Next Article in Journal
Nuclear Transit and HIV LTR Binding of NF-κB Subunits Held by IκB Proteins: Implications for HIV-1 Activation
Previous Article in Journal
IFI16 Inhibits Porcine Reproductive and Respiratory Syndrome Virus 2 Replication in a MAVS-Dependent Manner in MARC-145 Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mechanistic Insights into Chemoresistance Mediated by Oncogenic Viruses in Lymphomas

1
Department of Pathology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W Markham St, Little Rock, AR 72205, USA
2
Department of Biochemistry and Molecular Biology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W Markham St, Little Rock, AR 72205, USA
*
Author to whom correspondence should be addressed.
Submission received: 18 November 2019 / Revised: 9 December 2019 / Accepted: 12 December 2019 / Published: 16 December 2019
(This article belongs to the Section Animal Viruses)

Abstract

:
Viral lymphomagenesis induced by infection with oncogenic viruses, such as Kaposi’s sarcoma associated herpesvirus (KSHV), Epstein–Barr virus (EBV) and human T-cell leukemia virus (HTLV-1), represents a group of aggressive malignancies with a diverse range of pathological features. Combined chemotherapy remains the standard of care for these virus-associated lymphomas; however, frequent chemoresistance is a barrier to achieving successful long-term disease-free survival. There is increasing evidence that indicates virus-associated lymphomas display more resistance to cytotoxic chemotherapeutic agents than that observed in solid tumors. Although the tumor microenvironment and genetic changes, such as key oncogene mutations, are closely related to chemoresistance, some studies demonstrate that the components of oncogenic viruses themselves play pivotal roles in the multidrug chemoresistance of lymphoma cells. In this review, we summarize recent advances in the understanding of the mechanisms through which oncogenic viruses mediate lymphoma cell chemoresistance, with a particular focus on KSHV and EBV, two major oncogenic viruses. We also discuss the current challenges to overcome these obstacles in the treatment of virus-associated lymphomas.

1. Introduction

Virus-associated lymphomas represent a largely heterogeneous group of hematologic malignancies that are characterized by the uncontrolled growth of clonal lymphocytes [1]. The pathogenesis of virus-associated lymphomas is a complex process involving viral infections, autoimmune diseases, environmental and genetic factors, and exposure to chemical or other toxins [2,3]. Extensive published literature show a close interrelation between lymphomagenesis and infection by viruses, including Kaposi’s sarcoma associated herpesvirus (KSHV) [4], Epstein–Barr virus (EBV) [5], hepatitis C virus (HCV) [6], human T-cell leukemia virus (HTLV) [7], and human immunodeficiency virus (HIV) [8,9]. Interestingly, Dr. Robert Gallo’s group provided new evidence that some HIV-1 matrix protein variants (vp17s) can activate AKT signaling and promote growth of transformed B cells [10]. Moreover, vp17s are more frequently detected in plasma of HIV+ patients with non-Hodgkin’s lymphoma (NHL) compared to those patients without NHL [10]. These data taken together with other recent findings, including the persistence of HIV-associated proteins within lymphoid follicles, indicates a direct role of HIV-1 in promoting lymphomagenesis beyond immune suppression functions [11]. However, the HIV-1 genome is not detectable in these malignant B cells and the transforming ability of HIV-1 in B cells still needs further investigation. Thus, this review will focus on the two major oncogenic viruses that cause B-cell, T-cell, and NK-cell lymphomas: KSHV and EBV.
KSHV and EBV both belong to the γ-herpesviridae subfamily and exhibit two alternative life cycle programs during infection of host cells: The latent and lytic phases. Although there is accumulating data that collectively support the involvement of both latent and lytic programs in the development and maintenance of viral diseases [12,13], the latent transcripts may play a more crucial and prominent role in tumorigenesis [14]. The ability for prolonged latent infection increases the risk for cancer development, and until last year, there was little known about the mechanism behind the maintenance of persistent latency and prevention of the lytic phases. Suppression of genes that induce the lytic phase in both KSHV- and EBV-infected cells in culture and from infected patients can occur through Krüppel-associated box (KRAB) domain-zinc finger protein transcriptional repressors, stem cell zinc finger protein 1 (SZF1) and zinc finger protein 557 (ZNF557), which are both regulated by STAT3, revealing a new epigenome regulatory activity of STAT3 [15]. The less than ideal effectiveness of antiviral agents against lytic phases achieved in the clinic suggests there are additional, intractable functions of herpesvirus latent infection during treatment [16,17,18]. During the latency of KSHV, only a limited number of viral proteins are expressed, including Latency associated nuclear antigen (LANA), LANA-2, viral Cyclin (vCyclin), viral Fas-associated death domain-like interleukin-1β-converting enzyme-inhibitory protein (vFLIP), Kaposins, and at least 18 mature viral microRNAs [19]. Of note, some of these latent components are oncogenic and involved in immune escape and lymphomagenesis [20]. More recently, KSHV-encoded IL-6 was added to this list as B-cells in mice infected with KSHV were shown to upregulate activation-induced cytidine deaminase (AID) expression, an enzyme responsible for class switch recombination for antibody generation, that correlated with vIL-6 and an increase in class switching [21]. With AID implicated in misappropriated translocations and mutations in lymphoma, vIL-6 may also play a role in KSHV pathogenesis and the development of KSHV-lymphoproliferative disorders and primary effusion lymphoma (PEL), the main type of KSHV-associated lymphoma. PEL is a rare but rapidly progressive large B cell lymphoma clinically characterized by malignant effusion in body cavities usually without extracavitary tumor masses [22]. Approximately half of PEL patients have pre-existing or develop Kaposi’s sarcoma (KS, another cancer caused by KSHV) [23]. PEL is most commonly found in persons living with HIV and other immunocompromised individuals [24]. Interestingly, KHSV–EBV co-infection is also commonly found (60%–90% of cases) in PEL [25], although the role of this simultaneous infection in the pathogenesis of PEL remains unclear.
In contrast to the persistent latency of KHSV, EBV infection has three distinct latency patterns: Latency I, II, and III due to the expression of specific latent proteins seen in related diseases and/or cell lines, including EBNA1 (Epstein–Barr nuclear antigen 1), EBNA2, EBNA3A/3B/3C, LMP1/2, and some EBV-encoded small RNAs (EBERs) [26,27,28]. Latency states of EBV are identified according to the development of EBV-related tumors. EBV is considered as the epitome of a B lymphotropic virus, and as such, its infection is closely associated with various types of lymphomas: Classical Hodgkin lymphoma (cHL), Burkitt’s lymphoma (BL), T-cell/NK-cell lymphoma, diffuse large B-cell lymphoma (DLBCL), AIDS-associated lymphoma, post-transplant lymphoma (PTLD), and other rare B-cell and T-cell lymphoproliferative disorders [5,29,30]. There are different patterns of viral gene expression within these lymphomas. For instance, BL is always associated with latency I infection characterized by EBNA1 expression [31]. Latency II is associated with the expression of EBNA1, LMP1, and LMP2, usually seen in HL and T-cell NHL [32,33,34]. In contrast, the expression of all these latent components is present in latency III, occurring mainly in immunocompromised patients suffering from PTLD, AIDS-associated lymphoma, or in some lymphoblastoid cell lines [35].
Therapeutic regimens for virus-associated lymphomas include radiotherapy, chemotherapy, highly active antiretroviral therapy, antiviral agents, targeted therapy, specific cytotoxic T-cells, and immunotherapy [17,18,22,25,36,37]. Among these, conventional and cytotoxic chemotherapy is still the standard of care for most virus-associated lymphomas. However, most virus-associated lymphomas usually display higher rates of chemoresistance compared to solid tumors. Although there are complex factors that contribute to the chemoresistance of virus-associated lymphomas, the intrinsic viral proteins or components play a pivotal role in the chemoresistant phenotype of tumor cells. For instance, recent efforts to target EBNA1 with small molecules and inhibit its DNA binding activity proved useful in suppressing tumor growth in lymphoblastoid and patient-derived nasopharyngeal carcinoma models [38]. Resolving the mechanisms through which oncogenic viruses mediate chemoresistance may bring new insights and promising strategies to improve chemotherapy efficacy and allow for dose reduction to minimize the toxicity of chemotherapeutic agents in cancer patients. In this article, we will review the current advances in chemotherapeutic treatment for virus-associated lymphomas and the findings of oncogenic viral protein-mediated chemoresistance.

2. Current Status of Chemotherapy in Virus-Associated Lymphomas

2.1. KSHV-Associated Lymphoma

As mentioned above, PEL represents the major lymphoma etiologically related to KSHV. The prognosis for PEL remains poor, with a short median survival of approximately 6 months even under conventional chemotherapy [24]. Currently, there is no standard chemotherapeutic regimen that is universally accepted for the frontline treatment of PEL. The lack of a consensus in treatment is mostly due to the low incidence of this disease and inability to conduct large-scale randomized studies to guide treatment and management decisions. However, some classical cytotoxic agents are considered as the mainstay of treatment. Other targeted therapeutic agents are also documented for clinical therapy in this patient setting and are described in Table 1.
The chemotherapeutic regimens for PEL mainly include CHOP (cyclophosphamide, doxorubicin, vincristine, and prednisone)-like chemotherapy and two intensive chemotherapy cocktails, dose-adjusted EPOCH (DA-EPOCH; etoposide, prednisone, vincristine, cyclophosphamide, and doxorubicin) and CDE (cyclophosphamide, doxorubicin, etoposide) [17,22]. The mechanisms of action of these cytotoxic agents include the inhibition of DNA, RNA, or protein synthesis except prednisone, which is a glucocorticoid receptor agonist that controls inflammation [48,49,50,51]. Specifically, doxorubicin and etoposide interact with topoisomerase II to inhibit DNA synthesis [52]; cyclophosphamide directly cross-links guanine bases in DNA double-helix strands to prevent DNA and RNA synthesis [53]; and vincristine stops tubulin dimer polymerization and subsequent microtubule formation by binding to the tubulin protein for inhibition of mitosis and protein metabolism [54]. Of interest, PEL usually displays more resistance to CHOP-based regimens than DA-EPOCH [22,55,56].
Several clinical studies assessing PEL clinical outcomes have found that the addition of other chemotherapeutic agents may improve the chemotherapy efficacy. Despite the lack of statistical significance, one study involving 28 PEL patients reported that the 5-year overall survival rate of patients receiving a CHOP-like regimen with high-dose methotrexate (MTX) was 45.7% compared to 34.4% of CHOP alone [57,58]. Another recent study involving 12 HIV-NHL patients (10 high-risk patients with DLBCL, including one EBV+, one EBV+/HHV-8+ PEL, and one unclassifiable NHL) reported that the response rate in HIV-NHL treated with vorinostat, a histone deacetylase inhibitor, combined with rituximab-based EPOCH was 100% (complete 83% and partial 17%), with a one-year event-free survival of 83% (95% confidence interval, 51.6%–97.9%), indicating that this novel combination regimen is tolerable and efficacious in patients with aggressive HIV-NHL [59], including those cases with oncogenic virus infection. Although there is progress in alternative combinations of chemotherapy, the advent of targeted therapies has also led to investigations in PEL.
Previous reports demonstrate favorable PEL patient outcomes following treatment with selective targeted therapeutic drugs. In a case report, bortezomib, an inhibitor of the 26S proteasome and nuclear factor B (NF-κB), in combination with doxorubicin and rituximab, substantially improved the performance of an elderly HIV-seronegative man of Mediterranean origin who was diagnosed with PEL [60]. Another study reported that intrapleural injection of bleomycin without any systemic therapy achieved a durable remission after pleurodesis in two HIV-seronegative patients with PEL [61]. Recently, a case report showed that after one month of lenalidomide treatment (a multi-functional agent with anti-angiogenic and anti-osteoclastogenic effects, and immunomodulatory activity), an HIV-seronegative patient with PEL significantly improved and after 18 months, complete remission persistence was progressively achieved [62]. Yet, another study reported that two PEL patients treated with oral rapamycin showed a positive clinical response and the authors postulated that KSHV+ patients who received a transplant could also benefit from substituting rapamycin for cyclosporine A and offer protection against PEL [63]. Thus, the addition of targeted therapies is sometimes necessary to increase chemotherapy efficacy in PEL patients, implicating potential pathways involved in mechanisms for chemotherapy resistance.

2.2. EBV-Associated Lymphomas

Lymphomas associated with EBV infection include three major B-cell malignancies: cHL, BL and DLBCL; NK/T-cell lymphomas; and two types of lymphomas in the immunocompromised patient setting, PTLD- and HIV/AIDS-associated lymphomas [64]. Similar to KHSV, EBV-associated lymphomas are traditionally treated with the same strategy as EBV-negative lymphoma counterparts, such as the anthracycline-containing chemotherapy (e.g., CHOP) as shown in Table 1. However, the status of EBV infections within lymphomas may also affect the option of treatment regimens.
cHL is a distinct clinical and pathological entity with heterogeneous genetic and virological features, with regards to EBV infection [65]. An epidemiological investigation showed that the pooled prevalence of EBV infection in cHL was about 47.9%, which is significantly higher in Africa and Central and South America than other regions [66]. Treatment for cHL usually involves combined chemotherapy, sometimes followed by radiotherapy. Typical treatments for advanced stage cHL include MOPP (mustargen, oncovin, procarbazine, and prednisone) [67], COPP (cyclophosphamide, vincristine, procarbazine, and prednisone), or ABVD (doxorubicin, bleomycin, vinblastine, dacarbazine) [68], whereas those that are PET (positron emission tomography)-positive are escalated to BEACOPP (bleomycin, etoposide, doxorubicin, cyclophosphamide, vincristine, procarbazine, prednisone), representing a more aggressive therapy that was initially developed in Europe [69]. In a recent report about an analysis of long-term survival in two randomized clinical trials, 1282 cHL patients received eight alternating cycles of COPP and ABVD (COPP/ABVD), eight cycles of bBEACOPP, or eight cycles of eBEACOPP in HD9; and 1670 patients received eight cycles of eBEACOPP or four cycles of eBEACOPP plus four cycles of bBEACOPP, plus consolidation radiotherapy to initial bulk and residual disease or no radiotherapy in HD12 (ClinicalTrials.gov NCT00265031), and strongly supported the use of eBEACOPP in advanced stage cHL [70]. However, one question remains in these clinical studies regarding the unknown percentage of cases that are EBV positive.
BL is an interesting mature B-cell NHL that has distinct features with endemic, sporadic, and immunodeficiency-associated variants [71]. The endemic BL is associated with EBV infection in over 95% of cases [72]. Historically, CHOP alone or in combination with methotrexate, cytarabine, ifosfamide, and/or etoposide is used for patients with BL in different clinical settings [72,73], but the addition of rituximab, an anti-CD20 chimeric monoclonal antibody, may improve the survival outcome for these patients [74,75]. Similar to the effectiveness of immunomodulatory agents in KHSV-PEL, recent in vitro evidence using BL and EBV lymphoblastic cell lines showed that pomalidomide increased the surface expression of immune markers and led to an enhanced susceptibility to NK cell-mediated cytotoxicity. This therapeutic approach has the potential to increase patients’ immune response to viral-associated tumors and improve patient outcome [76].
In contrast to BL, EBV infection is not as prevalent in DLBCL; however, when DLBCL tissues are EBV positive, these tumors are typically seen in the elderly and thought to be associated with chronic EBV infection as well as severe immunosuppression or immunosenescence. Treatments in the era of chemoimmunotherapy of EBV-associated DLBCL have similar guidelines with EBV-negative DLBCL, although EBV-positive DLBCL show more resistance and patients usually exhibit a lower response rate to chemotherapy than EBV-negative DLBCL [77,78]. Although, CHOP alone or in combination with rituximab (R-CHOP) is used for EBV-associated DLBCL patients. More recent data suggest higher rates of response to R-CHOP at overall response (OR) rates of 50% to 90% and complete response (CR) rates of 30% to 70% compared to CHOP at OR rates of 30% to 80% and CR rates of 30% to 50%, indicating the addition of rituximab has clearly improved survival outcomes for EBV+ DLBCL [79,80,81,82].
Extranodal NK/T-cell lymphoma, nasal type (ENKL) is sometimes associated with EBV infection, although the prevalence is dependent on extranodal involvement and geographic diversity in incidence. In clinical practice, patients with ENKL are usually resistant to CHOP-based regimens. The prominent reason for ENKL resistance to anthracycline-containing chemotherapies is most likely attributable to the expression of P-glycoprotein (P-gp), which is significantly linked to multidrug resistance (MDR) [83]. Thus, new chemotherapies avoiding the use of anthracycline are being developed and tested in prospective clinical trials and thus far have markedly changed the management of ENKL. In support of these positive clinical outcomes, l-asparaginase (l-asp) and/or non-MDR-related agents are now prescribed for newly diagnosed advanced ENKL patients. Alternatively, concurrent chemoradiotherapy as the first treatment tested in a qualified clinical trial setting for ENKL is also reported to improve clinical outcome when using radiotherapy in combination with classical chemotherapy, such as DeVIC (dexamethasone, etoposide, ifosfamide, and carboplatin) [84,85], VIPD (etoposide, ifosfamide, cisplatin, and dexamethasone) [86], VIDL (etoposide, ifosfamide, dexamethasone, and l-asp), MIDLE (methotrexate, ifosfamide, dexamethasone, l-asp, and etoposide), SMILE (steroid = dexamethasone, methotrexate, ifosfamide, l-asp, and etoposide) [87,88], and GELOX (gemcitabine, l-asp, oxaliplatin) [89]. Recently, a study combined both of the new therapeutic strategies to treat adult ENKL with polyethylene glycol-conjugated asparaginase, and CHOP in combination with radiotherapy showed high overall survival (OS) rates of 100%, 90.61%, and 80.54% at 1, 2, and 3 years, respectively [90]. Although EBV-DNA sero-levels were not measured during this study, these regimens may also significantly benefit patients with EBV-associated ENKL. Moreover, a clinical study showed that the LOP regimen (l-asparaginase, vincristine, and dexamethasone) in combination with intensity-modulated conformal radiotherapy for nasal ENKL patients resulted in a higher overall remission rate, survival rate, and lower adverse reactions [91].
As mentioned above, EBV infection is also associated with two types of lymphomas in the immunocompromised patient setting. In AIDS-associated lymphomas, conventional chemotherapy in combination with combined antiretroviral (cART) therapy is required since antiretroviral therapy alone is inadequate for the treatment of the lymphoma [92]. For PTLDs, CHOP-like chemotherapy still remains the only option for many patients with aggressive PTLDs [26].

3. Involvement of KSHV-Encoded Proteins into Drug Resistance of Lymphomas

Various host and viral factors influence the sensitivity of virus-associated lymphomas to anticancer drugs. Despite overloading patients with cytotoxic chemotherapeutic agents to trigger cellular stress that should culminate in death of the malignant cells, it is impossible to eliminate oncogenic viruses in these patients. For instance, PEL cell lines display high heterogeneity in the drug response [93], and some recent studies found that this chemoresistance was promoted by the presence of specific viral proteins, particularly LANA and LANA2 (summarized in Figure 1A).

3.1. LANA

LANA is a master regulator of KSHV latency, and primarily functions to maintain latency, enable episome DNA replication and segregation, and regulates gene transcription. LANA has also been identified as an oncoprotein to steer immune evasion and facilitate tumorigenesis [94,95]. Our recent work highlighted an extended role of LANA in PEL chemoresistance [40,96]. We discovered that LANA contributed to PEL resistance to paclitaxel and doxorubicin by regulating the expression of Emmprin (Basigin; CD147), a multifunctional glycoprotein belonging to the immunoglobulin superfamily. This protein is involved in interactions with two key MDR cell surface proteins: A hyaluronan receptor known as the lymphatic vessel endothelial hyaluronan receptor-1 (LYVE-1) and a drug transporter ABCG2/BCRP [39,96]. In support, targeting Emmprin, LYVE-1, or BCRP by RNA interference promoted PEL cell apoptosis induced by chemotherapy while the overexpression of Emmprin greatly enhanced the resistance of chemosensitive tumor cells to paclitaxel and doxorubicin [39,96]. The mechanism for LANA-induced Emmprin is not fully understood, although conceivably, LANA may bind directly to the promoter of Emmprin or indirectly as a complex with other cellular transcription factors, such as Sp1 and Egr-2, for which the promoter of Emmprin contains putative binding sites [96,97]. Previous studies demonstrated that the drug resistance of malignant cells could be related to glycolysis and lactate secretion that require monocarboxylate transporters (MCTs) to efflux lactate resulting from glycolysis [98,99]. Interestingly, Emmprin interacted with various MCTs, and PEL cells preferentially utilize glycolysis as an energy resource for survival [100,101]. Therefore, further studies are of interest to determine whether Emmprin regulation of MCTs functions and lactate efflux may impact PEL chemoresistance.

3.2. LANA2

LANA2, a nucleocytoplasmic shuttling protein, is exclusively expressed within KSHV-infected B cells as a B-cell-specific latent protein [102]. Although LANA2 may not be detected using serums of KSHV+ patients [102], it still plays an important role in KSHV-induced tumorigenesis. Previous studies identified its oncogenic abilities based on the supportive data that LANA2 inhibited p53- and PKR-dependent apoptosis [102,103], and also inhibited virus-mediated transcriptional activity of interferon alpha promoter [104]. Meanwhile, LANA2 interacted with the pocket proteins, pRb, p107, and p130, and inhibits their sumoylation to regulate cell-cycle arrest [105]. Moreover, LANA2 inhibited PML-mediated transcriptional repression of the survivin gene by disrupting PML oncogenic domains [106]. Interestingly, a previous study found that LANA2 induced paclitaxel resistance of PEL cells through binding to polymerized microtubules to prevent microtubule stabilization caused by paclitaxel [41].

3.3. Other Viral Proteins

One study found that KSHV infection induced AKT hyperphosphorylation, bortezomib-resistance, and GLUT-1 plasma membrane exposure in a THP-1 monocytic cell line [107]. Moreover, KSHV latent infection in bladder cancer cells promoted drug resistance by reducing reactive oxygen species (ROS) [108]. However, it remains unclear which viral proteins are responsible for these observations. Unexpectedly, KSHV-encoded vFLIP expression conferred selective chemosensitization to bleomycin-mediated cytotoxicity in HEK293 cells [109]; however, the influence of vFLIP in PEL chemoresistance remains unclear.

4. Involvement of EBV Components into Drug Resistance of Lymphomas

EBV is not only an important pathogen etiologically related to many human cancers but also a considerable barrier to chemotherapy for EBV-associated malignancies. Previous reports indicated latent EBV infection interfered with the responses of BL-derived cells to cytotoxic drugs by activating the mitotic spindle assembly checkpoint [110]. Moreover, EBV infection may confer chemoresistance of gastric carcinoma in response to the chemotherapeutic agents, 5-Fluorouracil and docetaxel [111,112]. Although the underlying mechanisms are still unclear, some data suggest the contribution of EBERs to chemoresistance in EBV-associated gastric cancer cells [113]. Here, the roles of EBV components in the chemoresistance of virus-associated lymphomas are summarized (Figure 1B).

4.1. LMP1

LMP1, a member of the tumor necrosis factor receptor superfamily and constitutively active membrane protein, is considered as the principal viral oncoprotein critical for immortalization and transformation of B cells through mimicking the CD40 receptor signaling pathway [114,115,116]. Some studies suggest the involvement of LMP1 in chemoresistance to treatment for EBV-associated lymphomas. For instance, a study revealed that LMP1 conferred drug resistance of T-cell lymphoma cell lines in response to doxorubicin by upregulating the expression of SNF1/AMP kinase-related kinase, which subsequently increased the expression of anti-apoptotic factors, BCL6 and BIRC2 [43]. Another group reported LMP1-mediated chemoresistance for doxorubicin was also associated with an oncogenic gene, Pim-1, since LMP1 enhanced the expression of Pim-1 and translocated Pim-1 to the cytoplasm. This mechanism could lead to an increase in tumor cell survival [44]. In addition, LMP1 was found to mediate EBV-transformed B cell resistance to apoptosis and DNA damage through induction of autophagy [45,46,117,118]. For instance, there was a constitutively high level of autophagy in these apoptosis-resistant cell lines (mostly DLBCL and PTLD), which express high levels of the pro-autophagic protein, BECN1/Beclin 1, based on the activation of NF-κB signaling by LMP1 [45]. Another recent study reported that LIMD1 (LIM domain-containing protein 1) was induced and protected EBV-transformed cells from DNA damage-induced cell death through LMP1-activated IRF4 and NF-κB signaling and autophagy [46]. However, the clinical relevance of chemoresistance mediated by EBV/LPMP1-activated autophagy, as well as the translational potentials (e.g., autophagy inhibitors), still requires further investigations.
EBV-associated NK/T-cell lymphomas are highly resistant to CHOP, probably due to upregulation of P-gp, an energy-dependent efflux pump that excretes drugs from the cytoplasm to the extracellular space [119,120]. High expression of P-gp is also connected with ROS production, which is induced by EBV infection [121,122]. Previous work established that induction of intracellular ROS production under EBV latent infection elevated P-gp expression via the STAT1 pathway while downregulation of ROS by NecroX-5, an ROS scavenger, effectively attenuated P-gp-associated chemoresistance in EBV-positive NK/T-cell lymphoma [123]. These findings indicate ROS may represent a potential therapeutic target for reducing chemoresistance in this subtype of EBV+ lymphoma. Moreover, recent studies demonstrated that LMP1 and/or other viral components were also involved in P-gp dependent chemoresistance. LMP1 promoted excessive ROS production from EBV-related malignant cells through activation of NAD(P)H oxidases and induction of nuclear factor erythroid 2-related factor 2 (NRF2) [124,125]. Other studies showed that the expression of another EBV-associated protein, EBNA1, in either latent or lytic phases, regulated LMP1 expression [126], and upregulated cellular antioxidant defense to enable B-cell growth, transformation, and immortalization [122].

4.2. EBNA2

EBNA2 is one of the six viral nuclear proteins expressed in latently infected B lymphocytes [30]. EBNA2 is thought as the master regulator for viral latent transcripts and numerous host genes, and is required for the transformation and immortalization of EBV-infected cells [5]. EBNA2 expression in EBV-infected B lymphocytes is critical for lymphoblastoid cell growth and survival. An interesting report showed the involvement of EBNA2 in the doxorubicin resistance of EBV-associated B lymphoma cells occurred through increasing CCL3 and CCL4, which in turn activated the BTK and NF-κB pathways [42].

4.3. BHRF1

Another EBV-encoded protein, BHRF1 (BamHI-H rightward open reading frame 1), is a functional homologue of the human BCL2 anti-apoptotic protein. Overall, the levels and interactions between pro-apoptotic and pro-survival protein family members decide the fate of the intrinsic/mitochondrial apoptotic pathway and whether a cell will undergo apoptotic cell death [5]. Expression of BHRF1 in EBV infection is an important regulator that blocks host cell apoptosis and death by sequestering cellular pro-apoptotic proteins, such as BIM [127]. This function was correlated in a recent report to BHRF1 promoting chemoresistance in BL cells [45]. BHRF1 was shown to inhibit multiple cellular pro-apoptotic proteins, including BIM, BID, PUMA, and BAK, and prevent BL tumor cell apoptosis to etoposide and ionomycin. Loss-of-function assays using BHRF1 mutants in EBV-infected B-cell lymphomas also supported its role in chemoresistance and indicated that BHRF1-mediated anti-apoptosis was associated with mutants of G99A, R100D, and F72W [45]. The authors also found that one of the mutants, F72W, exhibited different effects on the response to etoposide and ionomycin. This differential effect may be due to the alternative mechanisms for inducing apoptosis specific to each chemotherapy agent. Ionomycin acts primarily through Bim [128] while etoposide activity is dependent on the activation of TP53 and concomitant upregulation of PUMA and NoxA [129].

5. HTLV-1

Human T-cell leukemia/lymphoma virus type 1 (HTLV-1) is a retrovirus that causes adult T-cell leukemia/lymphoma (ATLL), an aggressive form of T-cell malignancy with four major subtypes: A smoldering type, chronic type, lymphoma type, and leukemic type [130]. One of the viral proteins, Tax, is designated as the master modulator in contribution to HTLV-1-associated leukemogenic. Currently, the common first-line chemotherapies used to treat ATLL are the same as those used to treat other types of T-cell lymphomas, including CHOP, CHOEP (cyclophosphamide, doxorubicin, vincristine, etoposide, and prednisone), DA-EPOCH, and Hyper-CVAD (cyclophosphamide, vincristine, doxorubicin, and dexamethasone) [131]. Although ATLL displays intrinsic chemoresistance, the contribution of this oncogenic virus or viral proteins to tumor cell chemoresistance remains unknown.

6. Conclusions

In virus-associated lymphomas, the presence of viral proteins increases the complexity of the tumor microenvironment and induces the expression of oncogenes that can limit therapeutic efforts. Accumulating data demonstrate the involvement of viral components in multidrug chemoresistance of lymphoma cells and several mechanisms are potentially associated with oncogenic virus-mediated chemoresistance: (1) A reduction of drug accumulation in lymphoma cells by regulating the activities of cellular drug transporters, (2) the hijacking of chemotherapeutic agent cellular targets, (3) the regulation and/or interaction with key factors involved in chemo-induced cell death responses, and (4) the antagonizing of the anti-cancer effects of chemo-drugs by inducing oncogenic signaling during infection. Therefore, infection with oncogenic viruses clearly contributes to lymphomagenesis and the pathology of these diseases in addition to compromising the efficacy of chemotherapy. However, since to date the majority of these findings are derived from in vitro studies, the question remains of whether these identified and postulated mechanisms for viral-driven chemoresistance are the same as those that occur within the tumor of a patient. There are many challenges for successful treatment of virus-associated lymphomas that center around the inability to tease out the oncogenic activity of the virus and eradicate the virus itself. Most oncogenic viruses involved in the development of lymphomas are lacking vaccines or antiviral drugs to prevent their infections. Similarly, current antiviral drugs do not effectively target viruses during latent infection, especially for those oncogenic herpesviruses. As well, targeting the tumor proves difficult in the setting of virus infection since the mechanisms behind oncogenic virus-mediated chemoresistance still remain largely unclear. Furthermore, many patients with virus-associated lymphomas are immunosuppressed and/or immunodeficient, and these patients are already dose sensitive. Chemotherapy, especially high-dose treatments that are often necessary for this patient population, may cause additional severe toxicity and other side effects. Lastly, some chemo-drugs may induce viral lytic reactivation and facilitate virus dissemination. Overcoming these obstacles may help to improve the chemotherapy efficacy and prolong the survival time of patients with virus-associated lymphomas.

Author Contributions

Writing—original draft preparation, J.C.; writing—review and editing, S.K. and Z.Q.

Funding

This work was supported by NIH/NCI R01CA228166 to Z.Q. Support has also been provided in part by the Arkansas Bioscience Institute, the major research component of the Arkansas Tobacco Settlement Proceeds Act of 2000. S.K. is supported from the Center for Microbial Pathogenesis and Host Inflammatory Responses through the Center of Biomedical Research Excellence Award from the National Institutes of General Medical Sciences at the NIH (P20GM103625).

Conflicts of Interest

All the authors declare no conflict of interest. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  1. Castillo, J.J.; Reagan, J.L.; Bishop, K.D.; Apor, E. Viral lymphomagenesis: From pathophysiology to the rationale for novel therapies. Br. J. Haematol. 2014, 165, 300–315. [Google Scholar] [CrossRef] [Green Version]
  2. Alexander, D.D.; Mink, P.J.; Adami, H.O.; Chang, E.T.; Cole, P.; Mandel, J.S.; Trichopoulos, D. The non-Hodgkin lymphomas: A review of the epidemiologic literature. Int. J. Cancer 2007, 120, 1–39. [Google Scholar] [CrossRef]
  3. Grulich, A.E.; Vajdic, C.M. The epidemiology of non-Hodgkin lymphoma. Pathology 2005, 37, 409–419. [Google Scholar] [CrossRef]
  4. 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]
  5. Thompson, M.P.; Kurzrock, R. Epstein-barr virus and cancer. Clin. Cancer Res. 2004, 10, 803–821. [Google Scholar] [CrossRef] [Green Version]
  6. De Vita, S.; Sacco, C.; Sansonno, D.; Gloghini, A.; Dammacco, F.; Crovatto, M.; Santini, G.; Dolcetti, R.; Boiocchi, M.; Carbone, A.; et al. Characterization of overt B-cell lymphomas in patients with hepatitis C virus infection. Blood 1997, 90, 776–782. [Google Scholar] [CrossRef]
  7. Shimoyama, M. Diagnostic-criteria and classification of clinical subtypes of adult T-cell leukemia-lymphoma—A report from the Lymphoma-Study-Group (1984–1987). Br. J. Haematol. 1991, 79, 428–437. [Google Scholar] [CrossRef]
  8. Tarantul, V.Z. Virus-associated lymphomagenesis. Int. J. Biomed. Sci. 2006, 2, 101–113. [Google Scholar]
  9. Ziegler, J.L.; Beckstead, J.A.; Volberding, P.A.; Abrams, D.I.; Levine, A.M.; Lukes, R.J.; Gill, P.S.; Burkes, R.L.; Meyer, P.R.; Metroka, C.E.; et al. Non-Hodgkin’s lymphoma in 90 homosexual men. Relation to generalized lymphadenopathy and the acquired immunodeficiency syndrome. N. Engl. J. Med. 1984, 311, 565–570. [Google Scholar] [CrossRef]
  10. 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] [Green Version]
  11. 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]
  12. Manners, O.; Murphy, J.C.; Coleman, A.; Hughes, D.J.; Whitehouse, A. Contribution of the KSHV and EBV lytic cycles to tumourigenesis. Curr. Opin. Virol. 2018, 32, 60–70. [Google Scholar] [CrossRef]
  13. Hardie, D.R. Human gamma-herpesviruses: A review of 2 divergent paths to oncogenesis. Transfus. Apher. Sci. 2010, 42, 177–183. [Google Scholar] [CrossRef] [PubMed]
  14. Jha, H.C.; Banerjee, S.; Robertson, E.S. The role of gammaherpesviruses in cancer pathogenesis. Pathogens 2016, 5, 18. [Google Scholar] [CrossRef] [Green Version]
  15. Li, X.; Burton, E.M.; Koganti, S.; Zhi, J.; Doyle, F.; Tenenbaum, S.A.; Horn, B.; Bhaduri-McIntosh, S. KRAB-ZFP Repressors Enforce Quiescence of Oncogenic Human Herpesviruses. J. Virol. 2018, 92, e00298-18. [Google Scholar] [CrossRef] [Green Version]
  16. Gantt, S.; Casper, C. Human herpesvirus 8-associated neoplasms: The roles of viral replication and antiviral treatment. Curr. Opin. Infect. Dis. 2011, 24, 295–301. [Google Scholar] [CrossRef] [Green Version]
  17. Coen, N.; Duraffour, S.; Snoeck, R.; Andrei, G. KSHV targeted therapy: An update on inhibitors of viral lytic replication. Viruses 2014, 6, 4731–4759. [Google Scholar] [CrossRef] [Green Version]
  18. Pagano, J.S.; Whitehurst, C.B.; Andrei, G. Antiviral drugs for EBV. Cancers 2018, 10, 197. [Google Scholar] [CrossRef] [Green Version]
  19. Dittmer, D.; Lagunoff, M.; Renne, R.; Staskus, K.; Haase, A.; Ganem, D. A cluster of latently expressed genes in Kaposi’s sarcoma-associated herpesvirus. J. Virol. 1998, 72, 8309–8315. [Google Scholar]
  20. Greene, W.; Kuhne, K.; Ye, F.; Chen, J.; Zhou, F.; Lei, X.; Gao, S.J. Molecular biology of KSHV in relation to AIDS-associated oncogenesis. Cancer Treat. Res. 2007, 133, 69–127. [Google Scholar]
  21. Rosario, S.A.; Santiago, G.E.; Mesri, E.A.; Verdun, R.E. Kaposi’s sarcoma-associated herpesvirus-encoded viral IL-6 (vIL-6) enhances immunoglobulin class-switch recombination. Front. Microbiol. 2018, 9, 3119. [Google Scholar] [CrossRef] [PubMed]
  22. Okada, S.; Goto, H.; Yotsumoto, M. Current status of treatment for primary effusion lymphoma. Intractable Rare Dis. Res. 2014, 3, 65–74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Ansari, M.Q.; Dawson, D.B.; Nador, R.; Rutherford, C.; Schneider, N.R.; Latimer, M.J.; Picker, L.; Knowles, D.M.; McKenna, R.W. Primary body cavity-based AIDS-related lymphomas. Am. J. Clin. Pathol. 1996, 105, 221–229. [Google Scholar] [CrossRef] [Green Version]
  24. 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]
  25. Dunleavy, K.; Wilson, W.H. How I treat HIV-associated lymphoma. Blood 2012, 119, 3245–3255. [Google Scholar] [CrossRef]
  26. Carbone, A.; Gloghini, A.; Dotti, G. EBV-associated lymphoproliferative disorders: Classification and treatment. Oncologist 2008, 13, 577–585. [Google Scholar] [CrossRef]
  27. Calderwood, M.A.; Venkatesan, K.; Xing, L.; Chase, M.R.; Vazquez, A.; Holthaus, A.M.; Ewence, A.E.; Li, N.; Hirozane-Kishikawa, T.; Hill, D.E.; et al. Epstein-Barr virus and virus human protein interaction maps. Proc. Natl. Acad. Sci. USA 2007, 104, 7606–7611. [Google Scholar] [CrossRef] [Green Version]
  28. Hutzinger, R.; Feederle, R.; Mrazek, J.; Schiefermeier, N.; Balwierz, P.J.; Zavolan, M.; Polacek, N.; Delecluse, H.J.; Huttenhofer, A. Expression and processing of a small nucleolar RNA from the Epstein-Barr virus genome. PLoS Pathog. 2009, 5, e1000547. [Google Scholar] [CrossRef]
  29. Niedobitek, G.; Young, L.S. Epstein-Barr virus persistence and virus-associated tumours. Lancet 1994, 343, 333–335. [Google Scholar] [CrossRef]
  30. Rezk, S.A.; Weiss, L.M. Epstein-Barr virus-associated lymphoproliferative disorders. Hum. Pathol. 2007, 38, 1293–1304. [Google Scholar] [CrossRef] [Green Version]
  31. Onnis, A.; Navari, M.; Antonicelli, G.; Morettini, F.; Mannucci, S.; De Falco, G.; Vigorito, E.; Leoncini, L. Epstein-Barr nuclear antigen 1 induces expression of the cellular microRNA hsa-miR-127 and impairing B-cell differentiation in EBV-infected memory B cells. New insights into the pathogenesis of Burkitt lymphoma. Blood Cancer J. 2012, 2, e84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Ito, Y.; Kawamura, Y.; Iwata, S.; Kawada, J.; Yoshikawa, T.; Kimura, H. Demonstration of type II latency in T lymphocytes of Epstein-Barr Virus-associated hemophagocytic lymphohistiocytosis. Pediatr. Blood Cancer 2013, 60, 326–328. [Google Scholar] [CrossRef] [PubMed]
  33. Kis, L.L.; Gerasimcik, N.; Salamon, D.; Persson, E.K.; Nagy, N.; Klein, G.; Severinson, E.; Klein, E. STAT6 signaling pathway activated by the cytokines IL-4 and IL-13 induces expression of the Epstein-Barr virus-encoded protein LMP-1 in absence of EBNA-2: Implications for the type II EBV latent gene expression in Hodgkin lymphoma. Blood 2011, 117, 165–174. [Google Scholar] [CrossRef] [PubMed]
  34. Grywalska, E.; Rolinski, J. Epstein-Barr virus-associated lymphomas. Semin. Oncol. 2015, 42, 291–303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Niedobitek, G.; Young, L.S.; Herbst, H. Epstein-Barr virus infection and the pathogenesis of malignant lymphomas. Cancer Surv. 1997, 30, 143–162. [Google Scholar] [PubMed]
  36. Ramos, J.C.; Lossos, I.S. Newly emerging therapies targeting viral-related lymphomas. Curr. Oncol. Rep. 2011, 13, 416–426. [Google Scholar] [CrossRef] [Green Version]
  37. Ghosh, S.K.; Perrine, S.P.; Faller, D.V. Advances in virus-directed therapeutics against Epstein-Barr virus-associated malignancies. Adv. Virol. 2012, 2012, 509296. [Google Scholar] [CrossRef]
  38. 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]
  39. Qin, Z.; Dai, L.; Bratoeva, M.; Slomiany, M.G.; Toole, B.P.; Parsons, C. Cooperative roles for emmprin and LYVE-1 in the regulation of chemoresistance for primary effusion lymphoma. Leukemia 2011, 25, 1598–1609. [Google Scholar] [CrossRef] [Green Version]
  40. Qin, Z.; Dai, L.; Slomiany, M.G.; Toole, B.P.; Parsons, C. Direct activation of emmprin and associated pathogenesis by an oncogenic herpesvirus. Cancer Res. 2010, 70, 3884–3889. [Google Scholar] [CrossRef] [Green Version]
  41. Munoz-Fontela, C.; Marcos-Villar, L.; Hernandez, F.; Gallego, P.; Rodriguez, E.; Arroyo, J.; Gao, S.J.; Avila, J.; Rivas, C. Induction of paclitaxel resistance by the Kaposi’s sarcoma-associated herpesvirus latent protein LANA2. J. Virol. 2008, 82, 1518–1525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Kim, J.H.; Kim, W.S.; Hong, J.Y.; Ryu, K.J.; Kim, S.J.; Park, C. Epstein-Barr virus EBNA2 directs doxorubicin resistance of B cell lymphoma through CCL3 and CCL4-mediated activation of NF-κB and Btk. Oncotarget 2017, 8, 5361–5370. [Google Scholar] [PubMed] [Green Version]
  43. Kim, J.H.; Kim, W.S.; Park, C. SNARK, a novel downstream molecule of EBV latent membrane protein 1, is associated with resistance to cancer cell death. Leuk. Lymphoma 2008, 49, 1392–1398. [Google Scholar] [CrossRef] [PubMed]
  44. Kim, J.H.; Kim, W.S.; Yun, Y.; Park, C. Epstein-Barr virus latent membrane protein 1 increases chemo-resistance of cancer cells via cytoplasmic sequestration of Pim-1. Cell Signal 2010, 22, 1858–1863. [Google Scholar] [CrossRef] [PubMed]
  45. Pujals, A.; Favre, L.; Pioche-Durieu, C.; Robert, A.; Meurice, G.; Le Gentil, M.; Chelouah, S.; Martin-Garcia, N.; Le Cam, E.; Guettier, C.; et al. Constitutive autophagy contributes to resistance to TP53-mediated apoptosis in Epstein-Barr virus-positive latency III B-cell lymphoproliferations. Autophagy 2015, 11, 2275–2287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Wang, L.; Howell, M.E.A.; McPeak, B.; Riggs, K.; Kohne, C.; Yohanon, J.U.; Foxler, D.E.; Sharp, T.V.; Moorman, J.P.; Yao, Z.Q.; et al. LIMD1 is induced by and required for LMP1 signaling, and protects EBV-transformed cells from DNA damage-induced cell death. Oncotarget 2017, 9, 6282–6297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. 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. 2019. [Google Scholar] [CrossRef]
  48. Hall, A.G.; Tilby, M.J. Mechanisms of action of, and modes of resistance to, alkylating agents used in the treatment of haematological malignancies. Blood Rev. 1992, 6, 163–173. [Google Scholar] [CrossRef]
  49. Tacar, O.; Sriamornsak, P.; Dass, C.R. Doxorubicin: An update on anticancer molecular action, toxicity and novel drug delivery systems. J. Pharm. Pharmacol. 2013, 65, 157–170. [Google Scholar] [CrossRef]
  50. Pommier, Y.; Leo, E.; Zhang, H.L.; Marchand, C. DNA topoisomerases and their poisoning by anticancer and antibacterial drugs. Chem. Biol. 2010, 17, 421–433. [Google Scholar] [CrossRef] [Green Version]
  51. Pufall, M.A. Glucocorticoids and cancer. Adv. Exp. Med. Biol 2015, 872, 315–333. [Google Scholar] [PubMed] [Green Version]
  52. Nitiss, J.L. Targeting DNA topoisomerase II in cancer chemotherapy. Nat. Rev. Cancer 2009, 9, 338–350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Noll, D.M.; Mason, T.M.; Miller, P.S. Formation and repair of interstrand cross-links in DNA. Chem. Rev. 2006, 106, 277–301. [Google Scholar] [CrossRef] [PubMed]
  54. Stanton, R.A.; Gernert, K.M.; Nettles, J.H.; Aneja, R. Drugs That target dynamic microtubules: A new molecular perspective. Med. Res. Rev. 2011, 31, 443–481. [Google Scholar] [CrossRef] [Green Version]
  55. Wilson, W.H.; Grossbard, M.L.; Pittaluga, S.; Cole, D.; Pearson, D.; Drbohlav, N.; Steinberg, S.M.; Little, R.F.; Janik, J.; Gutierrez, M.; et al. Dose-adjusted EPOCH chemotherapy for untreated large B-cell lymphomas: A pharmacodynamic approach with high efficacy. Blood 2002, 99, 2685–2693. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. 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]
  57. Boulanger, E.; Gerard, 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]
  58. Guillet, S.; Gerard, 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]
  59. Ramos, J.C.; Sparano, J.A.; Rudek, M.A.; Moore, P.C.; Cesarman, E.; Reid, E.G.; Henry, D.; Ratner, L.; Aboulafia, D.; Lee, J.Y.; et al. Safety and preliminary efficacy of vorinostat with R-EPOCH in high-risk HIV-associated Non-Hodgkin’s lymphoma (AMC-075). Clin. Lymphoma Myeloma Leuk. 2018, 18, 180–190e2. [Google Scholar] [CrossRef]
  60. Siddiqi, T.; Joyce, R.M. A case of HIV-negative primary effusion lymphoma treated with bortezomib, pegylated liposomal doxorubicin, and rituximab. Clin. Lymphoma Myeloma 2008, 8, 300–304. [Google Scholar] [CrossRef]
  61. Yiakoumis, X.; Pangalis, G.A.; Kyrtsonis, M.C.; Vassilakopoulos, T.P.; Kontopidou, F.N.; Kalpadakis, C.; Korkolopoulou, P.; Levidou, G.; Androulaki, A.; Siakantaris, M.P.; et al. Primary effusion lymphoma in two HIV-negative patients successfully treated with pleurodesis as first-line therapy. Anticancer Res. 2010, 30, 271–276. [Google Scholar] [PubMed]
  62. 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] [PubMed] [Green Version]
  63. Sin, S.H.; Roy, D.; Wang, L.; Staudt, M.R.; Fakhari, F.D.; Patel, D.D.; Henry, D.; Harrington, W.J.; Damania, B.A.; Dittmer, D.P. Rapamycin is efficacious against primary effusion lymphoma (PEL) cell lines in vivo by inhibiting autocrine signaling. Blood 2007, 109, 2165–2173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Kim, H.J.; Ko, Y.H.; Kim, J.E.; Lee, S.S.; Lee, H.; Park, G.; Paik, J.H.; Cha, H.J.; Choi, Y.D.; Han, J.H.; et al. Epstein-Barr virus-associated lymphoproliferative disorders: Review and update on 2016 WHO classification. J. Pathol. Transl. Med. 2017, 51, 352–358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Carbone, A.; Gloghini, A. Epstein Barr virus-associated Hodgkin lymphoma. Cancers 2018, 10, 163. [Google Scholar] [CrossRef] [Green Version]
  66. Lee, J.H.; Kim, Y.; Choi, J.W.; Kim, Y.S. Prevalence and prognostic significance of Epstein-Barr virus infection in classical Hodgkin’s lymphoma: A meta-analysis. Arch. Med. Res. 2014, 45, 417–431. [Google Scholar] [CrossRef]
  67. Devita, V.T., Jr.; Simon, R.M.; Hubbard, S.M.; Young, R.C.; Berard, C.W.; Moxley, J.H., III; Frei, E., III; Carbone, P.P.; Canellos, G.P. Curability of advanced Hodgkin’s disease with chemotherapy: Long-term follow-up of MOPP-treated patients at the National Cancer Institute. Ann. Intern. Med. 1980, 92, 587–595. [Google Scholar] [CrossRef]
  68. Canellos, G.P.; Anderson, J.R.; Propert, K.J.; Nissen, N.; Cooper, M.R.; Henderson, E.S.; Green, M.R.; Gottlieb, A.; Peterson, B.A. Chemotherapy of advanced Hodgkin’s disease with MOPP, ABVD, or MOPP alternating with ABVD. N. Engl. J. Med. 1992, 327, 1478–1484. [Google Scholar] [CrossRef]
  69. Engert, A. ABVD or BEACOPP for advanced Hodgkin lymphoma. J. Clin. Oncol. 2016, 34, 1167–1169. [Google Scholar] [CrossRef] [Green Version]
  70. Von Tresckow, B.; Kreissl, S.; Goergen, H.; Brockelmann, P.J.; Pabst, T.; Fridrik, M.; Rummel, M.; Jung, W.; Thiemer, J.; Sasse, S.; et al. Intensive treatment strategies in advanced-stage Hodgkin’s lymphoma (HD9 and HD12): Analysis of long-term survival in two randomised trials. Lancet Haematol. 2018, 5, e462–e473. [Google Scholar] [CrossRef]
  71. Yustein, J.T.; Dang, C.V. Biology and treatment of Burkitt’s lymphoma. Curr. Opin. Hematol. 2007, 14, 375–381. [Google Scholar] [CrossRef] [PubMed]
  72. Brady, G.; MacArthur, G.J.; Farrell, P.J. Epstein-Barr virus and Burkitt lymphoma. J. Clin. Pathol. 2007, 60, 1397–1402. [Google Scholar] [CrossRef]
  73. Magrath, I.; Adde, M.; Shad, A.; Venzon, D.; Seibel, N.; Gootenberg, J.; Neely, J.; Arndt, C.; Nieder, M.; Jaffe, E.; et al. Adults and children with small non-cleaved-cell lymphoma have a similar excellent outcome when treated with the same chemotherapy regimen. J. Clin. Oncol. 1996, 14, 925–934. [Google Scholar] [CrossRef] [PubMed]
  74. Boue, F.; Gabarre, J.; Gisselbrecht, C.; Reynes, J.; Cheret, A.; Bonnet, F.; Billaud, E.; Raphael, M.; Lancar, R.; Costagliola, D. Phase II trial of CHOP plus rituximab in patients with HIV-associated non-Hodgkin’s lymphoma. J. Clin. Oncol. 2006, 24, 4123–4128. [Google Scholar] [CrossRef] [PubMed]
  75. Ribrag, V.; Koscielny, S.; Bosq, J.; Leguay, T.; Casasnovas, O.; Fornecker, L.M.; Recher, C.; Ghesquieres, H.; Morschhauser, F.; Girault, S.; et al. Rituximab and dose-dense chemotherapy for adults with Burkitt’s lymphoma: A randomised, controlled, open-label, phase 3 trial. Lancet 2016, 387, 2402–2411. [Google Scholar] [CrossRef]
  76. Davis, D.A.; Shrestha, P.; Aisabor, A.I.; Stream, A.; Galli, V.; Pise-Masison, C.A.; Tagawa, T.; Ziegelbauer, J.M.; Franchini, G.; Yarchoan, R. Pomalidomide increases immune surface marker expression and immune recognition of oncovirus-infected cells. Oncoimmunology 2019, 8, e1546544. [Google Scholar] [CrossRef] [Green Version]
  77. Park, S.; Lee, J.; Ko, Y.H.; Han, A.; Jun, H.J.; Lee, S.C.; Hwang, I.G.; Park, Y.H.; Ahn, J.S.; Jung, C.W.; et al. The impact of Epstein-Barr virus status on clinical outcome in diffuse large B-cell lymphoma. Blood 2007, 110, 972–978. [Google Scholar] [CrossRef]
  78. 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]
  79. Feugier, P.; Hoof, A.V.; Sebban, C.; Solal-Celigny, P.; Bouabdallah, R.; Fermé, C.; Christian, B.; Lepage, E.; Tilly, H.; Morschhauser, F.; et al. Long-term results of the R-CHOP study in the treatment of elderly patients with diffuse large B-cell lymphoma: A study by the Groupe d’etude des Lymphomes de l’adulte. J. Clin. Oncol. 2005, 23, 4117–4126. [Google Scholar] [CrossRef] [Green Version]
  80. Habermann, T.M.; Weller, E.A.; Morrison, V.A.; Gascoyne, R.D.; Cassileth, P.A.; Cohn, J.B.; Dakhil, S.R.; Woda, B.; Fisher, R.I.; Peterson, B.A.; et al. Rituximab-CHOP versus CHOP alone or with maintenance rituximab in older patients with diffuse large B-cell lymphoma. J. Clin. Oncol. 2006, 24, 3121–3127. [Google Scholar] [CrossRef]
  81. Pfreundschuh, M.; Kuhnt, E.; Trümper, L.; Österborg, A.; Trneny, M.; Shepherd, L.; Gill, D.S.; Walewski, J.; Pettengell, R.; Jaeger, U.; et al. CHOP-like chemotherapy with or without rituximab in young patients with good-prognosis diffuse large-B-cell lymphoma: 6-year results of an open-label randomised study of the MabThera International Trial (MInT) Group. Lancet Oncol. 2011, 12, 1013–1022. [Google Scholar] [CrossRef]
  82. Castillo, J.J.; Beltran, B.E.; Miranda, R.N.; Young, K.H.; Chavez, J.C.; Sotomayor, E.M. EBV-positive diffuse large B-cell lymphoma of the elderly: 2016 update on diagnosis, risk-stratification, and management. Am. J. Hematol. 2016, 91, 529–537. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Yamaguchi, M.; Miyazaki, K. Current treatment approaches for NK/T-cell lymphoma. J. Clin. Exp. Hematophol. 2017, 57, 98–108. [Google Scholar] [CrossRef] [Green Version]
  84. Yamaguchi, M.; Tobinai, K.; Oguchi, M.; Ishizuka, N.; Kobayashi, Y.; Isobe, Y.; Ishizawa, K.; Maseki, N.; Itoh, K.; Usui, N.; et al. Phase I/II study of concurrent chemoradiotherapy for localized nasal natural killer/T-cell lymphoma: Japan Clinical Oncology Group Study JCOG0211. J. Clin. Oncol. 2009, 27, 5594–5600. [Google Scholar] [CrossRef] [PubMed]
  85. Yamaguchi, M.; Tobinai, K.; Oguchi, M.; Ishizuka, N.; Kobayashi, Y.; Isobe, Y.; Ishizawa, K.; Maseki, N.; Itoh, K.; Usui, N.; et al. Concurrent chemoradiotherapy for localized nasal natural killer/T-cell lymphoma: An updated analysis of the Japan clinical oncology group study JCOG0211. J. Clin. Oncol. 2012, 30, 4044–4046. [Google Scholar] [CrossRef]
  86. Kim, S.J.; Kim, K.; Kim, B.S.; Kim, C.Y.; Suh, C.; Huh, J.; Lee, S.W.; Kim, J.S.; Cho, J.; Lee, G.W.; et al. Phase II trial of concurrent radiation and weekly cisplatin followed by VIPD chemotherapy in newly diagnosed, stage IE to IIE, nasal, extranodal NK/T-Cell Lymphoma: Consortium for Improving Survival of Lymphoma study. J. Clin. Oncol. 2009, 27, 6027–6032. [Google Scholar] [CrossRef]
  87. Yamaguchi, M.; Kwong, Y.L.; Kim, W.S.; Maeda, Y.; Hashimoto, C.; Suh, C.; Izutsu, K.; Ishida, F.; Isobe, Y.; Sueoka, E.; et al. Phase II study of SMILE chemotherapy for newly diagnosed stage IV, relapsed, or refractory extranodal natural killer (NK)/T-cell lymphoma, nasal type: The NK-cell tumor study group study. J. Clin. Oncol. 2011, 29, 4410–4416. [Google Scholar] [CrossRef]
  88. Kwong, Y.L.; Kim, W.S.; Lim, S.T.; Kim, S.J.; Tang, T.; Tse, E.; Leung, A.Y.; Chim, C.S. SMILE for natural killer/T-cell lymphoma: Analysis of safety and efficacy from the Asia lymphoma study group. Blood 2012, 120, 2973–2980. [Google Scholar] [CrossRef]
  89. Wang, L.; Wang, Z.H.; Chen, X.Q.; Wang, K.F.; Huang, H.Q.; Xia, Z.J. First-line combination of GELOX followed by radiation therapy for patients with stage IE/IIE ENKTL: An updated analysis with long-term follow-up. Oncol. Lett. 2015, 10, 1036–1040. [Google Scholar] [CrossRef] [Green Version]
  90. Zheng, W.; Gao, Y.H.; Ke, X.Y.; Zhang, W.J.; Su, L.P.; Ren, H.Y.; Lin, N.J.; Xie, Y.; Tu, M.F.; Liu, W.P.; et al. PEG-L-CHOP treatment is safe and effective in adult extranodal NK/T-cell lymphoma with a low rate of clinical hypersensitivity. BMC Cancer 2018, 18, 910. [Google Scholar] [CrossRef]
  91. Ai, W.B.; Jing, Q.C. Clinical observation of LOP chemotherapy combined with radiotherapy in the treatment of early nasal NK/T cell lymphoma. Lin chung er bi yan hou tou jing wai ke za zhi (J. Clin. Otorhinolaryngol. Head Neck Surg.) 2019, 33, 990–992. [Google Scholar]
  92. Kanakry, J.A.; Ambinder, R.F. EBV-related lymphomas: New approaches to treatment. Curr. Treat. Options Oncol. 2013, 14, 224–236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Otvos, R.; Skribek, H.; Kis, L.L.; Gloghini, A.; Markasz, L.; Flaberg, E.; Eksborg, S.; Konya, J.; Gergely, L.; Carbone, A.; et al. Drug sensitivity patterns of HHV8 carrying body cavity lymphoma cell lines. BMC Cancer 2011, 11, 441. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Wei, F.; Gan, J.; Wang, C.; Zhu, C.; Cai, Q. Cell cycle regulatory functions of the KSHV oncoprotein LANA. Front. Microbiol. 2016, 7, 334. [Google Scholar] [CrossRef] [Green Version]
  95. Lee, H.R.; Lee, S.; Chaudhary, P.M.; Gill, P.; Jung, J.U. Immune evasion by Kaposi’s sarcoma-associated herpesvirus. Future Microbiol. 2010, 5, 1349–1365. [Google Scholar] [CrossRef] [Green Version]
  96. Dai, L.; Bai, L.; Lu, Y.; Xu, Z.; Reiss, K.; Del Valle, L.; Kaleeba, J.; Toole, B.P.; Parsons, C.; Qin, Z. Emmprin and KSHV: New partners in viral cancer pathogenesis. Cancer Lett. 2013, 337, 161–166. [Google Scholar] [CrossRef]
  97. Dai, L.; Bratoeva, M.; Toole, B.P.; Qin, Z.Q.; Parsons, C. KSHV activation of VEGF secretion and invasion for endothelial cells is mediated through viral upregulation of emmprin-induced signal transduction. Int. J. Cancer 2012, 131, 834–843. [Google Scholar] [CrossRef] [Green Version]
  98. Pelicano, H.; Martin, D.S.; Xu, R.H.; Huang, P. Glycolysis inhibition for anticancer treatment. Oncogene 2006, 25, 4633–4646. [Google Scholar] [CrossRef] [Green Version]
  99. Gatenby, R.A.; Gillies, R.J. Why do cancers have high aerobic glycolysis? Nat. Rev. Cancer 2004, 4, 891–899. [Google Scholar] [CrossRef]
  100. Bhatt, A.P.; Jacobs, S.R.; Freemerman, A.J.; Makowski, L.; Rathmell, J.C.; Dittmer, D.P.; Damania, B. Dysregulation of fatty acid synthesis and glycolysis in non-Hodgkin lymphoma. Proc. Natl. Acad. Sci. USA 2012, 109, 11818–11823. [Google Scholar] [CrossRef] [Green Version]
  101. Delgado, T.; Carroll, P.A.; Punjabi, A.S.; Margineantu, D.; Hockenbery, D.M.; Lagunoff, M. Induction of the Warburg effect by Kaposi’s sarcoma herpesvirus is required for the maintenance of latently infected endothelial cells. Proc. Natl. Acad Sci. USA 2010, 107, 10696–10701. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Rivas, C.; Thlick, A.E.; Parravicini, C.; Moore, P.S.; Chang, Y. Kaposi’s sarcoma-associated herpesvirus LANA2 is a B-cell-specific latent viral protein that inhibits p53. J. Virol. 2001, 75, 429–438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Esteban, M.; García, M.A.; Domingo-Gil, E.; Arroyo, J.; Nombela, C.; Rivas, C. The latency protein LANA2 from Kaposi’s sarcoma-associated herpesvirus inhibits apoptosis induced by dsRNA-activated protein kinase but not RNase L activation. J. Gen. Virol. 2003, 84, 1463–1470. [Google Scholar] [CrossRef] [PubMed]
  104. Lubyova, B.; Pitha, P.M. Characterization of a novel human herpesvirus 8-encoded protein, vIRF-3, that shows homology to viral and cellular interferon regulatory factors. J. Virol. 2000, 74, 8194–8201. [Google Scholar] [CrossRef] [Green Version]
  105. Marcos-Villar, L.; Gallego, P.; Munoz-Fontela, C.; de la Cruz-Herrera, C.F.; Campagna, M.; Gonzalez, D.; Lopitz-Otsoa, F.; Rodriguez, M.S.; Rivas, C. Kaposi’s sarcoma-associated herpesvirus lana2 protein interacts with the pocket proteins and inhibits their sumoylation. Oncogene 2014, 33, 495–503. [Google Scholar] [CrossRef]
  106. Marcos-Villar, L.; Lopitz-Otsoa, F.; Gallego, P.; Munoz-Fontela, C.; Gonzalez-Santamaria, J.; Campagna, M.; Shou-Jiang, G.; Rodriguez, M.S.; Rivas, C. Kaposi’s sarcoma-associated herpesvirus protein LANA2 disrupts PML oncogenic domains and inhibits PML-mediated transcriptional repression of the survivin gene. J. Virol. 2009, 83, 8849–8858. [Google Scholar] [CrossRef] [Green Version]
  107. Gonnella, R.; Santarelli, R.; Farina, A.; Granato, M.; D’Orazi, G.; Faggioni, A.; Cirone, M. Kaposi sarcoma associated herpesvirus (KSHV) induces AKT hyperphosphorylation, bortezomib-resistance and GLUT-1 plasma membrane exposure in THP-1 monocytic cell line. J. Exp. Clin. Cancer Res. 2013, 32, 79. [Google Scholar] [CrossRef] [Green Version]
  108. Lee, S.; Jang, J.; Jeon, H.; Lee, J.; Yoo, S.M.; Park, J.; Lee, M.S. Latent Kaposi’s sarcoma-associated herpesvirus infection in bladder cancer cells promotes drug resistance by reducing reactive oxygen species. J. Microbiol. 2016, 54, 782–788. [Google Scholar] [CrossRef]
  109. Masuda, Y.; Noguchi, K.; Segawa, H.; Tanaka, N.; Katayama, K.; Mitsuhashi, J.; Sugimoto, Y. Novel regulatory role for Kaposi’s sarcoma-associated herpesvirus-encoded vFLIP in chemosensitization to bleomycin. Biochem. Biophys. Res. Commun. 2011, 415, 305–312. [Google Scholar] [CrossRef]
  110. Leao, M.; Anderton, E.; Wade, M.; Meekings, K.; Aday, M.J. Epstein-Barr virus-induced resistance to drugs that activate the mitotic spindle assembly checkpoint in Burkitt’s lymphoma cells. J. Virol. 2007, 81, 248–260. [Google Scholar] [CrossRef] [Green Version]
  111. Seo, J.S.; Kim, T.G.; Hong, Y.S.; Chen, J.Y.; Lee, S.K. Contribution of Epstein-Barr virus infection to chemoresistance of gastric carcinoma cells to 5-fluorouracil. Arch. Pharm. Res. 2011, 34, 635–643. [Google Scholar] [CrossRef] [PubMed]
  112. Shin, H.J.; Kim, D.N.; Lee, S.K. Association between Epstein-Barr virus infection and chemoresistance to docetaxel in gastric carcinoma. Mol. Cells 2011, 32, 173–179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Banerjee, A.S.; Pal, A.D.; Banerjee, S. Epstein-Barr virus-encoded small non-coding RNAs induce cancer cell chemoresistance and migration. Virology 2013, 443, 294–305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Dirmeier, U.; Neuhierl, B.; Kilger, E.; Reisbach, G.; Sandberg, M.L.; Hammerschmidt, W. Latent membrane protein 1 is critical for efficient growth transformation of human B cells by Epstein-Barr virus. Cancer Res. 2003, 63, 2982–2989. [Google Scholar] [PubMed]
  115. Saha, A.; Robertson, E.S. Mechanisms of B-Cell oncogenesis induced by Epstein-Barr virus. J. Virol. 2019, 93, e00238-19. [Google Scholar] [CrossRef] [Green Version]
  116. Zhang, B.; Kracker, S.; Yasuda, T.; Casola, S.; Vanneman, M.; Homig-Holzel, C.; Wang, Z.; Derudder, E.; Li, S.; Chakraborty, T.; et al. Immune surveillance and therapy of lymphomas driven by Epstein-Barr virus protein LMP1 in a mouse model. Cell 2012, 148, 739–751. [Google Scholar] [CrossRef] [Green Version]
  117. Lee, D.Y.; Sugden, B. The latent membrane protein 1 oncogene modifies B-cell physiology by regulating autophagy. Oncogene 2008, 27, 2833–2842. [Google Scholar] [CrossRef]
  118. Pratt, Z.L.; Zhang, J.Z.; Sugden, B. The latent membrane protein 1 (LMP1) oncogene of Epstein-Barr virus can simultaneously induce and inhibit apoptosis in B cells. J. Virol. 2012, 86, 4380–4393. [Google Scholar] [CrossRef] [Green Version]
  119. Yamaguchi, M.; Kita, K.; Miwa, H.; Nishii, K.; Oka, K.; Ohno, T.; Shirakawa, S.; Fukumoto, M. Frequent expression of P-glycoprotein/MDR1 by nasal T-cell lymphoma cells. Cancer 1995, 76, 2351–2356. [Google Scholar] [CrossRef]
  120. Yoshimori, M.; Takada, H.; Imadome, K.I.; Kurata, M.; Yamamoto, K.; Koyama, T.; Shimizu, N.; Fujiwara, S.; Miura, O.; Arai, A. P-glycoprotein is expressed and causes resistance to chemotherapy in EBV-positive T-cell lymphoproliferative diseases. Cancer Med. 2015, 4, 1494–1504. [Google Scholar] [CrossRef]
  121. Wartenberg, M.; Hoffmann, E.; Schwindt, H.; Grunheck, F.; Petros, J.; Arnold, J.R.; Hescheler, J.; Sauer, H. Reactive oxygen species-linked regulation of the multidrug resistance transporter P-glycoprotein in Nox-1 overexpressing prostate tumor spheroids. FEBS Lett. 2005, 579, 4541–4549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Wang, J.; Nagy, N.; Masucci, M.G. The Epstein–Barr virus nuclear antigen-1 upregulates the cellular antioxidant defense to enable B-cell growth transformation and immortalization. Oncogene 2019, 1–14. [Google Scholar] [CrossRef]
  123. Nam, Y.S.; Kim, N.; Song, Y.; Lee, J.S.; Jeon, Y.W.; Cho, S.G.; Im, K.-I. Down-regulation of intracellular reactive oxygen species attenuates P-glycoprotein-associated chemoresistance in Epstein-Barr virus-positive NK/T-cell lymphoma. Am. J. Transl. Res. 2019, 11, 1359. [Google Scholar] [PubMed]
  124. Sun, J.; Hu, C.Y.; Zhu, Y.H.; Sun, R.; Fang, Y.J.; Fan, Y.H.; Xu, F. LMP1 increases expression of NADPH oxidase (NOX) and its regulatory subunit p22 in NP69 nasopharyngeal cells and makes them sensitive to a treatment by a NOX inhibitor. PLoS ONE 2015, 10, e0134896. [Google Scholar] [CrossRef] [PubMed]
  125. Yun, S.-M.; Kim, Y.S.; Hur, D.Y. LMP1 and 2A induce the expression of Nrf2 through Akt signaling pathway in Epstein-Barr virus-transformed B cells. Transl. Oncol. 2019, 12, 775–783. [Google Scholar] [CrossRef] [PubMed]
  126. Gahn, T.A.; Sugden, B. An EBNA-1-dependent enhancer acts from a distance of 10 kilobase pairs to increase expression of the Epstein-Barr virus LMP gene. J. Virol. 1995, 69, 2633–2636. [Google Scholar]
  127. 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] [Green Version]
  128. Bouillet, P.; Metcalf, D.; Huang, D.C.; Tarlinton, D.M.; Kay, T.W.; Kontgen, F.; Adams, J.M.; Strasser, A. Proapoptotic Bcl-2 relative Bim required for certain apoptotic responses, leukocyte homeostasis, and to preclude autoimmunity. Science 1999, 286, 1735–1738. [Google Scholar] [CrossRef]
  129. Villunger, A.; Michalak, E.M.; Coultas, L.; Mullauer, F.; Bock, G.; Ausserlechner, M.J.; Adams, J.M.; Strasser, A. p53- and drug-induced apoptotic responses mediated by BH3-only proteins puma and noxa. Science 2003, 302, 1036–1038. [Google Scholar] [CrossRef] [Green Version]
  130. Mahieux, R.; Gessain, A. Adult T-cell leukemia/lymphoma and HTLV-1. Curr. Hematol. Malig. Rep. 2007, 2, 257–264. [Google Scholar] [CrossRef]
  131. Hermine, O.; Ramos, J.C.; Tobinai, K. A review of new findings in adult T-cell Leukemia-lymphoma: A focus on current and emerging treatment strategies. Adv. Ther. 2018, 35, 135–152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. The schematic of oncogenic virus-mediated chemoresistance in lymphomas. (A) The downstream chemoresistance pathways induced following B-cell infection with the Kaposi’s sarcoma associated herpesvirus (KSHV) driven by the LANA and LANA2 viral proteins. (B) The downstream chemoresistance pathways induced following B-cell infection with the Epstein–Barr virus (EBV) facilitated by the EBNA1, EBNA2, and BHRF1 EBV-encoded proteins.
Figure 1. The schematic of oncogenic virus-mediated chemoresistance in lymphomas. (A) The downstream chemoresistance pathways induced following B-cell infection with the Kaposi’s sarcoma associated herpesvirus (KSHV) driven by the LANA and LANA2 viral proteins. (B) The downstream chemoresistance pathways induced following B-cell infection with the Epstein–Barr virus (EBV) facilitated by the EBNA1, EBNA2, and BHRF1 EBV-encoded proteins.
Viruses 11 01161 g001
Table 1. Oncogenic virus-encoded proteins are involved in the chemoresistance of lymphoma cells based on in vitro studies.
Table 1. Oncogenic virus-encoded proteins are involved in the chemoresistance of lymphoma cells based on in vitro studies.
VirusesTranscriptsDrugs/AgentsLymphomasMechanismsRef.
KSHVLANADoxorubicinPELEmmprin, LYVE-1, BCRP and hyaluronan signaling[39,40]
PaclitaxelPELEmmprin, LYVE-1, BCRP and hyaluronan signaling[39]
LANA2PaclitaxelPELModulating microtubule dynamics to prevent microtubule stabilization induced by paclitaxel[41]
EBVEBNA2DoxorubicinDLBCLRegulating CCL3 and CCL4-mediated activation of NF-κB and Btk signaling[42]
LMP1DoxorubicinT-cell lymphomaUpregulating SNARK to increase expression of anti-apoptotic genes, such as BCL6 and BIRC2; Enhancing expression of Pim-1 and translocating Pim-1 to the cytoplasm[43,44]
Nutlin-3DLBCL/PTLDUpregulating pro-autophagic proteins such as Beclin 1 and Sestrin 1 through LMP1-activated NF-κB signaling[45]
IonomycinBLUpregulating LIMD1 through LMP1-activated IRF4 and NF-κB signaling as well as autophagy[46]
BHFR1EtoposideBLInteracting with the cellular pro-apoptotic BCL-2 proteins, BIM, BID, PUMA and BAK, to regulate apoptosis[47]
Ionomycin

Share and Cite

MDPI and ACS Style

Chen, J.; Kendrick, S.; Qin, Z. Mechanistic Insights into Chemoresistance Mediated by Oncogenic Viruses in Lymphomas. Viruses 2019, 11, 1161. https://0-doi-org.brum.beds.ac.uk/10.3390/v11121161

AMA Style

Chen J, Kendrick S, Qin Z. Mechanistic Insights into Chemoresistance Mediated by Oncogenic Viruses in Lymphomas. Viruses. 2019; 11(12):1161. https://0-doi-org.brum.beds.ac.uk/10.3390/v11121161

Chicago/Turabian Style

Chen, Jungang, Samantha Kendrick, and Zhiqiang Qin. 2019. "Mechanistic Insights into Chemoresistance Mediated by Oncogenic Viruses in Lymphomas" Viruses 11, no. 12: 1161. https://0-doi-org.brum.beds.ac.uk/10.3390/v11121161

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