• EBV-associated PCNSL typically has absent MYD88, CD79B, and PIM1 mutations, is rarely ABC cell of origin, and has intact HLA class I/II.

  • The tumor microenvironment in EBV-associated PCNSL adapts to tolerate expression of an immunogenic virus.

Primary central nervous system lymphoma (PCNSL) is confined to the brain, eyes, and cerebrospinal fluid without evidence of systemic spread. Rarely, PCNSL occurs in the context of immunosuppression (eg, posttransplant lymphoproliferative disorders or HIV [AIDS-related PCNSL]). These cases are poorly characterized, have dismal outcome, and are typically Epstein-Barr virus (EBV)-associated (ie, tissue-positive). We used targeted sequencing and digital multiplex gene expression to compare the genetic landscape and tumor microenvironment (TME) of 91 PCNSL tissues all with diffuse large B-cell lymphoma histology. Forty-seven were EBV tissue-negative: 45 EBV HIV PCNSL and 2 EBV HIV+ PCNSL; and 44 were EBV tissue-positive: 23 EBV+ HIV+ PCNSL and 21 EBV+ HIV PCNSL. As with prior studies, EBV HIV PCNSL had frequent MYD88, CD79B, and PIM1 mutations, and enrichment for the activated B-cell (ABC) cell-of-origin subtype. In contrast, these mutations were absent in all EBV tissue-positive cases and ABC frequency was low. Furthermore, copy number loss in HLA class I/II and antigen-presenting/processing genes were rarely observed, indicating retained antigen presentation. To counter this, EBV+ HIV PCNSL had a tolerogenic TME with elevated macrophage and immune-checkpoint gene expression, whereas AIDS-related PCNSL had low CD4 gene counts. EBV-associated PCNSL in the immunosuppressed is immunobiologically distinct from EBV HIV PCNSL, and, despite expressing an immunogenic virus, retains the ability to present EBV antigens. Results provide a framework for targeted treatment.

Primary central nervous system lymphoma (PCNSL) is a rare form of extranodal non-Hodgkin lymphoma (NHL) that is confined to the brain, eyes, and cerebrospinal fluid without evidence of systemic spread. It most commonly has diffuse large B-cell lymphoma (DLBCL) histology, has a median age of 65 years, and accounts for ∼1% of all cases of NHL.1,2  In recent years, understanding of the molecular pathogenesis of PCNSL has increased. Recurrent mutations have been identified in the B‐cell receptor (BCR) signaling axis and its downstream target: nuclear factor κ-light-chain enhancer of activated B cells (NF-κB). These principally involve MYD88, CD79B, and less often CARD11 and TNFAIP3.3-7  Phylogenetic analysis indicates MYD88 mutation is an early clonal event.8  Among other high-frequency mutations is the cell-cycle/adhesion gene PIM1.9,10 

Rarely, PCNSL occurs with immunosuppression (eg, posttransplant lymphoproliferative disorder [PTLD] or HIV [AIDS-related PCNSL]).11  Although no accurate figures exist, it has been estimated that overall PCNSL after immunosuppression accounts for <10% of PCNSL cases (<0.1% of NHL).12,13  These patients are typically younger than nonimmunosuppressed PCNSL, have dismal outcomes,13,14  and because (with a few worthy exceptions)15,16  clinical trials for PCNSL typically exclude patients with PTLD and HIV, the optimal management is unknown and there are no consensus guidelines. Because of its rarity, characterization of the immunobiological features of PCNSL after immunosuppression remains minimal, with the largest published series restricted to microRNA profiling of 9 cases.17  It is known however that the malignant B cells are typically tissue-positive for the lymphotropic Epstein-Barr virus (EBV), termed EBV-associated PCNSL.13 

Here, we present, to our knowledge, the first large-scale comparative data on the genetic and gene expression landscape of PCNSL subtypes, stratified by EBV tissue and HIV status. PCNSL tissues were subdivided into: EBV tissue-negative: EBV HIV PCNSL and EBV HIV+ PCNSL; and EBV tissue-positive: EBV+ HIV+ PCNSL and EBV+ HIV PCNSL. The findings provide a rationale for targeted therapy.

Samples

There were 91 samples in total (90 formalin-fixed paraffin-embedded [FFPE] patient samples and 1 cell line). FFPE samples were obtained retrospectively from Australia (60 samples), Germany (7 samples), and the United States (23 samples). Selection criteria were PCNSL cases with DLBCL histology in immunosuppressed and immunocompetent settings. For non-HIV cases, samples were obtained from hospitals that were tertiary referral centers for both lymphoma and organ transplantation. This ensured that PCNSL cases were enriched for PTLD. HIV cases (with the exception of samples from 3 Australian patients) were from US HIV biobanks. The consort diagram (Figure 1) provides details. Median age for EBV HIV PCNSL was 64 (range, 28-82) years, 61% male; for AIDS-related PCNSL 37 (range, 31-60) years, 100% male; and for EBV+ HIV PCNSL 55 (range, 30-71) years, 45% male. Clinical details are provided in the supplemental Data on the Blood Web site. In line with previous reports, EBV+ HIV PCNSL resulting from PTLD occurred late following transplant (mean >9 years) as distinct from systemic EBV+ PTLD.13  All tumors were classified as PCNSL with DLBCL histology according to the World Health Organization classification.1  Systemic staging was performed and only cases of isolated CNS lymphoma were included. EBV-tissue status was determined as published.17,18  There were: 44 EBV HIV PCNSL, 2 EBV HIV+ PCNSL, 23 EBV+ HIV+ PCNSL, and 21 EBV+ HIV PCNSL (18 with PTLD, 3 without PTLD/immunosuppression) FFPE samples. These were macrodissected to enrich for tumor content. Testing was based on quality and quantity of FFPE-extracted DNA/RNA, with all samples suitable for either sequencing and/or gene expression. The 44 EBV HIV PCNSL samples were supplemented with the human EBV HIV brain cell-line TK (JCRB1206, JCRB Cell-Bank, Japan), making 45. The study was approved by the relevant institutional regulatory boards in concordance with the Declaration of Helsinki.

Figure 1.

Consort diagram providing details of PCNSL subtype and sample testing performed. ACSRB, AIDS Cancer Specimen Resource Bank; AH, Austin Hospital; NNTC, NeuroAIDS National Tissue Collection; PAH, Princess Alexandra Hospital; WH, Westmead Hospital. *One of the 2 EBV HIV+ PCNSL tissues underwent sequencing only, and both had COO. **Brain TK cell-line underwent targeted sequencing only.

Figure 1.

Consort diagram providing details of PCNSL subtype and sample testing performed. ACSRB, AIDS Cancer Specimen Resource Bank; AH, Austin Hospital; NNTC, NeuroAIDS National Tissue Collection; PAH, Princess Alexandra Hospital; WH, Westmead Hospital. *One of the 2 EBV HIV+ PCNSL tissues underwent sequencing only, and both had COO. **Brain TK cell-line underwent targeted sequencing only.

Close modal

Sequencing

A customized, hybrid-capture SureSelect XT (Agilent, CA) panel of 54 genes was used to identify mutations in antigen presentation/processing (8 genes), immune function (7 genes), the BCR-dependent NF-κB pathway (8 genes), epigenetic regulation (10 genes), cell cycle/adhesion (12 genes), and B-cell differentiation (9 genes). These mutations were known to be of biological importance in EBV HIV PCNSL and/or tumor immunity, and were chosen on review of previous literature.3,4,6,7,19  The sequencing panel was also designed to enrich the targeted regions to obtain copy number variation (CNV) data for HLA class I/II alleles (known to be frequently absent in EBV HIV PCNSL),5,20,21  and the antigen presentation molecules CTSS, PSMB9, CIITA, CD80, B2M, CD58, and NLRC5. For mutations, the final probes covered 138 560 base pairs covering 97% breadth of coverage of the 532 targets. Polymerase chain reaction (PCR) assessment of PD-L1/PD-L2 copy number is outlined in the supplemental Data.

In SureSelectXT target enrichment, ultra-long 120-mer-biotinylated complementary RNA baits were hybridized with samples to capture region of interest and enrich them out of a next-generation sequencing genomic fragment. The KAPA Hyper Prep Kit were used in conjunction with Agilent SureSelect adapters and primers to improve the library quality and capture efficiency for FFPE samples with variable quality and limited quantity. Sequencing was performed using on an Illumina platform. The library pool was diluted and denatured according to the standard NextSeq protocol and sequenced to generate paired-end 76 base pair reads using a 150-cycle NextSeq500/550 Mid Output reagent Kit v2 (Illumina). After sequencing, fastq files were generated using bcl2fastq2 software (v2.18.0). The mean depth of coverage for the targeted DNA sequencing was >×200 for all subtypes: ×499 EBV HIV PCNSL, ×380 EBV HIV+ PCNSL, ×236 EBV HIV+ PCNSL, and ×1208 EBV+ HIV PCNSL. Mean coverage per EBV-tissue status, is 492 (EBV tissue-negative) and 666 (EBV tissue-positive). All samples had mean coverage of target genes of at least ×50.

Mutations were “called” by 2 software programs: MuTect2 and Lofreq. Only nonsynonymous mutations that were predicted to have a high impact on the protein were included. The mutations were discarded if (1) they had a variant allele frequency <5%, or (2) they were present in at least 1 the 16 surgically excised nonmalignant lymph nodes samples (taken during diagnostic workup in patients without lymphoma that were being evaluated for potential breast cancer), or (3) were present in paired germline tissue (available in 8 EBV HIV PCNSL and 3 EBV+ HIV PCNSL cases), or (4) they were present in the Genome Aggregation database (exomes release 2.0.2) at a frequency >1%. As expected, across subtypes, missense mutations (78.5%) predominated. Based on DNA availability, hotspot mutations detected by capture hybridization in MYD88 (1 hotspot, 8 cases) and CD79B (5 hotspots, 15 cases) underwent confirmatory Sanger sequencing of PCR-amplified gene products. In all 23 cases, the hotspots were validated. With regard to CNV, we used 3 tools to identify CNVs from the aligned read data including GATK, CNVPanelizer, and ONCOCN. Only CNVs identified by all 3 tools were included.

RNA quantification

Genes were digitally quantified for immune genes, Lymph2Cx and EBV genes using the nCounter platform (NanoString) as published.22-24  A targeted gene panel was chosen to permit analysis of selected clinically pertinent immune effectors (CD137, CD4, CD8, tumor necrosis factor-α [TNF-α]), macrophages (CD68, CD163), and immune-checkpoints (LAG-3, PD-1, PD-L1, PD-L2, TIM-3) similar to that previously outlined.22-24  EBV gene expression (EBER-1, LMP-1, EBNA2) to distinguish viral latency patterns (latency I: EBER+/LMP1/EBNA2; latency II: EBER+/LMP1+/EBNA2; latency III: EBER+/LMP1+/EBNA2+)18  and cell-of-origin (COO) categorization (by NanoString Lymph2Cx assay) was performed.25  Normalized data are provided in the supplemental Data.

Statistical analysis

Values between groups of data were tested for statistical significance using the 2-tailed Mann-Whitney tests. Categorical data were compared using Fisher exact test or χ2 test. All tests were 2-sided at the threshold of P = .05. Benjamini and Hochberg false discovery rate was used to compare combined EBV+ vs combined EBV tissues (threshold P = .05). All analyses were prepared using the GraphPad Prism platform (v7, GraphPad).

Comparison of the mutational landscapes across PCNSL subtypes

Figure 2A shows results of individual samples grouped by PCNSL subtypes. First, EBV tissue-negative PCNSL: EBV HIV PCNSL and EBV HIV+ PCNSL cases were analyzed. EBV HIV PCNSL was enriched in previously identified mutations.3-10  Mutations at high frequency (≥20%) include MYD88, CD79B, PIM1, KMT2D, TBL1XR1, TOX, and PRDM1. These involve BCR-NF-κB signaling, epigenetic regulation, cell cycle/adhesion, and B-cell differentiation. Mutations in antigen presentation/processing and immune function were less frequent. Only 1 EBV HIV+ PCNSL had DNA suitable for sequencing. Although conclusions should not be overinterpreted, it is notable that this case had multiple (8) mutations.

Figure 2.

Mutational landscape of PCNSL, according to EBV tissue and HIV serological status. (A) Each column in this plot represents an individual case (with mutation[s] in the displayed genes) of the final sequencing cohort (n = 79), across the 4 tissue subtypes: EBV HIV PCNSL, EBV HIV+ PCNSL, EBV+ HIV+ PCNSL, and EBV+ HIV PCNSL. Mutated genes constitute individual rows and are sorted according to their mutational frequencies of mutated cases as provided on the far right. Mutation types are color coded as indicated in the key; red* brain lymphoma TK cell-line, red** patients without PTLD/iatrogenic immunosuppression. (B) Stacked histograms show the percentage (percentages rounded to whole numbers) of cases with mutations in MYD88, CD79B, PIM1 by EBV-tissue and HIV serological status across the 3 main subtypes. Because EBV HIV+ PCNSL represented only 1 sequenced case, aggregate data are not shown. (C) Number of mutated genes observed using the targeted sequencing panel in EBV HIV PCNSL, EBV+ HIV+ PCNSL, and EBV+ HIV PCNSL, with P values for paired subtypes: *P ≤ .05; **P ≤ .01; ***P ≤ .001; ****P ≤ .0001.

Figure 2.

Mutational landscape of PCNSL, according to EBV tissue and HIV serological status. (A) Each column in this plot represents an individual case (with mutation[s] in the displayed genes) of the final sequencing cohort (n = 79), across the 4 tissue subtypes: EBV HIV PCNSL, EBV HIV+ PCNSL, EBV+ HIV+ PCNSL, and EBV+ HIV PCNSL. Mutated genes constitute individual rows and are sorted according to their mutational frequencies of mutated cases as provided on the far right. Mutation types are color coded as indicated in the key; red* brain lymphoma TK cell-line, red** patients without PTLD/iatrogenic immunosuppression. (B) Stacked histograms show the percentage (percentages rounded to whole numbers) of cases with mutations in MYD88, CD79B, PIM1 by EBV-tissue and HIV serological status across the 3 main subtypes. Because EBV HIV+ PCNSL represented only 1 sequenced case, aggregate data are not shown. (C) Number of mutated genes observed using the targeted sequencing panel in EBV HIV PCNSL, EBV+ HIV+ PCNSL, and EBV+ HIV PCNSL, with P values for paired subtypes: *P ≤ .05; **P ≤ .01; ***P ≤ .001; ****P ≤ .0001.

Close modal

Findings in EBV tissue-negative cases markedly contrasted with EBV+ HIV PCNSL and EBV+ HIV+ PCNSL (EBV tissue-positive) cases. In 44% of EBV+ HIV PCNSL, no mutation was detected. In the remaining, only 1 mutation occurred in >1 patient (KMT2C, n = 2). For EBV+ HIV+ PCNSL cases, 20% had no mutation present in the targeted panel, with KMT2D the most common mutation (25%), and CREBBP, CD36, and MYC mutations each occurring in 15%. Only 5 other mutations occurred in >1 patient (TNFAIP3, KMT2C, CDH22, NFKBIE). Only 30% of EBV+ HIV+ PCNSL had ≥3 mutations detected.

Next, we examined the frequency with which MYD88, CD79B, and PIM1 mutations cooccurred (Figure 2B). These 3 mutations are contained in the recently proposed “C5” and “MCD” DLBCL molecular classifications.26-28  In the EBV HIV PCNSL samples, >80% possessed at least 1 mutation in either/or MYD88, CD79B, and PIM1, with cooccurrence of these mutations in 69%. Similarly, in EBV HIV+ PCNSL, the single case sequenced had cooccurrence in MYD88 and PIM1. Strikingly, no MYD88, CD79B, and PIM1 mutations were seen in any of the EBV tissue-positive samples. Additionally, mutations in TBL1XR1, PRMD1, and TOX were also significantly overrepresented in EBV tissue-negative PCNSL by false discovery rate testing (supplemental Data).

The total number of mutated genes detected in the targeted panel were next compared (Figure 2C). There was a higher number of mutations observed in EBV HIV PCNSL compared with EBV+ HIV+ PCNSL and EBV+ HIV PCNSL cases (P = 2.0 × 10−9, and P = 3.7 × 10−15, respectively). EBV+ HIV PCNSL had a lower number of mutations compared with EBV+ HIV+ cases (P = .012).

COO differs by EBV-tissue status in PCNSL

To better understand the contrasting molecular underpinnings between the subtypes, we compared the COO classified by the Lymph2Cx assay. EBV HIV PCNSL (Figure 3A) mainly typed as activated B-cell (ABC) (69%). Fifty percent were ABC and 50% germinal center B cell (GCB) in the limited number (2 samples) of EBV HIV+ PCNSL tested. However, COO was only 25% and 11% ABC in EBV+ HIV PCNSL and EBV+ HIV+ PCNSL, respectively. ABC was higher in EBV HIV PCNSL compared with EBV-tissue positive PCNSL (P = .0001).

Figure 3.

Molecular COO and copy number alterations in HLA class I/II alleles and antigen presentation/processing genes, separated into the PCNSL subtypes. (A) Stacked histograms show the relationship between the molecular COO classification (using the Lymph2CX assay) across 3 subtypes. Because EBV HIV+ PCNSL represented only 2 COO cases, aggregate data are not shown. UC, unclassified. (B) Each column represents an individual case across the 4 PCNSL subtypes. Copy number altered genes constitute individual rows, with gains and losses shown in gold and blue, respectively. On the far right, percentages indicate the proportion of PCNSL tumors with copy number alterations of the specified gene; red* brain lymphoma TK cell-line, red** patients without PTLD/iatrogenic immunosuppression.

Figure 3.

Molecular COO and copy number alterations in HLA class I/II alleles and antigen presentation/processing genes, separated into the PCNSL subtypes. (A) Stacked histograms show the relationship between the molecular COO classification (using the Lymph2CX assay) across 3 subtypes. Because EBV HIV+ PCNSL represented only 2 COO cases, aggregate data are not shown. UC, unclassified. (B) Each column represents an individual case across the 4 PCNSL subtypes. Copy number altered genes constitute individual rows, with gains and losses shown in gold and blue, respectively. On the far right, percentages indicate the proportion of PCNSL tumors with copy number alterations of the specified gene; red* brain lymphoma TK cell-line, red** patients without PTLD/iatrogenic immunosuppression.

Close modal

Comparison of copy number alterations in antigen presentation/processing and HLA class I/II alleles across PCNSL subtypes

Copy number (CN) alterations in HLA I/II alleles were characterized (Figure 3B). As anticipated, CN loss was frequent in HLA class I/II alleles of EBV HIV PCNSL (43%). However, CN loss in HLA class I/II was seen in only 10% EBV+ HIV+ PCNSL and 13% EBV+ HIV PCNSL, respectively. The single EBV HIV+ PCNSL had no HLA class I/II CN loss.

CN alterations in genes involved in antigen presentation/processing were also tested. A total of 31% EBV HIV PCNSL had CN loss for genes involved with antigen presentation/processing. Except in 8%, CN loss in these genes did not cooccur (ie, they were mutually exclusive). No CN loss in any of these genes was seen in the remaining PCNSL subtypes. CN gain of CTSS is a feature of follicular lymphoma,29,30  and was seen in 9% of cases overall. A subset of cases (27 EBV HIV PCNSL and 6 EBV+ HIV PCNSL) had sufficient DNA available for PCR-based PD-L1, PD-L2 CN gain testing (supplemental Data). This showed there was no difference in CN gains between these PCNSL subtypes for PD-L1 or PD-L2 (supplemental Data; P = .87 and P = .08, respectively).

EBV tissue-positive PCNSL have distinct genetic pathway aberrations compared with EBV HIV PCNSL

To identify patterns within the 3 principal PCNSL subtypes (EBV HIV+ PCNSL was excluded from the aggregate analysis because n = 1), we compared mutations and/or CN loss categorized by pathways (Figure 4). First, EBV tissue-positive PCNSL subtypes were evaluated. There were no differences in genetic pathway aberrations between EBV+ HIV PCNSL and EBV+ HIV+ PCNSL for each of HLA class I/II CN loss, antigen presentation/processing (mutations combined with CN loss), or mutations in immune function, BCR-NF-κB, epigenetic regulation, cell cycle/adhesion, and B-cell differentiation. Next, EBV tissue-positive PCNSL was compared with EBV HIV PCNSL. There was a higher frequency of genetic pathway aberrations for all pathways (except immune function) in EBV HIV PCNSL relative to EBV tissue-positive PCNSL cases.

Figure 4.

Combined mutations and copy number loss separated into the 2 EBV-tissue positive PCNSL subtypes and EBVHIVPCNSL. Mutations and/or CN loss were categorized into pathway categories. CN loss is shown for HLA class I/II. For antigen presentation/processing mutations were combined with CN loss. Mutations for immune function, BCR-NF-κB signaling, epigenetic regulation, cell cycle/adhesion and B-cell differentiation. Percentages are shown for each PCNSL subtype, with P values for paired subtypes: *P ≤ .05; **P ≤ .01; ***P ≤ .001; ****P ≤ .0001. There were 42, 20, and 16 cases of EBV HIV, EBV+ HIV+, and EBV+ HIV PCNSL, respectively (data for the single EBV HIV PCNSL tissue is not shown).

Figure 4.

Combined mutations and copy number loss separated into the 2 EBV-tissue positive PCNSL subtypes and EBVHIVPCNSL. Mutations and/or CN loss were categorized into pathway categories. CN loss is shown for HLA class I/II. For antigen presentation/processing mutations were combined with CN loss. Mutations for immune function, BCR-NF-κB signaling, epigenetic regulation, cell cycle/adhesion and B-cell differentiation. Percentages are shown for each PCNSL subtype, with P values for paired subtypes: *P ≤ .05; **P ≤ .01; ***P ≤ .001; ****P ≤ .0001. There were 42, 20, and 16 cases of EBV HIV, EBV+ HIV+, and EBV+ HIV PCNSL, respectively (data for the single EBV HIV PCNSL tissue is not shown).

Close modal

Differential gene expression of the TME across PCNSL subtypes

To delineate differences in TME between the 3 principal PCNSL subtypes (as before, EBV HIV+ PCNSL was excluded from the aggregate analysis), we performed digital gene expression using a customized panel of immune genes (Figure 5). There was elevated TNF-α, CD68, CD163, PD-L1, PD-L2, LAG-3, and TIM-3 in EBV+ HIV PCNSL compared with all other PCNSL subtypes, broadly indicative of a tolerogenic TME. EBV+ HIV+ PCNSL cases had low levels of CD4 and macrophage markers CD68 and CD163 compared with non-AIDS-related PCNSL.

Figure 5.

The tumor microenvironment in the 2 EBV tissue-positive PCNSL subtypes and EBVHIVPCNSL. A targeted gene expression panel was chosen of selected clinically pertinent immune effectors, macrophages, and immune-checkpoint markers. Gene counts are shown for each PCNSL subtype, with P values for paired subtypes: *P ≤ .05; **P ≤ .01; ***P ≤ .001; ****P ≤ .0001. There were 34, 18, and 18 cases of EBV HIV, EBV+ HIV+, and EBV+ HIV PCNSL, respectively.

Figure 5.

The tumor microenvironment in the 2 EBV tissue-positive PCNSL subtypes and EBVHIVPCNSL. A targeted gene expression panel was chosen of selected clinically pertinent immune effectors, macrophages, and immune-checkpoint markers. Gene counts are shown for each PCNSL subtype, with P values for paired subtypes: *P ≤ .05; **P ≤ .01; ***P ≤ .001; ****P ≤ .0001. There were 34, 18, and 18 cases of EBV HIV, EBV+ HIV+, and EBV+ HIV PCNSL, respectively.

Close modal

EBV-latency profiling/categorization was performed in 36 EBV tissue-positive PCNSL with sufficient RNA as previously outlined.18  The frequencies in 18 EBV+ HIV PCNSL and 18 EBV+ HIV+ PCNSL of latency III/II/I were 56%/33%/11% and 78%/22%/0%, respectively.

Using a combination of targeted sequencing and digital multiplex gene expression applied to an international series of an extremely rare and poorly categorized lymphoma subtype, we demonstrate that the genetic landscape typically observed in EBV HIV PCNSL is distinct from EBV-associated (tissue-positive) PCNSL. Notably, mutations in the BCR-dependent NF-κB pathway, the cell cycle/adhesion pathway, CN loss in HLA class I/II and mutations/CN loss in the antigen presentation/processing pathways all rarely occur in EBV-tissue positive cases. Immune evasion in patients with EBV+ HIV+ PCNSL (AIDS-related PCNSL) was mediated by reduced CD4, whereas in EBV+ HIV PCNSL, the TME had increased expression of tolerogenic immune genes.

Among genetic aberrations known to be frequently observed in EBV HIV PCNSL are mutations in MYD88 and CD79B that activate NF-κB activation, which is a core pathway involved in the pathogenesis of PCNSL.4-8,26  Another central mechanism is mutation in the cell cycle/adhesion gene PIM1.9,10  Our observation that these genes frequently cooccurred and were enriched in the ABC COO,31  is in keeping with the recently proposed “C5” and “MCD” DLBCL molecular classifications.26-28  By marked contrast, in EBV tissue-positive cases, there were no mutations in PIM1, MYD88, and CD79B. Here, COO was typically either GCB or unclassified. This is similar to recent studies of systemic HIV+ and EBV+ DLBCL that used the Lymph2Cx classifier.32,33  Notably EBV induces an atypical germinal center reaction,34  and recent characterization of EBV-driven B-cell differentiation shows that segregation into GCB and ABC subtypes by a limited panel of immunohistochemical markers is overly simplistic.35  The combined mutational and COO data emphasize that the nature of the pathogenesis of PCNSL is tightly linked to presence or absence of EBV-tissue expression. This is consistent with Hodgkin lymphoma (HL), where EBV+ HL appears to have a lower mutation load than EBV HL.36  EBV is known to play a key role in the pathogenesis of several types of B-cell lymphomas, and to transform and immortalize B cells.37-39  In our series, EBV tissue-positive cases typically (≥89%) exhibited latency II or III gene profiles associated with expression of EBV viral genes that have an established capacity for inducting B-cell proliferation and prevention of senescence.40,41  We found only ∼30% EBV tissue-positive cases had a detectable mutation in the targeted exome panel. However, given that in plasmablastic lymphomas some genes appear to be more frequently mutated in EBV+ than in EBV cases,42  a comprehensive genomic analysis (eg, whole exome) is required to establish if other genetic aberrations are present, and to determine if these occur before and/or following EBV B-cell infection and malignant transformation.

EBV establishes a persistent infection and in the vast majority of individuals is asymptomatic. Lack of pathogenicity is in part maintained by a robust EBV-specific T-cell immune response.43  Both PTLD and AIDS are associated with a spectrum of B-cell lymphomas that are typically EBV associated.12,44-46  The emergence of EBV+ HIV+ PCNSL correlates with HIV-induced loss of EBV-specific CD4+ T cells rather than overall CD4+ T-cell loss.47  HIV was initially believed not to have oncogenic potential but rather increased cancer risk by allowing EBV to induce lymphomagenesis in coinfected subjects. More recently, this notion has been challenged.48  Interestingly, in our cohort, mutations were modestly but significantly more frequent in EBV+ HIV+ PCNSL than EBV+ HIV PCNSL. This is in keeping with various lines of evidence that suggest that HIV itself contributes to the accumulation of DNA mutations.49-52 

Immune evasion is a hallmark of cancer.53  Consistent with our observations in EBV HIV PCNSL, deletion involving the HLA locus has previously been observed.20  Losing the ability to present neoantigens through HLA loss facilitates immune evasion. By contrast, EBV tissue-positive cases typically had intact HLA and absent mutations/CN loss in antigen presentation/processing genes. To counter the presentation of an immunogenic virus, there were higher levels of genes for macrophage markers CD68/CD163 and immune checkpoints in EBV+ HIV PCNSL compared with other subtypes, perhaps as an adaptive response to counter the elevated levels of the antiviral cytokine TNF-α. Given that EBV+ HIV PCNSL mostly occurred in the setting of iatrogenic immunosuppression because of PTLD, the presence of a tolerogenic TME in these cases may appear counterintuitive. However, it is in line with previous observations comparing systemic DLBCL-PTLD with systemic DLBCL in the nonimmunocompromised, which have shown that immune gene expression clusters on the basis of EBV status rather than immune status. Specifically, EBV+ cases show both a tolerogenic TME and an increased T-cell signaling signature relative to EBV DLBCL tissues irrespective of iatrogenic immunosuppression.54  Similarly, studies restricted to systemic DLBCL in the nonimmunocompromised confirm that EBV+ DLBCL has a tolerogenic TME, and are associated with higher levels of antigen-presenting molecules.32,55  Put together, it seems likely that the alterations in the TME are modulated by the virus to promote tumor escape. Interestingly, the proportion of PD-L1, PD-L2 gene amplification was similar between EBV HIV PCNSL and EBV+ HIV PCNSL, suggesting that the increase in expression of these ligands in EBV+ HIV PCNSL may be due to macrophage expression, as previously observed.56  CD4+ T cells and CNS-associated macrophages are key targets of HIV-infection,57  and as expected EBV+ HIV+ PCNSL (AIDS-related PCNSL) had low levels of CD4 and macrophage markers compared with non-AIDS-related PCNSL.

The distinct immunobiology of EBV tissue-positive PCNSL has therapeutic implications (Figure 6). The BTK-inhibitor ibrutinib is known to cross the blood–brain barrier (BBB) and has impressive phase 1/2 efficacy in EBV HIV PCNSL,58-61  and may have beneficial effects on components of the TME in the non-PCNSL setting.62,63  Mutations such as CARD11 and PIM1 that are associated with complete or partial ibrutinib resistance in PCNSL and/or systemic DLBCL59,64  were rarely observed in EBV tissue-positive cases. However, the relevance of these mutations was established in the context of an intact B-cell receptor signaling complex, and it is unclear what the impact of absent CARD11 and PIM1 mutations makes on EBV-transformed B-cell sensitivity. Also, other BTK inhibitors are possibly better tolerated.65  Although upregulation of immune checkpoints is observed, checkpoint blockade is potentially contraindicated in PTLD because of the risk of graft rejection and graft-versus-host-disease. By contrast, checkpoint-blockade would not be contraindicated in EBV HIV PCNSL and AIDS-related PCNSL.10,56,66  The retained ability of malignant B cells to present antigens in EBV-associated PCNSL is notable. Restoration of EBV-specific T-cell immunity, has been shown to induce clinical response in EBV+ lymphomas.67-69  Adoptive transfer of EBV-specific third-party virus specific T cells (VST) chosen on the basis of the best HLA match and in vitro effector function has previously been shown to induce high response rates in EBV+ PTLD and can cross the BBB.70,71 

Figure 6.

Pathogenesis of EBV tissue-positive PCNSL in the immunosuppressed, and potential implications for immunotherapy. (A) Mutations in the NF-κB and cell-cycling pathways and deletion of HLA class I/II molecules in EBV HIV PCNSL. (B) Viral transformation in EBV tissue-positive PCNSL, with the loss of host immunity providing no selection pressure to prevent presentation of EBV-immunogenic peptides by HLA I/II molecules. (C) EBV-specific viral specific T cells (VST) cross the blood–brain barrier to target the EBV+ malignant B cell.

Figure 6.

Pathogenesis of EBV tissue-positive PCNSL in the immunosuppressed, and potential implications for immunotherapy. (A) Mutations in the NF-κB and cell-cycling pathways and deletion of HLA class I/II molecules in EBV HIV PCNSL. (B) Viral transformation in EBV tissue-positive PCNSL, with the loss of host immunity providing no selection pressure to prevent presentation of EBV-immunogenic peptides by HLA I/II molecules. (C) EBV-specific viral specific T cells (VST) cross the blood–brain barrier to target the EBV+ malignant B cell.

Close modal

In summary, EBV-associated PCNSL in the immunosuppressed is immunobiologically distinct from EBV HIV PCNSL, and, despite expressing an immunogenic virus, retains the ability to present EBV. Results support combination strategies that cross the BBB, block EBV-driven oncogenesis and target EBV antigens. Based on the cumulative biological data, a phase 1 Australasian Leukaemia/Lymphoma Group clinical trial incorporating EBV-specific third-party VST (ACTRN12618001541291) has commenced.

Presented in part in oral form at the 15th International Conference on Malignant Lymphoma, Lugano, Switzerland, June 12, 2019.

Study data is included in the supplemental file.

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

This work was supported by the Leukaemia Foundation and the Mater Foundation (M.K.G.), the University of Queensland Mai Lan Kunzy Scholarship (T.H.), a National Health and Medical Research Council Early Career Fellowship, and a Princess Alexandra Hospital Award and a Cancer Australia and Cancer Cure Grant (ID1161139) (C.K.). The Translational Research Institute is supported by the Australian government. Specimens were provided by AIDS and Cancer Specimen Resource, funded by the National Institutes of Health, National Cancer Institute (UM1CA181255). This publication was also made possible from National Institutes of Health funding through the National Institute of Mental Health and National Institute of Neurological Disorders and Stroke by the following grants: Manhattan HIV Brain Bank (U24MH100931), Texas NeuroAIDS Research Center (U24MH100930), National Neurological AIDS Bank (U24MH100929), California NeuroAIDS Tissue Network (U24MH100928), and Data Coordinating Center (U24MH100925). The contents are solely the responsibility of the authors and do not necessarily represent the official view of the NeuroAIDS National Tissue Collection or National Institutes of Health.

Contribution: M.K.G., T. H., and C. K. conceived and designed the study; M.K.G. provided financial support; M.K.G. provided administrative support; M.K.G., S.D., R.U.T., G.B., G.H., E.B., L.C., J.W., E.H., and C.K. provided study materials or patients; M.K.G., T.H., S.B., K.O., J.W.D.T., L.F., H.O., K.B., L.M.d.L., M.B.S., E.B., L.C., and C.K. collected and assembled data; M.K.G., T.H., S.C.L., S.B., K.O., J.W.T., F.V., V.M., J.G., C.G., E.B., L.C., L.M.R., A.M., K.B., B.C., and C.K. provided data analysis and interpretation; and all authors undertook manuscript writing, final approval of manuscript, and are accountable for all aspects of the work.

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

Correspondence: M. K. Gandhi, Level 7, Translational Research Institute, Brisbane, QLD 4102, Australia; e-mail: maher.gandhi@mater.uq.edu.au; and C. Keane, Level 7, Translational Research Institute, Brisbane, QLD 4102, Australia; e-mail: colm.keane@mater.uq.edu.au.

1.
Swerdlow
SH
,
Campo
E
,
Pileri
SA
, et al
.
The 2016 revision of the World Health Organization classification of lymphoid neoplasms
.
Blood
.
2016
;
127
(
20
):
2375
-
2390
.
2.
Villano
JL
,
Koshy
M
,
Shaikh
H
,
Dolecek
TA
,
McCarthy
BJ
.
Age, gender, and racial differences in incidence and survival in primary CNS lymphoma
.
Br J Cancer
.
2011
;
105
(
9
):
1414
-
1418
.
3.
Chapuy
B
,
Cheng
H
,
Watahiki
A
, et al
.
Diffuse large B-cell lymphoma patient-derived xenograft models capture the molecular and biological heterogeneity of the disease
.
Blood
.
2016
;
127
(
18
):
2203
-
2213
.
4.
Nakamura
T
,
Tateishi
K
,
Niwa
T
, et al
.
Recurrent mutations of CD79B and MYD88 are the hallmark of primary central nervous system lymphomas
.
Neuropathol Appl Neurobiol
.
2016
;
42
(
3
):
279
-
290
.
5.
Gonzalez-Aguilar
A
,
Idbaih
A
,
Boisselier
B
, et al
.
Recurrent mutations of MYD88 and TBL1XR1 in primary central nervous system lymphomas
.
Clin Cancer Res
.
2012
;
18
(
19
):
5203
-
5211
.
6.
Braggio
E
,
Van Wier
S
,
Ojha
J
, et al
.
Genome-wide analysis uncovers novel recurrent alterations in primary central nervous system lymphomas
.
Clin Cancer Res
.
2015
;
21
(
17
):
3986
-
3994
.
7.
Vater
I
,
Montesinos-Rongen
M
,
Schlesner
M
, et al
.
The mutational pattern of primary lymphoma of the central nervous system determined by whole-exome sequencing
.
Leukemia
.
2015
;
29
(
3
):
677
-
685
.
8.
Nayyar
N
,
White
MD
,
Gill
CM
, et al
.
MYD88 L265P mutation and CDKN2A loss are early mutational events in primary central nervous system diffuse large B-cell lymphomas
.
Blood Adv
.
2019
;
3
(
3
):
375
-
383
.
9.
Bödör
C
,
Alpár
D
,
Marosvári
D
, et al
.
Molecular subtypes and genomic profile of primary central nervous system lymphoma
.
J Neuropathol Exp Neurol
.
2020
;
79
(
2
):
176
-
183
.
10.
Ou
A
,
Sumrall
A
,
Phuphanich
S
, et al
.
Primary CNS lymphoma commonly expresses immune response biomarkers
.
Neurooncol Adv
.
2020
;
2
(
1
):
vdaa018
.
11.
Evens
AM
,
David
KA
,
Helenowski
I
, et al
.
Multicenter analysis of 80 solid organ transplantation recipients with post-transplantation lymphoproliferative disease: outcomes and prognostic factors in the modern era
.
J Clin Oncol
.
2010
;
28
(
6
):
1038
-
1046
.
12.
Coté
TR
,
Manns
A
,
Hardy
CR
,
Yellin
FJ
,
Hartge
P
;
AIDS/Cancer Study Group
.
Epidemiology of brain lymphoma among people with or without acquired immunodeficiency syndrome
.
J Natl Cancer Inst
.
1996
;
88
(
10
):
675
-
679
.
13.
Evens
AM
,
Choquet
S
,
Kroll-Desrosiers
AR
, et al
.
Primary CNS posttransplant lymphoproliferative disease (PTLD): an international report of 84 cases in the modern era
.
Am J Transplant
.
2013
;
13
(
6
):
1512
-
1522
.
14.
Kreisl
TN
,
Panageas
KS
,
Elkin
EB
,
Deangelis
LM
,
Abrey
LE
.
Treatment patterns and prognosis in patients with human immunodeficiency virus and primary central system lymphoma
.
Leuk Lymphoma
.
2008
;
49
(
9
):
1710
-
1716
.
15.
Lurain
K
,
Uldrick
TS
,
Ramaswami
R
, et al
.
Treatment of HIV-associated primary CNS lymphoma with antiretroviral therapy, rituximab, and high-dose methotrexate
.
Blood
.
2020
;
136
(
19
):
2229
-
2232
.
16.
Dugan
JP
,
Haverkos
BM
,
Villagomez
L
, et al
.
Complete and durable responses in primary central nervous system posttransplant lymphoproliferative disorder with zidovudine, ganciclovir, rituximab, and dexamethasone
.
Clin Cancer Res
.
2018
;
24
(
14
):
3273
-
3281
.
17.
Fink
SE
,
Gandhi
MK
,
Nourse
JP
, et al
.
A comprehensive analysis of the cellular and EBV-specific microRNAome in primary CNS PTLD identifies different patterns among EBV-associated tumors
.
Am J Transplant
.
2014
;
14
(
11
):
2577
-
2587
.
18.
Nourse
JP
,
Crooks
P
,
Keane
C
, et al
.
Expression profiling of Epstein-Barr virus-encoded microRNAs from paraffin-embedded formalin-fixed primary Epstein-Barr virus-positive B-cell lymphoma samples
.
J Virol Methods
.
2012
;
184
(
1-2
):
46
-
54
.
19.
Bruno
A
,
Boisselier
B
,
Labreche
K
, et al
.
Mutational analysis of primary central nervous system lymphoma
.
Oncotarget
.
2014
;
5
(
13
):
5065
-
5075
.
20.
Riemersma
SA
,
Jordanova
ES
,
Schop
RF
, et al
.
Extensive genetic alterations of the HLA region, including homozygous deletions of HLA class II genes in B-cell lymphomas arising in immune-privileged sites
.
Blood
.
2000
;
96
(
10
):
3569
-
3577
.
21.
Schwindt
H
,
Vater
I
,
Kreuz
M
, et al
.
Chromosomal imbalances and partial uniparental disomies in primary central nervous system lymphoma
.
Leukemia
.
2009
;
23
(
10
):
1875
-
1884
.
22.
Keane
C
,
Gould
C
,
Jones
K
, et al
.
The T-cell receptor repertoire influences the tumor microenvironment and is associated with survival in aggressive B-cell lymphoma
.
Clin Cancer Res
.
2017
;
23
(
7
):
1820
-
1828
.
23.
Tobin
JWD
,
Keane
C
,
Gunawardana
J
, et al
.
Progression of disease within 24 months in follicular lymphoma is associated with reduced intratumoral immune infiltration
.
J Clin Oncol
.
2019
;
37
(
34
):
3300
-
3309
.
24.
Keane
C
,
Vari
F
,
Hertzberg
M
, et al
.
Ratios of T-cell immune effectors and checkpoint molecules as prognostic biomarkers in diffuse large B-cell lymphoma: a population-based study
.
Lancet Haematol
.
2015
;
2
(
10
):
e445
-
e455
.
25.
Scott
DW
,
Wright
GW
,
Williams
PM
, et al
.
Determining cell-of-origin subtypes of diffuse large B-cell lymphoma using gene expression in formalin-fixed paraffin-embedded tissue
.
Blood
.
2014
;
123
(
8
):
1214
-
1217
.
26.
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):1292]
.
Nat Med
.
2018
;
24
(
5
):
679
-
690
.
27.
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
.
28.
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
.
29.
Bararia
D
,
Hildebrand
JA
,
Stolz
S
, et al
.
Cathepsin S alterations induce a tumor-promoting immune microenvironment in follicular lymphoma
.
Cell Rep
.
2020
;
31
(
5
):
107522
.
30.
Dheilly
E
,
Battistello
E
,
Katanayeva
N
, et al
.
Cathepsin S regulates antigen processing and T cell activity in non-Hodgkin lymphoma
.
Cancer Cell
.
2020
;
37
(
5
):
674
-
689
.
31.
Hilal
T
,
Maguire
A
,
Kosiorek
HE
,
Rimsza
LM
,
Rosenthal
AC
.
Clinical features and cell of origin subtyping using gene expression profiling in HIV-negative patients with primary central nervous system lymphoma
.
Leuk Lymphoma
.
2019
;
60
(
14
):
3581
-
3583
.
32.
Keane
C
,
Tobin
J
,
Gunawardana
J
, et al
.
The tumour microenvironment is immuno-tolerogenic and a principal determinant of patient outcome in EBV-positive diffuse large B-cell lymphoma
.
Eur J Haematol
.
2019
;
103
(
3
):
200
-
207
.
33.
Maguire
A
,
Chen
X
,
Wisner
L
, et al
.
Enhanced DNA repair and genomic stability identify a novel HIV-related diffuse large B-cell lymphoma signature
.
Int J Cancer
.
2019
;
145
(
11
):
3078
-
3088
.
34.
Thorley-Lawson
DA
,
Gross
A
.
Persistence of the Epstein-Barr virus and the origins of associated lymphomas
.
N Engl J Med
.
2004
;
350
(
13
):
1328
-
1337
.
35.
Cristino
AS
,
Nourse
J
,
West
RA
, et al
.
EBV microRNA-BHRF1-2-5p targets the 3’UTR of immune checkpoint ligands PD-L1 and PD-L2
.
Blood
.
2019
;
134
(
25
):
2261
-
2270
.
36.
Wienand
K
,
Chapuy
B
,
Stewart
C
, et al
.
Genomic analyses of flow-sorted Hodgkin Reed-Sternberg cells reveal complementary mechanisms of immune evasion
.
Blood Adv
.
2019
;
3
(
23
):
4065
-
4080
.
37.
Gandhi
MK
.
Epstein-Barr virus-associated lymphomas
.
Expert Rev Anti Infect Ther
.
2006
;
4
(
1
):
77
-
89
.
38.
Küppers
R
.
B cells under influence: transformation of B cells by Epstein-Barr virus
.
Nat Rev Immunol
.
2003
;
3
(
10
):
801
-
812
.
39.
Shannon-Lowe
C
,
Rickinson
A
.
The global landscape of EBV-associated tumors
.
Front Oncol
.
2019
;
9
:
713
.
40.
Ma
SD
,
Tsai
MH
,
Romero-Masters
JC
, et al
.
Latent membrane protein 1 (LMP1) and LMP2A collaborate to promote Epstein-Barr virus-induced B cell lymphomas in a cord blood-humanized mouse model but are not essential
.
J Virol
.
2017
;
91
(
7
):
e01928
-
16
.
41.
Farrell
PJ
.
Epstein-Barr virus and cancer
.
Annu Rev Pathol
.
2019
;
14
(
1
):
29
-
53
.
42.
Garcia-Reyero
J
,
Martinez Magunacelaya
N
,
Gonzalez de Villambrosia
S
, et al
.
Genetic lesions in MYC and STAT3 drive oncogenic transcription factor overexpression in plasmablastic lymphoma [published online ahead of print 9 April 2020]
.
Haematologica
.
doi:haematol.2020.251579
.
43.
Long
HM
,
Meckiff
BJ
,
Taylor
GS
.
The T-cell response to Epstein-Barr virus-new tricks from an old dog
.
Front Immunol
.
2019
;
10
:
2193
.
44.
Liu
Z
,
Yoshimi
A
,
Wang
J
, et al
.
Mutations in the RNA splicing factor SF3B1 promote tumorigenesis through MYC stabilization
.
Cancer Discov
.
2020
;
10
(
6
):
806
-
821
.
45.
Pantanowitz
L
,
Carbone
A
,
Dolcetti
R
.
Microenvironment and HIV-related lymphomagenesis
.
Semin Cancer Biol
.
2015
;
34
:
52
-
57
.
46.
Jones
K
,
Nourse
JP
,
Morrison
L
, et al
.
Expansion of EBNA1-specific effector T cells in posttransplantation lymphoproliferative disorders
.
Blood
.
2010
;
116
(
13
):
2245
-
2252
.
47.
Gasser
O
,
Bihl
FK
,
Wolbers
M
, et al;
Swiss HIV Cohort Study
.
HIV patients developing primary CNS lymphoma lack EBV-specific CD4+ T cell function irrespective of absolute CD4+ T cell counts
.
PLoS Med
.
2007
;
4
(
3
):
e96
.
48.
Dolcetti
R
,
Giagulli
C
,
He
W
, et al
.
Role of HIV-1 matrix protein p17 variants in lymphoma pathogenesis
.
Proc Natl Acad Sci USA
.
2015
;
112
(
46
):
14331
-
14336
.
49.
Bhatia
R
,
Ryscavage
P
,
Taiwo
B
.
Accelerated aging and human immunodeficiency virus infection: emerging challenges of growing older in the era of successful antiretroviral therapy
.
J Neurovirol
.
2012
;
18
(
4
):
247
-
255
.
50.
Grulich
AE
,
van Leeuwen
MT
,
Falster
MO
,
Vajdic
CM
.
Incidence of cancers in people with HIV/AIDS compared with immunosuppressed transplant recipients: a meta-analysis
.
Lancet
.
2007
;
370
(
9581
):
59
-
67
.
51.
Gleber-Netto
FO
,
Zhao
M
,
Trivedi
S
, et al;
Head and Neck Cancer Specialized Program of Research Excellence (SPORE) Human Immunodeficiency Virus (HIV) Supplement Consortium
.
Distinct pattern of TP53 mutations in human immunodeficiency virus-related head and neck squamous cell carcinoma
.
Cancer
.
2018
;
124
(
1
):
84
-
94
.
52.
Ziada
AS
,
Lu
MY
,
Ignas-Menzies
J
, et al;
CIHR team grant on cellular aging, HIV comorbidities in women, children (CARMA)
.
Mitochondrial DNA somatic mutation burden and heteroplasmy are associated with chronological age, smoking, and HIV infection
.
Aging Cell
.
2019
;
18
(
6
):
e13018
.
53.
Hanahan
D
,
Weinberg
RA
.
Hallmarks of cancer: the next generation
.
Cell
.
2011
;
144
(
5
):
646
-
674
.
54.
Morscio
J
,
Dierickx
D
,
Ferreiro
JF
, et al
.
Gene expression profiling reveals clear differences between EBV-positive and EBV-negative posttransplant lymphoproliferative disorders
.
Am J Transplant
.
2013
;
13
(
5
):
1305
-
1316
.
55.
Nicolae
A
,
Pittaluga
S
,
Abdullah
S
, et al
.
EBV-positive large B-cell lymphomas in young patients: a nodal lymphoma with evidence for a tolerogenic immune environment
.
Blood
.
2015
;
126
(
7
):
863
-
872
.
56.
Marcelis
L
,
Antoranz
A
,
Delsupehe
AM
, et al
.
In-depth characterization of the tumor microenvironment in central nervous system lymphoma reveals implications for immune-checkpoint therapy
.
Cancer Immunol Immunother
.
2020
;
69
(
9
):
1751
-
1766
.
57.
Aquaro
S
,
Caliò
R
,
Balzarini
J
,
Bellocchi
MC
,
Garaci
E
,
Perno
CF
.
Macrophages and HIV infection: therapeutical approaches toward this strategic virus reservoir
.
Antiviral Res
.
2002
;
55
(
2
):
209
-
225
.
58.
Soussain
C
,
Choquet
S
,
Blonski
M
, et al
.
Ibrutinib monotherapy for relapse or refractory primary CNS lymphoma and primary vitreoretinal lymphoma: Final analysis of the phase II “proof-of-concept” iLOC study by the Lymphoma Study Association (LYSA) and the French Oculo-cerebral Lymphoma (LOC) network
.
Eur J Cancer
.
2019
;
117
:
121
-
130
.
59.
Grommes
C
,
Pastore
A
,
Palaskas
N
, et al
.
Ibrutinib unmasks critical role of bruton tyrosine kinase in primary CNS lymphoma
.
Cancer Discov
.
2017
;
7
(
9
):
1018
-
1029
.
60.
Lionakis
MS
,
Dunleavy
K
,
Roschewski
M
, et al
.
Inhibition of B cell receptor signaling by ibrutinib in primary CNS lymphoma
.
Cancer Cell
.
2017
;
31
(
6
):
833
-
843
.
61.
Lewis
KL
,
Chin
CK
,
Manos
K
, et al
.
Ibrutinib for central nervous system lymphoma: the Australasian Lymphoma Alliance/MD Anderson Cancer Center experience
.
Br J Haematol
.
doi:10.1111/bjh.16946
.
62.
Ryan
CE
,
Sahaf
B
,
Logan
AC
, et al
.
Ibrutinib efficacy and tolerability in patients with relapsed chronic lymphocytic leukemia following allogeneic HCT
.
Blood
.
2016
;
128
(
25
):
2899
-
2908
.
63.
Stiff
A
,
Trikha
P
,
Wesolowski
R
, et al
.
Myeloid-derived suppressor cells express Bruton’s tyrosine kinase and can be depleted in tumor-bearing hosts by ibrutinib treatment
.
Cancer Res
.
2016
;
76
(
8
):
2125
-
2136
.
64.
Kuo
HP
,
Ezell
SA
,
Hsieh
S
, et al
.
The role of PIM1 in the ibrutinib-resistant ABC subtype of diffuse large B-cell lymphoma
.
Am J Cancer Res
.
2016
;
6
(
11
):
2489
-
2501
.
65.
Trotman
J
,
Opat
S
,
Gottlieb
D
, et al
.
Zanubrutinib for the treatment of patients with Waldenström macroglobulinemia: 3 years of follow-up
.
Blood
.
2020
;
136
(
18
):
2027
-
2037
.
66.
Alame
M
,
Pirel
M
,
Costes-Martineau
V
, et al
.
Characterisation of tumour microenvironment and immune checkpoints in primary central nervous system diffuse large B cell lymphomas
.
Virchows Arch
.
2020
;
476
(
6
):
891
-
902
.
67.
Bollard
CM
,
Rooney
CM
,
Heslop
HE
.
T-cell therapy in the treatment of post-transplant lymphoproliferative disease
.
Nat Rev Clin Oncol
.
2012
;
9
(
9
):
510
-
519
.
68.
Khanna
R
,
Moss
D
,
Gandhi
M
.
Technology insight: applications of emerging immunotherapeutic strategies for Epstein-Barr virus-associated malignancies
.
Nat Clin Pract Oncol
.
2005
;
2
(
3
):
138
-
149
.
69.
McLaughlin
LP
,
Bollard
CM
,
Keller
MD
.
Adoptive T cell therapy for Epstein-Barr virus complications in patients with primary immunodeficiency disorders
.
Front Immunol
.
2018
;
9
:
556
.
70.
Haque
T
,
Wilkie
GM
,
Jones
MM
, et al
.
Allogeneic cytotoxic T-cell therapy for EBV-positive posttransplantation lymphoproliferative disease: results of a phase 2 multicenter clinical trial
.
Blood
.
2007
;
110
(
4
):
1123
-
1131
.
71.
Gandhi
MK
,
Wilkie
GM
,
Dua
U
, et al
.
Immunity, homing and efficacy of allogeneic adoptive immunotherapy for posttransplant lymphoproliferative disorders
.
Am J Transplant
.
2007
;
7
(
5
):
1293
-
1299
.

Supplemental data

Sign in via your Institution