic features of PMBCL and distinguish it from DLBCL.16,17 The putative cell of origin is a thymic medullary B-cell that shares a similar phenotype, including MAL expression.18 A commercially available MAL antibody has 100% specificity in PMBCL, with expression in 72% vs 0% in DLBCL, and is superior to CD200 (specificity of 87%).19 Linked with aberrations of 9p24.1 is the expression of PDL1 (also known as B7-H1) and PDL2 in PMBCL tumors, the latter being a more specific marker for tumor cells, as PDL1 is also expressed in tumor-associated macrophages.10,20

Table 1.

Comparison of the clinical, pathologic, and genetic features of PMBCL and related entities

Clinical, pathologic, and genetic featuresPMBCLNonmediastinal PMBCL signature-positive DLBCLMGZLNScHL
Clinical and pathological features     
 Female:Male 2:1 1:1 1:3 1:1 
 Median age (y) ∼35 ∼66 ∼34-40 ∼28 
 Stages I and II, % 70-80 56 65 55 
 Mediastinal presentation, % All None 100 80 
 Bone marrow, % Rare 19 Rare Rare 
 Bulky disease (≥10 cm), % 70-80 30 54 
 Morphology Sheets of large cells; clear cells; no inflammatory polymorphous infiltrate Similar to DLBCL Broad cytologic appearance; may resemble PMBCL (25%); cHL (25%); intermediate (45%); composite (5%) Lacunar Hodgkin Reed-Sternberg cells; inflammatory polymorphous infiltrate 
 Sclerosis, % 70-100 (alveolar, fine bands) Negative Variable 100 (large bands, annular) 
 5-year OS, % ∼90-97 72 (2-year DSS)* ∼75 ∼85 
Immunohistochemistry, %     
 CD45 100 Not reported Positive Negative 
 CD30 70-85 (weak) 31 (weak) ∼85-100 (strong) 100 
 CD15 Negative Negative 58-80 75-85 
 CD20
 CD79a
 PAX-5
 MUM1 
100
100
100
75 
100
Not reported*
Not reported
19 
∼72-98 (weak)
67-75
98
Positive 
Very rare (minority)
Very rare
95 (weak)
100 
 MAL
 CD23 
60-70
85 
57
71 
30-49*
30-49 
19
Negative 
 TRAF1 expression
 Nuclear cRel 
60-70
60-70 
Not reported
Not reported 
Usually positive
Not reported 
84
92 
 PDL1
 PDL2 
71
72 
19
14 
∼80
∼30 
87 
Genetic aberrations, %     
 CIITA rearrangements
 9p.24.1 aberrations (JAK2, PDL)
 2p16.1 (Rel, BCL11A)
SOCS1 mutation/deletion
B2M mutations 
38
70
25-50
35-45
30-64 
Negative
33
Frequent
100
∼15 
30-37
60
33
40
32 
15
25-30
25
42
40 
Clinical, pathologic, and genetic featuresPMBCLNonmediastinal PMBCL signature-positive DLBCLMGZLNScHL
Clinical and pathological features     
 Female:Male 2:1 1:1 1:3 1:1 
 Median age (y) ∼35 ∼66 ∼34-40 ∼28 
 Stages I and II, % 70-80 56 65 55 
 Mediastinal presentation, % All None 100 80 
 Bone marrow, % Rare 19 Rare Rare 
 Bulky disease (≥10 cm), % 70-80 30 54 
 Morphology Sheets of large cells; clear cells; no inflammatory polymorphous infiltrate Similar to DLBCL Broad cytologic appearance; may resemble PMBCL (25%); cHL (25%); intermediate (45%); composite (5%) Lacunar Hodgkin Reed-Sternberg cells; inflammatory polymorphous infiltrate 
 Sclerosis, % 70-100 (alveolar, fine bands) Negative Variable 100 (large bands, annular) 
 5-year OS, % ∼90-97 72 (2-year DSS)* ∼75 ∼85 
Immunohistochemistry, %     
 CD45 100 Not reported Positive Negative 
 CD30 70-85 (weak) 31 (weak) ∼85-100 (strong) 100 
 CD15 Negative Negative 58-80 75-85 
 CD20
 CD79a
 PAX-5
 MUM1 
100
100
100
75 
100
Not reported*
Not reported
19 
∼72-98 (weak)
67-75
98
Positive 
Very rare (minority)
Very rare
95 (weak)
100 
 MAL
 CD23 
60-70
85 
57
71 
30-49*
30-49 
19
Negative 
 TRAF1 expression
 Nuclear cRel 
60-70
60-70 
Not reported
Not reported 
Usually positive
Not reported 
84
92 
 PDL1
 PDL2 
71
72 
19
14 
∼80
∼30 
87 
Genetic aberrations, %     
 CIITA rearrangements
 9p.24.1 aberrations (JAK2, PDL)
 2p16.1 (Rel, BCL11A)
SOCS1 mutation/deletion
B2M mutations 
38
70
25-50
35-45
30-64 
Negative
33
Frequent
100
∼15 
30-37
60
33
40
32 
15
25-30
25
42
40 

MGZL, B-cell lymphoma unclassifiable with features intermediate between diffuse large B-cell lymphoma and classic Hodgkin lymphoma. Estimates for MGZL (vs non-MGZL) are reported where possible.12,20,21,23,42,90-92 Frequencies are provided when available; immunohistochemistry frequencies may vary depending on the thresholds used. For GZL, frequencies vary for cHL-like and large B-cell lymphoma subtypes (eg, MAL and CD23 expression are higher in large B-cell lymphoma type GZL).

NScHL, nodular sclerosing cHL.

*

As a subtype of DLBCL, CD79a, and PAX5 would be expected to show frequencies of 100%. The survival estimate is based on only 16 cases and requires validation.

Of note, GZL is defined in the WHO classification as B-cell lymphoma, unclassifiable, with features intermediate between DLBCL and cHL. Approximately 60% to 70% of cases have mediastinal involvement (MGZL), and a diagnosis of MGZL is made if a tumor has overlapping genotypic and/or phenotypic features of cHL and PMBCL21,22 (Table 1).

Molecular signature of PMBCL

Clinical and pathological similarities between PMBCL and nodular sclerosis cHL have long been appreciated, and their pathogenetic relatedness was further supported by two gene expression–profiling studies that demonstrated striking similarities in their molecular signatures. Genes overexpressed in PMBCL included those involved in the JAK-STAT pathway (IL13RA, JAK2, and STAT1), as well as key components of NF-κB activation (TRAF1 and TFNAIP3), but expression of genes involved in B-cell receptor signaling were decreased.1,2 Interestingly, MGZL (PMBCL-like) has an intermediate gene expression profile between PMBCL and cHL, but a notably strong macrophage signature and downregulation of the B-cell program, which may be linked to poorer outcome.22,23

In practice, PMBCL is a clinicopathologic diagnosis that has inherent subjectivity. Integration of a robust PMBCL molecular classifier into routine diagnostics may guide therapy and would facilitate study comparison. Investigators from the Lymphoma/Leukemia Molecular Profiling Project used formalin-fixed, paraffin-embedded tissue from primary biopsies of PMBCL tumors to develop the Lymph3Cx Nanostring assay, incorporating genes to determine the DLBCL cell of origin24 along with a 30-gene PMBCL molecular classifier, with 24 genes overexpressed in PMBCL and 6 genes overexpressed in DLBCL (Figure 2).25 More recently, an expanded Nanostring-based assay, DLBCL90, combines previous DLBCL Nanostring assays (Lymph3Cx and the double-hit signature),26 as well as the reverse transcriptase multiplex ligation–dependent probe amplification classifier assay similarly distinguish DLBCL by cell of origin and PMBCL27; both have the potential to translate molecular classification into routine practice in PMBCL.

Figure 2.

Heat map of gene expression comparing PMBCL and DLBCL. Each column represents a case, and the discriminating gene features included in Lymph3Cx assay are in rows. The top 6 genes are overexpressed in DLBCL, and the remaining 24 genes show higher expression in PMBCL. Modified from Mottok et al.25 

Figure 2.

Heat map of gene expression comparing PMBCL and DLBCL. Each column represents a case, and the discriminating gene features included in Lymph3Cx assay are in rows. The top 6 genes are overexpressed in DLBCL, and the remaining 24 genes show higher expression in PMBCL. Modified from Mottok et al.25 

Close modal

Genomic landscape and pathogenic hallmarks of PMBCL

Molecular hallmarks of PMBCL include constitutive activation of the JAK-STAT and NF-κB pathways and genetic aberrations that lead to a phenotype of immune evasion, all shared features with cHL (Figure 3A-B). Mechanisms of NF-κB activation include copy number gains of REL (on 2p16.1) and inactivating mutations of negative regulators of NF-κB, including TNFAIP3 and NFKBIE.5,28,29 JAK-STAT pathway activation is achieved through multiple mechanisms, including paracrine activation by interleukin 13 (IL13) receptor–mediated signaling, constitutive activation through loss-of-function mutations in SOCS-1 (suppressor of cytokine signaling) and PTPN1 (protein tyrosine phosphatase), and gain-of-function mutations in STAT6 and IL4R.30-33 Mutations in these genes co-occur in ∼63% of cases, supporting an additive or synergistic cooperation.30 These findings were echoed in two integrative whole-genome sequencing studies where, in comparison with DLBCL,34,35 distinct recurrent genetic alterations emerged in PMBCL including, as expected, genes affected in the NF-κB and JAK-STAT signaling pathways, SOCS1, NFKBIE, and XPO1 (exportin 1)34 (Figure 4). Many of the involved genes were implicated as oncogenic drivers (eg, IL4R) and have also been observed in cHL34,35 (Figure 4).

Figure 3.

Key pathogenic hallmarks of PMBCL and pathway perturbation. (A) JAK-STAT and NFκB are key activation pathways in PMBCL leading to transcriptional deregulation and impaired immunogenicity. (B) Mechanisms of immune evasion in PMBCL, including PDL1/2 expression with resultant T-cell anergy, reduced antigenicity through MHC class 2 downregulation (in part, due to CIITA rearrangements and mutations), MHC class 1 defects caused by B2M mutations, and CD58 mutations/microdeletions leading to impaired T- and NK-cell interactions.8,32,35

Figure 3.

Key pathogenic hallmarks of PMBCL and pathway perturbation. (A) JAK-STAT and NFκB are key activation pathways in PMBCL leading to transcriptional deregulation and impaired immunogenicity. (B) Mechanisms of immune evasion in PMBCL, including PDL1/2 expression with resultant T-cell anergy, reduced antigenicity through MHC class 2 downregulation (in part, due to CIITA rearrangements and mutations), MHC class 1 defects caused by B2M mutations, and CD58 mutations/microdeletions leading to impaired T- and NK-cell interactions.8,32,35

Close modal
Figure 4.

Comparison of mutational frequencies between DLBCL, PMBCL, and cHL. (A) Comparison of mutation frequencies for putative PMBCL driver genes identified in Mottok et al34 and compared with DLBCL (Reddy et al42 and Chapuy et al43). EZH2(*) is the only gene with no significant difference in mutational frequency. (B) Mutational frequencies of PMBCL compared with cHL (Tiacci et al44). Mutational frequencies are comparable for most genes. Statistically different mutation frequencies are identified by an asterisk. Modified and reproduced from Mottok et al34 and, with permission, from Chapuy et al.43 

Figure 4.

Comparison of mutational frequencies between DLBCL, PMBCL, and cHL. (A) Comparison of mutation frequencies for putative PMBCL driver genes identified in Mottok et al34 and compared with DLBCL (Reddy et al42 and Chapuy et al43). EZH2(*) is the only gene with no significant difference in mutational frequency. (B) Mutational frequencies of PMBCL compared with cHL (Tiacci et al44). Mutational frequencies are comparable for most genes. Statistically different mutation frequencies are identified by an asterisk. Modified and reproduced from Mottok et al34 and, with permission, from Chapuy et al.43 

Close modal

These and other studies have also illuminated a diverse array of mechanisms that contribute to the immune escape phenotype of PMBCL. CD274 (PDL1) and PDCD1LG2 (PDL2) are concurrently amplified with JAK2 at 9p24.1, leading to PDL1/2 expression and the resultant T-cell anergy10,36,37 (Figure 3B). Interestingly, STAT-dependent expression of PDL1 has also been identified, supporting the notion of cooperation between these pathways31 (Figure 3A).

Impaired antigen presentation through MHC defects also contributes to the immune-privileged phenotype in PMBCL (Figure 3B). Similar to cHL, downregulation of MHC Class 2 molecules is associated with inferior outcome in PMBCL9 and is linked to genetic aberrations at the CIITA (an MHC Class 2 master transcriptional regulator) locus,8 which in composite, occur in ∼70% of PMBCL cases.38 Mutations in β-2-microglobulin (B2M) are prominent in PMBCL and cHL and lead to impaired MHC class 1–mediated antigen presentation (Figure 3B). Similarly, CD58 mutations, a shared feature with cHL39 and DLBCL,40 are thought to impair interactions with NK and T cells. Mutations in immune response (IRF) genes, including a hotspot mutation IRF4, as well as mutations in downstream targets collectively occur in more than half of PMBCL cases and affect antigen presentation, as well as T-cell, NK-cell, and macrophage responses.34 

DLBCL with a PMBCL-like signature

Rarely, DLBCL cases with a non-mediastinal location can have a molecular signature reminiscent of PMBCL.41 In a recent study, 5.8% of DLBCL cases had a molecular signature of PMBCL in the absence of mediastinal involvement.42 In comparison with bona fide PMBCL, these patients were older (median age, 66 years), with equal sex predominance, but MAL (57%) and CD23 (71%) protein expression were frequent. although CD30 was positive in only 17% of the patients42 (Table 1). Similar to PMBCL, aberrations in genes involving JAK-STAT signaling were common, including, SOCS1 mutations in all cases, and frequent mutations in STAT6, IL4R, and DUSP2 as well as copy number alterations at 9p24.1 with resultant transcript levels in affected genes (eg, PDL1/2, JAK2) was observed. Mutations in genes involved in the immune response were also found (eg, CD8, HLA-A, and HLA-C), and the IL4R mutation location was different from those found in bona fide PMBCL, suggesting alternate mechanisms leading to an immune evasion phenotype.42 Two key studies of DLBCL have defined a new classification that integrates genetic features.43-45 Cluster 4 defined by Chapuy and colleagues, includes recurrent mutations similar to those of non-mediastinal PMBCL signature-positive tumors, except that SOCS1 mutations were not reported, as they did not pass MutsigCV filtering thresholds.43 Lacy and colleagues applied targeted sequencing to define a SOCS/SGK1 DLBCL molecular group that also shared a similar mutation profile, providing further evidence that non-mediastinal PMBCL signature-positive tumors represent a true entity with unique biology, which may have therapeutic implications.46 

Frontline therapy in PMBCL

There are no randomized controlled studies defining the optimal frontline treatment for PMBCL, and as a result, international guidelines vary (supplemental Table 1, available on the Blood Web site). Data are largely derived from non-randomized prospective and retrospective studies, both of which can be subject to selection bias. An acute presentation may necessitate urgent chemotherapy, which may preclude trial enrollment, whereas retrospective studies rely on comprehensive inclusion as well as center diagnosis and may be affected by diverse treatment approaches. Given that PMBCL is a clinicopathologic diagnosis, diagnostic certainty may affect study comparisons, and to date, there have been few studies that applied gold-standard molecular diagnostics. Regardless, because patients often are younger, balancing cure and the potential for long-term toxicities, such as secondary malignancies and cardiovascular complications, are important treatment considerations in PMBCL.

Most studies support the notion that the addition of rituximab to CHOP/CHOP-like (cyclophosphamide, doxorubicin, vincristine, and prednisone) chemotherapy adds a magnitude of benefit similar to that shown in DLBCL.47-50 This was not as clear in a study evaluating R-MACOPB (methotrexate, doxorubicin, cyclophosphamide, prednisolone, and bleomycin with rituximab) or R-VACOPB (etoposide, doxorubicin, cyclophosphamide, vincristine, and prednisone, bleomycin with rituximab), with 69% receiving RT, where historical comparisons to V/MACOPB+RT appeared similar51 (Table 2). In a separate study, there was a trend toward improved progression-free survival (PFS; P = .06) of R-VACOPB over VACOPB but no difference in overall survival (P = .2). In addition, PFS with R-VACOPB was similar to that with CHOP+rituximab (R-CHOP; P = .3)52 (Table 2), suggesting that rituximab may somewhat negate the benefit of dose intensity. None of these studies are definitive, given the small number of patients enrolled. Taking all study results together, R-CHOP or R-CHOP-like therapy yields a 2-year PFS of ∼80% and a 2-year OS of ∼90% in most of the analyses; however, the majority of studies have also incorporated routine mediastinal RT, particularly if CHOP was used (Table 2), which may have long-term secondary complications.

Table 2.

Prospective and retrospective studies evaluating R-CHOP(like) and DA-EPOCHR chemotherapy in PMBCL

PMBCL studies R-chemotherapyStudy size, n; Median age, y (range)Study typePFS/EFS, % (y)OS, % (y)% RTTreatment comparison/study comment
Prospective 
 R-CHOP14/ICE±R
 (MSKCC 01-142) Moskowitz et al
 (abstract), 201093  
54
34 (19-59) 
Prospective + per protocol 78 (3) 88 (3) First 28 patients treated during the study and 26 treated per protocol.*
Planned switch to ICE. 
 R-CHOP/R-CHOEP (MINT study group),
 Rieger et al, 201149  
44
36 (27-43) 
Prospective subgroup 78 (3) 89 (3) 73 R-CHO(E)P vs CHO(E)P
EFS P = .012
OS P = .158
0 or 1 aaIPI factor* 
 DA-EPOCHR (NCI)
 DA-EPOCHR (Stanford)
 Dunleavy et al, 201354  
51
30 (23-51)
16
33 (23-68) 
Prospective
Retrospective 
93 (5)
100 
97 (5)
100 
4
Subset with PMBCL within phase 2 study of  aggressive BCLs.*
Series of PMBCL treated with DA-EPOCHR at Stanford 2007-12 reported with phase 2 study. 
 R-MACOPB/R-VACOPB (84%)
 or R-CHOP IELSG-26
 Martelli et al, 201468  
125
33 (range, NR) 
Prospective
observational 
86 (5) 92 (5) 92 Purpose was to evaluate CR by Deauville criteria.* 
 R-CHOP14/21 (NCI)
 Gleeson et al, 201694  
50
38.5 (22-78) 
Prospective
subgroup 
80 (5) 84 (5) 58 R-CHOP21 vs R-CHOP14
PFS P = .10; OS P = .06
Unadjusted analysis* 
 DA-EPOCH-R (pediatric)
 Burke et al, 202195  
47
15 (range NR) 
Prospective 69 (2) 82 (2) No patients received consolidative RT.* 
 R-CHOP21/14 (UNFOLDER trial)
 Held et al (abstract), 202096 § 
131
34 (range NR) 
Prospective subgroup 93 (3) 97 (3) 62.5 R-CHOP21 vs R-CHOP14 (no difference)
Pooled 3-y event rates;*
RT did not impact PFS (P = .25). 
Retrospective 
 R-V/MACOPB
 Zinzani et al, 200951  
45
38 (17-66) 
Retrospective 84 (5) 80 (5) 71 No difference in R-V/MACOPB vs historical  estimates without R.* 
 R-CHOP
 Ahn et al, 201097  
21
30 (17-79) 
Retrospective 79 (2) 83 (2) 62 R-CHOP vs CHOP
PFS P = .043
OS P = .08 
 R-CHOP
 Vassilakopoulos et al, 201248  
76
31.5 (17-73) 
Retrospective 81 (5) 89 (5) 76 R-CHOP vs CHOP
FFP P < .0001
OS P = .003
Early treatment failure (<6 m), 9%* 
 R-CHOP
 Xu et al, 201350  
39
29 (13-54) 
Retrospective 77 (5) 84 (5) 76 R-CHOP vs CHOP
PFS P = .012
OS P = .011 
 R-CHOP
 Aoki et al, 201415 ǁ 
187
31.5 (17-77) 
Retrospective 71 (4) 90 (4) 34 R-chemo all vs CHOP PFS P = .001
OS P = .001
DA-EPOCHR; 2nd/3rd generation HDC/SCT (vs  R-CHOP no difference)
Treatment selection criteria unknown.* 
 R-CHOP
 Soumerai et al, 201498  
63
37 (20-82) 
Retrospective 68 (5) 79 (5) 77 PD during R-CHOP 18%*
Male predominance (60%)* 
 R-VACOPB
 Avigdor et al, 201452  
40
33 (range NR) 
Retrospective 83 (5) NR R-VACOPB vs VACOPB
PFS P = .06
OS P = .2
R-VACOPB vs R-CHOP
P = .3 
 R-MACOPB
 Zinzani et al, 201566  
74
34 (18-63) 
Retrospective 88 (10) 82 (10) 69 RT vs no RT
P = .85
RT only in PET-positive (IHP).* 
 R-CHOP
 DA-EPOCHR
 Pinnix et al, 201599  
50
35 (19-70)
25 
Retrospective ∼90 (estimate)
83 
NR
NR 
90
20 
R-CHOP vs DA-EPOCHR (and R-HCVAD)
PFS P = .35
Treatment selection criteria unknown.* 
 R-CHOP-RICE
 Goldschmidt et al, 2016100  
24
34 (18-60) 
Retrospective 87 (5) 100 (5) 12 R-CHOP-RICE vs other protocols, R CHOP/R-M/  VACOPB/DA-EPOCHR
PFS P = .35
OS P = .31
78% other protocols received RT.* 
 R-CHOP/R-CHOEP
 Lisenko et al, 201747  
45
38 (19-64) 
Retrospective 95 92 91 R-CHO(E)P vs CHOP
PFS P = .001
OS P = .023 
 DA-EPOCH-R (Adult)
 DA-EPOCHR (Pediatric)
 Giulino-Roth et al, 201714  
118
34 (21-70)
38
16 (9-20) 

Retrospective 
87 (3)
81 (3) 
97 (3)
91 (3) 
16
11 
Adult vs pediatric; EFS P = .34 
 R-CHOP
 DA-EPOCHR
 Shah et al, 201858  
56
37 (20-77)
76
34 (18-69) 
Retrospective 76 (2)
85 (2) 
89 (2)
91 (2) 
59
13 
R-CHOP vs DA-EPOCHR
PFS P = .28
OS P = .83
Treatment selection criteria unknown.* 
 R-CHOP
 Malenda et al, 202059  
25
37 (18-80) 
Retrospective 87 (1) 100 (1) 25 R-CHOP vs DA-EPOCHR
PFS P = .20
OS P = .66
Treatment selection criteria unknown.* 
 R-CHOP
 R-CHOP + RT
 Messmer et al, 2019101  
16
36 (23-52)
10
40.5 (19-60) 
Retrospective 93 (3)
100 (3) 
100 (3)
100 (3) 
0
100 
RT vs no RT
P = .85
RT criteria unknown.* 
 R-CHOP
 R-CHOP + RT
 DA-EPOCHR
 Chan et al, 2019102  
41
28 (11-72)
37
26 (14-48)
46
27 (16-51) 
Retrospective 56.5(5)
88 (5)
90 (5) 
76 (5)
94 (5)
94 (5) 
0
100
R-CHOP vs DA-EPOCHR
PFS P = .012
OS P = .01
RT vs no RT with R-CHOP
PFS P = .04
Treatment selection criteria unknown.* 
 R-CHOP
 Zhou et al, 202057 # 
89
33 (11-64) 
Retrospective ∼60 (5) estimate ∼70 (5) estimate 84 (5) R-CHOP vs DA-EPOCHR/R-HCVAD
PFS P = .048
OS P = .0067
Treatement selection criteria unknown.* 
 R-CHOP
 Hayden et al, 202013  
159
36 (19-84) 
Retrospective 80 (5) 89 (5) 28 PET-guided use of consolidative RT (no difference in outcome vs routine RT).* 
 R-CHOP14/21
 R-CHOEP14
 Wästerlid et al, 2021103 ** 
17
49 (18-83)
90
35 (18-74) 
Registry NR 74 (5) RS
95 (5) RS 
14
18 
R-CHOEP14/R-HyperCVAD/DA-EPOCHR
Treatment selection criteria unknown (not formally compared)* 
PMBCL studies R-chemotherapyStudy size, n; Median age, y (range)Study typePFS/EFS, % (y)OS, % (y)% RTTreatment comparison/study comment
Prospective 
 R-CHOP14/ICE±R
 (MSKCC 01-142) Moskowitz et al
 (abstract), 201093  
54
34 (19-59) 
Prospective + per protocol 78 (3) 88 (3) First 28 patients treated during the study and 26 treated per protocol.*
Planned switch to ICE. 
 R-CHOP/R-CHOEP (MINT study group),
 Rieger et al, 201149  
44
36 (27-43) 
Prospective subgroup 78 (3) 89 (3) 73 R-CHO(E)P vs CHO(E)P
EFS P = .012
OS P = .158
0 or 1 aaIPI factor* 
 DA-EPOCHR (NCI)
 DA-EPOCHR (Stanford)
 Dunleavy et al, 201354  
51
30 (23-51)
16
33 (23-68) 
Prospective
Retrospective 
93 (5)
100 
97 (5)
100 
4
Subset with PMBCL within phase 2 study of  aggressive BCLs.*
Series of PMBCL treated with DA-EPOCHR at Stanford 2007-12 reported with phase 2 study. 
 R-MACOPB/R-VACOPB (84%)
 or R-CHOP IELSG-26
 Martelli et al, 201468  
125
33 (range, NR) 
Prospective
observational 
86 (5) 92 (5) 92 Purpose was to evaluate CR by Deauville criteria.* 
 R-CHOP14/21 (NCI)
 Gleeson et al, 201694  
50
38.5 (22-78) 
Prospective
subgroup 
80 (5) 84 (5) 58 R-CHOP21 vs R-CHOP14
PFS P = .10; OS P = .06
Unadjusted analysis* 
 DA-EPOCH-R (pediatric)
 Burke et al, 202195  
47
15 (range NR) 
Prospective 69 (2) 82 (2) No patients received consolidative RT.* 
 R-CHOP21/14 (UNFOLDER trial)
 Held et al (abstract), 202096 § 
131
34 (range NR) 
Prospective subgroup 93 (3) 97 (3) 62.5 R-CHOP21 vs R-CHOP14 (no difference)
Pooled 3-y event rates;*
RT did not impact PFS (P = .25). 
Retrospective 
 R-V/MACOPB
 Zinzani et al, 200951  
45
38 (17-66) 
Retrospective 84 (5) 80 (5) 71 No difference in R-V/MACOPB vs historical  estimates without R.* 
 R-CHOP
 Ahn et al, 201097  
21
30 (17-79) 
Retrospective 79 (2) 83 (2) 62 R-CHOP vs CHOP
PFS P = .043
OS P = .08 
 R-CHOP
 Vassilakopoulos et al, 201248  
76
31.5 (17-73) 
Retrospective 81 (5) 89 (5) 76 R-CHOP vs CHOP
FFP P < .0001
OS P = .003
Early treatment failure (<6 m), 9%* 
 R-CHOP
 Xu et al, 201350  
39
29 (13-54) 
Retrospective 77 (5) 84 (5) 76 R-CHOP vs CHOP
PFS P = .012
OS P = .011 
 R-CHOP
 Aoki et al, 201415 ǁ 
187
31.5 (17-77) 
Retrospective 71 (4) 90 (4) 34 R-chemo all vs CHOP PFS P = .001
OS P = .001
DA-EPOCHR; 2nd/3rd generation HDC/SCT (vs  R-CHOP no difference)
Treatment selection criteria unknown.* 
 R-CHOP
 Soumerai et al, 201498  
63
37 (20-82) 
Retrospective 68 (5) 79 (5) 77 PD during R-CHOP 18%*
Male predominance (60%)* 
 R-VACOPB
 Avigdor et al, 201452  
40
33 (range NR) 
Retrospective 83 (5) NR R-VACOPB vs VACOPB
PFS P = .06
OS P = .2
R-VACOPB vs R-CHOP
P = .3 
 R-MACOPB
 Zinzani et al, 201566  
74
34 (18-63) 
Retrospective 88 (10) 82 (10) 69 RT vs no RT
P = .85
RT only in PET-positive (IHP).* 
 R-CHOP
 DA-EPOCHR
 Pinnix et al, 201599  
50
35 (19-70)
25 
Retrospective ∼90 (estimate)
83 
NR
NR 
90
20 
R-CHOP vs DA-EPOCHR (and R-HCVAD)
PFS P = .35
Treatment selection criteria unknown.* 
 R-CHOP-RICE
 Goldschmidt et al, 2016100  
24
34 (18-60) 
Retrospective 87 (5) 100 (5) 12 R-CHOP-RICE vs other protocols, R CHOP/R-M/  VACOPB/DA-EPOCHR
PFS P = .35
OS P = .31
78% other protocols received RT.* 
 R-CHOP/R-CHOEP
 Lisenko et al, 201747  
45
38 (19-64) 
Retrospective 95 92 91 R-CHO(E)P vs CHOP
PFS P = .001
OS P = .023 
 DA-EPOCH-R (Adult)
 DA-EPOCHR (Pediatric)
 Giulino-Roth et al, 201714  
118
34 (21-70)
38
16 (9-20) 

Retrospective 
87 (3)
81 (3) 
97 (3)
91 (3) 
16
11 
Adult vs pediatric; EFS P = .34 
 R-CHOP
 DA-EPOCHR
 Shah et al, 201858  
56
37 (20-77)
76
34 (18-69) 
Retrospective 76 (2)
85 (2) 
89 (2)
91 (2) 
59
13 
R-CHOP vs DA-EPOCHR
PFS P = .28
OS P = .83
Treatment selection criteria unknown.* 
 R-CHOP
 Malenda et al, 202059  
25
37 (18-80) 
Retrospective 87 (1) 100 (1) 25 R-CHOP vs DA-EPOCHR
PFS P = .20
OS P = .66
Treatment selection criteria unknown.* 
 R-CHOP
 R-CHOP + RT
 Messmer et al, 2019101  
16
36 (23-52)
10
40.5 (19-60) 
Retrospective 93 (3)
100 (3) 
100 (3)
100 (3) 
0
100 
RT vs no RT
P = .85
RT criteria unknown.* 
 R-CHOP
 R-CHOP + RT
 DA-EPOCHR
 Chan et al, 2019102  
41
28 (11-72)
37
26 (14-48)
46
27 (16-51) 
Retrospective 56.5(5)
88 (5)
90 (5) 
76 (5)
94 (5)
94 (5) 
0
100
R-CHOP vs DA-EPOCHR
PFS P = .012
OS P = .01
RT vs no RT with R-CHOP
PFS P = .04
Treatment selection criteria unknown.* 
 R-CHOP
 Zhou et al, 202057 # 
89
33 (11-64) 
Retrospective ∼60 (5) estimate ∼70 (5) estimate 84 (5) R-CHOP vs DA-EPOCHR/R-HCVAD
PFS P = .048
OS P = .0067
Treatement selection criteria unknown.* 
 R-CHOP
 Hayden et al, 202013  
159
36 (19-84) 
Retrospective 80 (5) 89 (5) 28 PET-guided use of consolidative RT (no difference in outcome vs routine RT).* 
 R-CHOP14/21
 R-CHOEP14
 Wästerlid et al, 2021103 ** 
17
49 (18-83)
90
35 (18-74) 
Registry NR 74 (5) RS
95 (5) RS 
14
18 
R-CHOEP14/R-HyperCVAD/DA-EPOCHR
Treatment selection criteria unknown (not formally compared)* 

Estimates are rounded; complete remission.

aaIPI, age-adjusted International Prognostic Index; CVAD, cyclophosphamide, vincristine, doxorubicin, dexamethasone; HCVAD hyper-CVAD; ICE, ifosfamide, carboplatin, and etoposide; IHP International Harmonization Project; NCI, National Cancer Institute; NR not reported; RS, relative survival; Tx, treatment.

*

Comment on treatment comparison.

R-CHOP (n=21), RCHOEP (n=23), RCHOP/RCHOEP (n=1).

Median age for all patients.

§

Abstract only; P-values not provided.

ǁ

The study included 345 patients (CHOP [n = 47]; R-CHOP [n = 187]; DA-EPOCHR [n = 9]; second/third generation [n = 45]; high-dose chemotherapy/ASCT [n = 57]).

Median age for all patients; outcome for cohort: 5-year PFS 91%, 5-year OS 99%; PFS estimated for R-CHOP.

#

Median age for all patients; 19 patients were treated with HCVAD (vs R-CHOP PFS, P = .0088; OS, P = .095).

**

R-CHOP (n = 3); R-CHOP14 (n = 14); not shown DA-EPOCHR (n = 11); 5-year RS, 82%; R-HCVAD (n = 16), 5-year RS 100%.

DA-EPOCHR (dose-adjusted etoposide, prednisone, vincristine, cyclophosphamide, doxorubicin, rituximab) was developed for aggressive lymphomas to overcome drug resistance and integrates three important modifications over R-CHOP: (1) the addition of etoposide with added activity and synergy; (2) continuous infusion of etoposide, vincristine, and doxorubicin, with in vitro data supporting less drug resistance; and (3) individualized dose adjustment based on the previous cycle neutrophil nadir with growth factor support.53 The subset of 51 patients classified as PMBCL enrolled in the phase 2 National Cancer Institute (NCI) study of DA-EPOCHR in aggressive lymphomas, demonstrated excellent outcomes, with a 5-year event-free survival (EFS) of 93% and OS of 97%, with only 2 patients receiving consolidative RT (Table 2).54 In the same report, a retrospective series of 16 PMBCL patients from Stanford treated with DA-EPOCHR was included, and with a median follow-up of 37 months, all patients were alive and event free.54 In the largest retrospective series of DA-EPOCHR treated PMBCL patients, Guilino-Roth et al evaluated156 patients, most of whom were adults (n = 118)14 (Table 1). For all patients, the 3-year EFS was 86%, and OS was 95%. Considering only adult patients (≥21 years, with a median age of 34 years [range, 21-70]), the 5-year EFS was 87.4%, and the OS was 97%, with 16% having received mediastinal RT. Pediatric patients had a 3-year EFS of 81% and 91% (vs adult patients, P = .34) but were more likely to have dose escalation to at least level 4 (54% vs 33%; P = .03). The authors concluded that DA-EPOCHR is a reasonable approach for PMBCL of all ages. In contrast, in a phase 2 of the Children’s Oncology Group (COG) and European Intergroup for Childhood Non-Hodgkin Lymphoma of 46 pediatric/adolescent patients with PMBCL (median age, 15.4 years) were treated with DA-EPOCHR and demonstrated a 2-year EFS of 69.6% and OS of 82%,55 with 48% advancing to at least dose level 4. In addition, 41% received pre-phase COP (cyclophosphamide, vincristine, and prednisolone) 1 week before the study.55 The recently reported CALGB 50303 phase 3 study comparing DA-EPOCHR to R-CHOP as a frontline therapy for DLBCL, including morphologic subtypes and PMBCL, failed to show a benefit of the intensive regimen. For the 35 patients with PMBCL, PFS events were similar, (DA-EPOCHR, 2 of 15 [13%] vs R-CHOP 3 of 20 [15%]), with no patients receiving RT; but, importantly, the study was not designed to evaluate subtypes, and the overall trial results cannot be applied to PMBCL56 (Table 2).

Three retrospective studies have compared outcomes of therapy with DA-EPOCHR and R-CHOP, typically with RT, in PMBCL, with one demonstrating superiority of DA-EPOCHR,57 and the others showing no difference.58,59 Interpretation remains challenging, as the selection criteria for each regimen is unclear, and the studies are underpowered to detect small differences. In the CALGB 50303 study, given that DA-EPOCHR was designed to push chemotherapy doses, it was associated with increased acute hematologic toxicity, including febrile neutropenia, compared with R-CHOP (35% vs 18%; P < .001), and also was noted to have higher frequencies of non-hematologic toxicities, including high-grade neuropathy (grades 3 and 4, 10.8% vs 3.3%).56 A separate study demonstrated that 40% of patients with lymphoma treated with DA-EPOCHR had central line complications, including extravasation from port insertion, and the researchers endorsed a peripherally inserted central catheter (PICC) for that reason.60 Collectively, the 2-year PFS is 81% to 93% and the 2-year OS is 92% to 97%, in adult patients with PMBCL treated with DA-EPOCHR (Table 2). Although cross-trial comparisons remain limited, there is likely a proportion of patients in whom DA-EPOCHR is warranted, especially those with aggressive presentations, including advanced stage with extrathoracic involvement.

Consolidative RT and use of positron emission tomographic scan in PMBCL

Consolidative mediastinal RT is still frequently delivered after R-CHOP; however, it is unclear whether RT affects the relapse risk or impacts OS, especially for those in complete remission (CR). Retrospective studies have shown conflicting results and are subject to bias caused by variable inclusion criteria, and RT was not administered by uniform criteria. For example, a SEER study of 250 patients with stages I or II PMBCL diagnosed from 2001 through 2012 reported a superior 5-year OS in patients who underwent RT vs those who did not (90% vs 79%; P = .029).61 However, treatment and response data are unknown, and earlier patients may not have received rituximab, which was only approved by the US Food and Drug Administration (FDA) in 2006. In contrast, a separate SEER study of 258 cases, including all stages of PMBCL (diagnosed 2006 through 2011) failed to show a difference (5-year OS, treated with RT 82.5% vs nontreated 78.6%; P = .47), despite the no-RT group having a numerically higher proportion of patients with advanced-stage disease (31 vs 19%).62 

One key benefit of DA-EPOCHR is less reliance on mediastinal RT. Although the long-term effects of modern RT doses remain unknown,63 especially if guided by positron emission tomography (PET), avoidance of RT is relevant in younger patients, given the potential for long-term cardiovascular complications and secondary malignancies, as shown in Hodgkin lymphoma.64,65 Few studies have evaluated the outcome using R-CHOP or R-CHOP–like therapies alone to treat PMBCL and whether a PET scan can be used to guide the use of RT. This approach was first evaluated after MACOPB, where a similar 10-year PFS (∼90%; P = .85) was reported, regardless of end-of-treatment (EOT) PET scan status, with only those with a positive scan (by the International Harmonization Project) receiving RT.66 The International Extranodal Lymphoma Study Group (IELSG) evaluated EOT response by PET scan using the more granular Deauville (D) 5-point scale67 after R-VACOPB/R-MACOPB chemotherapy and consolidative RT (IELSG26)68 (Table 3). This study established that D3 (FDG uptake greater than the mediastinum but less than in the liver) should be included as a complete metabolic response at EOT.68 We recently reported the outcome of PMBCL in British Columbia, applying Provincial Lymphoma Tumor Group–endorsed guidelines, whereby an EOT PET scan was used to guide consolidative RT after R-CHOP therapy. Patients with an EOT PET-positive scan were referred for RT, and those with a negative PET scan were observed, regardless of initial disease bulk.13 Overall, for the 113 PET-evaluable population, the 5-year time to progression (TTP) was 82% and the 5-year OS was 94%, with 99% of patients with EOT PET-negative scans receiving chemotherapy alone. For the patients with available Deauville-scored PET scans, the PET-negative (D1-3, DX) and PET-positive (D4, D5) rates were 71% and 29%, respectively (Table 3). In the PET-negative cases, the 5-year TTP and OS were 91% and 97%, respectively, and ultimately, only 11% received RT as a result of reassignment of earlier scans according to the Deauville criteria (Figure 5; Table 3). The negative predictive value was also high in other studies after R-CHOP without RT (92% to 97%)69,70 or DA-EPOCHR (95% to 100%)14,71,72 (Figure 5; Table 3) and increased further if extramediastinal recurrence, including CNS relapse, were excluded.13,69 Larger studies are needed to determine whether the negative predictive value is higher with DA-EPOCHR than with R-CHOP. Importantly, the IELSG37 phase 3 trial is evaluating the role of consolidative RT in PMBCL with an EOT PET-negative scan after investigator’s-choice rituximab chemotherapy, but has not yet been reported (registered at www.clincaltrials.com as NCT01599559).

Figure 5.

Outcome of PMBCL by EOT PET Deauville score. Outcomes after R-CHOP (A) and after DA-EPOCHR (B-C). Modified and reproduced from Hayden et al,13 Pinnix et al,71 and, with permission, from Guilino-Roth et al.14 

Figure 5.

Outcome of PMBCL by EOT PET Deauville score. Outcomes after R-CHOP (A) and after DA-EPOCHR (B-C). Modified and reproduced from Hayden et al,13 Pinnix et al,71 and, with permission, from Guilino-Roth et al.14 

Close modal
Table 3.

Studies evaluating outcomes in PMBCL by EOT PET scan using Deauville criteria

StudyTreatmentFrequency EOT PET-neg/PET-pos %PET-neg D1-3 PFS, % (y)PET-pos D4-D5 PFS, % (y)Frequency PET pos D4, %PET-pos D4 PFS, % (y)Frequency PET-pos D5, %PET-pos D5 PFS, %(y)
Martelli et al, 201468 * R-MACOPB 70/30 99 (5) 68 (5) 21 NR NR 
Vassilakopoulos et al, 2016 69  R-CHOP 71/29 96 (3) 61 (3) 15 65 (3) 14 57 (3) 
Giulino-Roth et al, 201714  DA-EPOCHR 75/25 95 (3) 55 (3) 15 ∼78 (3) 11 ∼30 (3) 
Melani et al, 201872 § DA-EPOCHR 69/31 96 (8) 71 (8) 21 NR 10 50 (8) 
Pinnix et al, 201871 ǁ DA-EPOCHR 62/38 100 (2) 51 (2) NR NR 14 22 (2) 
Hayden et al, 202013  R-CHOP 71/29 92 (5) 68 (5) 17 87 (5) 12 33 (5) 
Vassilakopoulos et al, 202170 # R-CHOP 73/27 92-97 (5) NR 16 82 (5) 11 44 (5) 
StudyTreatmentFrequency EOT PET-neg/PET-pos %PET-neg D1-3 PFS, % (y)PET-pos D4-D5 PFS, % (y)Frequency PET pos D4, %PET-pos D4 PFS, % (y)Frequency PET-pos D5, %PET-pos D5 PFS, %(y)
Martelli et al, 201468 * R-MACOPB 70/30 99 (5) 68 (5) 21 NR NR 
Vassilakopoulos et al, 2016 69  R-CHOP 71/29 96 (3) 61 (3) 15 65 (3) 14 57 (3) 
Giulino-Roth et al, 201714  DA-EPOCHR 75/25 95 (3) 55 (3) 15 ∼78 (3) 11 ∼30 (3) 
Melani et al, 201872 § DA-EPOCHR 69/31 96 (8) 71 (8) 21 NR 10 50 (8) 
Pinnix et al, 201871 ǁ DA-EPOCHR 62/38 100 (2) 51 (2) NR NR 14 22 (2) 
Hayden et al, 202013  R-CHOP 71/29 92 (5) 68 (5) 17 87 (5) 12 33 (5) 
Vassilakopoulos et al, 202170 # R-CHOP 73/27 92-97 (5) NR 16 82 (5) 11 44 (5) 

Estimates (∼) are rounded. Frequencies of D4 and D5 reflect proportions in the entire cohort.

EOT, end of treatment; NR, not reported; neg, negative; PD, progressive disease; PFS progression free survival; pos, positive.

*

EOT PET scan for central review was performed on 115 patients (4 with early PD excluded); RT in 94% of PET-neg; PFS estimates NR separately in D4 and D5; however, in categories D4, 5 of 24 (21%), and in D5, and 6 of 10 (60%) patients relapsed/progressed.

Included only responding patients with PMBCL; RT in 49%; 12% received R-CHOP14; FFP reported.

Excluded 2 patients with early PD before EOT PET scan.

§

Survival estimates not reported separately for D4; however, 1 of 17 relapsed/progressed patient (6%) had EFS.

ǁ

Included only patients with pre-PET and EOT PET scans.

Excluded 1 patient with early PD before EOT PET; RT in 11% (due to reassignment of earlier scans according to Deauville criteria; 2 patients had DA-EP OCHR; TTP reported).

#

Extended results of prior study: 231 responding patients; PET-neg, RT in 72%, RT (D1-2 47% vs D3 96%; 5-year FFP 100% vs 92%; mediastinal only 100% vs 96%. P = .16); PET-pos RT in 89% (D4 97% vs D5 80%); FFP reported.

In contrast, for the ∼30% of patients with a positive EOT PET scan, the positive predictive value (PPV) was very low (Table 3). However, this is highly driven by the EOT Deauville score with the PPV of an EOT score of D4 notably poor (Figure 5), regardless of frontline therapy; however, outside of studies using DA-EPOCHR, most patients received RT. In the BC Cancer study of primarily R-CHOP–treated PMBCL, overall, the PET-positive cases (D4, D5) had an inferior 5-year TTP (68%) compared with PET-negative cases but it was far superior in D4 vs D5 cases where the 5-year TTP was 87% vs 33%, respectively. The D4 EOT PET-positive cases had outcomes similar to those of the PET-negative cases and, although 4 patients were observed without subsequent relapse, overall, 72% underwent RT.13 Of note, in this study, an EOT PET-positive scan was more frequent in those with bulky disease at diagnosis and led to a greater proportion receiving consolidative RT (33% [bulky] vs 16% [non-bulky]; P = .06). A separate study from Greece demonstrated a less favorable outcome of D4 (3-year freedom from progression [FFP], 65%; Table 3) after R-CHOP, where 15 of 16 patients received RT, and the investigators found that a maximum standardized uptake value (SUV) of ≥5 was predictive of relapse. An extension of this study to include 231 patients with PMBCL treated with R-CHOP by EOT PET using the Deauville criteria,, and noted a 5-year FFP of 82% in D4 patients, of which 28 of 29, had received RT. After DA-EPOCHR therapy, favorable outcomes are consistently observed in D4 cases, with most not including RT.14,71,72 In an expanded study of 93 patients from the NCI phase 2 study and Stanford series, only 1 of 17 patients with a D4 EOT scan relapsed and none received RT.72 Taken together, the data support observation with serial PET scans after DA-EPOCHR with a D4 score. However, further studies are needed to determine whether this approach is appropriate after R-CHOP, with repeat biopsy favored when possible.

In contrast to EOT D4 scans, for the 11% to 14% of patients with a EOT D5 scan (Table 3), the PPV is much higher, and outcome is poor, regardless of frontline therapy, with a 5-year TTP/PFS of 22% to 57%13,14,69,71 (Figure 5), with consolidative RT less effective in one study (5-year TTP 92% D4 vs 57% D5).13 The variability in outcome may be related to the inclusion of both marked fluorodeoxyglucose (FDG) uptake and new lesions (ie, progressive disease) in the D5 definition. Regardless, events are typically early, and most of the patients have refractory disease. It is important also to note that some PET-focused studies included only responding patients,70,73 and others may have excluded a minority of very high-risk patients who may have progressed at an earlier time point (Table 5).

In general, most R-CHOP-like–treated patients can be effectively treated without RT; however, overall, RT use is still more frequent than with DA-EPOCH, in part because of the lack of certainty of whether a D4 score represents false-positive disease. In younger patients with bulky disease, DA-EPOCHR may still be preferred in centers with expertise, as it largely avoids mediastinal RT.

PET radiomics in PMBCL

PET radiomics has emerged as an evolving field whereby baseline quantitative 18FDGPET/computed tomographic (CT) parameters can be important predictors of outcome. Volume-based metabolic assessment including maximum standard uptake volume, metabolic tumor volume, and total lesion glycolysis (TLG) were correlated with prognosis in patients in the IELSG study in those with staging PET scans available (n = 103).74 TLG, which combines tumor volume and metabolism on baseline PET/CT scan, emerged as the most robust predictor of outcome, where the 5-year PFS was 64% vs 99% in those with a high vs low TLG, respectively, with similar findings in a separate study using DA-EPOCHR (high TLG, 2-year PFS 60% vs low TLG, 2-year PFS 95%; P = .006).71 Building on this result, the IELSG evaluated metabolic heterogeneity (MH), and found that, in multivariate analysis, only MH and TLG were associated with a shorter PFS, and combining these 2 factors identified 10% of all patients with a 5-year PFS of only 11%75 (Figure 6). Given the challenges of applying clinical risk models to PMBCL, this approach represents a potential means of identifying a subset that may need an alternate treatment approach.

Figure 6.

PFS by combined MH (metabolic heterogeneity) and TLG (total lesion glycolysis). Low risk (MH and TLG low); intermediate risk (either low MH and high TLG or high MH and low TLG); high risk (high MH and high TLG). Modified and reproduced from Ceriani et al.75 

Figure 6.

PFS by combined MH (metabolic heterogeneity) and TLG (total lesion glycolysis). Low risk (MH and TLG low); intermediate risk (either low MH and high TLG or high MH and low TLG); high risk (high MH and high TLG). Modified and reproduced from Ceriani et al.75 

Close modal

Relapsed/refractory PMBCL

Almost all treatment failures in PMBCL occur within the first 2 years, with the exception of rare, late CNS relapse, which occurs in ∽2.5% to 4.5% overall.13,15,71 Management of relapsed/refractory (R/R) PMBCL parallels DLBCL, where salvage therapy is administered, followed by high-dose chemotherapy and autologous stem cell transplant (ASCT), if a response is documented. There have been limited studies evaluating the outcome of R/R PMBCL, particularly in the rituximab treatment era, and very few have evaluated the intent-to-transplant (ITTx) population (Table 4). An earlier study of 37 patients with R/R PMBCL from Princess Margaret Cancer Centre (PMCC), almost entirely in the era before rituximab, demonstrated a transplant rate of only 22% and 2-year OS of only 15% in ITTx R/R PMBCL. For the transplant recipients, the 2-year OS was 67%. In contrast, a recent study of 51 patients with R/R PMBCL from Memorial Sloan Kettering Cancer Centre (MSKCC) demonstrated an 85% transplant rate and a 3-year PFS and OS of 57% and 61% in the ITTx population, respectively, with only slightly better rates in those who underwent transplant (60% and 65%, respectively).76 The investigators credited the excellent outcomes to the integration of mediastinal RT in 90% of patients before transplant.76 Outcomes were also more favorable after ASCT in other series with 5-year OS 57% to 70%,13,77,78 but a 5-year OS of only 48% in the ITTx group, suggesting salvage failure in some cases13 (Table 4). Overall, patients with refractory disease seem to fare worse and may benefit from alternate treatment approaches.

Table 4.

Select studies evaluating the outcome of relapsed/refractory PMBCL

Studyn (% rituximab primary treatment) Year of primary diagnosisOutcomes in intention to transplantPost-ASCT outcomes% Salvage RT pre- vs post- ASCT
PFS, % (y)OS, % (y)ASCT rate, %PFS/EFS, % (y)OS, % (y)
Kuruvilla et al, 2005104  37
3
1995-2004 
15 (2) 15 (2)
Rel 29 (2)
Ref 31 (2) 
22 57 (2) 67 (2) NR 
Aoki et al, 201577  44
66
1996-2012 
NR NR NR 61 (4)* 70 (4)*
Rel 73 (4)
Ref 65 (4) 
NR 
Avivi et al, EBMT, 201878  86
85
2000-2012 
NR NR NR 62 (5)
Rel 64 (5)
Ref 39 (5) 
71 (5)
77 (5)
41 (5) 
30 pre 
Vardhana et al, 201876  60
40
1989-2014 
57 (3) 61 (3) 85 60 (3) 65 (3) 90 pre 
Hayden et al, 202013  31
100
2001-2018 
NR 48 (5) Rel 78
Ref 75 
NR 57 (5)
Rel 71 (5)
Ref 50 (5) 
36 post 
Studyn (% rituximab primary treatment) Year of primary diagnosisOutcomes in intention to transplantPost-ASCT outcomes% Salvage RT pre- vs post- ASCT
PFS, % (y)OS, % (y)ASCT rate, %PFS/EFS, % (y)OS, % (y)
Kuruvilla et al, 2005104  37
3
1995-2004 
15 (2) 15 (2)
Rel 29 (2)
Ref 31 (2) 
22 57 (2) 67 (2) NR 
Aoki et al, 201577  44
66
1996-2012 
NR NR NR 61 (4)* 70 (4)*
Rel 73 (4)
Ref 65 (4) 
NR 
Avivi et al, EBMT, 201878  86
85
2000-2012 
NR NR NR 62 (5)
Rel 64 (5)
Ref 39 (5) 
71 (5)
77 (5)
41 (5) 
30 pre 
Vardhana et al, 201876  60
40
1989-2014 
57 (3) 61 (3) 85 60 (3) 65 (3) 90 pre 
Hayden et al, 202013  31
100
2001-2018 
NR 48 (5) Rel 78
Ref 75 
NR 57 (5)
Rel 71 (5)
Ref 50 (5) 
36 post 

Outcomes are for >1 CR/PR and refractory disease.

NR, not reported; ref, refractory; rel, relapsed.

*

Five patients underwent allo-SCT.

Includes 16 patients who underwent transplant in first remission (included in overall PFS and OS estimate only).

New approaches in relapsed/refractory PMBCL

PD1 inhibitors in PMBCL

Proof of principle of the efficacy of PD1 inhibitors in PMBCL first came from the phase 1 KEYNOTE-013 study, which evaluated pembrolizumab across cohorts with R/R lymphoma, including PMBCL.79 In the updated analysis, the objective response rate (ORR) in R/R PMBCL was 48% (CR 33%), and similar results were observed in the subsequent phase 2 study, the KEYNOTE-170 trial, with an ORR of 45% and a CR of 13%. With a median follow-up of 29.1 months (KEYNOTE-013) and 12.5 months (KEYNOTE-170), the median PFS was 10.4 and 5.5 months, respectively, and the median duration of response (DOR) was not yet reached in either study.80 Pembrolizumab was used as a bridge to transplant in 9 patients. Grade 3/4 treatment-related adverse events (AEs) occurred in 23% of patients, and neutropenia was the most common high-grade AE (13.5%). This data led to FDA approval of pembrolizumab in R/R PMBCL for the treatment of adult and pediatric patients who have relapsed after 2 or more lines of therapy.

The PD1 inhibitor nivolumab was combined with the anti-CD30 antibody drug conjugate brentuximab vedotin (BV) in the CheckMate 436 study, which included a cohort of patients with R/R PMBCL.81 Despite an ORR of only 13% in a prior study with BV alone in R/R PMBCL,82 the ORR was 73% and CR was 37%, which reflects the potential synergy of PD1 therapy combined with other systemic agents.81 Half of the responding patients (n = 11) underwent subsequent SCT and none have relapsed. Grade 3 and 4 treatment-related AEs were higher (53%) than with single-agent PD1 inhibitor, with 30% neutropenia and 10% peripheral neuropathy (27% overall). IRAEs were reported in 10 patients (grade 3; n = 3). Studies are underway integrating PD1 inhibitors in the up-front setting in PMBCL (supplemental Table 2).

Cellular therapies in relapsed/refractory PMBCL

CAR T-cell therapy is a form of cellular therapy using patients’ autologous T lymphocytes that are genetically engineered to express an anti-CD19 single-chain fragment on the cell surface, linked to costimulatory proteins required for T-cell activation. CAR T-cell therapy has changed the landscape of treatment of R/R CD19+ aggressive large B-cell lymphomas, including PMBCL, with seemingly curative potential at a stage of disease where the OS is typically <6 months.83 The ZUMA-1 and TRANSCEND NHL001 studies tested axicabtagene ciloleucel (axi-cel) and tisagenlecleucel products, respectively, in R/R aggressive large B-cell lymphomas (including PMBCL) after failure of at least 2 lines of therapy.84 Response assessment in the ZUMA-1 study initially grouped PMBCL (n = 8) with transformed follicular lymphoma, but regardless, the ORR was 71% with CR of 12% in the initial report.84 For all 119 patients in the study, the ORR was 83%, with 58% CR and a median PFS and DOR of 5.6 and 11.1 months, respectively, and the 2-year PFS was ∼40%.85 In a subsequent report with a median follow-up of 27 months, ongoing responses were maintained in 5 of 8 cases of PMBCL.85 Similarly, in the TRANSCEND study the ORR was 73% and CR was 53% for all enrolled patients, and in the 15 patients with R/R PMBCL (14 evaluable), ORR was 79% (CR not stated). Median PFS was 6.8 months for all patients and 1-year PFS was 44% in all patients, 65% for patients in CR and median DOR not yet reached.86 These data led to FDA approval of both agents and, if available, CAR T-cell therapy is the preferred option in R/R PMBCL after failure of 2 lines of therapy, including ASCT.

Other novel therapies/treatment approaches

Given that most cases of PMBCL are CD30+, they were included in a phase 1/2 study of CD30+ B-cell lymphomas evaluating BV and CHP (cyclophosphamide, doxorubicin, prednisone; CHP+BV).87 In the 22 patients with PMBCL, 2-year PFS was 86% and 2-year OS was 100%, with no difference made by the addition of consolidative RT (P = .95). Overall, 11 of 14 (79%) were confirmed molecular PMBCL by Lymph3x, which implies that there is some imprecision in clinicopathologic diagnoses of PMBCL.87 The antibody drug conjugate loncastuximab (ADCT-402) was evaluated in R/R B-cell NHL, and although only 2 cases of PMBCL were included, the ORR for all patients was 42% and the CR was 23%, with some durable responses, albeit with moderate hematologic toxicity (71% thrombocytopenia, 59% neutropenia), leading to recent FDA approval. Bispecific antibodies are under investigation across a range of B-cell lymphomas but have not specifically included PMBCL. Despite evidence of aberrant activation of the JAK/STAT pathway in PMBCL, use of JAK2 inhibitors has not been fruitful to date. A pilot study evaluated ruxolitinib in R/R cHL and R/R PMBCL and none of the patients with PMBCL responded.88 Whether combination therapy with PD1 inhibitor will translate into clinical benefit remains unknown.89 

Summary

There have been significant scientific advances over the past 20 years in dissecting the disease biology of PMBCL. Integral to disease pathogenesis is an immune-privileged phenotype that has led to pivotal studies and approval of PD1 inhibitors in R/R PMBCL. Although most patients have an excellent outcome with R-chemoimmunotherapy, ∼10% of patients have refractory disease, regardless of primary therapy, providing a strong rationale to integrate PD1 inhibitors in the up-front setting. Finally, further study of radiomics and biomarkers at diagnosis may help to identify patients who would benefit from alternative treatment approaches.

The author acknowledges Jane Rowlands for figure creation, Christian Steidl and David Scott for manuscript review, and Jeffrey Craig and Graham Slack for supplying the histology figure and for manuscript review.

Conflict-of-interest disclosure: K.J.S. has received honoraria and consulting fees from Seattle Genetics, Bristol Myers Squibb, Merck, Novartis, Kyowa, Janssen, and AstraZeneca and consulting fees from Servier; has been a member of the Data Safety Monitoring Committee of Regeneron; and has served on the Steering Committee of Beigene.

Correspondence: Kerry J. Savage, Centre for Lymphoid Cancer, BC Cancer, 600 West 10th Ave, Vancouver, BC V5Z 4E6, Canada; e-mail: ksavage@bccancer.bc.ca.

The online version of this article contains a data supplement.

1.
Savage
KJ
,
Monti
S
,
Kutok
JL
, et al
.
The molecular signature of mediastinal large B-cell lymphoma differs from that of other diffuse large B-cell lymphomas and shares features with classical Hodgkin lymphoma
.
Blood.
2003
;
102
(
12
):
3871
-
3879
.
2.
Rosenwald
A
,
Wright
G
,
Leroy
K
, et al
.
Molecular diagnosis of primary mediastinal B cell lymphoma identifies a clinically favorable subgroup of diffuse large B cell lymphoma related to Hodgkin lymphoma
.
J Exp Med.
2003
;
198
(
6
):
851
-
862
.
3.
Gaulard
P
,
Harris
NL
,
Pileri
SA
, et al
. Primary mediastinal (thymic) large B-cell lymphoma. In:
Swerdlow
SH
,
Campo
E
,
Jaffe
ES
, et al, eds.
WHO Classification of Tumours of Haematopoetic and Lymphoid Tissues.
4th ed.
Lyon, France
:
International Agency of Research on Cancer (IARC)
;
2008
:250-253.
4.
Joos
S
,
Küpper
M
,
Ohl
S
, et al
.
Genomic imbalances including amplification of the tyrosine kinase gene JAK2 in CD30+ Hodgkin cells
.
Cancer Res.
2000
;
60
(
3
):
549
-
552
5.
Joos
S
,
Otaño-Joos
MI
,
Ziegler
S
, et al
.
Primary mediastinal (thymic) B-cell lymphoma is characterized by gains of chromosomal material including 9p and amplification of the REL gene
.
Blood.
1996
;
87
(
4
):
1571
-
1578
.
6.
Bentz
M
,
Barth
TF
,
Brüderlein
S
, et al
.
Gain of chromosome arm 9p is characteristic of primary mediastinal B-cell lymphoma (MBL): comprehensive molecular cytogenetic analysis and presentation of a novel MBL cell line
.
Genes Chromosomes Cancer.
2001
;
30
(
4
):
393
-
401
.
7.
Twa
DD
,
Steidl
C
.
Structural genomic alterations in primary mediastinal large B-cell lymphoma
.
Leuk Lymphoma.
2015
;
56
(
8
):
2239
-
2250
.
8.
Steidl
C
,
Shah
SP
,
Woolcock
BW
, et al
.
MHC class II transactivator CIITA is a recurrent gene fusion partner in lymphoid cancers
.
Nature.
2011
;
471
(
7338
):
377
-
381
.
9.
Roberts
RA
,
Wright
G
,
Rosenwald
AR
, et al
.
Loss of major histocompatibility class II gene and protein expression in primary mediastinal large B-cell lymphoma is highly coordinated and related to poor patient survival
.
Blood.
2006
;
108
(
1
):
311
-
318
.
10.
Shi
M
,
Roemer
MG
,
Chapuy
B
, et al
.
Expression of programmed cell death 1 ligand 2 (PD-L2) is a distinguishing feature of primary mediastinal (thymic) large B-cell lymphoma and associated with PDCD1LG2 copy gain
.
Am J Surg Pathol.
2014
;
38
(
12
):
1715
-
1723
.
11.
Tanaka
Y
,
Maeshima
AM
,
Nomoto
J
, et al
.
Expression pattern of PD-L1 and PD-L2 in classical Hodgkin lymphoma, primary mediastinal large B-cell lymphoma, and gray zone lymphoma
.
Eur J Haematol.
2018
;
100
(
5
):
511
-
517
.
12.
Gaulard
P
,
Harris
NL
,
Pileri
SA
, et al
. Primary mediastinal (Thymic) large B-cell lymphoma. In:
Swerdlow
SH
,
Campo
E
,
Harris
NL
, et al, eds.
WHO Classification of Tumours of Haematopoetic and Lymphoid Tissues.
Revised 4th ed.
Lyon, France
:
International Agency for Research on Cancer (IARC)
;
2017
:
314
-
316
.
13.
Hayden
AR
,
Tonseth
P
,
Lee
DG
, et al
.
Outcome of primary mediastinal large B-cell lymphoma using R-CHOP: impact of a PET-adapted approach
.
Blood.
2020
;
136
(
24
):
2803
-
2811
.
14.
Giulino-Roth
L
,
O’Donohue
T
,
Chen
Z
, et al
.
Outcomes of adults and children with primary mediastinal B-cell lymphoma treated with dose-adjusted EPOCH-R
.
Br J Haematol.
2017
;
179
(
5
):
739
-
747
.
15.
Aoki
T
,
Izutsu
K
,
Suzuki
R
, et al
.
Prognostic significance of pleural or pericardial effusion and the implication of optimal treatment in primary mediastinal large B-cell lymphoma: a multicenter retrospective study in Japan
.
Haematologica.
2014
;
99
(
12
):
1817
-
1825
.
16.
Dorfman
DM
,
Shahsafaei
A
,
Alonso
MA
.
Utility of CD200 immunostaining in the diagnosis of primary mediastinal large B cell lymphoma: comparison with MAL, CD23, and other markers
.
Mod Pathol.
2012
;
25
(
12
):
1637
-
1643
.
17.
Rodig
SJ
,
Abramson
JS
,
Pinkus
GS
, et al
.
Heterogeneous CD52 expression among hematologic neoplasms: implications for the use of alemtuzumab (CAMPATH-1H)
.
Clin Cancer Res.
2006
;
12
(
23
):
7174
-
7179
.
18.
Moldenhauer
G
,
Popov
SW
,
Wotschke
B
, et al
.
AID expression identifies interfollicular large B cells as putative precursors of mature B-cell malignancies
.
Blood.
2006
;
107
(
6
):
2470
-
2473
.
19.
Gentry
M
,
Bodo
J
,
Durkin
L
,
Hsi
ED
.
Performance of a commercially available MAL antibody in the diagnosis of primary mediastinal large B-cell lymphoma
.
Am J Surg Pathol.
2017
;
41
(
2
):
189
-
194
.
20.
Chen
BJ
,
Chapuy
B
,
Ouyang
J
, et al
.
PD-L1 expression is characteristic of a subset of aggressive B-cell lymphomas and virus-associated malignancies
.
Clin Cancer Res.
2013
;
19
(
13
):
3462
-
3473
.
21.
Sarkozy
C
,
Hung
SS
,
Chavez
EA
, et al
.
Mutational landscape of gray zone lymphoma
.
Blood.
2021
;
137
(
13
):
1765
-
1776
.
22.
Pittaluga
S
,
Nicolae
A
,
Wright
GW
, et al
.
Gene expression profiling of mediastinal gray zone lymphoma and its relationship to primary mediastinal B-cell lymphoma and classical Hodgkin lymphoma
.
Blood Cancer Discov.
2020
;
1
(
2
):
155
-
161
.
23.
Sarkozy
C
,
Chong
L
,
Takata
K
, et al
.
Gene expression profiling of gray zone lymphoma
.
Blood Adv.
2020
;
4
(
11
):
2523
-
2535
.
24.
Scott
DW
,
Mottok
A
,
Ennishi
D
, et al
.
Prognostic significance of diffuse large B-cell lymphoma cell of origin determined by digital gene expression in formalin-fixed paraffin-embedded tissue biopsies
.
J Clin Oncol.
2015
;
33
(
26
):
2848
-
2856
.
25.
Mottok
A
,
Wright
G
,
Rosenwald
A
, et al
.
Molecular classification of primary mediastinal large B-cell lymphoma using routinely available tissue specimens
.
Blood.
2018
;
132
(
22
):
2401
-
2405
.
26.
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
.
27.
Bobée
V
,
Ruminy
P
,
Marchand
V
, et al
.
Determination of molecular subtypes of diffuse large B-cell lymphoma using a reverse transcriptase multiplex ligation-dependent probe amplification classifier: a CALYM study
.
J Mol Diagn.
2017
;
19
(
6
):
892
-
904
.
28.
Schmitz
R
,
Hansmann
ML
,
Bohle
V
, et al
.
TNFAIP3 (A20) is a tumor suppressor gene in Hodgkin lymphoma and primary mediastinal B cell lymphoma
.
J Exp Med.
2009
;
206
(
5
):
981
-
989
.
29.
Mansouri
L
,
Noerenberg
D
,
Young
E
, et al
.
Frequent NFKBIE deletions are associated with poor outcome in primary mediastinal B-cell lymphoma
.
Blood.
2016
;
128
(
23
):
2666
-
2670
.
30.
Viganò
E
,
Gunawardana
J
,
Mottok
A
, et al
.
Somatic IL4R mutations in primary mediastinal large B-cell lymphoma lead to constitutive JAK-STAT signaling activation
.
Blood.
2018
;
131
(
18
):
2036
-
2046
.
31.
Green
MR
,
Monti
S
,
Rodig
SJ
, et al
.
Integrative analysis reveals selective 9p24.1 amplification, increased PD-1 ligand expression, and further induction via JAK2 in nodular sclerosing Hodgkin lymphoma and primary mediastinal large B-cell lymphoma
.
Blood.
2010
;
116
(
17
):
3268
-
3277
.
32.
Gunawardana
J
,
Chan
FC
,
Telenius
A
, et al
.
Recurrent somatic mutations of PTPN1 in primary mediastinal B cell lymphoma and Hodgkin lymphoma
.
Nat Genet.
2014
;
46
(
4
):
329
-
335
.
33.
Ritz
O
,
Guiter
C
,
Castellano
F
, et al
.
Recurrent mutations of the STAT6 DNA binding domain in primary mediastinal B-cell lymphoma
.
Blood.
2009
;
114
(
6
):
1236
-
1242
.
34.
Mottok
A
,
Hung
SS
,
Chavez
EA
, et al
.
Integrative genomic analysis identifies key pathogenic mechanisms in primary mediastinal large B-cell lymphoma
.
Blood.
2019
;
134
(
10
):
802
-
813
.
35.
Chapuy
B
,
Stewart
C
,
Dunford
AJ
, et al
.
Genomic analyses of PMBL reveal new drivers and mechanisms of sensitivity to PD-1 blockade
.
Blood.
2019
;
134
(
26
):
2369
-
2382
.
36.
Chong
LC
,
Twa
DD
,
Mottok
A
, et al
.
Comprehensive characterization of programmed death ligand structural rearrangements in B-cell non-Hodgkin lymphomas
.
Blood.
2016
;
128
(
9
):
1206
-
1213
.
37.
Twa
DD
,
Chan
FC
,
Ben-Neriah
S
, et al
.
Genomic rearrangements involving programmed death ligands are recurrent in primary mediastinal large B-cell lymphoma
.
Blood.
2014
;
123
(
13
):
2062
-
2065
.
38.
Mottok
A
,
Steidl
C
.
Genomic alterations underlying immune privilege in malignant lymphomas
.
Curr Opin Hematol.
2015
;
22
(
4
):
343
-
354
.
39.
Schneider
M
,
Schneider
S
,
Zühlke-Jenisch
R
, et al
.
Alterations of the CD58 gene in classical Hodgkin lymphoma
.
Genes Chromosomes Cancer.
2015
;
54
(
10
):
638
-
645
.
40.
Challa-Malladi
M
,
Lieu
YK
,
Califano
O
, et al
.
Combined genetic inactivation of β2-Microglobulin and CD58 reveals frequent escape from immune recognition in diffuse large B cell lymphoma
.
Cancer Cell.
2011
;
20
(
6
):
728
-
740
.
41.
Yuan
J
,
Wright
G
,
Rosenwald
A
, et al;
Lymphoma Leukemia Molecular Profiling Project (LLMPP)
.
Identification of primary mediastinal large B-cell lymphoma at nonmediastinal sites by gene expression profiling
.
Am J Surg Pathol.
2015
;
39
(
10
):
1322
-
1330
.
42.
Duns
G
,
Viganò
E
,
Ennishi
D
, et al
.
Characterization of DLBCL with a PMBL gene expression signature
.
Blood.
2021
;
138
(
2
):
136
-
148
.
43.
Chapuy
B
,
Stewart
C
,
Dunford
AJ
, et al
.
Molecular subtypes of diffuse large B cell lymphoma are associated with distinct pathogenic mechanisms and outcomes [published correction appears in Nat Med. 2018;24(8):129 and Nat Med. 2018;24(8):1290]
.
Nat Med.
2018
;
24
(
5
):
679
-
690
.
44.
Schmitz
R
,
Wright
GW
,
Huang
DW
, et al
.
Genetics and pathogenesis of diffuse large B-cell lymphoma
.
N Engl J Med.
2018
;
378
(
15
):
1396
-
1407
.
45.
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
.
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.
Lisenko
K
,
Dingeldein
G
,
Cremer
M
, et al
.
Addition of rituximab to CHOP-like chemotherapy in first line treatment of primary mediastinal B-cell lymphoma
.
BMC Cancer.
2017
;
17
(
1
):
359
.
48.
Vassilakopoulos
TP
,
Pangalis
GA
,
Katsigiannis
A
, et al
.
Rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone with or without radiotherapy in primary mediastinal large B-cell lymphoma: the emerging standard of care
.
Oncologist.
2012
;
17
(
2
):
239
-
249
.
49.
Rieger
M
,
Österborg
A
,
Pettengell
R
, et al;
MabThera International Trial (MInT) Group
.
Primary mediastinal B-cell lymphoma treated with CHOP-like chemotherapy with or without rituximab: results of the Mabthera International Trial Group study
.
Ann Oncol.
2011
;
22
(
3
):
664
-
670
.
50.
Xu
LM
,
Fang
H
,
Wang
WH
, et al
.
Prognostic significance of rituximab and radiotherapy for patients with primary mediastinal large B-cell lymphoma receiving doxorubicin-containing chemotherapy
.
Leuk Lymphoma.
2013
;
54
(
8
):
1684
-
1690
.
51.
Zinzani
PL
,
Stefoni
V
,
Finolezzi
E
, et al
.
Rituximab combined with MACOP-B or VACOP-B and radiation therapy in primary mediastinal large B-cell lymphoma: a retrospective study
.
Clin Lymphoma Myeloma.
2009
;
9
(
5
):
381
-
385
.
52.
Avigdor
A
,
Sirotkin
T
,
Kedmi
M
, et al
.
The impact of R-VACOP-B and interim FDG-PET/CT on outcome in primary mediastinal large B cell lymphoma
.
Ann Hematol.
2014
;
93
(
8
):
1297
-
1304
.
53.
Wilson
WH
,
Grossbard
ML
,
Pittaluga
S
, et al
.
Dose-adjusted EPOCH chemotherapy for untreated large B-cell lymphomas: a pharmacodynamic approach with high efficacy
.
Blood.
2002
;
99
(
8
):
2685
-
2693
.
54.
Dunleavy
K
,
Pittaluga
S
,
Maeda
LS
, et al
.
Dose-adjusted EPOCH-rituximab therapy in primary mediastinal B-cell lymphoma
.
N Engl J Med.
2013
;
368
(
15
):
1408
-
1416
.
55.
Burke
JS
,
Hoppe
RT
,
Cibull
ML
,
Dorfman
RF
.
Cutaneous malignant lymphoma: a pathologic study of 50 cases with clinical analysis of 37
.
Cancer.
1981
;
47
(
2
):
300
-
310
.
56.
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
.
57.
Zhou
H
,
Xu-Monette
ZY
,
Xiao
L
, et al
.
Prognostic factors, therapeutic approaches, and distinct immunobiologic features in patients with primary mediastinal large B-cell lymphoma on long-term follow-up
.
Blood Cancer J.
2020
;
10
(
5
):
49
.
58.
Shah
NN
,
Szabo
A
,
Huntington
SF
, et al
.
R-CHOP versus dose-adjusted R-EPOCH in frontline management of primary mediastinal B-cell lymphoma: a multi-centre analysis
.
Br J Haematol.
2018
;
180
(
4
):
534
-
544
.
59.
Malenda
A
,
Kołkowska-Leśniak
A
,
Puła
B
, et al
.
Outcomes of treatment with dose-adjusted EPOCH-R or R-CHOP in primary mediastinal large B-cell lymphoma
.
Eur J Haematol.
2020
;
104
(
1
):
59
-
66
.
60.
David
RJ
,
Baran
A
,
Loh
KP
, et al
.
Complications associated with dose-adjusted EPOCH-rituximab therapy for non-Hodgkin lymphoma
.
Clin Lymphoma Myeloma Leuk.
2018
;
18
(
12
):
781
-
787
.
61.
Jackson
MW
,
Rusthoven
CG
,
Jones
BL
,
Kamdar
M
,
Rabinovitch
R
.
Improved survival with radiation therapy in stage I-II primary mediastinal B cell lymphoma: a surveillance, epidemiology, and end results database analysis
.
Int J Radiat Oncol Biol Phys.
2016
;
94
(
1
):
126
-
132
.
62.
Giri
S
,
Bhatt
VR
,
Pathak
R
,
Bociek
RG
,
Vose
JM
,
Armitage
JO
.
Role of radiation therapy in primary mediastinal large B-cell lymphoma in rituximab era: a US population-based analysis
.
Am J Hematol.
2015
;
90
(
11
):
1052
-
1054
.
63.
Conway
JL
,
Connors
JM
,
Tyldesley
S
, et al
.
Secondary breast cancer risk by radiation volume in women with Hodgkin lymphoma
.
Int J Radiat Oncol Biol Phys.
2017
;
97
(
1
):
35
-
41
.
64.
Bhakta
N
,
Liu
Q
,
Yeo
F
, et al
.
Cumulative burden of cardiovascular morbidity in paediatric, adolescent, and young adult survivors of Hodgkin’s lymphoma: an analysis from the St Jude Lifetime Cohort Study
.
Lancet Oncol.
2016
;
17
(
9
):
1325
-
1334
.
65.
Schaapveld
M
,
Aleman
BM
,
van Eggermond
AM
, et al
.
Second cancer risk up to 40 years after treatment for Hodgkin’s lymphoma
.
N Engl J Med.
2015
;
373
(
26
):
2499
-
2511
.
66.
Zinzani
PL
,
Broccoli
A
,
Casadei
B
, et al
.
The role of rituximab and positron emission tomography in the treatment of primary mediastinal large B-cell lymphoma: experience on 74 patients
.
Hematol Oncol.
2015
;
33
(
4
):
145
-
150
.
67.
Barrington
SF
,
Mikhaeel
NG
,
Kostakoglu
L
, et al
.
Role of imaging in the staging and response assessment of lymphoma: consensus of the International Conference on Malignant Lymphomas Imaging Working Group
.
J Clin Oncol.
2014
;
32
(
27
):
3048
-
3058
.
68.
Martelli
M
,
Ceriani
L
,
Zucca
E
, et al
.
[18F]fluorodeoxyglucose positron emission tomography predicts survival after chemoimmunotherapy for primary mediastinal large B-cell lymphoma: results of the International Extranodal Lymphoma Study Group IELSG-26 Study [published correction appears in J Clin Oncol. 2016;34(21):2562]
.
J Clin Oncol.
2014
;
32
(
17
):
1769
-
1775
.
69.
Vassilakopoulos
TP
,
Pangalis
GA
,
Chatziioannou
S
, et al
.
PET/CT in primary mediastinal large B-cell lymphoma responding to rituximab-CHOP: An analysis of 106 patients regarding prognostic significance and implications for subsequent radiotherapy
.
Leukemia.
2016
;
30
(
1
):
238
-
242
.
70.
Vassilakopoulos
TP
,
Papageorgiou
SG
,
Angelopoulou
MK
, et al
.
Positron emission tomography after response to rituximab-CHOP in primary mediastinal large B-cell lymphoma: impact on outcomes and radiotherapy strategies
.
Ann Hematol.
2021
;
100
(
9
):
2279
-
2292
.
71.
Pinnix
CC
,
Ng
AK
,
Dabaja
BS
, et al
.
Positron emission tomography-computed tomography predictors of progression after DA-R-EPOCH for PMBCL
.
Blood Adv.
2018
;
2
(
11
):
1334
-
1343
.
72.
Melani
C
,
Advani
R
,
Roschewski
M
, et al
.
End-of-treatment and serial PET imaging in primary mediastinal B-cell lymphoma following dose-adjusted EPOCH-R: a paradigm shift in clinical decision making
.
Haematologica.
2018
;
103
(
8
):
1337
-
1344
.
73.
Vassilakopoulos
TP
,
Papageorgiou
S
,
Pangalis
GA
, et al
.
Response to ‘PET after response to R-CHOP in primary mediastinal large B-cell lymphoma’
.
Leukemia.
2016
;
30
(
8
):
1800
-
1801
.
74.
Ceriani
L
,
Martelli
M
,
Gospodarowicz
MK
, et al
.
Positron emission tomography/computed tomography assessment after immunochemotherapy and irradiation using the Lugano classification criteria in the IELSG-26 study of primary mediastinal B-cell lymphoma
.
Int J Radiat Oncol Biol Phys.
2017
;
97
(
1
):
42
-
49
.
75.
Ceriani
L
,
Milan
L
,
Martelli
M
, et al
.
Metabolic heterogeneity on baseline 18FDG-PET/CT scan is a predictor of outcome in primary mediastinal B-cell lymphoma
.
Blood.
2018
;
132
(
2
):
179
-
186
.
76.
Vardhana
S
,
Hamlin
PA
,
Yang
J
, et al
.
Outcomes of relapsed and refractory primary mediastinal (Thymic) large B cell lymphoma treated with second-line therapy and intent to transplant
.
Biol Blood Marrow Transplant.
2018
;
24
(
10
):
2133
-
2138
.
77.
Aoki
T
,
Shimada
K
,
Suzuki
R
, et al
.
High-dose chemotherapy followed by autologous stem cell transplantation for relapsed/refractory primary mediastinal large B-cell lymphoma
.
Blood Cancer J.
2015
;
5
(
12
):
e372
.
78.
Avivi
I
,
Boumendil
A
,
Finel
H
, et al
.
Autologous stem cell transplantation for primary mediastinal B-cell lymphoma: long-term outcome and role of post-transplant radiotherapy. A report of the European Society for Blood and Marrow Transplantation
.
Bone Marrow Transplant.
2018
;
53
(
8
):
1001
-
1009
.
79.
Zinzani
PL
,
Ribrag
V
,
Moskowitz
CH
, et al
.
Safety and tolerability of pembrolizumab in patients with relapsed/refractory primary mediastinal large B-cell lymphoma
.
Blood.
2017
;
130
(
3
):
267
-
270
.
80.
Armand
P
,
Rodig
S
,
Melnichenko
V
, et al
.
Pembrolizumab in relapsed or refractory primary mediastinal large B-cell lymphoma
.
J Clin Oncol.
2019
;
37
(
34
):
3291
-
3299
.
81.
Zinzani
PL
,
Santoro
A
,
Gritti
G
, et al
.
Nivolumab combined with brentuximab vedotin for relapsed/refractory primary mediastinal large B-cell lymphoma: efficacy and safety from the Phase II CheckMate 436 Study
.
J Clin Oncol.
2019
;
37
(
33
):
3081
-
3089
.
82.
Zinzani
PL
,
Pellegrini
C
,
Chiappella
A
, et al
.
Brentuximab vedotin in relapsed primary mediastinal large B-cell lymphoma: results from a phase 2 clinical trial
.
Blood.
2017
;
129
(
16
):
2328
-
2330
.
83.
Crump
M
,
Neelapu
SS
,
Farooq
U
, et al
.
Outcomes in refractory diffuse large B-cell lymphoma: results from the international SCHOLAR-1 study [published correction appears in Blood. 2018;131(5):587-588]
.
Blood.
2017
;
130
(
16
):
1800
-
1808
.
84.
Neelapu
SS
,
Locke
FL
,
Bartlett
NL
, et al
.
Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma
.
N Engl J Med.
2017
;
377
(
26
):
2531
-
2544
.
85.
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
.
86.
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
.
87.
Svoboda
J
,
Bair
SM
,
Landsburg
DJ
, et al
.
Brentuximab vedotin in combination with rituximab, cyclophosphamide, doxorubicin, and prednisone as frontline treatment for patients with CD30-positive B-cell lymphomas
.
Haematologica.
 
2021
;
106
(
6
):
1705
-
1713
.
88.
Kim
SJ
,
Yoon
DH
,
Kang
HJ
, et al
.
Ruxolitinib shows activity against Hodgkin lymphoma but not primary mediastinal large B-cell lymphoma
.
BMC Cancer.
2019
;
19
(
1
):
1080
.
89.
Nijland
M
,
van Meerten
T
,
Seitz
A
, et al
.
Combined PD-1 and JAK1/2 inhibition in refractory primary mediastinal B-cell lymphoma
.
Ann Hematol.
2018
;
97
(
5
):
905
-
907
.
90.
Eberle
FC
,
Salaverria
I
,
Steidl
C
, et al
.
Gray zone lymphoma: chromosomal aberrations with immunophenotypic and clinical correlations
.
Mod Pathol.
2011
;
24
(
12
):
1586
-
1597
.
91.
Sarkozy
C
,
Molina
T
,
Ghesquières
H
, et al
.
Mediastinal gray zone lymphoma: clinico-pathological characteristics and outcomes of 99 patients from the Lymphoma Study Association
.
Haematologica.
2017
;
102
(
1
):
150
-
159
.
92.
Duns
G
,
Viganò
E
,
Ennishi
D
, et al
.
Diffuse large B-cell lymphomas with a molecular PMBCL expression signature represent a distinct molecular subtype associated with poor clinical outcome [abstract]
.
Blood.
2019
;
134
(
suppl_1
). Abstract 922.
93.
Moskowitz
C
,
Hamlin
PA
Jr
,
Maragulia
J
,
Meikle
J
,
Zelenetz
AD
.
Sequential dose-dense RCHOP followed by ICE consolidation (MSKCC protocol 01–142) without radiotherapy for patients with primary mediastinal large B cell lymphoma [abstract]
.
Blood.
2010
;
116
(
21
). Abstract 420.  
94.
Gleeson
M
,
Hawkes
EA
,
Cunningham
D
, et al
.
Rituximab, cyclophosphamide, doxorubicin, vincristine and prednisolone (R-CHOP) in the management of primary mediastinal B-cell lymphoma: a subgroup analysis of the UK NCRI R-CHOP 14 versus 21 trial
.
Br J Haematol.
2016
;
175
(
4
):
668
-
672
.
95.
Burke
GAA
, Minard-Colin V, Auperin A, et al
.
Dose-adjusted etoposide, doxorubicin, andcyclophosphamide with vincristine and prednisone plus rituximab therapy in children and adolescents with primary mediastinal B-cell lymphoma: a multicenter phase II trial
.
J Clin Oncol
.
2021
;
39
(
33
):
3716
-
3724
.
96.
Held
G
,
Thurner
L
,
Pöschel
V
, et al
.
Role of radiotherapy and dose-densificaiton of R-CHOP in primary mediastinal large B-cell lymphoma: a subgroup analysis of the UNFOLDER trial of the German Lymphoma Alliance (GLA) [abstract]
.
EHA Library
2020
. Abstract S230.
97.
Ahn
HK
,
Kim
SJ
,
Yun
J
, et al
.
Improved treatment outcome of primary mediastinal large B-cell lymphoma after introduction of rituximab in Korean patients
.
Int J Hematol.
2010
;
91
(
3
):
456
-
463
.
98.
Soumerai
JD
,
Hellmann
MD
,
Feng
Y
, et al
.
Treatment of primary mediastinal B-cell lymphoma with rituximab, cyclophosphamide, doxorubicin, vincristine and prednisone is associated with a high rate of primary refractory disease
.
Leuk Lymphoma.
2014
;
55
(
3
):
538
-
543
.
99.
Pinnix
CC
,
Dabaja
B
,
Ahmed
MA
, et al
.
Single-institution experience in the treatment of primary mediastinal B cell lymphoma treated with immunochemotherapy in the setting of response assessment by 18fluorodeoxyglucose positron emission tomography
.
Int J Radiat Oncol Biol Phys.
2015
;
92
(
1
):
113
-
121
.
100.
Goldschmidt
N
,
Kleinstern
G
,
Orevi
M
, et al
.
Favorable outcome of primary mediastinal large B-cell lymphoma patients treated with sequential RCHOP-RICE regimen without radiotherapy
.
Cancer Chemother Pharmacol.
2016
;
77
(
5
):
1053
-
1060
.
101.
Messmer
M
,
Tsai
HL
,
Varadhan
R
, et al
.
R-CHOP without radiation in frontline management of primary mediastinal B-cell lymphoma
.
Leuk Lymphoma.
2019
;
60
(
5
):
1261
-
1265
.
102.
Chan
EHL
,
Koh
LP
,
Lee
J
, et al
.
Real world experience of R-CHOP with or without consolidative radiotherapy vs DA-EPOCH-R in the first-line treatment of primary mediastinal B-cell lymphoma
.
Cancer Med.
2019
;
8
(
10
):
4626
-
4632
.
103.
Wästerlid
T
,
Hasselblom
S
,
Joelsson
J
, et al
.
Real-world data on treatment and outcomes of patients with primary mediastinal large B-cell lymphoma: a Swedish lymphoma register study
.
Blood Cancer J.
2021
;
11
(
5
):
100
.
104.
Kuruvilla
J
,
Nagy
T
,
Pintilie
M
,
Keating
A
,
Crump
M
.
Outcomes of salvage chemotherapy and autologous stem cell transplanatation for relapsed or refractory primary medistianal large B-cell lymphoma (PMLCL) are inferior to diffuse large B-cell lymphoma [abstract]
.
Blood.
2008
;
49
(
7
):
1329
-
1336
.

Supplemental data

Sign in via your Institution