Next Article in Journal
Management of VEGFR-Targeted TKI for Thyroid Cancer
Next Article in Special Issue
Impact of Tumour Epstein–Barr Virus Status on Clinical Outcome in Patients with Classical Hodgkin Lymphoma (cHL): A Review of the Literature and Analysis of a Clinical Trial Cohort of Children with cHL
Previous Article in Journal
Galectins in Glioma: Current Roles in Cancer Progression and Future Directions for Improving Treatment
Previous Article in Special Issue
New Look of EBV LMP1 Signaling Landscape
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Clinical and Therapeutic Implications of Epstein–Barr Virus in HIV-Related Lymphomas

by
Miriam Verdu-Bou
1,
Gustavo Tapia
2,
Agueda Hernandez-Rodriguez
3 and
Jose-Tomas Navarro
1,4,5,*
1
Lymphoid Neoplasms Group, Josep Carreras Leukaemia Research Institute, Can Ruti Campus, 08916 Badalona, Spain
2
Department of Pathology, Germans Trias i Pujol Hospital, Universitat Autònoma de Barcelona, 08916 Badalona, Spain
3
Department of Microbiology, Germans Trias i Pujol Hospital, Universitat Autònoma de Barcelona, 08916 Badalona, Spain
4
Department of Hematology, Institut Català d’Oncologia-Germans Trias i Pujol Hospital, 08916 Badalona, Spain
5
Department of Medicine, Universitat Autònoma de Barcelona, 08916 Badalona, Spain
*
Author to whom correspondence should be addressed.
Cancers 2021, 13(21), 5534; https://doi.org/10.3390/cancers13215534
Submission received: 20 October 2021 / Accepted: 29 October 2021 / Published: 4 November 2021
(This article belongs to the Special Issue Epstein-Barr Virus Infection in Cancer)

Abstract

:

Simple Summary

Epstein–Barr virus (EBV) is involved in lymphomagenesis, especially lymphomas affecting populations with immunodeficiencies, such as people living with HIV (PLWH). The pathogenic roles of EBV in lymphomas arising in PLWH are mediated by several viral proteins, as well as cooperation between EBV and HIV. The presence of EBV in these lymphomas conditionate some of their epidemiological, pathological, and clinical characteristics, as well as their prognosis. In this article, the authors review the different EBV-associated lymphomas affecting PLWH, analyzing the influence of EBV on the epidemiology, etiopathogenesis, clinical features, treatment, diagnosis and prognosis of each lymphoma subtype. Furthermore, new EBV-targeted therapies currently under development for some lymphomas are discussed.

Abstract

The incidence of lymphomas is increased in people living with HIV (PLWH). Aggressive B-cell non-Hodgkin lymphomas (NHLs) are the most common and are considered an AIDS-defining cancer (ADC). Although Hodgkin lymphoma (HL) is not considered an ADC, its incidence is also increased in PLWH. Among all HIV-related lymphomas (HRL), the prevalence of Epstein–Barr virus (EBV) is high. It has been shown that EBV is involved in different lymphomagenic mechanisms mediated by some of its proteins, contributing to the development of different lymphoma subtypes. Additionally, cooperation between both HIV and EBV can lead to the proliferation of aberrant B-cells, thereby being an additional lymphomagenic mechanism in EBV-associated HRL. Despite the close relationship between EBV and HRL, the impact of EBV on clinical aspects has not been extensively studied. These lymphomas are treated with the same therapeutic regimens as the general population in combination with cART. Nevertheless, new therapeutic strategies targeting EBV are promising for these lymphomas. In this article, the different types of HRL are extensively reviewed, focusing on the influence of EBV on the epidemiology, pathogenesis, clinical presentation, and pathological characteristics of each lymphoma subtype. Moreover, novel therapies targeting EBV and future strategies to treat HRL harboring EBV are discussed.

1. Introduction

People living with HIV (PLWH) are at higher risk of developing aggressive B-cell lymphomas than the general population, and B-cell non-Hodgkin lymphoma (NHL) is currently the most frequent AIDS-defining cancer (ADC) [1,2]. Among NHL, diffuse large B-cell lymphoma (DLBCL) is the most common, including primary central nervous system lymphoma (PCNSL), followed by Burkitt lymphoma (BL). The frequency of other lymphoma subtypes, such as plasmablastic lymphoma (PBL) and primary effusion lymphoma (PEL) is lower, but they more frequently affect PLWH [3]. While the incidence of aggressive B-cell NHL has decreased in PLWH since the widespread use of combined antiretroviral therapy (cART), lymphoma is still an important cause of morbidity and mortality among this population [4,5]. On the other hand, although Hodgkin lymphoma (HL) is not considered an ADC, its incidence increased among PLWH during the first years after the introduction of cART [3,6], and eventually has remained stable or with only a slight increase [7,8].
The pathogenesis of HIV-related lymphomas is complex and influenced by immunosuppression and coinfection with oncogenic viruses, mainly human herpes virus 8 (HHV-8) and Epstein–Barr virus (EBV), two human γ-herpesviruses that establish latency in the host B-cell nucleus. However, the prevalence of EBV is higher in HIV-related lymphomas (HRLs) and is therefore implicated in the pathogenesis of these lymphomas [9,10,11,12]. Some components of the virus related to the latent state, such as EBV latent membrane proteins (LMP1s and LMP2A/B), EBV-nuclear antigens (EBNA1, EBNA2, EBNA3A, EBNA3B, EBNA3C, EBNA leader protein (EBNA-LP), EBV-encoded small RNAs (EBERs), and microRNAs (miRNAs) play important roles in lymphomagenesis [13]. The EBV latency type differs among HRLs, suggesting that EBV may regulate different processes related to B-cell transformation and evasion of the immune system [14,15,16,17,18,19]. The EBV-encoded miRNAs have a role in the lymphomagenesis, regulating different cellular pathways such as apoptosis, proliferation, immune recognition, and microenvironment [20]. EBV-miRNAs are located in two regions of the viral genome: the BamHI-A region rightward transcript (BART) and BamHI-H rightward fragment 1 (BHRF1) [21]. The expression of viral miRNAs differs according to the latency type; while BART miRNAs are expressed in all latency types (mainly in latency I and II), BHRF1–3 miRNAs are expressed almost exclusively in the latency III type [21]. During the latent state, EBV can intermittently reactivate, expressing lytic genes that are also involved in lymphoma development [22].
Furthermore, cooperation between HIV and EBV in HRLs has been speculated, with HIV likely contributing to the generation of a permissive microenvironment for EBV infection, and the differentiation and survival of infected B-cells [18,23,24].
In addition to the presence of EBV in lymphoma tissue, the virus can be detected in peripheral blood of patients with lymphoma. Some studies have suggested the usefulness of EBV load as a lymphoma biomarker with diagnostic and prognostic implications in PLWH [19,25,26,27].
There is great interest and a need to develop new EBV-targeted therapies for the treatment of patients with EBV-driven lymphomas. Multiple EBV-target strategies have been studied in HRLs in preclinical studies, but few clinical trials have been carried out [28].
This article is a review of the state of the art of the implications of EBV in different clinical, epidemiological, and etiopathogenical aspects of HRL, as well as a description of the EBV-targeted therapies currently under development for the treatment of these lymphomas.

2. Implications of Epstein–Barr Virus in the Different HIV-Related Lymphoma Types

2.1. Diffuse Large B-Cell Lymphoma

Diffuse large B-cell lymphoma is an aggressive B-cell neoplasm that can be classified, according to the cell-of-origin (COO), into germinal center B-cell (GCB) and activated B-cell (ABC) [29]. Two entities have classically been considered, systemic DLBCL and DLBCL with exclusive involvement of the central nervous system (Figure 1) [30]. Although the WHO classification reserves the term primary DLBCL of the central nervous system (PCNSL) for immunocompetent patients, PLWH typically have DLBCL with exclusive involvement of the CNS, which is considered an AIDS-defining condition, thus the term PCNSL is commonly found in the literature in the HIV setting [31].

2.1.1. Epidemiology

Systemic DLBCL is the most common NHL in both PLWH and the general population. Although its incidence in PLWH has diminished since the widespread use of cART, DLBCL is still an important cause of morbidity and mortality among this population [15,32,33,34]. EBV infection is present in 30–50% of HIV-related DLBCL cases, a frequency higher than that of the general population [15,19,35,36,37,38]. According to the COO, EBV is more frequently found in ABC-DLBCL (44–74%) than in GCB-DLBCL (13–25%) [15,37,39]. To our knowledge, in PLWH with DLBCL, no differences regarding age and gender have been reported between EBV-positive and EBV-negative cases [19,37,39,40].
PCNSL is an infrequent lymphoma affecting 15 per 100,000 persons-year in PLWH [32]. HIV is considered a risk factor in this lymphoma, but its incidence has dramatically decreased in the cART era [31,32,41,42]. Nearly all cases (80–100%) of HIV-related PCNSL are associated with EBV [27,43,44,45,46].

2.1.2. Etiopathogenesis

As reported by Arvey et al., the frequency of EBV latency types differs in PLWH depending on the DLBCL subtype [15]. They described that 76% of GCB-DLBCLs are associated with latency type I (LMP1−, EBNA2−), 12% with latency type II (LMP1+, EBNA2−), and 12% with latency III (LMP1+, EBNA2+) (Table 1). On the other hand, in ABC-DLBCLs, either latency types II or III were found in 30% of cases each, and latency type I in 37% of cases. These data suggest that EBV could be involved in different pathogenic mechanisms depending on the DLBCL COO subtype.
HIV-related DLBCL associated with EBV also presents high expression of Blimp1, a transcriptional repressor of TP53, which confers the capacity to escape from apoptosis and deregulate B-cell differentiation [19]. Furthermore, EBV-positive HIV-related DLBCL frequently expresses CD30, which is an NF-кB promoter [19]. It has been demonstrated that LMP1 activates the NF-кB signaling pathway and induces chronic B-cell activation, both being typical deregulated pathways in the ABC subtype [19,37,47]. Thus, EBV could have a synergistic effect on the lymphomagenesis of the HIV-related ABC subtype.
The expression of miR-BHRF1–3 has been observed in an increased frequency in primary cell lines and frozen samples from HIV-related DLBCL [48]. This miRNA targets C-X-C motif chemokine ligand 11 (CXCL-11), providing the cells the ability to escape from the immune system. The expression of different miR-BHRF1s have been also confirmed in frozen samples of HIV-related DLBCL [49]. In addition, these patients showed an elevated expression of miR-BARTs 15, 10-3p, 11-3p, and 14-3p compared with other EBV-related lymphomas in immunocompetent individuals [49].
Additionally, miR-BHRF1–2 inhibits PRDM1, preventing apoptosis and cell cycle arrest in lymphoblastoid cell lines (LCL) [50]. This EBV-miRNA could be a synergistic mechanism of the downregulation of PRDM1 in GCB-DLBCL. Furthermore, some EBV-miRNAs such as miR-BART3, miR-BART9, and miR-BART17-5p can downregulate BCL6, an NF-кB repressor [51].
The expression of EBV-lytic genes has been also described in lymphomas, and emerging studies of its impact on the lymphomagenesis are in progress [22]. Cohen et al. have reported an expression of the lytic proteins BZLF1, BHRF1, and BLLF1 in immunocompetent EBV-related DLBCL that could deregulate cellular pathways and contribute to the lymphomagenesis [52].
Additionally, HIV could be involved in the pathogenesis of DLBCL in PLWH. In this regard, Liapis et al. reported increased infiltration of CD8+ cytotoxic T-lymphocytes (CTL) in DLBCL tumors with expression of LMP1 and the HIV-1 p24 protein, which is related to active HIV replication [53]. Furthermore, it has been postulated that the HIV-1-matrix protein p17 persists after antiretroviral drug suppression and acts as a cytokine for T-cell activation and promotes angiogenesis [54,55]. In addition, p17 also can increase the expression of LMP1 in primary EBV-infected B-cell lymphocytes, as described by Martorelli et al. [16]. Both p17 and LMP1 are involved in the cell growth mediated by the Akt/ERK and STAT signaling pathways, suggesting cooperation between HIV and EBV in the proliferation of malignant B-cells and the lymphomagenesis of DLBCL [56].
In HIV-related PCNSL, EBV infection affects 80–100% of cases [57,58]. In these lymphomas, EBV latency III (including LMPs, EBNAs, and EBERs expression) is the most frequent latency type [59]. This lymphoma usually affects PLWH at advanced stages of immunosuppression, with very low CD4+ lymphocyte counts (median < 50 cells/µL) and with loss of immune response mediated by CTLs [58,60]. Although EBV does not replicate in the tissue of the central nervous system (CNS), the lax immune response produced by HIV facilitates EBV infection and the migration of infected B-cells to the CNS [58,61].

2.1.3. Impact of EBV on Clinical Features and Prognosis

Systemic HIV-related DLBCL usually has extranodal involvement and an advanced stage at diagnosis (III or IV) [19]. These patients usually present median CD4+ lymphocyte counts of 100–223 cells/µL [40,53,62]. The impact of EBV on the clinical features and prognosis of DLBCL in PLWH have been scarcely studied. Chao et al. found no differences in most of the clinical variables according to EBV status, but they found a significant reduction in CD4+ lymphocyte counts in EBV-positive compared with EBV-negative cases (mean 128 cells/µL vs. 248 cells/µL). These authors found that the period of time between HIV infection and the development of DLBCL tended to be shorter in EBV-positive compared to EBV-negative cases [19]. In addition, there was a trend toward a higher frequency of ABC cases in EBV-positive cases [19,63].
The impact of EBV on prognosis in HRL is a matter of controversy. In a study by Chao et al., the presence of EBV in tumoral tissue was associated with a worse overall survival (OS) [19]. However, Chadburn et al. described that the presence of EBV in lymphoma cells was not associated with poorer survival or low CD4+ lymphocyte counts [63].
PCNSL usually occurs in patients with advanced immunosuppressed status and low CD4+ lymphocyte counts (<50 cells/µL), and frequently with an AIDS-defining illness before lymphoma diagnosis [31,45,64,65,66]. In this regard, low CD4+ lymphocyte counts and high HIV load are associated with a worse outcome in patients with PCNSL [31,42]. Given that EBV coinfection is detected in nearly all HIV-PCNSL patients, the detection of EBV-DNA in cerebrospinal fluid (CSF) is a quick diagnostic tool for HIV-related PCNSL diagnosis, having a high sensitivity and specificity [45,67]. Thus, the elevated frequency of EBV infection and the advanced immunodeficiency conditions caused by HIV suggest that EBV plays a relevant role in the lymphomagenesis of PCNSL.

2.1.4. Treatment

Patients with systemic HIV-related DLBCL are treated with the same regimens as the general population. The gold standard treatment used in DLBCL is rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP). Additionally, it is highly recommended to add cART concomitantly with the immunochemotherapy because this strategy has been demonstrated to improve the outcome of these patients [68,69]. The recommended treatment for relapsed/resistant patients is the same as that used in the general population, and therefore they should receive second-line regimens containing drugs not included in the frontline treatment, followed by autologous stem cell transplantation (ASCT) (Table 2) [70].
There is no gold standard treatment for patients with HIV-related PCNSL. Patients with a good general condition currently receive induction treatment with high-dose methotrexate-based polychemotherapy in combination with cART [71,72]. In some patients, ASCT can be considered. Radiotherapy may be considered in patients without response to chemotherapy treatments [73].

2.2. Burkitt Lymphoma

Burkitt lymphoma is an aggressive NHL with a germinal center (GC) origin that can be classified as follows depending on the epidemiological characteristics: endemic BL, sporadic BL, and immunodeficiency-associated BL, which is mainly associated with HIV infection (Figure 1) [29,74].

2.2.1. Epidemiology

Similar to DLBCL, PLWH have a higher incidence of BL than the general population, and BL comprises 20–30% of HRL [29,75]. HIV-related BL is more frequent among men (86.7%) with a median age of 39 years and is associated with EBV in 30–60% of cases [76,77,78,79,80]. This entity mainly develops in individuals at an early stage of HIV infection with a moderate reduction of CD4+ lymphocyte counts (200–270 cells/µL) [77,81]. Unlike DLBCL, the introduction of cART did not lead to a decrease in the number of BL cases diagnosed in PLWH, and the incidence has remained stable in recent years [82,83]. Regarding pathologic features, plasmacytoid differentiation is characteristic of HIV-related BL, and is found in 50–70% of EBV-infected cases [57,77,84].

2.2.2. Etiopathogenesis

The causes and mechanisms that lead to the development of BL have not been completely elucidated. The translocation of MYC with the immunoglobulin heavy-chain loci (IgH) and inactivation of p53 are hallmarks of the pathogenesis of BL [80,85]. In HIV-related BL, MYC translocations and TP53 mutations are detected in 90–100% and 60% of cases, respectively [78,80]. The translocation of MYC leads to constitutive activation of MYC, increasing proliferation, so these tumors are characterized by their high proliferative rates [57].
Among cases with EBV coinfection, latency type I (EBERs+ and EBNA1+ expression) is the most common EBV latency type (90%), although it is possible to find latency type II (EBERs+, EBNA1+, LMP1+) in a few cases (10%) (Table 1) [15,59]. LMP1 and EBNA2 have antagonistic effects to Myc in the phenotype of BL B-cells. This could be the cause of the reduced frequency of latency type II and the absence of latency type III in this lymphoma [86].
The EBNA1 protein not only maintains the EBV episomal genome in the host cells and regulates extrachromosomal replication, but may also deregulate host gene expression [17,87]. This protein has an antiapoptotic effect, since it decreases p53 stability and thus favors the survival of tumoral cells [88,89]. Furthermore, Zhang et al. described a hypermethylation of PRDM1 (the gene that encodes the Blimp1 protein) exclusively in EBV-positive BL, inactivating this gene [90]. The overexpression of this gene results in cell cycle arrest, thus the inactivation of PRDM1 could be beneficial for cell growth and contribute to lymphomagenesis of BL in EBV-infected cases (Figure 2) [90].
Additionally, MYC could alter immune system response by reducing the activity of the NF-кB pathway. This effect, in combination with the poor antigenic property of EBNA1, leads EBV-infected B-cells to acquire the ability to escape from immune system recognition [75]. Moreover, the absence of the immunogenic proteins EBNA2 and LMP1 in EBV-positive HIV-related BL could be a self-defense mechanism of the virus to remain in host cells and escape the immune system [11,16].
The EBV lytic protein, BHRF1, confers protection from apoptosis in EBV-positive BL cell lines, since it is a homolog of the antiapoptotic protein BCL-2 and negatively regulates the proapoptotic protein Bim [91,92]. This phenomenon can avoid the cell death associated with genetic alterations in MYC, being a cooperative mechanism of Myc-driven lymphomagenesis and favoring the chemoresistance [92]. Another lytic protein, BZLF1, directly inhibits p53 in BL cell lines and could be an alternative mechanism for the inhibition of this protein, in addition to the inactivating mutations of TP53 [93].
Moreover, EBV-miR-BARTs are upregulated in EBV-positive HIV-related BL and could deregulate the host gene and miRNA expression in these tumors [17]. In particular, EBV-negative immunodeficiency-BL presents a downregulated expression of miR-BART6-3p compared with EBV-positive cases. This miRNA may have an impact on the proliferation, cell growth, and apoptosis in the BL cell line, downregulating the expression of PTEN, a negative regulator of the Akt/PI3K signaling pathway [94,95]. In addition, miR-BART6-3p regulates the expression of the IL-6 receptor (IL-6R), being able to contribute to immune system evasion [94]. Furthermore, it has been postulated that EBV-miRNAs have a role in the microenvironment regulation, reducing the innate and adaptative immune response in EBV-positive tumors, similar to BL (Figure 2) [96].
Moreover, EBERs induce the IL-10 expression in EBV-BL cell lines, suggesting an EBV involvement in the tumor growth [97]. They bind protein kinase RNA-activated (PKR), a protein involved in interferon-α (IFN-α)-mediated apoptosis, avoiding its phosphorylation [98]. Therefore, EBERs can be also involved in apoptosis resistance.

2.2.3. Impact of EBV on Clinical Features and Prognosis

To our knowledge, studies of the clinical features of EBV-positive BL specifically focused on PLWH have not been carried out. However, HIV-related BL has been more extensively studied. Patients usually develop BL early in HIV disease, and they still maintain a moderate amount of CD4+ lymphocyte counts (greater than 200 cells/µL), often without a history of opportunistic infections (Table 1) [81]. This lymphoma presents an aggressive clinical behavior in advanced stages (III and IV), with nodal and bone marrow involvement [29,99,100,101]. If relapse occurs, it is usually seen within the first year after response and can be associated with CNS involvement [102]. In the cART era, the prognosis of HIV-related BL has improved, and similar responses and survival probabilities are now achieved compared with those of non-HIV-infected individuals [103,104].

2.2.4. Treatment

The treatment of HIV-related BL is based on intensive chemotherapeutic schemes as in the general population, but in combination with cART [84]. The chemotherapeutic strategies include cyclophosphamide, vincristine, doxorubicin, methotrexate/ifosfamide, etoposide, cytarabine (CODOX-M/IVAC) [105]; cyclophosphamide, methotrexate, dexamethasone, ifosfamide, vincristine, etoposide, cytarabine, doxorubicin, and vindesine in combination with rituximab (Burkimab) [103]; cyclophosphamide, vincristine, doxorubicin, and dexamethasone with methotrexate and high-dose cytarabine (hyper-CVAD); or dose-adjusted etoposide, prednisone, vincristine, cyclophosphamide, and doxorubicin with rituximab (DA-EPOCH-R) [106]. There is no standard second-line treatment for this lymphoma, and the few refractory/relapsed patients should be treated with experimental strategies (Table 2).

2.3. Plasmablastic Lymphoma

Plasmablastic lymphoma is a B-cell NHL that most commonly occurs in PLWH and is characterized by loss of GC B-cell markers and the expression of plasma cell markers and features of an activated B-cell (Figure 1) [29].

2.3.1. Epidemiology

PBL is an uncommon lymphoma with an aggressive clinical course, with a median OS of 6–11 months [29,107]. This lymphoma presents more frequently in men (80%) with a median age of 39–46 years. It is closely related to HIV infection, representing around 2–12% of HRL [108,109,110,111,112] and in most cases the cells are also infected by EBV (75–100%) [113,114,115].

2.3.2. Etiopathogenesis

The genetic and molecular characterization of PBL has not been clearly described. Nevertheless, recent studies have revealed that dysregulating mutations in the JAK-STAT and RAS-MAPK pathways are genetic signatures of PBL. These mutations mostly affect STAT3, TP53, and RAS family members, and with less frequency affect MYC, EP300, CARD11, SOCS1, and TET2 [116,117,118,119]. Additionally, MYC translocation is the most frequent genetic alteration found [114,120,121], being more frequent in HIV-related PBL than in PBL in the general population (78% vs. 44%). In addition, it is more common in EBV-positive than in EBV-negative PBL (57–74 vs. 20–43%) [118,121,122]. The high frequency of EBV among HIV-related PBL reported in many studies indicates a strong association between the two viruses, and EBV latency I is the most common pattern (Table 1), but the latency III pattern has been observed as well [108,109,114,121,123,124]. Additionally, some studies describe different genetic alterations depending on the EBV status. Mutations in TP53 are more frequent among EBV-negative compared with EBV-positive PBL [116,117], while mutations in STAT3 and SOCS1 are more common in EBV-positive cases [116]. Garcia-Reyero et al. reported that EBV-related PBL cases presented greater genomic stability than EBV-negative cases (87.5% vs. 54%), as well as a different mutational profile [118]. In this regard, PRDM1 and STAT3 mutations are only detected in EBV-related PBL, and most are also HIV-related [118]. Blimp1 is a transcriptional repressor of MYC in transformation of a B-cell to a plasma cell, so coexpression of both proteins would not be expected in this lymphoma [125]. However, both proteins are usually expressed in HIV-related PBL [122]. In this regard, Montes-Moreno et al. described that PRDM1 mutations affect functional domains involved in the regulation of other genes, which could avoid the negative regulation of Myc, but not B-cell terminal differentiation. Thus, PRDM1 mutations would reinforce Myc-driven lymphomagenesis [122]. This could also explain the discontinuation of plasmacytic differentiation at the plasmablast stage [126].
Given the biological similarities shared between PBL and BL extramedullary plasmacytoma (EMPC), a recent study revealed different EBV-miRNAs expression among these groups. A total of 38 EBV-miR-BARTs were upregulated in PBL compared with EMPC, while 19 EBV-miR-BARTs were downregulated in PBL compared with BL, suggesting that EBV-miRNAs could be useful for the differential diagnosis of PBL [127].
The expression of EBV lytic proteins in lymphomas could contribute to the lymphomagenesis. In this regard, PBL shows expression of EBV proteins related with lytic cycle such as BZLF-1/ZEBRA, BHRF-1/Ea-R, and BMRF-1/Ea-D, but absence of BLLF1/gp50 expression [127]. Other authors have recently described a higher expression of the lytic genes BALF4 and BALF5 in HIV-related PBL in EBV-positive cases [119].
EBV-related PBL shows an increased number of infiltrating T-cells, natural killer (NK) cells, and protumorigenic M2-macrophages, compared to EBV-negative PBL, and in turn presents higher activation of immune evasion mechanisms. In this regard, EBV-related PBL present an overexpression of programmed cell death ligand 1 (PD-L1) that is associated with immune evasion by tumoral cells [128]. These mechanisms could lead to the construction of a favorable microenvironment for lymphoma development.

2.3.3. Impact of EBV on Clinical Features and Prognosis

Extranodal involvement is very frequent in PBL in PLWH (95% of cases) at diagnosis, and in these patients the lymphoma usually presents at advanced stages (III or IV) [108,109,112,115,120,129]. The oral cavity is the most frequent extranodal site (48–58%) [29,108,113,129]. Schommers et al. reported that some clinical features such as age older than 60, high serum lactate dehydrogenase, an Eastern Cooperative Oncology Group (ECOG) score > 2, and an International Prognostic Index (IPI) score of 2–5 are risk factors associated with worse outcomes [26]. The translocation of MYC has been associated with shorter OS compared to patients with non-rearranged MYC [114]. In one study, PLWL with MYC gene rearrangements were associated with a 6-fold increased risk of death [130]. Patients with EBV-related PBL have a worse prognosis than EBV-negative cases [112]. The elevated incidence of EBV infection could be useful for the differential diagnosis with other B-cell neoplasm with similar features.
While the prognosis of PBL has improved since the introduction of cART, these patients have a poor outcome, with a median OS of 10–15 months [107,108,129]. In this regard, the median CD4+ lymphocyte count at diagnosis is 63–165 cells/µL, indicating a status of important immunodeficiency (Table 1) [26,131,132]. Low CD4+ lymphocyte counts and the absence of cART treatment prior to lymphoma are considered factors of worse prognosis [112]. The impact of EBV on the prognosis of PBL is controversial, with some studies not reporting an influence of EBV infection on OS [26,108,124,129], whereas others have reported better outcomes in EBV-positive than in EBV-negative PBL in PLWH [114,124,133].

2.3.4. Treatment

There is no standard treatment for PBL, although intensive treatment is recommended. PLWH patients are currently treated with the same regimens as those of the general population, in combination with cART, such as CODOX-M/IVAC, hyper-CVAD, or DA-EPOCH [112]. Plasmablastic differentiation of neoplastic cells has led to testing drugs against multiple myeloma. The proteasome inhibitor bortezomib has been used alone and in combination with other drugs. In this sense, the combination with DA-EPOCH has shown good results in a retrospective series, with a 5-year OS of 63% [134].
The role of ASCT in the treatment of PBL has not been well studied. It could be used as a first- or second-line treatment, although further studies are needed to confirm possible beneficial effects (Table 2) [135,136].

2.4. Primary Effusion Lymphoma

Primary effusion lymphoma is a rare and aggressive NHL appearing mainly in HIV-infected patients in the form of body cavity effusion, although there are also solid forms. The lymphoma cells have a plasma cell phenotype and are positive for HHV-8 [29], and elevated percentage of cases are also positive for EBV (Figure 1) [11,29,35,137,138]. The presence of EBV in the cells is a useful diagnostic tool to differentiate this lymphoma from HHV-8-positive DLBCL, not otherwise specified (NOS), a similar entity that overlaps histological features, since this entity is EBV-negative [29].

2.4.1. Epidemiology

This lymphoma occurs mainly in men (90%) with a median age of 50–55 years and advanced AIDS disease. Two-thirds of the patients are HIV-infected (representing 3–5% of HIV-NHLs), and in this group the median age is 40–45 years, and around 70 years in HIV-negative individuals [29,138,139,140]. In addition, it is also closely related to EBV, which accounts for 70–100% of HIV-PEL [11,29,35,137,138].
This lymphoma also occurs in patients with other immunodeficiencies, such as liver cirrhosis and recipients of solid organ transplants. Moreover, some cases of PEL have been reported in elderly patients with positive serology for HHV-8 without any cause of immunodeficiency [141].
Approximately one-third of the patients have another HHV-8-related disease, such as Kaposi’s sarcoma, multicentric Castleman disease, or both [142].

2.4.2. Etiopathogenesis

The neoplastic cells of PEL show low expression of mature B-cell genes and high expression of post-GC markers [29]. In addition, expression profile studies have revealed overexpression of plasma cell genes, indicating that the COO is a post-GC B-cell at an advanced stage of B-cell differentiation (Figure 1) [143].
HHV-8 is a DNA virus strongly implicated in the pathogenesis of PEL. This virus prevents apoptosis and activates NF-кB, promoting proliferation and indicating that HHV-8 is involved in PEL survival [144,145]. This virus has been related to the immune evasion system since it can increase the expression of PD-L1 [142]. In addition, most individuals with PEL are also coinfected by EBV. Latency I, with expression of EBNA1 and EBERs, is the most frequent subtype (90–92%), although it is possible to detect latency II (8%) with LMP1 expression in some cases (Table 1) [11,15,138,140]. The role of EBV in the pathogenesis of PEL remains unclear; however, the MAPK signaling pathway seems to differentiate EBV-positive and EBV-negative PEL, and together with HHV-8, could contribute to the proliferation and evasion of programmed cell death [10,57]. Curiously, Roy et al. have reported that EBV-negative PEL cell lines present more copy number variations than EBV-positive, thus indicating that EBV could maintain the genomic stability of host cells [146]. Nevertheless, the common expression of latency type I, without expression of immunogenic EBV proteins, suggests that EBV is not the only mechanism responsible for the pathogenesis of PEL [35,147,148]. In PEL cell lines, some EBV-miR-BARTs are expressed, but these cells do not express EBV-miR-BHRF1s. This miRNAs expression pattern is characteristic of EBV latency I [149]. It has been speculated that EBV cooperates with HHV-8 in the infection of B-cells, and HIV may contribute to lymphomagenesis, generating a permissive microenvironment [150].
In a mice model with both HHV-8 and EBV infection, HHV-8 increases the expression of EBV-lytic genes, especially BZLF1 and BALF2, which lead to increase tumor growth. A high expression of EBV lytic genes has been also described in EBV and HHV-8 dual-infected patients with lymphoproliferative disorders, including PEL [151]. On the other hand, the EBV-protein EBNA-1 contributes to the survival of HHV-8 infected B-cells in PEL [152]. These results suggest a cooperation of both viruses (HHV-8 and EBV) in the lymphomagenesis of PEL.

2.4.3. Clinical Features and Prognosis

Effusion(s) of body cavities (pleural, peritoneal, and/or pericardial) without tumor mass is the typical presentation of PEL [11,35]. By definition, all cases with this presentation (effusion) are at stage IV [139,140,153,154]. In addition, these patients usually present advanced AIDS disease with low CD4+ lymphocyte counts (median 98–133 cells/µL) (Table 1) [137,138,139,155]. Patients present with symptoms related to the affected cavity, with the pleura being the most frequent, followed by peritoneal and pericardial. Solid forms of PEL present with symptoms related to the affected organ, with the gastrointestinal tract being the most frequent [137,138].
The prognosis of PEL is poor, with a median OS of less than 12 months [29,137,154,156,157]. The prognostic factors of HIV-related PEL are controversial. Some authors report that the administration of cART before PEL development is a prognostic factor, since patients that received cART have a longer OS [137,158]. However, other authors did not observe this prolonged OS with cART administration [84]. In this regard, Boulanger et al. studied a series of 28 patients and reported that a bad performance status and absence of cART before lymphoma diagnosis were factors that had a negative influence on prognosis [137]. Furthermore, Lurain et al. described, in HIV-related PEL, an association between elevated interleukin-6 (IL-6) levels and shorter OS, while EBV infection correlated with longer survival [159].

2.4.4. Treatment

There is no standard first-line treatment for PEL, and the available therapies present very poor results. Treatment with CHOP has achieved responses of 20–50% [155], while DA-EPOCH increases the complete response ratio and prolongs OS in patients with HIV-related PEL [140,159,160]. There is no established second-line treatment, but ASCT can be a therapeutic option, given the poor prognosis of this lymphoma [161,162]. Importantly, all of these treatments should be administered in combination with cART (Table 2).

2.5. Hodgkin Lymphoma

Hodgkin lymphoma is divided into two subtypes with different morphological and immunophenotypic features: nodular lymphocyte predominant HL and classic HL (cHL), which is related to PLWH [29,163,164,165]. In this lymphoma, Hodgkin Reed–Sternberg cells (HRS) are characteristically observed in a heterogeneous background of lymphocytes, eosinophils, neutrophils, macrophages, and plasma cells.

2.5.1. Epidemiology

The incidence of HL in PLWH is around 50 per 100,000 cases/year, and has increased since the introduction of cART, with the risk being 5–25 times higher than in the general population [29,166]. The incidence of cHL has remained stable after the increase observed in the first years of the cART era [7,8,167]. At cHL diagnosis, CD4+ lymphocyte counts are moderately decreased (median between 150–260 cells/μL), indicating a non-severe immunosuppression (Table 1) [6,8,167,168,169,170,171]. Coinfection with EBV occurs in 90–100% of cases, compared to 30–40% in HIV-negative patients) [172]. In HIV-related cHL, mixed cellularity is the most common subtype, followed by lymphocyte depletion and nodular sclerosis [29,35,173].

2.5.2. Etiopathogenesis

The malignant cells of cHL, which are HRS cells, seem to be GC-derived B-cells, as they carry somatic hypermutations, and this phenomenon is performed exclusively in the GC [174]. Therefore, aberrant mutations in BCR appear, and they are present in nearly all cHL. Unlike what occurs in normal conditions, these cells can escape from apoptosis. These events point to an origin in a preapoptotic GC-derived B-cell (Figure 1) [174,175].
Virtually all HIV-related cHLs are coinfected by EBV, and are latency II type, with expression of EBNA1, LMP1, and LMP2, as well as EBER and BART miRNAs. (Table 1) [176,177,178,179]. In this context, besides promoting cell cycle and inhibiting apoptosis, the LMP2 protein mimics BCR signaling and enables the survival of these cells in the absence of functional host BCR expression [179,180,181,182]. LMP1 promotes diverse signaling pathways involved in cell cycle progression, proliferation, and apoptosis, such as NF-кB, MAPK, PI3K/Akt, and JAK/STAT, and mimics CD40, a receptor necessary for B-cell activation [183,184]. In addition, EBV induces the overexpression of PD-L1 in HRS, leading to escape from immune response [185,186]. All of these mechanisms contribute to the proliferation and tumor progression of EBV-infected HRS. At the same time, HIV proteins such as gp120 and Tat reinforce chronic B-cell activation and promote the production of cytokines involved in the promotion of B-cell proliferation [187].
How the microenvironment influences the pathogenesis of cHL is not well understood. EBV-positive cHL frequently presents mixed cellularity with increased infiltration of NK cells, macrophages (M1 proinflammatory and M2 protumorigenic), CD4+ T-cells, and CTLs [188]. Although this tumor exhibits an increased infiltration of CTLs, EBV could inhibit their response by contributing to a permissive immunologic microenvironment [189]. Under immunodeficiency conditions, in cHL there is a depletion of CD4+ T-cells and an increased infiltration of M1 macrophages, and hence, HIV may also contribute to this permissive microenvironment [190]. Macrophage phenotypes could be regulated via EBV. Although the role of EBV-miRNAs has been scarcely studied so far in HL, EBV-BART miRNAs can be transferred to macrophages via exosomes, inducing the transformation of macrophages into a proinflammatory phenotype [191]. EBV-BART13-3p is the most expressed EBV-miRNA in HL, and this fact could transform macrophages and contribute to lymphoma microenvironment (Figure 2) [49].

2.5.3. Impact of EBV on Clinical Features and Prognosis

Classical HL affecting PLWH is virtually always EBV-positive and is characterized by aggressive clinical characteristics, such as advanced stages (III/IV), bone marrow and multiple nodal involvement, and B symptoms. The proportion of males affected (80–98%) is higher than in HIV-negative subjects, and some studies have shown that the age at diagnosis is higher in PLWH, with a median age of 40–44 years, than in the general population [166,192,193,194].
The prognosis of HL in PLWH is similar to that of the general population using the same therapeutic strategies in both groups. The presence of EBV in the lymphoma cells has not been reported to have any impact on prognosis.

2.5.4. Treatment

The treatment of cHL in PLWH is the same as that of the general population, based on chemotherapy with or without radiotherapy, in localized stages, and chemotherapy in advanced stages. The treatments most currently used are doxorubicin, bleomycin, vinblastine, and dacarbazine (ABVD) and bleomycin, etoposide, doxorubicin, cyclophosphamide, vincristine, procarbazine, and prednisone (BEACOPP) [195,196]. Relapses of cHL in PLWH can be treated with second-line drugs followed by ASCT (Table 2) [197,198].

3. Implications of EBV Load in HIV-Related Lymphoma

Several studies have evaluated the impact of EBV load on the diagnosis and prognosis of NHL and HL in the immunocompetent population [199,200]. In this regard, EBV loads measured in plasma and peripheral blood mononuclear cells (PBMCs) can serve as a diagnostic tool and could also have prognostic impact on lymphomas [201,202,203,204,205,206,207]. Fewer studies have been performed measuring EBV in peripheral blood of PLWH to determine its value as a diagnostic tool and prognostic factor. In this regard, the detection of EBV load has been reported to be more frequent among lymphomas in PLWH than in the general population [25,208,209]. Moreover, some studies, including ours, have suggested that EBV load measured in plasma could be a useful tool for the diagnosis of NHL and HL in PLWH, since EBV has been detected in plasma of these patients at lymphoma diagnosis [210,211]. This fact has also been demonstrated in PBMCs and serum in HRL [212]. In our recent study by Muncunill et al., we reported a strong association between the detection of EBV load in plasma and HIV-related lymphoma, since EBV was significantly less frequently detected in both HIV-negative patients with lymphoma and HIV-infected individuals without lymphoma [25,213]. This association has also been reported in whole blood (WB) [214]. Additionally, EBV loads measured either in plasma [25,210,211,214] or in PBMCs [215] could be useful to anticipate the development of lymphoma in PLWH. In contrast, EBV load measured in PBMCs was not useful for predicting the development of HIV-related NHL (DLBCL included) in the study of Van Baarle et al. [216]. Different studies suggest that EBV load measured in plasma [25,211,213,217], WB [217], PBMCs, and serum [212] can be used as a follow-up biomarker in HRL. All these techniques can be used to detect the presence of EBV in peripheral blood and have demonstrated to be useful for the follow-up of lymphomas in PLWH.
Some studies have pointed out that the presence of EBV in peripheral blood could be used as a prognostic factor in HRL. Individuals with high levels of EBV load in plasma have shown worse OS [25,209,210,211]. In this regard, Muncunill et al. reported a negative impact of high EBV load on the survival of HIV-related NHL treated without rituximab. However, this difference in outcome was not observed in patients treated with rituximab [25]. On the other hand, the impact of plasma EBV load in HL of PLWH remains controversial. In an HIV-cohort published by Muncunill et al., high EBV load in plasma had a negative influence on the prognosis of HL patients [25]. In contrast, in other cohorts the EBV plasma load was not described as having any prognostic impact on the survival of HIV-related HL [217,218,219].
In summary, the results of the different studies point out that the EBV load in peripheral blood may be a diagnostic tool and a biomarker for HRL. Further studies with larger cohorts of NHL and HL in PLWH are needed to clarify the usefulness of EBV in peripheral blood as a prognostic factor in these patients.

4. General Recommendations for the Treatment of Lymphomas in PLWH

Several studies have shown similar responses and prognoses in all lymphoma subtypes, regardless of HIV infection, when patients are treated with standard therapies. Therefore, the same chemotherapeutic strategies used in the general population are currently recommended for PLWH [168]. As previously stated in the different sections of the article, EBV-positive lymphomas in PLWH are currently treated following the general recommendations for lymphomas in these patients. Moreover, the same additional measures and supportive care recommended for treatment of lymphomas in PLWH should be applied to EBV-positive cases.
Most authors recommend concomitant administration of cART during lymphoma treatment due to the evidence that better outcomes are obtained with this approach [220]. Nevertheless, the synergistic toxicity and the drug–drug interactions between multiagent chemotherapy and cART, especially protease inhibitors, should be considered before starting lymphoma treatment [221,222,223]. Thus, interdisciplinary collaboration between hemato-oncologists and HIV specialists is the best way for the optimal treatment of both lymphoma and HIV infection while minimizing the risk of adverse outcomes for the patients.
Although there is a lack of studies on the efficacy of CNS prophylaxis in lymphomas affecting PLWH, patients with lymphomas at high risk of CNS involvement should receive prophylaxis as recommended for the general population. Therefore, patients with BL, MYC-rearranged high-grade lymphomas, PBL, as well as DLBCL with risk factors for meningeal involvement, should receive additional intrathecal prophylaxis or high-dose methotrexate, depending on the patient tolerability and the experience of the team [224,225,226,227].
Primary infectious prophylaxis using colony-stimulating factors, such as G-CSF given after every cycle of chemotherapy, is highly recommended to prevent neutropenia and dose reductions [73,228,229]. Moreover, common infections affecting PLWH should be prevented because chemotherapy decreases the CD4+ lymphocyte. In this regard, prophylaxis against Pneumocystis jirovecci [222,230,231] is recommended, and Mycobacterium avium complex should also be prevented in patients with CD4+ lymphocytes lower than 50/µL [73,229,232]. Moreover, hepatitis B and C infections are common among HIV-infected individuals. In these cases, concomitant treatment with antiviral therapy against HBV and HCV must be considered [73,223].

5. EBV-Targeted Therapies

EBV status is currently not a differential factor for the choice of treatment in HRL, and patients are treated with the same therapeutic strategies independently of the presence of EBV in lymphoma. Nevertheless, given that EBV is involved in the pathogenesis of HRL, different strategies targeting EBV could improve the treatment of these patients. In this regard, preclinical studies have evaluated different strategies in lymphoma cell lines based on protein inhibitors of signaling pathways deregulated by EBV, such as BCR signaling, PI3K, JAK/STAT, MAPK, NF-кB, cell cycle, and apoptosis [28]. Other studies have focused on the design of drugs against EBV antigens, such as EBNA1 [233,234]. Moreover, some strategies are based on immunotherapy, including PD-1/PD-L1 antibodies, monoclonal antibodies, or T-cell receptor-modified T-cell therapies, among others [235,236,237,238,239,240]. Lastly, several studies attempting to develop a vaccine against EBV have been performed. However, there is still no commercial vaccine against this virus [241].
Despite the extensive in vitro studies of EBV targets, we still have a long wait until the development and approval of EBV-specific therapies in EBV-related lymphomas. Only a few clinical trials on EBV-related lymphomas have been performed so far (Table 2). Some have evaluated drugs based on the combination of nucleoside analogs that inhibits viral DNA polymerase in the lytic phase of viral replication, such as ganciclovir (GCV) and zidovudine (azidotimidine) in combination with immunomodulatory IL-2 and cART in EBV-positive HIV-related PCNSL [242,243]. The results revealed better OS and could be effective for the treatment of these patients. Specifically, GCV reduced the EBV-DNA load in CSF of patients with HIV-PCNSL, improving survival [244]. Unfortunately, latent EBV tumors do not express the EBV-thymidine kinase (TK), and for this reason, GCV may be ineffective. Thus, other strategies are focused on the induction of the EBV lytic cycle and EBV-TK [245]. In this regard, clinical trials using arginine butyrate in combination with GCV in refractory EBV-related lymphomas have shown good tolerability and antitumoral response (10 of 15 patients; 4 complete and 6 partial responses), although these results should be confirmed in a larger cohort [246,247]. On the other hand, the inhibition of histone deacetylases (HDACs) also may induce lytic infection from the latency state. Some HDAC inhibitors such as vorinostat/suberoylanilide hydroxamic acid (SAHA) have been tested in HL and NHL [248,249]. These studies showed that SAHA could have a positive effect on HL and DLBCL, leading to partial response or stable disease, as well as a modest effect on relapsed DLBCL, although these results require validation. Currently, an ongoing phase I/II clinical trial is testing the combination of HDAC inhibitors and valganciclovir in relapsed/refractory EBV-related lymphomas.
Regarding immune evasion, PD-L1 is overexpressed in diverse EBV-related lymphomas, and therefore, strategies targeting PD-1/PD-L1 interaction are of great importance and could be a very useful target for the treatment of EBV-related HRL. A clinical trial of immunocompetent HL patients revealed that the blockage of PD-1, using the monoclonal antibody pembrolizumab, could be useful for the treatment of HL [250]. Currently, several clinical trials are evaluating the effect of PD-1/PD-L1 inhibitors, such as nivolumab, pembrolizumab, toripalimab, and sintilimab, on EBV-related NHL.
Lastly, EBV-specific T-cells (EBVST) that recognize specific EBV antigens presented by infected B-cells are another EBV-targeted therapeutic strategy. LMP1/LMP2- or LMP2-specific CTLs administered to 50 patients with EBV-related NHL or HL showed promising results in patients with risk or refractory/relapsed disease, with most achieving a 2-year event-free survival. On the other hand, 62% of patients with active lymphoma achieved complete or partial response with EBVST administration [251]. EBNA1-specific CTL administration in patients with post-transplant proliferative disease after stem cell transplantation seems to restore T-cell immune response against EBV [252]. Transfusion of EBVST may restore immune response in EBV-related lymphoma patients and results in a promising therapy to eliminate EBV-infected B-cells and avoid possible relapses in EBV-positive HRL.

6. Conclusions

In summary, EBV is involved in the lymphomagenesis of the different HRL subtypes mediated by several viral molecules. Close cooperation between EBV and HIV, as well as HHV-8 in some lymphoma subtypes, seems to be an additional lymphomagenic mechanism in which HIV may favor a permissive microenvironment for EBV infection and the development of lymphoma. EBV load in peripheral blood can be used as a lymphoma biomarker in PLWH. Diverse EBV-targeted therapies have reported promising results for the treatment of EBV-related lymphomas. Nevertheless, most studies did not include PLWH, and therefore further clinical trials are needed to confirm these results in HRL. Given the postulated cooperation between EBV and HIV in the etiopathogenesis of these lymphomas, the combination of new therapeutic strategies against both viruses should be considered in order to improve the adverse outcomes that a great proportion of these patients still have.

Author Contributions

Conceptualization, M.V.-B., G.T., A.H.-R., J.-T.N.; writing-original draft preparation, M.V.-B., J.-T.N.; figures design, M.V.-B., G.T.; writing-review and editing, G.T., A.H.-R., J.-T.N.; supervision, J.-T.N.; project administration and funding acquisition, J.-T.N. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported in part by the Instituto de Salud Carlos III, Ministerio de Economia y Competividad, Spain (PI19/01588); by Gilead Sciences S.L., Spain (GLD19/00121); and by funds from Josep Carreras International Foundation and “la Caixa” Foundation.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Horner, M.-J.; Shiels, M.S.; Pfeiffer, R.M.; Engels, E.A. Deaths Attributable to Cancer in the US Human Immunodeficiency Virus Population during 2001–2015. Clin. Infect. Dis. 2021, 72, e224–e231. [Google Scholar] [CrossRef] [PubMed]
  2. Poizot-Martin, I.; Lions, C.; Allavena, C.; Huleux, T.; Bani-Sadr, F.; Cheret, A.; Rey, D.; Duvivier, C.; Jacomet, C.; Ferry, T.; et al. Spectrum and Incidence Trends of AIDS- and Non–AIDS-Defining Cancers between 2010 and 2015 in the French Dat’AIDS Cohort. Cancer Epidemiol. Prev. Biomark. 2021, 30, 554–563. [Google Scholar] [CrossRef] [PubMed]
  3. Vangipuram, R.; Tyring, S.K. AIDS-Associated Malignancies. In HIV/AIDS-Associated Viral Oncogenesis; Cancer Treatment and Research; Meyers, C., Ed.; Springer International Publishing: Cham, Switzerland, 2019; pp. 1–21. ISBN 978-3-030-03502-0. [Google Scholar]
  4. Palella, F.J.J.; Baker, R.K.; Moorman, A.C.; Chmiel, J.S.; Wood, K.C.; Brooks, J.T.; Holmberg, S.D.; Investigators, H.O.S. Mortality in the Highly Active Antiretroviral Therapy Era: Changing Causes of Death and Disease in the HIV Outpatient Study. JAIDS J. Acquir. Immune Defic. Syndr. 2006, 43, 27–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Lim, S.T.; Levine, A.M. Recent Advances in Acquired Immunodeficiency Syndrome (AIDS)-Related Lymphoma. CA Cancer J. Clin. 2005, 55, 229–241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Goedert, J.J.; Bower, M. Impact of Highly Effective Antiretroviral Therapy on the Risk for Hodgkin Lymphoma among People with Human Immunodeficiency Virus Infection. Curr. Opin. Oncol. 2012, 24, 531–536. [Google Scholar] [CrossRef] [Green Version]
  7. Powles, T.; Robinson, D.; Stebbing, J.; Shamash, J.; Nelson, M.; Gazzard, B.; Mandelia, S.; Møller, H.; Bower, M. Highly Active Antiretroviral Therapy and the Incidence of Non–AIDS-Defining Cancers in People with HIV Infection. J. Clin. Oncol. 2008, 27, 884–890. [Google Scholar] [CrossRef]
  8. Biggar, R.J.; Jaffe, E.S.; Goedert, J.J.; Chaturvedi, A.; Pfeiffer, R.; Engels, E.A. Hodgkin Lymphoma and Immunodeficiency in Persons with HIV/AIDS. Blood 2006, 108, 3786–3791. [Google Scholar] [CrossRef]
  9. Coleman, C.B.; Lang, J.; Sweet, L.A.; Smith, N.A.; Freed, B.M.; Pan, Z.; Haverkos, B.; Pelanda, R.; Rochford, R. Epstein-Barr Virus Type 2 Infects T Cells and Induces B Cell Lymphomagenesis in Humanized Mice. J. Virol. 2018, 92, e00813-18. [Google Scholar] [CrossRef] [Green Version]
  10. Shannon-Lowe, C.; Rickinson, A.B.; Bell, A.I. Epstein–Barr Virus-Associated Lymphomas. Philos. Trans. R. Soc. B Biol. Sci. 2017, 372, 20160271. [Google Scholar] [CrossRef]
  11. Linke-Serinsöz, E.; Fend, F.; Quintanilla-Martinez, L. Human Immunodeficiency Virus (HIV) and Epstein-Barr Virus (EBV) Related Lymphomas, Pathology View Point. Semin. Diagn. Pathol. 2017, 34, 352–363. [Google Scholar] [CrossRef]
  12. Verma, M. Epigenetic Regulation of HIV, AIDS, and AIDS-Related Malignancies. In Cancer Epigenetics: Risk Assessment, Diagnosis, Treatment, and Prognosis; Methods in Molecular Biology; Verma, M., Ed.; Springer: New York, NY, USA, 2015; pp. 381–403. [Google Scholar]
  13. Mesri, E.A.; Feitelson, M.; Munger, K. Human Viral Oncogenesis: A Cancer Hallmarks Analysis. Cell Host Microbe 2014, 15, 266–282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. De Paoli, P.; Carbone, A. Microenvironmental Abnormalities Induced by Viral Cooperation: Impact on Lymphomagenesis. Semin. Cancer Biol. 2015, 34, 70–80. [Google Scholar] [CrossRef] [Green Version]
  15. Arvey, A.; Ojesina, A.I.; Pedamallu, C.S.; Ballon, G.; Jung, J.; Duke, F.; Leoncini, L.; De Falco, G.; Bressman, E.; Tam, W.; et al. The Tumor Virus Landscape of AIDS-Related Lymphomas. Blood 2015, 125, e14–e22. [Google Scholar] [CrossRef] [Green Version]
  16. Dolcetti, R.; Gloghini, A.; Caruso, A.; Carbone, A. A Lymphomagenic Role for HIV beyond Immune Suppression? Blood 2016, 127, 1403–1409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Navari, M.; Etebari, M.; De Falco, G.; Ambrosio, M.R.; Gibellini, D.; Leoncini, L.; Piccaluga, P.P. The Presence of Epstein-Barr Virus Significantly Impacts the Transcriptional Profile in Immunodeficiency-Associated Burkitt Lymphoma. Front. Microbiol. 2015, 6, 556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Moir, S.; Fauci, A.S. Insights into B Cells and HIV-Specific B-Cell Responses in HIV-Infected Individuals. Immunol. Rev. 2013, 254, 207–224. [Google Scholar] [CrossRef] [PubMed]
  19. Chao, C.; Silverberg, M.J.; Martínez-Maza, O.; Chi, M.; Abrams, D.I.; Haque, R.; Zha, H.D.; McGuire, M.; Xu, L.; Said, J. Epstein-Barr Virus Infection and Expression of B-Cell Oncogenic Markers in HIV-Related Diffuse Large B-Cell Lymphoma. Clin. Cancer Res. 2012, 18, 4702–4712. [Google Scholar] [CrossRef] [Green Version]
  20. Navari, M.; Etebari, M.; Ibrahimi, M.; Leoncini, L.; Piccaluga, P.P. Pathobiologic Roles of Epstein–Barr Virus-Encoded MicroRNAs in Human Lymphomas. Int. J. Mol. Sci. 2018, 19, 1168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Forte, E.; Luftig, M.A. The Role of MicroRNAs in Epstein-Barr Virus Latency and Lytic Reactivation. Microbes Infect. Inst. Pasteur 2011, 13, 1156–1167. [Google Scholar] [CrossRef] [Green Version]
  22. Rosemarie, Q.; Sugden, B. Epstein–Barr Virus: How Its Lytic Phase Contributes to Oncogenesis. Microorganisms 2020, 8, 1824. [Google Scholar] [CrossRef]
  23. Pietersma, F.; Piriou, E.; Baarle, D. van Immune Surveillance of EBV-Infected B Cells and the Development of Non-Hodgkin Lymphomas in Immunocompromised Patients. Leuk. Lymphoma 2008, 49, 1028–1041. [Google Scholar] [CrossRef]
  24. Haas, A.; Zimmermann, K.; Oxenius, A. Antigen-Dependent and -Independent Mechanisms of T and B Cell Hyperactivation during Chronic HIV-1 Infection. J. Virol. 2011, 85, 12102–12113. [Google Scholar] [CrossRef] [Green Version]
  25. Muncunill, J.; Baptista, M.-J.; Hernandez-Rodríguez, Á.; Dalmau, J.; Garcia, O.; Tapia, G.; Moreno, M.; Sancho, J.-M.; Martínez-Picado, J.; Feliu, E.; et al. Plasma Epstein-Barr Virus Load as an Early Biomarker and Prognostic Factor of Human Immunodeficiency Virus–Related Lymphomas. Clin. Infect. Dis. 2019, 68, 834–843. [Google Scholar] [CrossRef] [PubMed]
  26. Schommers, P.; Wyen, C.; Hentrich, M.; Gillor, D.; Zoufaly, A.; Jensen, B.; Bogner, J.R.; Thoden, J.; Wasmuth, J.C.; Fätkenheuer, G.; et al. Poor Outcome of HIV-Infected Patients with Plasmablastic Lymphoma: Results from the German AIDS-Related Lymphoma Cohort Study. AIDS 2013, 27, 842–845. [Google Scholar] [CrossRef]
  27. Kaulen, L.D.; Galluzzo, D.; Hui, P.; Barbiero, F.; Karschnia, P.; Huttner, A.; Fulbright, R.; Baehring, J.M. Prognostic Markers for Immunodeficiency-Associated Primary Central Nervous System Lymphoma. J. Neurooncol. 2019, 144, 107–115. [Google Scholar] [CrossRef] [PubMed]
  28. Pei, Y.; Wong, J.H.Y.; Robertson, E.S. Targeted Therapies for Epstein-Barr Virus-Associated Lymphomas. Cancers 2020, 12, 2565. [Google Scholar] [CrossRef]
  29. Swerdlow, S.H.; Campo, E.; Harris, N.L.; Jaffe, E.S.; Pileri, S.A.; Stein, H.; Thiele, J. (Eds.) WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues, 4th ed.; International Agency for Research on Cancer (IARC): Geneva, Switzerland, 2017; Volume 2, ISBN 978-92-832-4494-3. [Google Scholar]
  30. Ferreri, A.J.M.; Holdhoff, M.; Nayak, L.; Rubenstein, J.L. Evolving Treatments for Primary Central Nervous System Lymphoma. Am. Soc. Clin. Oncol. Educ. Book 2019, 39, 454–466. [Google Scholar] [CrossRef] [PubMed]
  31. Franca, R.A.; Travaglino, A.; Varricchio, S.; Russo, D.; Picardi, M.; Pane, F.; Pace, M.; Del Basso De Caro, M.; Mascolo, M. HIV Prevalence in Primary Central Nervous System Lymphoma: A Systematic Review and Meta-Analysis. Pathol.-Res. Pract. 2020, 216, 153192. [Google Scholar] [CrossRef]
  32. Engels, E.A.; Biggar, R.J.; Hall, H.I.; Cross, H.; Crutchfield, A.; Finch, J.L.; Grigg, R.; Hylton, T.; Pawlish, K.S.; McNeel, T.S.; et al. Cancer Risk in People Infected with Human Immunodeficiency Virus in the United States. Int. J. Cancer 2008, 123, 187–194. [Google Scholar] [CrossRef]
  33. Coghill, A.E.; Shiels, M.S.; Suneja, G.; Engels, E.A. Elevated Cancer-Specific Mortality Among HIV-Infected Patients in the United States. J. Clin. Oncol. 2015, 33, 2376–2383. [Google Scholar] [CrossRef]
  34. Baptista, M.J.; Garcia, O.; Morgades, M.; Gonzalez-Barca, E.; Miralles, P.; Lopez-Guillermo, A.; Abella, E.; Moreno, M.; Sancho, J.-M.; Feliu, E.; et al. HIV-Infection Impact on Clinical–Biological Features and Outcome of Diffuse Large B-Cell Lymphoma Treated with R-CHOP in the Combination Antiretroviral Therapy Era. AIDS 2015, 29, 811–818. [Google Scholar] [CrossRef] [PubMed]
  35. Cesarman, E. Gammaherpesvirus and Lymphoprolliferative Disorders in Immunocompromised Patients. Cancer Lett. 2011, 305, 163–174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Shindiapina, P.; Ahmed, E.H.; Mozhenkova, A.; Abebe, T.; Baiocchi, R.A. Immunology of EBV-Related Lymphoproliferative Disease in HIV-Positive Individuals. Front. Oncol. 2020, 10, 1723. [Google Scholar] [CrossRef]
  37. Morton, L.M.; Kim, C.J.; Weiss, L.M.; Bhatia, K.; Cockburn, M.; Hawes, D.; Wang, S.S.; Chang, C.; Altekruse, S.F.; Engels, E.A.; et al. Molecular Characteristics of Diffuse Large B-Cell Lymphoma in Human Immunodeficiency Virus-Infected and -Uninfected Patients in the Pre-Highly Active Antiretroviral Therapy and Pre-Rituximab Era. Leuk. Lymphoma 2014, 55, 551–557. [Google Scholar] [CrossRef] [Green Version]
  38. Dunleavy, K.; Little, R.F.; Pittaluga, S.; Grant, N.; Wayne, A.S.; Carrasquillo, J.A.; Steinberg, S.M.; Yarchoan, R.; Jaffe, E.S.; Wilson, W.H. The Role of Tumor Histogenesis, FDG-PET, and Short-Course EPOCH with Dose-Dense Rituximab (SC-EPOCH-RR) in HIV-Associated Diffuse Large B-Cell Lymphoma. Blood 2010, 115, 3017–3024. [Google Scholar] [CrossRef] [Green Version]
  39. Baptista, M.J.; Tapia, G.; Morgades, M.; Muncunill, J.; Muñoz-Marmol, A.-M.; Montoto, S.; Gribben, J.G.; Calaminici, M.; Martinez, A.; Gonzalez-Farre, B.; et al. Using the Lymph2Cx Assay for Assessing Cell-of-Origin Subtypes of HIV-Related Diffuse Large B-Cell Lymphoma. Leuk. Lymphoma 2019, 60, 1087–1091. [Google Scholar] [CrossRef]
  40. Besson, C.; Lancar, R.; Prevot, S.; Algarte-Genin, M.; Delobel, P.; Bonnet, F.; Meyohas, M.-C.; Partisani, M.; Oberic, L.; Gabarre, J.; et al. Outcomes for HIV-Associated Diffuse Large B-Cell Lymphoma in the Modern Combined Antiretroviral Therapy Era. AIDS 2017, 31, 2493–2501. [Google Scholar] [CrossRef]
  41. Diamond, C.; Taylor, T.H.; Aboumrad, T.; Anton-Culver, H. Changes in Acquired Immunodeficiency Syndrome-Related Non-Hodgkin Lymphoma in the Era of Highly Active Antiretroviral Therapy. Cancer 2006, 106, 128–135. [Google Scholar] [CrossRef]
  42. Matinella, A.; Lanzafame, M.; Bonometti, M.A.; Gajofatto, A.; Concia, E.; Vento, S.; Monaco, S.; Ferrari, S. Neurological Complications of HIV Infection in Pre-HAART and HAART Era: A Retrospective Study. J. Neurol. 2015, 262, 1317–1327. [Google Scholar] [CrossRef]
  43. González-Aguilar, A.; Soto-Hernández, J.L. The Management of Primary Central Nervous System Lymphoma Related to AIDS in the HAART Era. Curr. Opin. Oncol. 2011, 23, 648–653. [Google Scholar] [CrossRef] [PubMed]
  44. Ivers, L.C.; Kim, A.Y.; Sax, P.E. Predictive Value of Polymerase Chain Reaction of Cerebrospinal Fluid for Detection of Epstein-Barr Virus to Establish the Diagnosis of HIV-Related Primary Central Nervous System Lymphoma. Clin. Infect. Dis. 2004, 38, 1629–1632. [Google Scholar] [CrossRef] [PubMed]
  45. Cingolani, A.; De Luca, A.; Ammassari, A.; Antinori, A.; Ortona, L.; Larocca, L.M.; Scerrati, M. Minimally Invasive Diagnosis of Acquired Immunodeficiency Syndrome-Related Primary Central Nervous System Lymphoma. JNCI J. Natl. Cancer Inst. 1998, 90, 364–369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Cinque, P.; Vago, L.; Dahl, H.; Brytting, M.; Terreni, M.R.; Fornara, C.; Racca, S.; Castagna, A.; Monforte, A.D.; Wahren, B.; et al. Polymerase Chain Reaction on Cerebrospinal Fluid for Diagnosis of Virus-Associated Opportunistic Diseases of the Central Nervous System in HIV-Infected Patients. AIDS 1996, 10, 951–958. [Google Scholar] [CrossRef]
  47. Crombie, J.L.; LaCasce, A.S. Epstein Barr Virus Associated B-Cell Lymphomas and Iatrogenic Lymphoproliferative Disorders. Front. Oncol. 2019, 9, 109. [Google Scholar] [CrossRef] [Green Version]
  48. Xia, T.; O’Hara, A.; Araujo, I.; Barreto, J.; Carvalho, E.; Sapucaia, J.B.; Ramos, J.C.; Luz, E.; Pedroso, C.; Manrique, M.; et al. EBV MicroRNAs in Primary Lymphomas and Targeting of CXCL-11 by Ebv-Mir-BHRF1-3. Cancer Res. 2008, 68, 1436–1442. [Google Scholar] [CrossRef] [Green Version]
  49. Sakamoto, K.; Sekizuka, T.; Uehara, T.; Hishima, T.; Mine, S.; Fukumoto, H.; Sato, Y.; Hasegawa, H.; Kuroda, M.; Katano, H. Next–generation Sequencing of MiRNAs in Clinical Samples of Epstein–Barr Virus–associated B–cell Lymphomas. Cancer Med. 2017, 6, 605–618. [Google Scholar] [CrossRef]
  50. Ma, J.; Nie, K.; Redmond, D.; Liu, Y.; Elemento, O.; Knowles, D.M.; Tam, W. EBV-MiR-BHRF1-2 Targets PRDM1/Blimp1: Potential Role in EBV Lymphomagenesis. Leukemia 2016, 30, 594–604. [Google Scholar] [CrossRef] [Green Version]
  51. Martín-Pérez, D.; Vargiu, P.; Montes-Moreno, S.; León, E.A.; Rodríguez-Pinilla, S.M.; Lisio, L.D.; Martínez, N.; Rodríguez, R.; Mollejo, M.; Castellvi, J.; et al. Epstein-Barr Virus MicroRNAs Repress BCL6 Expression in Diffuse Large B-Cell Lymphoma. Leukemia 2012, 26, 180–183. [Google Scholar] [CrossRef] [PubMed]
  52. Cohen, M.; Vistarop, A.G.; Huaman, F.; Narbaitz, M.; Metrebian, F.; De Matteo, E.; Preciado, M.V.; Chabay, P.A. Epstein-Barr Virus Lytic Cycle Involvement in Diffuse Large B Cell Lymphoma. Hematol. Oncol. 2018, 36, 98–103. [Google Scholar] [CrossRef]
  53. Liapis, K.; Clear, A.; Owen, A.; Coutinho, R.; Greaves, P.; Lee, A.M.; Montoto, S.; Calaminici, M.; Gribben, J.G. The Microenvironment of AIDS-Related Diffuse Large B-Cell Lymphoma Provides Insight into the Pathophysiology and Indicates Possible Therapeutic Strategies. Blood 2013, 122, 424–433. [Google Scholar] [CrossRef] [Green Version]
  54. Fiorentini, S.; Marini, E.; Caracciolo, S.; Caruso, A. Functions of the HIV-1 Matrix Protein P17. New Microbiol. 2006, 29, 1–10. [Google Scholar]
  55. Dolcetti, R.; Giagulli, C.; He, W.; Selleri, M.; Caccuri, F.; Eyzaguirre, L.M.; Mazzuca, P.; Corbellini, S.; Campilongo, F.; Marsico, S.; et al. Role of HIV-1 Matrix Protein P17 Variants in Lymphoma Pathogenesis. Proc. Natl. Acad. Sci. USA 2015, 112, 14331–14336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Martorelli, D.; Muraro, E.; Mastorci, K.; Col, J.D.; Faè, D.A.; Furlan, C.; Giagulli, C.; Caccuri, F.; Rusnati, M.; Fiorentini, S.; et al. A Natural HIV P17 Protein Variant Up-Regulates the LMP-1 EBV Oncoprotein and Promotes the Growth of EBV-Infected B-Lymphocytes: Implications for EBV-Driven Lymphomagenesis in the HIV Setting. Int. J. Cancer 2015, 137, 1374–1385. [Google Scholar] [CrossRef]
  57. Gloghini, A.; Dolcetti, R.; Carbone, A. Lymphomas Occurring Specifically in HIV-Infected Patients: From Pathogenesis to Pathology. Semin. Cancer Biol. 2013, 23, 457–467. [Google Scholar] [CrossRef] [PubMed]
  58. Brandsma, D.; Bromberg, J.E.C. Chapter 14—Primary CNS lymphoma in HIV infection. In Handbook of Clinical Neurology; The Neurology of HIV Infection; Brew, B.J., Ed.; Elsevier: Amsterdam, The Netherlands, 2018; Volume 152, pp. 177–186. [Google Scholar] [CrossRef]
  59. Bibas, M.; Antinori, A. EBV and HIV-Related Lymphoma. Mediterr. J. Hematol. Infect. Dis. 2009, 1, e2009032. [Google Scholar] [CrossRef] [PubMed]
  60. Gasser, O.; Bihl, F.K.; Wolbers, M.; Loggi, E.; Steffen, I.; Hirsch, H.H.; Günthard, H.F.; Walker, B.D.; Brander, C.; Battegay, M.; et al. HIV Patients Developing Primary CNS Lymphoma Lack EBV-Specific CD4+ T Cell Function Irrespective of Absolute CD4+ T Cell Counts. PLOS Med. 2007, 4, e96. [Google Scholar] [CrossRef] [Green Version]
  61. Rios, A. HIV-Related Hematological Malignancies: A concise review. Clin. Lymphoma Myeloma Leuk. 2014, 14, S96–S103. [Google Scholar] [CrossRef]
  62. Maguire, A.; Chen, X.; Wisner, L.; Malasi, S.; Ramsower, C.; Kendrick, S.; Barrett, M.T.; Glinsmann–Gibson, B.; McGrath, M.; Rimsza, L.M. Enhanced DNA Repair and Genomic Stability Identify a Novel HIV–related Diffuse Large B–cell Lymphoma Signature. Int. J. Cancer 2019, 145, 3078–3088. [Google Scholar] [CrossRef]
  63. Chadburn, A.; Chiu, A.; Lee, J.Y.; Chen, X.; Hyjek, E.; Banham, A.H.; Noy, A.; Kaplan, L.D.; Sparano, J.A.; Bhatia, K.; et al. Immunophenotypic Analysis of AIDS-Related Diffuse Large B-Cell Lymphoma and Clinical Implications in Patients from AIDS Malignancies Consortium Clinical Trials 010 and 034. J. Clin. Oncol. 2009, 27, 5039. [Google Scholar] [CrossRef] [Green Version]
  64. Bayraktar, S.; Bayraktar, U.D.; Ramos, J.C.; Stefanovic, A.; Lossos, I.S. Primary CNS Lymphoma in HIV Positive and Negative Patients: Comparison of Clinical Characteristics, Outcome and Prognostic Factors. J. Neurooncol. 2011, 101, 257–265. [Google Scholar] [CrossRef] [PubMed]
  65. Gupta, N.K.; Nolan, A.; Omuro, A.; Reid, E.G.; Wang, C.-C.; Mannis, G.; Jaglal, M.; Chavez, J.C.; Rubinstein, P.G.; Griffin, A.; et al. Long-Term Survival in AIDS-Related Primary Central Nervous System Lymphoma. Neuro-Oncology 2017, 19, 99–108. [Google Scholar] [CrossRef] [Green Version]
  66. Gopal, S.; Martin, K.E.; Richards, K.L.; Eron, J.J. Clinical Presentation, Treatment, and Outcomes among 65 Patients with HIV-Associated Lymphoma Treated at the University of North Carolina, 2000–2010. AIDS Res. Hum. Retrovir. 2012, 28, 798–805. [Google Scholar] [CrossRef] [Green Version]
  67. De Luca, A.; Antinori, A.; Cingolani, A.; Larocca, L.M.; Linzalone, A.; Ammassari, A.; Scerrati, M.; Roselli, R.; Tamburrini, E.; Ortona, L. Evaluation of Cerebrospinal Fluid EBV–DNA and IL–10 as Markers for in Vivo Diagnosis of AIDS–related Primary Central Nervous System Lymphoma. Br. J. Haematol. 1995, 90, 844–849. [Google Scholar] [CrossRef] [PubMed]
  68. Ribera, J.-M.; Oriol, A.; Morgades, M.; González–Barca, E.; Miralles, P.; López–Guillermo, A.; Gardella, S.; López, A.; Abella, E.; García, M. Safety and Efficacy of Cyclophosphamide, Adriamycin, Vincristine, Prednisone and Rituximab in Patients with Human Immunodeficiency Virus-Associated Diffuse Large B-Cell Lymphoma: Results of a Phase II Trial. Br. J. Haematol. 2008, 140, 411–419. [Google Scholar] [CrossRef]
  69. Liu, Y.; Barta, S.K. Diffuse Large B-Cell Lymphoma: 2019 Update on Diagnosis, Risk Stratification, and Treatment. Am. J. Hematol. 2019, 94, 604–616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Duarte, R.F.; Labopin, M.; Bader, P.; Basak, G.W.; Bonini, C.; Chabannon, C.; Corbacioglu, S.; Dreger, P.; Dufour, C.; Gennery, A.R.; et al. Indications for Haematopoietic Stem Cell Transplantation for Haematological Diseases, Solid Tumours and Immune Disorders: Current Practice in Europe, 2019. Bone Marrow Transplant. 2019, 54, 1525–1552. [Google Scholar] [CrossRef] [PubMed]
  71. Grommes, C.; Rubenstein, J.L.; DeAngelis, L.M.; Ferreri, A.J.M.; Batchelor, T.T. Comprehensive Approach to Diagnosis and Treatment of Newly Diagnosed Primary CNS Lymphoma. Neuro-Oncology 2019, 21, 296–305. [Google Scholar] [CrossRef]
  72. Han, C.H.; Batchelor, T.T. Diagnosis and Management of Primary Central Nervous System Lymphoma. Cancer 2017, 123, 4314–4324. [Google Scholar] [CrossRef]
  73. Miralles, P.; Navarro, J.T.; Berenguer, J.; Gómez Codina, J.; Kwon, M.; Serrano, D.; Díez-Martín, J.L.; Villà, S.; Rubio, R.; Menárguez, J.; et al. GESIDA/PETHEMA Recommendations on the Diagnosis and Treatment of Lymphomas in Patients Infected by the Human Immunodeficiency Virus. Med. Clin. 2018, 151, 39.e1–39.e17. [Google Scholar] [CrossRef]
  74. Graham, B.S.; Lynch, D.T. Burkitt Lymphoma. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2021. [Google Scholar]
  75. God, J.M.; Haque, A. Burkitt Lymphoma: Pathogenesis and Immune Evasion. J. Oncol. 2010, 2010, e516047. [Google Scholar] [CrossRef]
  76. Burkitt, D.; O’conor, G.T. Malignant Lymphoma in African Children. I. A Clinical Syndrome. Cancer 1961, 14, 258–269. [Google Scholar] [CrossRef]
  77. Davi, F.; Delecluse, H.J.; Guiet, P.; Gabarre, J.; Fayon, A.; Gentilhomme, O.; Felman, P.; Bayle, C.; Berger, F.; Audouin, J.; et al. Burkitt-like Lymphomas in AIDS Patients: Characterization within a Series of 103 Human Immunodeficiency Virus-Associated Non-Hodgkin’s Lymphomas. Burkitt’s Lymphoma Study Group. J. Clin. Oncol. 1998, 16, 3788–3795. [Google Scholar] [CrossRef] [PubMed]
  78. Lenze, D.; Leoncini, L.; Hummel, M.; Volinia, S.; Liu, C.G.; Amato, T.; De Falco, G.; Githanga, J.; Horn, H.; Nyagol, J.; et al. The Different Epidemiologic Subtypes of Burkitt Lymphoma Share a Homogenous Micro RNA Profile Distinct from Diffuse Large B-Cell Lymphoma. Leukemia 2011, 25, 1869–1876. [Google Scholar] [CrossRef] [Green Version]
  79. Mbulaiteye, S.M.; Pullarkat, S.T.; Nathwani, B.N.; Weiss, L.M.; Rao, N.; Emmanuel, B.; Lynch, C.F.; Hernandez, B.; Neppalli, V.; Hawes, D.; et al. Epstein-Barr Virus Patterns in U.S. Burkitt Lymphoma Tumors from the SEER Residual Tissue Repository during 1979–2009. APMIS Acta Pathol. Microbiol. Immunol. Scand. 2014, 122, 5–15. [Google Scholar] [CrossRef] [Green Version]
  80. Carbone, A.; Gloghini, A. AIDS-Related Lymphomas: From Pathogenesis to Pathology. Br. J. Haematol. 2005, 130, 662–670. [Google Scholar] [CrossRef] [PubMed]
  81. Guech-Ongey, M.; Simard, E.P.; Anderson, W.F.; Engels, E.A.; Bhatia, K.; Devesa, S.S.; Mbulaiteye, S.M. AIDS-Related Burkitt Lymphoma in the United States: What Do Age and CD4 Lymphocyte Patterns Tell Us about Etiology and/or Biology? Blood 2010, 116, 5600–5604. [Google Scholar] [CrossRef] [Green Version]
  82. Lim, S.T.; Karim, R.; Nathwani, B.N.; Tulpule, A.; Espina, B.; Levine, A.M. AIDS-Related Burkitt’s Lymphoma Versus Diffuse Large-Cell Lymphoma in the Pre–Highly Active Antiretroviral Therapy (HAART) and HAART Eras: Significant Differences in Survival with Standard Chemotherapy. J. Clin. Oncol. 2005, 23, 4430–4438. [Google Scholar] [CrossRef] [PubMed]
  83. Gopal, S.; Patel, M.R.; Yanik, E.L.; Cole, S.R.; Achenbach, C.J.; Napravnik, S.; Burkholder, G.A.; Reid, E.G.; Rodriguez, B.; Deeks, S.G.; et al. Temporal Trends in Presentation and Survival for HIV-Associated Lymphoma in the Antiretroviral Therapy Era. J. Natl. Cancer Inst. 2013, 105, 1221–1229. [Google Scholar] [CrossRef] [PubMed]
  84. Dunleavy, K.; Wilson, W.H. How I Treat HIV-Associated Lymphoma. Blood 2012, 119, 3245–3255. [Google Scholar] [CrossRef]
  85. Carbone, A. Emerging Pathways in the Development of AIDS-Related Lymphomas. Lancet Oncol. 2003, 4, 22–29. [Google Scholar] [CrossRef]
  86. Pajic, A.; Staege, M.S.; Dudziak, D.; Schuhmacher, M.; Spitkovsky, D.; Eissner, G.; Brielmeier, M.; Polack, A.; Bornkamm, G.W. Antagonistic Effects of C-Myc and Epstein-Barr Virus Latent Genes on the Phenotype of Human B Cells. Int. J. Cancer 2001, 93, 810–816. [Google Scholar] [CrossRef]
  87. Humme, S.; Reisbach, G.; Feederle, R.; Delecluse, H.-J.; Bousset, K.; Hammerschmidt, W.; Schepers, A. The EBV Nuclear Antigen 1 (EBNA1) Enhances B Cell Immortalization Several Thousandfold. Proc. Natl. Acad. Sci. USA 2003, 100, 10989–10994. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Allday, M.J. How Does Epstein–Barr Virus (EBV) Complement the Activation of Myc in the Pathogenesis of Burkitt’s Lymphoma? Semin. Cancer Biol. 2009, 19, 366–376. [Google Scholar] [CrossRef] [Green Version]
  89. Drotar, M.E.; Silva, S.; Barone, E.; Campbell, D.; Tsimbouri, P.; Jurvansu, J.; Bhatia, P.; Klein, G.; Wilson, J.B. Epstein-Barr Virus Nuclear Antigen-1 and Myc Cooperate in Lymphomagenesis. Int. J. Cancer 2003, 106, 388–395. [Google Scholar] [CrossRef]
  90. Zhang, T.; Ma, J.; Nie, K.; Yan, J.; Liu, Y.; Bacchi, C.E.; Queiroga, E.M.; Gualco, G.; Sample, J.T.; Orazi, A.; et al. Hypermethylation of the Tumor Suppressor Gene PRDM1/Blimp-1 Supports a Pathogenetic Role in EBV-Positive Burkitt Lymphoma. Blood Cancer J. 2014, 4, e261. [Google Scholar] [CrossRef]
  91. Desbien, A.L.; Kappler, J.W.; Marrack, P. The Epstein–Barr Virus Bcl-2 Homolog, BHRF1, Blocks Apoptosis by Binding to a Limited Amount of Bim. Proc. Natl. Acad. Sci. USA 2009, 106, 5663–5668. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Fitzsimmons, L.; Cartlidge, R.; Chang, C.; Sejic, N.; Galbraith, L.C.A.; Suraweera, C.D.; Croom-Carter, D.; Dewson, G.; Tierney, R.J.; Bell, A.I.; et al. EBV BCL-2 Homologue BHRF1 Drives Chemoresistance and Lymphomagenesis by Inhibiting Multiple Cellular pro-Apoptotic Proteins. Cell Death Differ. 2020, 27, 1554–1568. [Google Scholar] [CrossRef]
  93. Zhang, Q.; Gutsch, D.; Kenney, S. Functional and Physical Interaction between P53 and BZLF1: Implications for Epstein-Barr Virus Latency. Mol. Cell. Biol. 1994, 14, 1929–1938. [Google Scholar] [CrossRef] [Green Version]
  94. Ambrosio, M.R.; Navari, M.; Di Lisio, L.; Leon, E.A.; Onnis, A.; Gazaneo, S.; Mundo, L.; Ulivieri, C.; Gomez, G.; Lazzi, S.; et al. The Epstein Barr-Encoded BART-6-3p MicroRNA Affects Regulation of Cell Growth and Immuno Response in Burkitt Lymphoma. Infect. Agents Cancer 2014, 9, 12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Zhou, L.; Bu, Y.; Liang, Y.; Zhang, F.; Zhang, H.; Li, S. Epstein-Barr Virus (EBV)-BamHI-A Rightward Transcript (BART)-6 and Cellular MicroRNA-142 Synergistically Compromise Immune Defense of Host Cells in EBV-Positive Burkitt Lymphoma. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2016, 22, 4114–4120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Ungerleider, N.; Bullard, W.; Kara, M.; Wang, X.; Roberts, C.; Renne, R.; Tibbetts, S.; Flemington, E.K. EBV MiRNAs Are Potent Effectors of Tumor Cell Transcriptome Remodeling in Promoting Immune Escape. PLOS Pathog. 2021, 17, e1009217. [Google Scholar] [CrossRef]
  97. Kitagawa, N.; Goto, M.; Kurozumi, K.; Maruo, S.; Fukayama, M.; Naoe, T.; Yasukawa, M.; Hino, K.; Suzuki, T.; Todo, S.; et al. Epstein–Barr Virus-Encoded Poly(A) RNA Supports Burkitt’s Lymphoma Growth through Interleukin-10 Induction. EMBO J. 2000, 19, 6742–6750. [Google Scholar] [CrossRef] [Green Version]
  98. Nanbo, A.; Inoue, K.; Adachi-Takasawa, K.; Takada, K. Epstein–Barr Virus RNA Confers Resistance to Interferon-α-Induced Apoptosis in Burkitt’s Lymphoma. EMBO J. 2002, 21, 954–965. [Google Scholar] [CrossRef] [PubMed]
  99. Pannone, G.; Zamparese, R.; Pace, M.; Pedicillo, M.C.; Cagiano, S.; Somma, P.; Errico, M.E.; Donofrio, V.; Franco, R.; De Chiara, A.; et al. The Role of EBV in the Pathogenesis of Burkitt’s Lymphoma: An Italian Hospital Based Survey. Infect. Agents Cancer 2014, 9, 34. [Google Scholar] [CrossRef] [Green Version]
  100. Casulo, C.; Friedberg, J.W. Burkitt Lymphoma- a Rare but Challenging Lymphoma. Best Pract. Res. Clin. Haematol. 2018, 31, 279–284. [Google Scholar] [CrossRef] [PubMed]
  101. Blum, K.A.; Lozanski, G.; Byrd, J.C. Adult Burkitt Leukemia and Lymphoma. Blood 2004, 104, 3009–3020. [Google Scholar] [CrossRef]
  102. Blinder, V.S.; Chadburn, A.; Furman, R.R.; Mathew, S.; Leonard, J.P. Review: Improving Outcomes for Patients with Burkitt Lymphoma and HIV. AIDS Patient Care STDs 2008, 22, 175–187. [Google Scholar] [CrossRef] [PubMed]
  103. Ribera, J.-M.; García, O.; Grande, C.; Esteve, J.; Oriol, A.; Bergua, J.; González-Campos, J.; Vall-llovera, F.; Tormo, M.; Hernández-Rivas, J.-M.; et al. Dose-Intensive Chemotherapy Including Rituximab in Burkitt’s Leukemia or Lymphoma Regardless of Human Immunodeficiency Virus Infection Status. Cancer 2013, 119, 1660–1668. [Google Scholar] [CrossRef]
  104. Oriol, A.; Ribera, J.-M.; Esteve, J.; Sanz, M.-A.; Brunet, S.; Garcia-Boyero, R.; Fernández-Abellán, P.; Martí, J.-M.; Abella, E.; Sánchez-Delgado, M.; et al. Lack of Influence of Human Immunodeficiency Virus Infection Status in the Response to Therapy and Survival of Adult Patients with Mature B-Cell Lymphoma or Leukemia. Results of the PETHEMA-LAL3/97 Study. Haematologica 2003, 88, 445–453. [Google Scholar]
  105. Barnes, J.A.; LaCasce, A.S.; Feng, Y.; Toomey, C.E.; Neuberg, D.; Michaelson, J.S.; Hochberg, E.P.; Abramson, J.S. Evaluation of the Addition of Rituximab to CODOX-M/IVAC for Burkitt’s Lymphoma: A Retrospective Analysis. Ann. Oncol. 2011, 22, 1859–1864. [Google Scholar] [CrossRef]
  106. Dunleavy, K.; Pittaluga, S.; Shovlin, M.; Steinberg, S.M.; Cole, D.; Grant, C.; Widemann, B.; Staudt, L.M.; Jaffe, E.S.; Little, R.F.; et al. Low-Intensity Therapy in Adults with Burkitt’s Lymphoma. N. Engl. J. Med. 2013, 369, 1915–1925. [Google Scholar] [CrossRef] [Green Version]
  107. Florindez, J.A.; Alderuccio, J.P.; Reis, I.M.; Lossos, I.S. Survival Analysis in Treated Plasmablastic Lymphoma Patients: A Population-Based Study. Am. J. Hematol. 2020, 95, 1344–1351. [Google Scholar] [CrossRef]
  108. Castillo, J.J.; Winer, E.S.; Stachurski, D.; Perez, K.; Jabbour, M.; Milani, C.; Colvin, G.; Butera, J.N. Clinical and Pathological Differences between Human Immunodeficiency Virus-Positive and Human Immunodeficiency Virus-Negative Patients with Plasmablastic Lymphoma. Leuk. Lymphoma 2010, 51, 2047–2053. [Google Scholar] [CrossRef]
  109. Tchernonog, E.; Faurie, P.; Coppo, P.; Monjanel, H.; Bonnet, A.; Génin, M.A.; Mercier, M.; Dupuis, J.; Bijou, F.; Herbaux, C.; et al. Clinical Characteristics and Prognostic Factors of Plasmablastic Lymphoma Patients: Analysis of 135 Patients from the LYSA Group. Ann. Oncol. 2017, 28, 843–848. [Google Scholar] [CrossRef]
  110. Delecluse, H.J.; Anagnostopoulos, I.; Dallenbach, F.; Hummel, M.; Marafioti, T.; Schneider, U.; Huhn, D.; Schmidt-Westhausen, A.; Reichart, P.A.; Gross, U.; et al. Plasmablastic Lymphomas of the Oral Cavity: A New Entity Associated with the Human Immunodeficiency Virus Infection. Blood 1997, 89, 1413–1420. [Google Scholar] [CrossRef]
  111. Carbone, A. AIDS-Related Non-Hodgkin’s Lymphomas: From Pathology and Molecular Pathogenesis to Treatment. Hum. Pathol. 2002, 33, 392–404. [Google Scholar] [CrossRef] [PubMed]
  112. Fernández-Álvarez, R.; Sancho, J.-M.; Ribera, J.-M. Plasmablastic Lymphoma. Med. Clín. Engl. Ed. 2016, 147, 399–404. [Google Scholar] [CrossRef]
  113. Castillo, J.J.; Bibas, M.; Miranda, R.N. The Biology and Treatment of Plasmablastic Lymphoma. Blood 2015, 125, 2323–2330. [Google Scholar] [CrossRef] [Green Version]
  114. Morscio, J.; Dierickx, D.; Nijs, J.; Verhoef, G.; Bittoun, E.; Vanoeteren, X.; Wlodarska, I.; Sagaert, X.; Tousseyn, T. Clinicopathologic Comparison of Plasmablastic Lymphoma in HIV-Positive, Immunocompetent, and Posttransplant Patients: Single-Center Series of 25 Cases and Meta-Analysis of 277 Reported Cases. Am. J. Surg. Pathol. 2014, 38, 875–886. [Google Scholar] [CrossRef] [PubMed]
  115. Teruya-Feldstein, J.; Chiao, E.; Filippa, D.A.; Lin, O.; Comenzo, R.; Coleman, M.; Portlock, C.; Noy, A. CD20-Negative Large-Cell Lymphoma with Plasmablastic Features: A Clinically Heterogenous Spectrum in Both HIV-Positive and -Negative Patients. Ann. Oncol. 2004, 15, 1673–1679. [Google Scholar] [CrossRef]
  116. Ramis-Zaldivar, J.E.; Gonzalez-Farre, B.; Nicolae, A.; Pack, S.; Clot, G.; Nadeu, F.; Mottok, A.; Horn, H.; Song, J.Y.; Fu, K.; et al. MAP-Kinase and JAK-STAT Pathways Dysregulation in Plasmablastic Lymphoma. Haematologica 2020, 106, 2682–2693. [Google Scholar] [CrossRef] [PubMed]
  117. Frontzek, F.; Staiger, A.M.; Zapukhlyak, M.; Xu, W.; Bonzheim, I.; Borgmann, V.; Sander, P.; Baptista, M.J.; Heming, J.-N.; Berning, P.; et al. Molecular and Functional Profiling Identifies Therapeutically Targetable Vulnerabilities in Plasmablastic Lymphoma. Nat. Commun. 2021, 12, 5183. [Google Scholar] [CrossRef] [PubMed]
  118. Garcia-Reyero, J.; Magunacelaya, N.M.; de Villambrosia, S.G.; Loghavi, S.; Mediavilla, A.G.; Tonda, R.; Beltran, S.; Gut, M.; Gonzalez, A.P.; D’Ámore, E.; et al. Genetic Lesions in MYC and STAT3 Drive Oncogenic Transcription Factor Overexpression in Plasmablastic Lymphoma. Haematologica 2020, 106, 1120–1128. [Google Scholar] [CrossRef] [Green Version]
  119. Liu, Z.; Filip, I.; Gomez, K.; Engelbrecht, D.; Meer, S.; Lalloo, P.N.; Patel, P.; Perner, Y.; Zhao, J.; Wang, J.; et al. Genomic Characterization of HIV-Associated Plasmablastic Lymphoma Identifies Pervasive Mutations in the JAK–STAT Pathway. Blood Cancer Discov. 2020, 1, 112–125. [Google Scholar] [CrossRef]
  120. Lopez, A.; Abrisqueta, P. Plasmablastic Lymphoma: Current Perspectives. Blood Lymphat. Cancer Targets Ther. 2018, 8, 63–70. [Google Scholar] [CrossRef] [Green Version]
  121. Valera, A.; Balagué, O.; Colomo, L.; Martínez, A.; Delabie, J.; Taddesse-Heath, L.; Jaffe, E.S.; Campo, E. IG/MYC Rearrangements Are the Main Cytogenetic Alteration in Plasmablastic Lymphomas. Am. J. Surg. Pathol. 2010, 34, 1686–1694. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Montes-Moreno, S.; Martinez-Magunacelaya, N.; Zecchini-Barrese, T.; Villambrosía, S.G.; de Linares, E.; Ranchal, T.; Rodriguez-Pinilla, M.; Batlle, A.; Cereceda-Company, L.; Revert-Arce, J.B.; et al. Plasmablastic Lymphoma Phenotype Is Determined by Genetic Alterations in MYC and PRDM1. Mod. Pathol. 2017, 30, 85–94. [Google Scholar] [CrossRef] [Green Version]
  123. Schommers, P.; Gillor, D.; Hentrich, M.; Wyen, C.; Wolf, T.; Oette, M.; Zoufaly, A.; Wasmuth, J.-C.; Bogner, J.R.; Müller, M.; et al. Incidence and Risk Factors for Relapses in HIV-Associated Non-Hodgkin Lymphoma as Observed in the German HIV-Related Lymphoma Cohort Study. Haematologica 2018, 103, 857–864. [Google Scholar] [CrossRef]
  124. Loghavi, S.; Alayed, K.; Aladily, T.N.; Zuo, Z.; Ng, S.-B.; Tang, G.; Hu, S.; Yin, C.C.; Miranda, R.N.; Medeiros, L.J.; et al. Stage, Age, and EBV Status Impact Outcomes of Plasmablastic Lymphoma Patients: A Clinicopathologic Analysis of 61 Patients. J. Hematol. Oncol. 2015, 8, 65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Lin, Y.; Wong, K.; Calame, K. Repression of C-Myc Transcription by Blimp-1, an Inducer of Terminal B Cell Differentiation. Science 1997, 276, 596–599. [Google Scholar] [CrossRef] [PubMed]
  126. Shaffer, A.L.; Lin, K.-I.; Kuo, T.C.; Yu, X.; Hurt, E.M.; Rosenwald, A.; Giltnane, J.M.; Yang, L.; Zhao, H.; Calame, K.; et al. Blimp-1 Orchestrates Plasma Cell Differentiation by Extinguishing the Mature B Cell Gene Expression Program. Immunity 2002, 17, 51–62. [Google Scholar] [CrossRef] [Green Version]
  127. Ambrosio, M.R.; Mundo, L.; Gazaneo, S.; Picciolini, M.; Vara, P.S.; Sayed, S.; Ginori, A.; Lo Bello, G.; Del Porro, L.; Navari, M.; et al. MicroRNAs Sequencing Unveils Distinct Molecular Subgroups of Plasmablastic Lymphoma. Oncotarget 2017, 8, 107356–107373. [Google Scholar] [CrossRef] [Green Version]
  128. Gravelle, P.; Péricart, S.; Tosolini, M.; Fabiani, B.; Coppo, P.; Amara, N.; Traverse-Gléhen, A.; Van Acker, N.; Brousset, P.; Fournie, J.-J.; et al. EBV Infection Determines the Immune Hallmarks of Plasmablastic Lymphoma. Oncoimmunology 2018, 7, e1486950. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Castillo, J.; Pantanowitz, L.; Dezube, B.J. HIV-Associated Plasmablastic Lymphoma: Lessons Learned from 112 Published Cases. Am. J. Hematol. 2008, 83, 804–809. [Google Scholar] [CrossRef] [PubMed]
  130. Castillo, J.J.; Furman, M.; Beltrán, B.E.; Bibas, M.; Bower, M.; Chen, W.; Díez-Martín, J.L.; Liu, J.J.; Miranda, R.N.; Montoto, S.; et al. Human Immunodeficiency Virus-Associated Plasmablastic Lymphoma. Cancer 2012, 118, 5270–5277. [Google Scholar] [CrossRef] [PubMed]
  131. Castillo, J.J.; Winer, E.S.; Stachurski, D.; Perez, K.; Jabbour, M.; Milani, C.; Colvin, G.; Butera, J.N. Prognostic Factors in Chemotherapy-Treated Patients with HIV-Associated Plasmablastic Lymphoma. Oncologist 2010, 15, 293–299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Koizumi, Y.; Uehira, T.; Ota, Y.; Ogawa, Y.; Yajima, K.; Tanuma, J.; Yotsumoto, M.; Hagiwara, S.; Ikegaya, S.; Watanabe, D.; et al. Clinical and Pathological Aspects of Human Immunodeficiency Virus-Associated Plasmablastic Lymphoma: Analysis of 24 Cases. Int. J. Hematol. 2016, 104, 669–681. [Google Scholar] [CrossRef]
  133. Laurent, C.; Fabiani, B.; Do, C.; Tchernonog, E.; Cartron, G.; Gravelle, P.; Amara, N.; Malot, S.; Palisoc, M.M.; Copie-Bergman, C.; et al. Immune-Checkpoint Expression in Epstein-Barr Virus Positive and Negative Plasmablastic Lymphoma: A Clinical and Pathological Study in 82 Patients. Haematologica 2016, 101, 976–984. [Google Scholar] [CrossRef] [Green Version]
  134. Castillo, J.J.; Guerrero–Garcia, T.; Baldini, F.; Tchernonog, E.; Cartron, G.; Ninkovic, S.; Cwynarski, K.; Dierickx, D.; Tousseyn, T.; Lansigan, F.; et al. Bortezomib plus EPOCH Is Effective as Frontline Treatment in Patients with Plasmablastic Lymphoma. Br. J. Haematol. 2019, 184, 679–682. [Google Scholar] [CrossRef] [Green Version]
  135. Al-Malki, M.M.; Castillo, J.J.; Sloan, J.M.; Re, A. Hematopoietic Cell Transplantation for Plasmablastic Lymphoma: A Review. Biol. Blood Marrow Transplant. 2014, 20, 1877–1884. [Google Scholar] [CrossRef] [Green Version]
  136. Cattaneo, C.; Re, A.; Ungari, M.; Peli, A.; Casari, S.; Castelnuovo, F.; Fisogni, S.; Lonardi, S.; Pellegrini, V.; Petullà, M.; et al. Plasmablastic Lymphoma among Human Immunodeficiency Virus-Positive Patients: Results of a Single Center’s Experience. Leuk. Lymphoma 2015, 56, 267–269. [Google Scholar] [CrossRef]
  137. Boulanger, E.; Gérard, L.; Gabarre, J.; Molina, J.-M.; Rapp, C.; Abino, J.-F.; Cadranel, J.; Chevret, S.; Oksenhendler, E. Prognostic Factors and Outcome of Human Herpesvirus 8–Associated Primary Effusion Lymphoma in Patients with AIDS. J. Clin. Oncol. 2005, 23, 4372–4380. [Google Scholar] [CrossRef] [PubMed]
  138. Hu, Z.; Pan, Z.; Chen, W.; Shi, Y.; Wang, W.; Yuan, J.; Wang, E.; Zhang, S.; Kurt, H.; Mai, B.; et al. Primary Effusion Lymphoma: A Clinicopathological Study of 70 Cases. Cancers 2021, 13, 878. [Google Scholar] [CrossRef] [PubMed]
  139. Chadburn, A.; Hyjek, E.; Mathew, S.; Cesarman, E.; Said, J.; Knowles, D.M. KSHV-Positive Solid Lymphomas Represent an Extra-Cavitary Variant of Primary Effusion Lymphoma. Am. J. Surg. Pathol. 2004, 28, 1401–1416. [Google Scholar] [CrossRef]
  140. Narkhede, M.; Arora, S.; Ujjani, C. Primary Effusion Lymphoma: Current Perspectives. OncoTargets Ther. 2018, 11, 3747–3754. [Google Scholar] [CrossRef] [Green Version]
  141. Antar, A.; El Hajj, H.; Jabbour, M.; Khalifeh, I.; EL-Merhi, F.; Mahfouz, R.; Bazarbachi, A. Primary Effusion Lymphoma in an Elderly Patient Effectively Treated by Lenalidomide: Case Report and Review of Literature. Blood Cancer J. 2014, 4, e190. [Google Scholar] [CrossRef] [Green Version]
  142. Shimada, K.; Hayakawa, F.; Kiyoi, H. Biology and Management of Primary Effusion Lymphoma. Blood 2018, 132, 1879–1888. [Google Scholar] [CrossRef] [Green Version]
  143. Jenner, R.G.; Maillard, K.; Cattini, N.; Weiss, R.A.; Boshoff, C.; Wooster, R.; Kellam, P. Kaposi’s Sarcoma-Associated Herpesvirus-Infected Primary Effusion Lymphoma Has a Plasma Cell Gene Expression Profile. Proc. Natl. Acad. Sci. USA 2003, 100, 10399–10404. [Google Scholar] [CrossRef] [Green Version]
  144. Asou, H.; Said, J.W.; Yang, R.; Munker, R.; Park, D.J.; Kamada, N.; Koeffler, H.P. Mechanisms of Growth Control of Kaposi’s Sarcoma–Associated Herpes Virus–Associated Primary Effusion Lymphoma Cells. Blood 1998, 91, 2475–2481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Liu, L.; Eby, M.T.; Rathore, N.; Sinha, S.K.; Kumar, A.; Chaudhary, P.M. The Human Herpes Virus 8-Encoded Viral FLICE Inhibitory Protein Physically Associates with and Persistently Activates the IκB Kinase Complex. J. Biol. Chem. 2002, 277, 13745–13751. [Google Scholar] [CrossRef] [Green Version]
  146. Roy, D.; Sin, S.-H.; Damania, B.; Dittmer, D.P. Tumor Suppressor Genes FHIT and WWOX Are Deleted in Primary Effusion Lymphoma (PEL) Cell Lines. Blood 2011, 118, e32–e39. [Google Scholar] [CrossRef] [Green Version]
  147. Horenstein, M.G.; Nador, R.G.; Chadburn, A.; Hyjek, E.M.; Inghirami, G.; Knowles, D.M.; Cesarman, E. Epstein-Barr Virus Latent Gene Expression in Primary Effusion Lymphomas Containing Kaposi’s Sarcoma–Associated Herpesvirus/Human Herpesvirus-8. Blood 1997, 90, 1186–1191. [Google Scholar] [CrossRef] [Green Version]
  148. Cesarman, E.; Chang, Y.; Moore, P.S.; Said, J.W.; Knowles, D.M. Kaposi’s Sarcoma-Associated Herpesvirus-like DNA Sequences in AIDS-Related Body-Cavity-Based Lymphomas. N. Engl. J. Med. 1995, 332, 1186–1191. [Google Scholar] [CrossRef]
  149. Cai, X.; Schäfer, A.; Lu, S.; Bilello, J.P.; Desrosiers, R.C.; Edwards, R.; Raab-Traub, N.; Cullen, B.R. Epstein–Barr Virus MicroRNAs Are Evolutionarily Conserved and Differentially Expressed. PLoS Pathog. 2006, 2, e23. [Google Scholar] [CrossRef] [Green Version]
  150. Gloghini, A.; Volpi, C.C.; Gualeni, A.V.; Dolcetti, R.; Bongarzone, I.; Paoli, P.D.; Carbone, A. Multiple Viral Infections in Primary Effusion Lymphoma: A Model of Viral Cooperation in Lymphomagenesis. Expert Rev. Hematol. 2017, 10, 505–514. [Google Scholar] [CrossRef]
  151. McHugh, D.; Caduff, N.; Barros, M.H.M.; Rämer, P.C.; Raykova, A.; Murer, A.; Landtwing, V.; Quast, I.; Styles, C.T.; Spohn, M.; et al. Persistent KSHV Infection Increases EBV-Associated Tumor Formation In Vivo via Enhanced EBV Lytic Gene Expression. Cell Host Microbe 2017, 22, 61–73.e7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Bigi, R.; Landis, J.T.; An, H.; Caro-Vegas, C.; Raab-Traub, N.; Dittmer, D.P. Epstein-Barr Virus Enhances Genome Maintenance of Kaposi Sarcoma-Associated Herpesvirus. Proc. Natl. Acad. Sci. USA 2018, 115, E11379–E11387. [Google Scholar] [CrossRef] [Green Version]
  153. Olszewski, A.J.; Fallah, J.; Castillo, J.J. Human Immunodeficiency Virus-Associated Lymphomas in the Antiretroviral Therapy Era: Analysis of the National Cancer Data Base. Cancer 2016, 122, 2689–2697. [Google Scholar] [CrossRef] [PubMed]
  154. El–Fattah, M.A. Clinical Characteristics and Survival Outcome of Primary Effusion Lymphoma: A Review of 105 Patients. Hematol. Oncol. 2017, 35, 878–883. [Google Scholar] [CrossRef] [PubMed]
  155. Simonelli, C.; Spina, M.; Cinelli, R.; Talamini, R.; Tedeschi, R.; Gloghini, A.; Vaccher, E.; Carbone, A.; Tirelli, U. Clinical Features and Outcome of Primary Effusion Lymphoma in HIV-Infected Patients: A Single-Institution Study. J. Clin. Oncol. 2003, 21, 3948–3954. [Google Scholar] [CrossRef]
  156. Castillo, J.J.; Shum, H.; Lahijani, M.; Winer, E.S.; Butera, J.N. Prognosis in Primary Effusion Lymphoma Is Associated with the Number of Body Cavities Involved. Leuk. Lymphoma 2012, 53, 2378–2382. [Google Scholar] [CrossRef]
  157. Guillet, S.; Gérard, L.; Meignin, V.; Agbalika, F.; Cuccini, W.; Denis, B.; Katlama, C.; Galicier, L.; Oksenhendler, E. Classic and Extracavitary Primary Effusion Lymphoma in 51 HIV-Infected Patients from a Single Institution. Am. J. Hematol. 2016, 91, 233–237. [Google Scholar] [CrossRef] [Green Version]
  158. Hocqueloux, L.; Agbalika, F.; Oksenhendler, E.; Molina, J.-M. Long-Term Remission of an AIDS-Related Primary Effusion Lymphoma with Antiviral Therapy. AIDS 2001, 15, 280–282. [Google Scholar] [CrossRef]
  159. Lurain, K.; Polizzotto, M.N.; Aleman, K.; Bhutani, M.; Wyvill, K.M.; Gonçalves, P.H.; Ramaswami, R.; Marshall, V.A.; Miley, W.; Steinberg, S.M.; et al. Viral, Immunologic, and Clinical Features of Primary Effusion Lymphoma. Blood 2019, 133, 1753–1761. [Google Scholar] [CrossRef] [Green Version]
  160. Jessamy, K.; Ojevwe, F.O.; Doobay, R.; Naous, R.; Yu, J.; Lemke, S.M. Primary Effusion Lymphoma: Is Dose-Adjusted-EPOCH Worthwhile Therapy? Case Rep. Oncol. 2016, 9, 273–279. [Google Scholar] [CrossRef]
  161. Won, J.-H.; Han, S.-H.; Bae, S.-B.; Kim, C.-K.; Lee, N.-S.; Lee, K.-T.; Park, S.-K.; Hong, D.-S.; Lee, D.-W.; Park, H.-S. Successful Eradication of Relapsed Primary Effusion Lymphoma with High-Dose Chemotherapy and Autologous Stem Cell Transplantation in a Patient Seronegative for Human Immunodeficiency Virus. Int. J. Hematol. 2006, 83, 328–330. [Google Scholar] [CrossRef] [PubMed]
  162. Bryant, A.; Milliken, S. Successful Reduced-Intensity Conditioning Allogeneic HSCT for HIV-Related Primary Effusion Lymphoma. Biol. Blood Marrow Transplant. 2008, 14, 601–602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Xicoy, B.; Ribera, J.-M.; Miralles, P.; Berenguer, J.; Rubio, R.; Mahillo, B.; Valencia, M.-E.; Abella, E.; López-Guillermo, A.; Sureda, A.; et al. Results of Treatment with Doxorubicin, Bleomycin, Vinblastine and Dacarbazine and Highly Active Antiretroviral Therapy in Advanced Stage, Human Immunodeficiency Virus-Related Hodgkin’s Lymphoma. Haematologica 2007, 92, 191–198. [Google Scholar] [CrossRef] [Green Version]
  164. Díez-Martín, J.L.; Balsalobre, P.; Re, A.; Michieli, M.; Ribera, J.M.; Canals, C.; Conde, E.; Rosselet, A.; Gabriel, I.; Varela, R.; et al. Comparable Survival between HIV+ and HIV- Non-Hodgkin and Hodgkin Lymphoma Patients Undergoing Autologous Peripheral Blood Stem Cell Transplantation. Blood 2009, 113, 6011–6014. [Google Scholar] [CrossRef] [Green Version]
  165. Ruiz, M.; Parsons, C.; Cole, J. Characterization of HIV-Associated Hodgkin’s Lymphoma in HIV-Infected Patients: A Single-Center Experience. J. Int. Assoc. Physicians AIDS Care Chic. Ill 2002 2012, 11, 234–238. [Google Scholar] [CrossRef] [PubMed]
  166. Shiels, M.S.; Koritzinsky, E.; Clarke, C.A.; Suneja, G.; Morton, L.M.; Engels, E.A. Prevalence of HIV Infection among U.S. Hodgkin Lymphoma Cases. Cancer Epidemiol. Biomark. Prev. 2014, 23, 274–281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Clifford, G.M.; Polesel, J.; Rickenbach, M.; Dal Maso, L.; Keiser, O.; Kofler, A.; Rapiti, E.; Levi, F.; Jundt, G.; Fisch, T.; et al. Cancer Risk in the Swiss HIV Cohort Study: Associations with Immunodeficiency, Smoking, and Highly Active Antiretroviral Therapy. J. Natl. Cancer Inst. 2005, 97, 425–432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Re, A.; Cattaneo, C.; Rossi, G. Hiv and Lymphoma: From Epidemiology to Clinical Management. Mediterr. J. Hematol. Infect. Dis. 2019, 11, e2019004. [Google Scholar] [CrossRef] [PubMed]
  169. Hoffmann, C.; Hentrich, M.; Gillor, D.; Behrens, G.; Jensen, B.; Stoehr, A.; Esser, S.; Lunzen, J. van Krznaric, I.; Müller, M.; et al. Hodgkin Lymphoma Is as Common as Non-Hodgkin Lymphoma in HIV-Positive Patients with Sustained Viral Suppression and Limited Immune Deficiency: A Prospective Cohort Study. HIV Med. 2015, 16, 261–264. [Google Scholar] [CrossRef] [Green Version]
  170. Lanoy, E.; Rosenberg, P.S.; Fily, F.; Lascaux, A.-S.; Martinez, V.; Partisani, M.; Poizot-Martin, I.; Rouveix, E.; Engels, E.A.; Costagliola, D.; et al. HIV-Associated Hodgkin Lymphoma during the First Months on Combination Antiretroviral Therapy. Blood 2011, 118, 44–49. [Google Scholar] [CrossRef] [Green Version]
  171. Bohlius, J.; Schmidlin, K.; Boué, F.; Fätkenheuer, G.; May, M.; Caro-Murillo, A.M.; Mocroft, A.; Bonnet, F.; Clifford, G.; Paparizos, V.; et al. HIV-1–Related Hodgkin Lymphoma in the Era of Combination Antiretroviral Therapy: Incidence and Evolution of CD4+ T-Cell Lymphocytes. Blood 2011, 117, 6100–6108. [Google Scholar] [CrossRef] [Green Version]
  172. Dolcetti, R.; Boiocchi, M.; Gloghini, A.; Carbone, A. Pathogenetic and Histogenetic Features of HIV-Associated Hodgkin’s Disease. Eur. J. Cancer 2001, 37, 1276–1287. [Google Scholar] [CrossRef]
  173. Rapezzi, D.; Ugolini, D.; Ferraris, A.M.; Racchi, O.; Gaetani, G.F. Histological Subtypes of Hodgkin’s Disease in the Setting of HIV Infection. Ann. Hematol. 2001, 80, 340–344. [Google Scholar] [CrossRef]
  174. Küppers, R.; Rajewsky, K.; Zhao, M.; Simons, G.; Laumann, R.; Fischer, R.; Hansmann, M.L. Hodgkin Disease: Hodgkin and Reed-Sternberg Cells Picked from Histological Sections Show Clonal Immunoglobulin Gene Rearrangements and Appear to Be Derived from B Cells at Various Stages of Development. Proc. Natl. Acad. Sci. USA 1994, 91, 10962–10966. [Google Scholar] [CrossRef] [Green Version]
  175. Weniger, M.A.; Küppers, R. Molecular Biology of Hodgkin Lymphoma. Leukemia 2021, 35, 968–981. [Google Scholar] [CrossRef]
  176. Chang, K.; Albujar, P.; Chen, Y.; Johnson, R.; Weiss, L. High Prevalence of Epstein-Barr Virus in the Reed-Sternberg Cells of Hodgkin’s Disease Occurring in Peru. Blood 1993, 81, 496–501. [Google Scholar] [CrossRef]
  177. Audouin, J.; Diebold, J.; Pallesen, G. Frequent Expression of Epstein-Barr Virus Latent Membrane Protein-1 in Tumour Cells of Hodgkin’s Disease in HIV-Positive Patients. J. Pathol. 1992, 167, 381–384. [Google Scholar] [CrossRef] [PubMed]
  178. Naresh, K.N.; Johnson, J.; Srinivas, V.; Soman, C.S.; Saikia, T.; Advani, S.H.; Badwe, R.A.; Dinshaw, K.A.; Muckaden, M.; Magrath, I.; et al. Epstein-Barr Virus Association in Classical Hodgkin’s Disease Provides Survival Advantage to Patients and Correlates with Higher Expression of Proliferation Markers in Reed-Sternberg Cells. Ann. Oncol. 2000, 11, 91–96. [Google Scholar] [CrossRef] [PubMed]
  179. Young, L.S.; Murray, P.G. Epstein–Barr Virus and Oncogenesis: From Latent Genes to Tumours. Oncogene 2003, 22, 5108–5121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Caldwell, R.G.; Wilson, J.B.; Anderson, S.J.; Longnecker, R. Epstein-Barr Virus LMP2A Drives B Cell Development and Survival in the Absence of Normal B Cell Receptor Signals. Immunity 1998, 9, 405–411. [Google Scholar] [CrossRef] [Green Version]
  181. Portis, T.; Dyck, P.; Longnecker, R. Epstein-Barr Virus (EBV) LMP2A Induces Alterations in Gene Transcription Similar to Those Observed in Reed-Sternberg Cells of Hodgkin Lymphoma. Blood 2003, 102, 4166–4178. [Google Scholar] [CrossRef]
  182. Cen, O.; Longnecker, R. Latent Membrane Protein 2 (LMP2). Curr. Top. Microbiol. Immunol. 2015, 391, 151–180. [Google Scholar] [CrossRef]
  183. Kieser, A.; Sterz, K.R. The Latent Membrane Protein 1 (LMP1). Curr. Top. Microbiol. Immunol. 2015, 391, 119–149. [Google Scholar] [CrossRef]
  184. Kilger, E.; Kieser, A.; Baumann, M.; Hammerschmidt, W. Epstein-Barr Virus-Mediated B-Cell Proliferation Is Dependent upon Latent Membrane Protein 1, Which Simulates an Activated CD40 Receptor. EMBO J. 1998, 17, 1700–1709. [Google Scholar] [CrossRef] [Green Version]
  185. Green, M.R.; Rodig, S.; Juszczynski, P.; Ouyang, J.; Sinha, P.; O’Donnell, E.; Neuberg, D.; Shipp, M.A. Constitutive AP-1 Activity and EBV Infection Induce PD-L1 in Hodgkin Lymphomas and Post-Transplant Lymphoproliferative Disorders: Implications for Targeted Therapy. Clin. Cancer Res. 2012, 18, 1611–1618. [Google Scholar] [CrossRef] [Green Version]
  186. Armand, P. Immune Checkpoint Blockade in Hematologic Malignancies. Blood 2015, 125, 3393–3400. [Google Scholar] [CrossRef]
  187. Carbone, A.; Gloghini, A.; Caruso, A.; Paoli, P.D.; Dolcetti, R. The Impact of EBV and HIV Infection on the Microenvironmental Niche Underlying Hodgkin Lymphoma Pathogenesis. Int. J. Cancer 2017, 140, 1233–1245. [Google Scholar] [CrossRef]
  188. Wu, R.; Sattarzadeh, A.; Rutgers, B.; Diepstra, A.; van den Berg, A.; Visser, L. The Microenvironment of Classical Hodgkin Lymphoma: Heterogeneity by Epstein–Barr Virus Presence and Location within the Tumor. Blood Cancer J. 2016, 6, e417. [Google Scholar] [CrossRef] [PubMed]
  189. Dukers, D.F.; Meij, P.; Vervoort, M.B.H.J.; Vos, W.; Scheper, R.J.; Meijer, C.J.L.M.; Bloemena, E.; Middeldorp, J.M. Direct Immunosuppressive Effects of EBV-Encoded Latent Membrane Protein 1. J. Immunol. 2000, 165, 663–670. [Google Scholar] [CrossRef]
  190. Hartmann, S.; Jakobus, C.; Rengstl, B.; Döring, C.; Newrzela, S.; Brodt, H.-R.; Wolf, T.; Hansmann, M.-L. Spindle-Shaped CD163+ Rosetting Macrophages Replace CD4+ T-Cells in HIV-Related Classical Hodgkin Lymphoma. Mod. Pathol. 2013, 26, 648–657. [Google Scholar] [CrossRef] [PubMed]
  191. Higuchi, H.; Yamakawa, N.; Imadome, K.-I.; Yahata, T.; Kotaki, R.; Ogata, J.; Kakizaki, M.; Fujita, K.; Lu, J.; Yokoyama, K.; et al. Role of Exosomes as a Proinflammatory Mediator in the Development of EBV-Associated Lymphoma. Blood 2018, 131, 2552–2567. [Google Scholar] [CrossRef] [Green Version]
  192. Thompson, L.D.R.; Fisher, M.S.I.; Chu, W.S.; Nelson, A.; Abbondanzo, S.L. HIV-Associated Hodgkin Lymphoma: A Clinicopathologic and Immunophenotypic Study of 45 Cases. Am. J. Clin. Pathol. 2004, 121, 727–738. [Google Scholar] [CrossRef]
  193. Olszewski, A.J.; Castillo, J.J. Outcomes of HIV-Associated Hodgkin Lymphoma in the Era of Antiretroviral Therapy. AIDS 2016, 30, 787–796. [Google Scholar] [CrossRef] [PubMed]
  194. Westmoreland, K.D.; Stanley, C.C.; Montgomery, N.D.; Kaimila, B.; Kasonkanji, E.; El-Mallawany, N.K.; Wasswa, P.; Mtete, I.; Butia, M.; Itimu, S.; et al. Hodgkin Lymphoma, HIV, and Epstein–Barr Virus in Malawi: Longitudinal Results from the Kamuzu Central Hospital Lymphoma Study. Pediatr. Blood Cancer 2017, 64, e26302. [Google Scholar] [CrossRef] [Green Version]
  195. Hentrich, M.; Berger, M.; Wyen, C.; Siehl, J.; Rockstroh, J.K.; Müller, M.; Fätkenheuer, G.; Seidel, E.; Nickelsen, M.; Wolf, T.; et al. Stage-Adapted Treatment of HIV-Associated Hodgkin Lymphoma: Results of a Prospective Multicenter Study. J. Clin. Oncol. 2012, 30, 4117–4123. [Google Scholar] [CrossRef]
  196. Hartmann, P.; Rehwald, U.; Salzberger, B.; Franzen, C.; Sieber, M.; Wöhrmann, A.; Diehl, V. BEACOPP Therapeutic Regimen for Patients with Hodgkin’s Disease and HIV Infection. Ann. Oncol. 2003, 14, 1562–1569. [Google Scholar] [CrossRef] [PubMed]
  197. Krishnan, A.; Molina, A.; Zaia, J.; Smith, D.; Vasquez, D.; Kogut, N.; Falk, P.M.; Rosenthal, J.; Alvarnas, J.; Forman, S.J. Durable Remissions with Autologous Stem Cell Transplantation for High-Risk HIV-Associated Lymphomas. Blood 2005, 105, 874–878. [Google Scholar] [CrossRef]
  198. Spitzer, T.R.; Ambinder, R.F.; Lee, J.Y.; Kaplan, L.D.; Wachsman, W.; Straus, D.J.; Aboulafia, D.M.; Scadden, D.T. Dose-Reduced Busulfan, Cyclophosphamide, and Autologous Stem Cell Transplantation for Human Immunodeficiency Virus–Associated Lymphoma: AIDS Malignancy Consortium Study 020. Biol. Blood Marrow Transplant. 2008, 14, 59–66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. Tisi, M.C.; Cupelli, E.; Santangelo, R.; Maiolo, E.; Alma, E.; Giachelia, M.; Martini, M.; Bellesi, S.; D’Alò, F.; Voso, M.T.; et al. Whole Blood EBV-DNA Predicts Outcome in Diffuse Large B-Cell Lymphoma. Leuk. Lymphoma 2016, 57, 628–634. [Google Scholar] [CrossRef]
  200. Okamoto, A.; Yanada, M.; Miura, H.; Inaguma, Y.; Tokuda, M.; Morishima, S.; Kanie, T.; Yamamoto, Y.; Mizuta, S.; Akatsuka, Y.; et al. Prognostic Significance of Epstein–Barr Virus DNA Detection in Pretreatment Serum in Diffuse Large B–cell Lymphoma. Cancer Sci. 2015, 106, 1576–1581. [Google Scholar] [CrossRef]
  201. Kanakry, J.A.; Li, H.; Gellert, L.L.; Lemas, M.V.; Hsieh, W.; Hong, F.; Tan, K.L.; Gascoyne, R.D.; Gordon, L.I.; Fisher, R.I.; et al. Plasma Epstein-Barr Virus DNA Predicts Outcome in Advanced Hodgkin Lymphoma: Correlative Analysis from a Large North American Cooperative Group Trial. Blood 2013, 121, 3547–3553. [Google Scholar] [CrossRef] [Green Version]
  202. Kanakry, J.A.; Hegde, A.M.; Durand, C.M.; Massie, A.B.; Greer, A.E.; Ambinder, R.F.; Valsamakis, A. The Clinical Significance of EBV DNA in the Plasma and Peripheral Blood Mononuclear Cells of Patients with or without EBV Diseases. Blood 2016, 127, 2007–2017. [Google Scholar] [CrossRef]
  203. Kanakry, J.; Ambinder, R. The Biology and Clinical Utility of EBV Monitoring in Blood. In Epstein Barr Virus Volume 2: One Herpes Virus: Many Diseases; Current Topics in Microbiology and Immunology; Münz, C., Ed.; Springer International Publishing: Cham, Switzerland, 2015; pp. 475–499. [Google Scholar]
  204. Lu, T.-X.; Liang, J.-H.; Miao, Y.; Fan, L.; Wang, L.; Qu, X.-Y.; Cao, L.; Gong, Q.-X.; Wang, Z.; Zhang, Z.-H.; et al. Epstein-Barr Virus Positive Diffuse Large B-Cell Lymphoma Predict Poor Outcome, Regardless of the Age. Sci. Rep. 2015, 5, 12168. [Google Scholar] [CrossRef]
  205. Gandhi, M.K.; Lambley, E.; Burrows, J.; Dua, U.; Elliott, S.; Shaw, P.J.; Prince, H.M.; Wolf, M.; Clarke, K.; Underhill, C.; et al. Plasma Epstein-Barr Virus (EBV) DNA Is a Biomarker for EBV-Positive Hodgkin’s Lymphoma. Clin. Cancer Res. 2006, 12, 460–464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Gallagher, A.; Armstrong, A.A.; MacKenzie, J.; Shield, L.; Khan, G.; Lake, A.; Proctor, S.; Taylor, P.; Clements, G.B.; Jarrett, R.F. Detection of Epstein-Barr Virus (EBV) Genomes in the Serum of Patients with EBV-Associated Hodgkin’s Disease. Int. J. Cancer 1999, 84, 442–448. [Google Scholar] [CrossRef]
  207. Hohaus, S.; Santangelo, R.; Giachelia, M.; Vannata, B.; Massini, G.; Cuccaro, A.; Martini, M.; Cesarini, V.; Cenci, T.; D’Alo, F.; et al. The Viral Load of Epstein–Barr Virus (EBV) DNA in Peripheral Blood Predicts for Biological and Clinical Characteristics in Hodgkin Lymphoma. Clin. Cancer Res. 2011, 17, 2885–2892. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Martelius, T.; Anttila, V.-J.; Nihtinen, A.; Lappalainen, M.; Aalto, S.M.; Hedman, K. Clinical Characteristics, Outcome and the Role of Viral Load in Nontransplant Patients with Epstein--Barr Viraemia. Clin. Microbiol. Infect. 2010, 16, 657–662. [Google Scholar] [CrossRef] [Green Version]
  209. Montgomery, N.D.; Randall, C.; Painschab, M.; Seguin, R.; Kaimila, B.; Kasonkanji, E.; Zuze, T.; Krysiak, R.; Sanders, M.K.; Elliott, A.; et al. High Pretreatment Plasma Epstein–Barr Virus (EBV) DNA Level Is a Poor Prognostic Marker in HIV–associated, EBV–negative Diffuse Large B–cell Lymphoma in Malawi. Cancer Med. 2019, 9, 552–561. [Google Scholar] [CrossRef] [Green Version]
  210. Navarro, J.-T.; Hernández, Á.; Rodríguez-Manzano, J.; Mate, J.-L.; Grau, J.; Morgades, M.; Martró, E.; Tural, C.; Ribera, J.-M.; Matas, L. Plasma Epstein-Barr viral load measurement as a diagnostic marker of lymphoma in HIV-infected patients. Med. Clínica 2010, 135, 485–490. [Google Scholar] [CrossRef] [PubMed]
  211. Baptista, M.J.; Hernandez-Rodriguez, A.; Martinez-Caceres, E.; Morgades, M.; Martinez-Picado, J.; Sirera, G.; Sancho, J.-M.; Feliu, E.; Ribera, J.-M.; Navarro, J.-T. Epstein-Barr Viral Loads and Serum Free Light Chains Levels Are Potential Follow-up Markers of HIV-Related Lymphomas. Leuk. Lymphoma 2017, 58, 211–213. [Google Scholar] [CrossRef]
  212. Tanaka, P.Y.; Ohshima, K.; Matsuoka, M.; Sabino, E.C.; Ferreira, S.C.; Nishya, A.S.; de Oliveira Costa, R.; Calore, E.E.; Perez, N.M.; Pereira, J. Epstein–Barr Viral Load Is Associated to Response in AIDS-Related Lymphomas. Indian J. Hematol. Blood Transfus. 2014, 30, 191–194. [Google Scholar] [CrossRef] [Green Version]
  213. Fan, H.; Kim, S.C.; Chima, C.O.; Israel, B.F.; Lawless, K.M.; Eagan, P.A.; Elmore, S.; Moore, D.T.; Schichman, S.A.; Swinnen, L.J.; et al. Epstein–Barr Viral Load as a Marker of Lymphoma in AIDS Patients. J. Med. Virol. 2005, 75, 59–69. [Google Scholar] [CrossRef]
  214. Amiel, C.; LeGoff, J.; Lescure, F.X.; Coste-Burel, M.; Deback, C.; Fafi-Kremer, S.; Gueudin, M.; Lafon, M.-E.; Leruez-Ville, M.; Mengelle, C.; et al. Epstein-Barr Virus Load in Whole Blood Correlates with HIV Surrogate Markers and Lymphoma: A French National Cross-Sectional Study. JAIDS J. Acquir. Immune Defic. Syndr. 2009, 50, 427–429. [Google Scholar] [CrossRef]
  215. Leruez-Ville, M.; Seng, R.; Morand, P.; Boufassa, F.; Boue, F.; Deveau, C.; Rouzioux, C.; Goujard, C.; Seigneurin, J.M.; Meyer, L. Blood Epstein–Barr Virus DNA Load and Risk of Progression to AIDS-Related Systemic B Lymphoma. HIV Med. 2012, 13, 479–487. [Google Scholar] [CrossRef] [PubMed]
  216. van Baarle, D.; Wolthers, K.C.; Hovenkamp, E.; Niesters, H.G.M.; Osterhaus, A.D.M.E.; Miedema, F.; van Oers, M.H.J. Absolute Level of Epstein-Barr Virus DNA in Human Immunodeficiency Virus Type 1 Infection Is Not Predictive of AIDS-Related Non-Hodgkin Lymphoma. J. Infect. Dis. 2002, 186, 405–409. [Google Scholar] [CrossRef] [Green Version]
  217. Lupo, J.; Germi, R.; Lancar, R.; Algarte-Genin, M.; Hendel-Chavez, H.; Taoufik, Y.; Mounier, N.; Partisani, M.; Bonnet, F.; Meyohas, M.-C.; et al. Prospective Evaluation of Blood Epstein–Barr Virus DNA Load and Antibody Profile in HIV-Related Non-Hodgkin Lymphomas. AIDS 2021, 35, 861–868. [Google Scholar] [CrossRef]
  218. Ul-Haq, I.; Dalla Pria, A.; Suardi, E.; Pinato, D.J.; Froeling, F.; Forni, J.; Randell, P.; Bower, M. Blood Epstein–Barr Virus DNA Does Not Predict Outcome in Advanced HIV-Associated Hodgkin Lymphoma. Med. Oncol. 2018, 35, 53. [Google Scholar] [CrossRef] [Green Version]
  219. Lupo, J.; Germi, R.; Lancar, R.; Algarte-Genin, M.; Hendel-Chavez, H.; Taoufik, Y.; Mounier, N.; Partisani, M.; Bonnet, F.; Meyohas, M.-C.; et al. Epstein–Barr Virus Biomarkers Have No Prognostic Value in HIV-Related Hodgkin Lymphoma in the Modern Combined Antiretroviral Therapy Era. AIDS 2019, 33, 993–1000. [Google Scholar] [CrossRef]
  220. Barta, S.K.; Xue, X.; Wang, D.; Tamari, R.; Lee, J.Y.; Mounier, N.; Kaplan, L.D.; Ribera, J.-M.; Spina, M.; Tirelli, U.; et al. Treatment Factors Affecting Outcomes in HIV-Associated Non-Hodgkin Lymphomas: A Pooled Analysis of 1546 Patients. Blood 2013, 122, 3251–3262. [Google Scholar] [CrossRef] [Green Version]
  221. Little, R.F.; Dunleavy, K. Update on the Treatment of HIV-Associated Hematologic Malignancies. Hematology 2013, 2013, 382–388. [Google Scholar] [CrossRef] [PubMed]
  222. Mayer, K.H.; Torres, H.A.; Mulanovich, V. Management of HIV Infection in Patients with Cancer Receiving Chemotherapy. Clin. Infect. Dis. 2014, 59, 106–114. [Google Scholar] [CrossRef]
  223. Noy, A. Optimizing Treatment of HIV-Associated Lymphoma. Blood 2019, 134, 1385–1394. [Google Scholar] [CrossRef]
  224. Hill, Q.A.; Owen, R.G. CNS Prophylaxis in Lymphoma: Who to Target and What Therapy to Use. Blood Rev. 2006, 20, 319–332. [Google Scholar] [CrossRef]
  225. Sancho, J.-M.; Ribera, J.-M. Recaída en el sistema nervioso central en el linfoma B difuso de célula grande: Factores de riesgo. Med. Clín. 2016, 146, 74–80. [Google Scholar] [CrossRef]
  226. Fletcher, C.D.; Kahl, B.S. Central Nervous System Involvement in Diffuse Large B-Cell Lymphoma: An Analysis of Risks and Prevention Strategies in the Post-Rituximab Era. Leuk. Lymphoma 2014, 55, 2228–2240. [Google Scholar] [CrossRef]
  227. Mazhar, D.; Stebbing, J.; Bower, M. Non-Hodgkin’s Lymphoma and the CNS: Prophylaxis and Therapy in Immunocompetent and HIV-Positive Individuals. Expert Rev. Anticancer. Ther. 2006, 6, 335–341. [Google Scholar] [CrossRef]
  228. Smith, T.J.; Khatcheressian, J.; Lyman, G.H.; Ozer, H.; Armitage, J.O.; Balducci, L.; Bennett, C.L.; Cantor, S.B.; Crawford, J.; Cross, S.J.; et al. 2006 Update of Recommendations for the Use of White Blood Cell Growth Factors: An Evidence-Based Clinical Practice Guideline. J. Clin. Oncol. 2006, 24, 3187–3205. [Google Scholar] [CrossRef] [Green Version]
  229. Re, A.; Cattaneo, C.; Montoto, S. Treatment Management of Haematological Malignancies in People Living with HIV. Lancet Haematol. 2020, 7, e679–e689. [Google Scholar] [CrossRef]
  230. Bower, M.; Palfreeman, A.; Alfa-Wali, M.; Bunker, C.; Burns, F.; Churchill, D.; Collins, S.; Cwynarski, K.; Edwards, S.; Fields, P.; et al. British HIV Association Guidelines for HIV-Associated Malignancies 2014. HIV Med. 2014, 15 (Suppl. 2), 1–92. [Google Scholar] [CrossRef]
  231. Reid, E.; Suneja, G.; Ambinder, R.F.; Ard, K.; Baiocchi, R.; Barta, S.K.; Carchman, E.; Cohen, A.; Gupta, N.; Johung, K.L.; et al. Cancer in People Living with HIV, Version 1.2018, NCCN Clinical Practice Guidelines in Oncology. JNCCN J. Natl. Compr. Cancer Netw. 2018, 16, 986–1017. [Google Scholar] [CrossRef] [Green Version]
  232. Navarro, J.-T.; Moltó, J.; Tapia, G.; Ribera, J.-M. Hodgkin Lymphoma in People Living with HIV. Cancers 2021, 13, 4366. [Google Scholar] [CrossRef]
  233. Thompson, S.; Messick, T.; Schultz, D.C.; Reichman, M.; Lieberman, P.M. Development of a High-Throughput Screen for Inhibitors of Epstein-Barr Virus EBNA1. J. Biomol. Screen. 2010, 15, 1107–1115. [Google Scholar] [CrossRef] [Green Version]
  234. Messick, T.E.; Smith, G.R.; Soldan, S.S.; McDonnell, M.E.; Deakyne, J.S.; Malecka, K.A.; Tolvinski, L.; van den Heuvel, A.P.J.; Gu, B.-W.; Cassel, J.A.; et al. Structure-Based Design of Small-Molecule Inhibitors of EBNA1 DNA Binding Blocks Epstein-Barr Virus Latent Infection and Tumor Growth. Sci. Transl. Med. 2019, 11, eaau5612. [Google Scholar] [CrossRef]
  235. Israel, B.F.; Gulley, M.; Elmore, S.; Ferrini, S.; Feng, W.; Kenney, S.C. Anti-CD70 Antibodies: A Potential Treatment for EBV CD70-Expressing Lymphomas. Mol. Cancer Ther. 2005, 4, 2037–2044. [Google Scholar] [CrossRef] [Green Version]
  236. Schaft, N.; Lankiewicz, B.; Drexhage, J.; Berrevoets, C.; Moss, D.J.; Levitsky, V.; Bonneville, M.; Lee, S.P.; McMichael, A.J.; Gratama, J.-W.; et al. T Cell Re-Targeting to EBV Antigens Following TCR Gene Transfer: CD28-Containing Receptors Mediate Enhanced Antigen-Specific IFNgamma Production. Int. Immunol. 2006, 18, 591–601. [Google Scholar] [CrossRef]
  237. Shen, R.R.; Pham, C.D.; Wu, M.; Munson, D.J.; Aftab, B.T. CD19 Chimeric Antigen Receptor (CAR) Engineered Epstein-Barr Virus (EBV) Specific T Cells—An off-the-Shelf, Allogeneic CAR T-Cell Immunotherapy Platform. Cytotherapy 2019, 21, S11. [Google Scholar] [CrossRef]
  238. Quan, L.; Chen, X.; Liu, A.; Zhang, Y.; Guo, X.; Yan, S.; Liu, Y. PD-1 Blockade Can Restore Functions of T-Cells in Epstein-Barr Virus-Positive Diffuse Large B-Cell Lymphoma In Vitro. PLoS ONE 2015, 10, e0136476. [Google Scholar] [CrossRef]
  239. Kim, S.-J.; Hyeon, J.; Cho, I.; Ko, Y.H.; Kim, W.S. Comparison of Efficacy of Pembrolizumab between Epstein-Barr Virus‒Positive and ‒Negative Relapsed or Refractory Non-Hodgkin Lymphomas. Cancer Res. Treat. 2019, 51, 611–622. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  240. Cho, H.-I.; Kim, U.-H.; Shin, A.-R.; Won, J.-N.; Lee, H.-J.; Sohn, H.-J.; Kim, T.-G. A Novel Epstein-Barr Virus-Latent Membrane Protein-1-Specific T-Cell Receptor for TCR Gene Therapy. Br. J. Cancer 2018, 118, 534–545. [Google Scholar] [CrossRef] [Green Version]
  241. van Zyl, D.G.; Mautner, J.; Delecluse, H.-J. Progress in EBV Vaccines. Front. Oncol. 2019, 9, 104. [Google Scholar] [CrossRef] [Green Version]
  242. Aboulafia, D.M.; Ratner, L.; Miles, S.A.; Harrington, W.J. AIDS Associated Malignancies Clinical Trials Consortium Antiviral and Immunomodulatory Treatment for AIDS-Related Primary Central Nervous System Lymphoma: AIDS Malignancies Consortium Pilot Study 019. Clin. Lymphoma Myeloma 2006, 6, 399–402. [Google Scholar] [CrossRef]
  243. Raez, L.; Cabral, L.; Cai, J.P.; Landy, H.; Sfakianakis, G.; Byrne, G.E.; Hurley, J.; Scerpella, E.; Jayaweera, D.; Harrington, W.J. Treatment of AIDS-Related Primary Central Nervous System Lymphoma with Zidovudine, Ganciclovir, and Interleukin 2. AIDS Res. Hum. Retrovir. 1999, 15, 713–719. [Google Scholar] [CrossRef] [PubMed]
  244. Bossolasco, S.; Falk, K.I.; Ponzoni, M.; Ceserani, N.; Crippa, F.; Lazzarin, A.; Linde, A.; Cinque, P. Ganciclovir Is Associated with Low or Undetectable Epstein-Barr Virus DNA Load in Cerebrospinal Fluid of Patients with HIV-Related Primary Central Nervous System Lymphoma. Clin. Infect. Dis. 2006, 42, e21–e25. [Google Scholar] [CrossRef] [Green Version]
  245. Faller, D.V.; Mentzer, S.J.; Perrine, S.P. Induction of the Epstein–Barr Virus Thymidine Kinase Gene with Concomitant Nucleoside Antivirals as a Therapeutic Strategy for Epstein–Barr Virus–Associated Malignancies. Curr. Opin. Oncol. 2001, 13, 360–367. [Google Scholar] [CrossRef]
  246. Perrine, S.P.; Hermine, O.; Small, T.; Suarez, F.; O’Reilly, R.; Boulad, F.; Fingeroth, J.; Askin, M.; Levy, A.; Mentzer, S.J.; et al. A Phase 1/2 Trial of Arginine Butyrate and Ganciclovir in Patients with Epstein-Barr Virus–Associated Lymphoid Malignancies. Blood 2007, 109, 2571–2578. [Google Scholar] [CrossRef] [Green Version]
  247. Faller, D.V.; Ghosh, S.; Feldman, T.; Lerner, A.; Smith, J.; Goy, A.; Berenson, R.; Perrine, S. Short-Term Exposure to Arginine Butyrate, in Combination with Ganciclovir, Is as Effective as Continuous Exposure for Virus-Targeted Therapy of EBV-Positive Lymphomas. Blood 2009, 114, 4754. [Google Scholar] [CrossRef]
  248. O’Connor, O.A.; Heaney, M.L.; Schwartz, L.; Richardson, S.; Willim, R.; MacGregor-Cortelli, B.; Curly, T.; Moskowitz, C.; Portlock, C.; Horwitz, S.; et al. Clinical Experience with Intravenous and Oral Formulations of the Novel Histone Deacetylase Inhibitor Suberoylanilide Hydroxamic Acid in Patients with Advanced Hematologic Malignancies. J. Clin. Oncol. 2006, 24, 166–173. [Google Scholar] [CrossRef]
  249. Crump, M.; Coiffier, B.; Jacobsen, E.D.; Sun, L.; Ricker, J.L.; Xie, H.; Frankel, S.R.; Randolph, S.S.; Cheson, B.D. Oral Vorinostat (Suberoylanilide Hydroxamic Acid, SAHA) in Relapsed Diffuse Large B-Cell Lymphoma (DLBCL): Final Results of a Phase II Trial. J. Clin. Oncol. 2007, 25, 18511. [Google Scholar] [CrossRef]
  250. Armand, P.; Shipp, M.A.; Ribrag, V.; Michot, J.-M.; Zinzani, P.L.; Kuruvilla, J.; Snyder, E.S.; Ricart, A.D.; Balakumaran, A.; Rose, S.; et al. Programmed Death-1 Blockade with Pembrolizumab in Patients with Classical Hodgkin Lymphoma After Brentuximab Vedotin Failure. J. Clin. Oncol. 2016, 34, 3733–3739. [Google Scholar] [CrossRef] [PubMed]
  251. Bollard, C.M.; Gottschalk, S.; Torrano, V.; Diouf, O.; Ku, S.; Hazrat, Y.; Carrum, G.; Ramos, C.; Fayad, L.; Shpall, E.J.; et al. Sustained Complete Responses in Patients with Lymphoma Receiving Autologous Cytotoxic T Lymphocytes Targeting Epstein-Barr Virus Latent Membrane Proteins. J. Clin. Oncol. 2014, 392, 798–808. [Google Scholar] [CrossRef]
  252. Icheva, V.; Kayser, S.; Wolff, D.; Tuve, S.; Kyzirakos, C.; Bethge, W.; Greil, J.; Albert, M.H.; Schwinger, W.; Nathrath, M.; et al. Adoptive Transfer of Epstein-Barr Virus (EBV) Nuclear Antigen 1–Specific T Cells As Treatment for EBV Reactivation and Lymphoproliferative Disorders After Allogeneic Stem-Cell Transplantation. J. Clin. Oncol. 2013, 31, 39–48. [Google Scholar] [CrossRef] [PubMed]
Figure 1. EBV-associated HIV-related lymphoma development during B-cell differentiation in the germinal center. Burkitt lymphoma (BL) and germinal center B-cell diffuse large B-cell lymphoma (GCB-DLBCL) have a germinal center (GC) origin. Primary central nervous system lymphoma (PCNSL) has a late B-cell origin with both ABC and GC B-cell features. Hodgkin lymphoma (HL) is originated in a preapoptotic GC B-cell. ABC-DLBCL develops after the activation of the B-cell through the interaction with antigen-presenting cells. After this event, cells can differentiate into memory B-cells or plasma cells. During plasma cell differentiation, activated B-cells are first differentiated to plasmablast, which is the cell of origin of plasmablastic lymphoma (PBL). Primary effusion lymphoma (PEL) is originated in post-GC B-cells with plasmablastic differentiation.
Figure 1. EBV-associated HIV-related lymphoma development during B-cell differentiation in the germinal center. Burkitt lymphoma (BL) and germinal center B-cell diffuse large B-cell lymphoma (GCB-DLBCL) have a germinal center (GC) origin. Primary central nervous system lymphoma (PCNSL) has a late B-cell origin with both ABC and GC B-cell features. Hodgkin lymphoma (HL) is originated in a preapoptotic GC B-cell. ABC-DLBCL develops after the activation of the B-cell through the interaction with antigen-presenting cells. After this event, cells can differentiate into memory B-cells or plasma cells. During plasma cell differentiation, activated B-cells are first differentiated to plasmablast, which is the cell of origin of plasmablastic lymphoma (PBL). Primary effusion lymphoma (PEL) is originated in post-GC B-cells with plasmablastic differentiation.
Cancers 13 05534 g001
Figure 2. EBV genes and noncoding RNAs are involved in diverse lymphomagenic processes. EBV can prevent apoptosis and promote cell growth and proliferation via latency and lytic genes, miRNAs, and EBERs. These EBV components can also protect infected B-cells from the immune cells’ recognition via latency genes, miRNAs, and EBERs. Moreover, miRNAs can also alter the microenvironment, inducing the transformation of tumor-associated macrophages. Latency genes and EBERs are also involved in activation and transformation of B-cells. EBV may also induce chemoresistance mediated by lytic genes.
Figure 2. EBV genes and noncoding RNAs are involved in diverse lymphomagenic processes. EBV can prevent apoptosis and promote cell growth and proliferation via latency and lytic genes, miRNAs, and EBERs. These EBV components can also protect infected B-cells from the immune cells’ recognition via latency genes, miRNAs, and EBERs. Moreover, miRNAs can also alter the microenvironment, inducing the transformation of tumor-associated macrophages. Latency genes and EBERs are also involved in activation and transformation of B-cells. EBV may also induce chemoresistance mediated by lytic genes.
Cancers 13 05534 g002
Table 1. Lymphomas affecting PLWH. The percentage of EBV, latency type, and the CD4+ lymphocyte counts for each lymphoma are shown. BL: Burkitt lymphoma; PBL: plasmablastic lymphoma; DLBCL: diffuse large B-cell lymphoma; EBV: Epstein–Barr virus; HL: Hodgkin lymphoma; PCNSL: primary central nervous system lymphoma; PEL: primary effusion lymphoma.
Table 1. Lymphomas affecting PLWH. The percentage of EBV, latency type, and the CD4+ lymphocyte counts for each lymphoma are shown. BL: Burkitt lymphoma; PBL: plasmablastic lymphoma; DLBCL: diffuse large B-cell lymphoma; EBV: Epstein–Barr virus; HL: Hodgkin lymphoma; PCNSL: primary central nervous system lymphoma; PEL: primary effusion lymphoma.
LymphomaEBV FrequencyEBV Latency TypeCD4+ Lymphocyte Counts (Cells/µL)
DLBCL30–50%I/II/III100–223
PCNSL80–100%III<50
BL30–60%I200–270
PBL75–100%I63–165
PEL70–100%I98–133
HL90–100%II150–200
Table 2. Current therapeutic regimens of HRL and EBV-targeted therapies. (A). Chemotherapeutic regimens of 1st and 2nd line in combination with cART for the treatment of HRL associated with EBV infection. (B). Diverse EBV-targeted therapeutic strategies in preclinical studies (left) and EBV-targeted therapies tested in clinical trials in EBV-related lymphomas (right). ABVD: doxorubicin, bleomycin, vinblastine, and dacarbazine; ASCT: autologous stem cell transplantation; AZT: azidotimidine; BEACOPP: bleomycin, etoposide, doxorubicin, cyclophosphamide, vincristine, procarbazine, prednisone; BL: Burkitt lymphoma; Burkimab: cyclophosphamide, methotrexate, dexamethasone, ifosfamide, vincristine, etoposide, cytarabine, doxorubicin, vindesine; cART: combined antiretroviral therapy; CAR-T: chimeric antigen receptor T-cells; CHOP: cyclophosphamide, doxorubicin, vincristine, and prednisone; CODOX-M/IVAC: cyclophosphamide, doxorubicin, vincristine, methotrexate/ifosfamide, etoposide, cytarabine; CTLs: cytotoxic T-lymphocytes; DA-EPOCH: dose-adjusted etoposide, prednisone, vincristine, cyclophosphamide, and doxorubicin; DHAP: dexamethasone, cytarabine, and cisplatin; DHAX: dexamethasone, cytarabine, and oxaliplatin; DLBCL: diffuse large B-cell lymphoma; EBNA1: EBV-nuclear antigen 1; EBV: Epstein–Barr virus; EBVST: EBV-specific T-cells; ESHAP: etoposide, cisplatin, methylprednisolone, and cytarabine; GCV: ganciclovir; GDP: gemcitabine, dexamethasone, and oxaliplatin; GEMOX: gemcitabine and oxaliplatin; HDAC: histone deacetylase; HD-MTX: high dose methotrexate; HIV: human immunodeficiency virus; HL: Hodgkin lymphoma; Hyper-CVAD: cyclophosphamide, doxorubicin, vincristine, dexamethasone with methotrexate and high-dose cytarabine; ICE: ifosfamide, etoposide, and carboplatin; IC-HL: immunocompetent Hodgkin lymphoma; LMP1: latent membrane protein-1; LMP2: latent membrane protein-2; NHL: non-Hodgkin lymphoma; PBL: plasmablastic lymphoma; PCNSL: primary central nervous system lymphoma; PD-1: programmed death 1; PD-L1: programmed death ligand 1; PEL: primary effusion lymphoma; PTLD: post-transplant lymphoproliferative disorder; R: rituximab; SAHA: suberoylanilide hydroxamic acid; SCT: stem cell transplantation; THP-COP: pirarubicin, cyclophosphamide, vincristine, and prednisolone.
Table 2. Current therapeutic regimens of HRL and EBV-targeted therapies. (A). Chemotherapeutic regimens of 1st and 2nd line in combination with cART for the treatment of HRL associated with EBV infection. (B). Diverse EBV-targeted therapeutic strategies in preclinical studies (left) and EBV-targeted therapies tested in clinical trials in EBV-related lymphomas (right). ABVD: doxorubicin, bleomycin, vinblastine, and dacarbazine; ASCT: autologous stem cell transplantation; AZT: azidotimidine; BEACOPP: bleomycin, etoposide, doxorubicin, cyclophosphamide, vincristine, procarbazine, prednisone; BL: Burkitt lymphoma; Burkimab: cyclophosphamide, methotrexate, dexamethasone, ifosfamide, vincristine, etoposide, cytarabine, doxorubicin, vindesine; cART: combined antiretroviral therapy; CAR-T: chimeric antigen receptor T-cells; CHOP: cyclophosphamide, doxorubicin, vincristine, and prednisone; CODOX-M/IVAC: cyclophosphamide, doxorubicin, vincristine, methotrexate/ifosfamide, etoposide, cytarabine; CTLs: cytotoxic T-lymphocytes; DA-EPOCH: dose-adjusted etoposide, prednisone, vincristine, cyclophosphamide, and doxorubicin; DHAP: dexamethasone, cytarabine, and cisplatin; DHAX: dexamethasone, cytarabine, and oxaliplatin; DLBCL: diffuse large B-cell lymphoma; EBNA1: EBV-nuclear antigen 1; EBV: Epstein–Barr virus; EBVST: EBV-specific T-cells; ESHAP: etoposide, cisplatin, methylprednisolone, and cytarabine; GCV: ganciclovir; GDP: gemcitabine, dexamethasone, and oxaliplatin; GEMOX: gemcitabine and oxaliplatin; HDAC: histone deacetylase; HD-MTX: high dose methotrexate; HIV: human immunodeficiency virus; HL: Hodgkin lymphoma; Hyper-CVAD: cyclophosphamide, doxorubicin, vincristine, dexamethasone with methotrexate and high-dose cytarabine; ICE: ifosfamide, etoposide, and carboplatin; IC-HL: immunocompetent Hodgkin lymphoma; LMP1: latent membrane protein-1; LMP2: latent membrane protein-2; NHL: non-Hodgkin lymphoma; PBL: plasmablastic lymphoma; PCNSL: primary central nervous system lymphoma; PD-1: programmed death 1; PD-L1: programmed death ligand 1; PEL: primary effusion lymphoma; PTLD: post-transplant lymphoproliferative disorder; R: rituximab; SAHA: suberoylanilide hydroxamic acid; SCT: stem cell transplantation; THP-COP: pirarubicin, cyclophosphamide, vincristine, and prednisolone.
(A) Chemotherapeutic Treatment in HIV-Related Lymphomas
Lymphoma1st Line2nd Line
DLBCLR-CHOPR-ESHAP, R-ICE, R-GEMOX; followed by ASCT
PCNSLHD-MTXASCT, Radiotherapy
BLCODOX-M/IVAC
Burkimab
Hyper-CVAD
DA-EPOCH-R
R-DHAP, R-DHAX, R-GDP, R-GDP, R-GEMOX
PBLCODOX- M/IVAC
Hyper-CVAD
DA-EPOCH-R
Bortezomib
THP-COP, ESHAP, ICE and ASCT
PELDA-EPOCH
CHOP
ASCT, radiotherapy, bortezomib
HLABVD
BEACOPP
ESHAP, DHAP, ICE; followed by ASCT
(B) EBV-Targeted Therapies
Preclinical Studies
Small molecule inhibitors•   Targeting host factors and signaling pathways
•   Targeting EBV antigens
Immunotherapy•   PD-1/PD-L1 antibodies
•   Monoclonal antibodies
•   EBVST
•   T-cell receptor-modified T-cell therapy
•   CAR-T
Clinical Trials
Antiretroviral drugs •   GCV and AZT in combination with immunomodulatory IL-2 and cART in EBV+ HIV-related PCNSL
Induction of lytic infection from latency state •   Arginine butyrate in combination with GCV in refractory EBV-related lymphomas
•   Inhibition of histone deacetylase (HDAC), such as SAHA in HL and NHL
Inhibition of PD-1/PD-L1•   Prembrolizumab in IC-HL
EBVST•   LMP1/LMP2- or LMP2-specific CTLs administered in EBV-related NHL or HL
•  EBNA-specific CTL in patients with EBV-related PTLD after SCT
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Verdu-Bou, M.; Tapia, G.; Hernandez-Rodriguez, A.; Navarro, J.-T. Clinical and Therapeutic Implications of Epstein–Barr Virus in HIV-Related Lymphomas. Cancers 2021, 13, 5534. https://doi.org/10.3390/cancers13215534

AMA Style

Verdu-Bou M, Tapia G, Hernandez-Rodriguez A, Navarro J-T. Clinical and Therapeutic Implications of Epstein–Barr Virus in HIV-Related Lymphomas. Cancers. 2021; 13(21):5534. https://doi.org/10.3390/cancers13215534

Chicago/Turabian Style

Verdu-Bou, Miriam, Gustavo Tapia, Agueda Hernandez-Rodriguez, and Jose-Tomas Navarro. 2021. "Clinical and Therapeutic Implications of Epstein–Barr Virus in HIV-Related Lymphomas" Cancers 13, no. 21: 5534. https://doi.org/10.3390/cancers13215534

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop