Next Article in Journal
Current State of AIDS-Related Malignant Lymphoma
Next Article in Special Issue
Advances in Epstein–Barr Virus Detection: From Traditional Methods to Modern Technologies
Previous Article in Journal
Importance of Target Gene Locus on the Stability of Recombinant Viruses in the Baculovirus Expression System
Previous Article in Special Issue
CRISPR/Cas13-Mediated Inhibition of EBNA1 for Suppression of Epstein–Barr Virus Transcripts and DNA Load in Nasopharyngeal Carcinoma Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Association and Interaction of Epstein–Barr Virus with SARS-CoV-2 Infection—A Review

1
Department of Microbiology, School of Medical Sciences and Research, Sharda University, Greater Noida 201306, Uttar Pradesh, India
2
Department of Neurosurgery, Max Superspeciality Hospital, Patparganj, New Delhi 110092, India
*
Author to whom correspondence should be addressed.
Viruses 2025, 17(7), 903; https://doi.org/10.3390/v17070903
Submission received: 10 May 2025 / Revised: 23 June 2025 / Accepted: 25 June 2025 / Published: 26 June 2025
(This article belongs to the Special Issue EBV and Disease: New Perspectives in the Post COVID-19 Era)

Abstract

Despite the significant decrease in SARS-CoV-2-related mortality, COVID-19 continues to impose a high public health burden due to the high rate of post-COVID-19 pathological conditions, broadly termed Long COVID, that continue for any period of time and are generally multisystemic. However, recent studies have strengthened the evidence that the reactivation of the Epstein–Barr virus (EBV) in the post-COVID-19 era has significantly contributed to the exacerbation and prolongation of Long COVID symptoms. The mechanism and pathophysiology of EBV reactivation in Long COVID patients still need further exploration due to limited studies. This review summarises the various studies linking EBV reactivation in Long COVID along with its pathophysiology and novel therapeutics for EBV in a post-COVID-19 era.

1. Introduction

The Epstein–Barr virus (EBV) is a double-stranded DNA virus included in the herpes family and is also called human herpesvirus 4 (HHV-4) [1]. It is a leading cause of infectious mononucleosis and was the first human oncogenic virus to be identified, specifically in Burkitt lymphoma cells [2]. It is globally present in 90% of the population [3] and is associated with various lymphoproliferative disorders including Burkitt’s lymphoma, Hodgkin’s lymphoma, T and natural killer (NK) cell lymphomas, nasopharyngeal carcinoma, gastric adenocarcinoma, post-transplant lymphoproliferative disease and AIDS-associated lymphoblastoma [3,4].
EBV exhibits a biphasic life cycle, comprising a latent phase where the virus remains asymptomatic and a lytic phase that leads to the production of infectious virions [2]. This virus can switch from latent to lytic phases, a process known as EBV reactivation, which can be triggered by various stimuli, including psychological stress and immunosuppression [3].
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) caused the novel coronavirus disease 2019 (COVID-19) pandemic that produced an unprecedented burden on approximately 185 countries [5]. The pandemic further led to the development of Long COVID, which is defined as new or persistent symptoms that begin three months after a probable or confirmed SARS-CoV-2 infection and last for at least two months, with no other plausible explanation [6]. Although a number of studies have suggested the role of autoimmune factors and viral persistence in the development of Long COVID, the role of EBV in both COVID-19 and Long COVID needs to be explored. This review highlights the association of EBV with SARS-CoV-2 with special emphasis on EBV reactivation and immune dysregulation in Long COVID, EBV epidemiology and pathogenesis in COVID-19, and novel therapeutics for EBV in a post-COVID-19 era.

2. Mechanism of EBV Reactivation

The immediate–early (IE) genes BZLF1 (Z, Zta, ZEBRA, EB1) and BRLF1 (R, Rta) are encoded by EBV and are essential for initiating lytic reactivation [7], wherein BZLF1 has been speculated to be more important because more efficient reactivation can be achieved by exogenous expression of BZLF1 as compared to BRLF1 [8]. Reactivation occurs in the following three stages [9]:
Stage 1—BZLF1 and BRLF1 are immediately transcribed after the viral entry within host cells, causing significant upregulation in the expression of the EBV lytic phase by not only activating the viral lytic cascade but also synergistically activating their own promoters, leading to the expression of early-stage genes like DNA polymerase catalytic subunits BALF1, BMRF1, and BALF2.
Stage 2—The viral genome is replicated.
Stage 3—Late genes are expressed, and their encoded proteins form the structural components of the virus, including the capsid, viral antigens, and glycoproteins. This is followed by the assembly of the components and genome, after which the virus buds out from the host cell.
Various pathways have been implicated in the reactivation process, including the Phosphoinositide 3-kinase (PI3K) pathway, the c-Jun N-terminal Kinase (JNK) pathway, and the mitogen-activated protein kinase (MAPK) pathway [2,10]. EBV reactivation at the cellular level involves the induction of one of the proteins PI3K, MAPK, or PKC followed by the induction of the transcription factors Sp1, MEF2D, AP1, and c-jun, which bind to the BZLF1-promotor region. BZLF1 transcriptional activation, triggered by this process, marks the beginning of the EBV lytic cycle. Alternatively, a double-stranded DNA break derived from damaged mitochondria or nuclei also triggers the same pathway, leading to Sp1 induction or possibly causing an AKT activation cascade [2].

3. EBV Reactivation and SARS-CoV-2 Infection

EBV initially infects resting B cells, which then become a reservoir for the virus as latently infected memory B cells in the peripheral blood, expressing latent membrane protein 2 (LMP-2) and EBV nuclear antigens (EBNAs). SARS-CoV-2 infection reactivates EBV-infected B cells that express LMP-2 and EBNA-1, resulting in severe systemic inflammation [11]. Although various studies have been conducted that demonstrate EBV reactivation cases ranging from 13% to 65% among Long COVID patients [12,13,14,15,16,17,18,19,20,21,22,23], as shown in Table 1, there has been limited literature on the actual mechanism of EBV lytic reactivation in Long COVID cases. There has been little published data that depict the results of the different aspects of the still unknown mechanism.
The various proposed mechanisms involved in EBV reactivation in Long COVID are as follows.

3.1. EBV Reactivation Induced by Drugs Used for COVID-19 Treatment

Recent data has indicated that certain drugs used for the treatment of COVID-19 patients, such as azithromycin, chloroquine diphosphate, hydroxychloroquine sulfate, and nafamostat mesylate [24,25], increased viral lytic gene expression via the activation of MAPK and NF-kB signalling pathways [26].
Recently, Chen J. et al. conducted a study where remdesivir; which was authorised by the United States Food and Drug Administration (FDA) for COVID-19 treatment; increased the expression of viral lytic genes, such as BZLF1 (immediate early gene) and BHFR1 (early gene), in all three EBV+ lymphoma cell lines; i.e., RPMI 6666 (Hodgkin’s lymphoma), Akata (Burkitt’s lymphoma), and VAL (diffuse large B cell lymphoma), as quantified by qRT-PCR [27]. Remdesivir also reduced STAT3 but increased p38 MAPK phosphorylation from EBV+ lymphoma cells, which are two signalling pathways that are associated with EBV reactivation [28,29]. Hence, remdesivir treatment requires the continuous monitoring of viral loads and risk assessment of developing EBV-associated malignancies, even after patients have fully recovered from COVID-19. The association of some important anti-SARS-CoV-2 drugs with EBV reactivation is shown in Table 2 [26,27,28,29,30,31].
Fluvoxamine is a selective serotonin reuptake inhibitor (SSRI) and a σ-1 receptor (S1R) agonist [32] that inhibits acid sphingomyelinase (ASM) activity and the formation of the ceramide-enriched membrane domain. It also attenuates SARS-CoV-2 cell entry and decreases SARS-CoV-2 replication as well as subsequent endoplasmic reticulum (ER) stress and inflammation [33]. S1R agonists inhibit the splicing of mRNA-encoding X-box binding protein-1 (XBP1) by preventing inositol-requiring enzyme 1α (IRE1) activity; hence reducing XBP1 activation, which modulates the ER stress response pathway and reduces cytokine storm [34]. ER stress and unfolded protein response upregulate latent membrane protein 1 (LMP1), which enhances the production of interleukin-8, thus inducing EBV lytic gene expression [35,36].

3.2. EBV Reactivation Induced by Host Proteins Interacting with SARS-CoV-2 Virus

Recent reports have shown the interaction between various SARS-CoV-2 proteins and host proteins. Interestingly, these host proteins listed in Table 3 [37,38,39,40,41,42,43,44,45,46,47,48,49,50,51] have been shown to play a vital role in EBV lytic reactivation or the maintenance of EBV inside cells.
Some important host proteins linked with the SARS-CoV-2 virus and their role in EBV reactivation are explained below.

3.2.1. Role of BRD4 Host Protein in EBV Reactivation

BRD4 host protein linked to SARS-CoV-2 E-protein has been shown to interact with EBNA1 through N-terminal sequences to mediate EBV transcriptional activation. Also, BRD4 is specifically localised to the FR enhancer element regulated by EBNA1, which further enhances transcriptional activation by EBNA1 [52]. BRD4 is a member of the Bromodomain and Extra-Terminal (BET) domain proteins that uses its bromodomains to bind to acetylated histones H3 and H4 and associate with interphase chromatin and mitotic chromosomes, thus leading to cell cycle progression and the development of viral oncogenes [53]. JQ1, a BET inhibitor, prevents EBV reactivation by blocking the production of the BZLF1 protein and subsequently inhibiting the transcription of downstream lytic genes. JQ1 achieves this by interfering with the recruitment of BRD4 to the BZLF1 promoter. It binds to the recognition pocket for acetylated lysine residues of BRD4 and hence competitively inhibits the BRD4-histone binding and recruitment of transcriptional complexes to the BZLF1 promoter [54].

3.2.2. Role of UPF1 Host Protein in EBV Reactivation

UPF1 is a crucial protein in the nonsense-mediated decay (NMD) pathway that mediates the degradation of BRLF1 transcripts, thus acting as a negative regulator of EBV reactivation [38]. NMD is a cellular surveillance mechanism that degrades mRNAs containing premature stop codons and prevents the production of harmful proteins [55].

3.2.3. Role of Anoctamin 2 (ANO2) Host Protein in EBV Reactivation

ANO2 is an ion channel expressed in the central nervous system (CNS). ANO2 antibodies recognise a fragment of EBNA1 via the mechanism of molecular mimicry, thereby causing CNS inflammation through T cells that are reactive with the same protein and triggering EBV reactivation, particularly in patients with multiple sclerosis [40].

3.2.4. Role of Insulin-Degrading Enzyme (IDE) Host Protein in EBV Reactivation

IDE has a more prominent role as a cellular receptor for varicella zoster virus (VZV) where it interacts with VZV glycoprotein E (gE), and this interaction is crucial for VZV infection and cell-to-cell spread [56]. Although the role of IDE in EBV reactivation has not yet been properly studied, the fact that it interacts with a key viral protein (gE) in a closely related virus suggests a potentially similar role in EBV reactivation. IDE, however, is known to interact with amyloid-beta peptides, which are implicated in mitochondrial dysfunction and neuroinflammation, leading to fatigue and neurological symptoms experienced by ME/CFS patients [57].

3.2.5. Role of HDAC2 Host Protein in EBV Reactivation

HDAC2, a histone deacetylase, removes acetyl groups from histone proteins associated with viral promoters, thus repressing viral gene transcription. HDAC inhibitors (HDACi) block the activity of HDACs, thus increasing histone acetylation levels, potentially leading to the activation of viral genes and the initiation of the EBV lytic cycle [58].

3.2.6. Role of PGE2 Host Protein in EBV Reactivation

Prostaglandin E2 (PGE2), produced by the enzyme COX-2, has a convincing positive correlation with LMP1 protein, which, in turn, can upregulate COX-2, leading to accelerated lymph node metastasis in nasopharyngeal carcinoma. COX-2 can also be upregulated by EBV latent antigen EBNA3C. PGE2 signalling acts through its receptors, EP1 and EP4, which are involved in mediating inflammation and cancer progression [45].

3.2.7. Role of Eukaryotic Translation Initiation Factor 4E (eIF4E) Host Protein in EBV Reactivation

The transcription of eIF4E is stimulated by the LMP1 protein, leading to nasopharyngeal carcinoma. eIF4E is significantly reduced by knocking down LMP1 and c-Myc, thereby inhibiting tumour proliferation, migration, and invasion. EIF4H also interacts with certain viral proteins, such as BZLF1 (also known as Zta or ZEBRA), which is a key regulator of the EBV lytic cycle [47].

3.2.8. Role of RIPK1 Host Protein in EBV Reactivation

RIPK1 is a protein kinase involved in both cell death and survival pathways. LMP1 increases the K48 and K63 ubiquitination of RIPK1, which leads to the activation of NF-κB, a signalling pathway that promotes cell survival and replication and shifts RIPK1 signalling away from necroptosis (cell death) [59].

3.3. EBV Lytic Replication Induces ACE2 Expression and Enhances SARS-CoV-2 Virus Entry

The spike protein of SARS-CoV-2 mediates viral entry into the cells by binding to the ACE2 receptor on epithelial cells. Verma D. et al. observed that EBV lytic gene expression increases ACE2 expression in EBV-infected epithelial cells. Changes in cellular gene expression after EBV lytic replication were analysed and a 500-fold increase in ACE2 mRNA levels was observed 24 h after lytic induction, which remained elevated for two days. Additionally, Verma D. et al. also observed that the ACE2 promoter further enhances EBV lytic expression as it contains response elements for Zta, an EBV transcriptional activator, which preferentially acts on methylated promoters, allowing them to reactivate epigenetically silenced EBV promoters from latency, hence leading to EBV entry into the lytic cycle of replication. Furthermore, SARS-CoV-2 virus entry into the cells was measured using a luciferase assay and it was found that EBV replication led to a 5--to-6-fold increase in SARS-CoV-2 entry. This indicates that subclinical EBV replication and lytic EBV gene expression may enhance the efficiency and extent of SARS-CoV-2 infection in humans [60].

3.4. Other Hypothetical Proposed Mechanisms of EBV Reactivation

3.4.1. Trogocytosis-Induced EBV Reactivation

Human primary B lymphocytes are the major reservoirs for EBV, and it seems quite reasonable that SARS-CoV-2 infection can trigger EBV reactivation in EBV-positive B cells. However, the irony here is that the SARS-CoV-2 virus attaches to the ACE2 receptor, which is not expressed well in B cells [61]. This irony has been adequately solved through a biological phenomenon known as trogocytosis, wherein cells share membrane-associated proteins, viral receptors, and viral particles during cell–cell conjugation. Previous studies have demonstrated that NK cells acquired a receptor for EBV from EBV-infected B cells [62], dendritic cells acquired HIV-1 from infected T cells [63], and B cells acquired membrane-bound α2,3 sialic acid receptor molecules from monocytes via trogocytosis [64]. Although direct studies related to SARS-CoV-2 are lacking, it can be speculated that EBV-infected B cells might be able to steal an ACE2 receptor via trogocytosis and then become susceptible to SARS-CoV2 infection.

3.4.2. Exosome-Mediated EBV Reactivation

The second possible explanation of EBV reactivation during Long COVID is based on the role of exosomes that are usually released by a host cell during the course of any viral infection, thus carrying viral and host components that can trigger an immune response. Barberis E. et al. performed a proteomic analysis of plasma exosomes and found the presence of SARS-CoV-2 RNA in host exosomal cargo using a reverse transcription–droplet digital polymerase chain reaction (RT–ddPCR), which can be speculated to have helped SARS-CoV-2 in spreading cell-to-cell infection via an endocytosis route [65]. Similarly, it can be speculated that exosomes containing SARS-CoV-2 RNA can spread infection from healthy B cells to B cells already infected with EBV.

4. Inflammation—A Common Thread Binding EBV and COVID-19

SARS-CoV-2 is known to cause the activation of the NLRP3 inflammasome [66,67], which in turn stimulates EBV reactivation [68]. Recent studies have demonstrated that EBV promotes the production of the inflammatory cytokines interleukin (IL)-1β and tumour necrosis factor-α (TNF-α) as well as the chemokines IL-8 and monocyte chemoattractant protein-1 (MCP-1) through the TLR9–MyD88–NF-κB pathway, which are also major cytokines implicated in Long COVID and other inflammatory diseases, hence linking EBV, inflammation, and COVID-19 [69]. Figure 1 depicts the inflammatory cascade associated with the co-infection of EBV and SARS-CoV-2 infection, leading to persistent symptoms of Long COVID.
An inflammatory state is further maintained by the following actions of innate and adaptive immune cells [69]:
(a)
There is an increase in neutrophils that maintain an inflammatory state by producing cytokines.
(b)
Macrophages become coated with myelin and adopt a foamy morphology. Foamy macrophages can maintain inflammation via EBV exosomes, cytokine production, and recruitment of more macrophages.
(c)
Monocytes maintain inflammation by carrying viral infection long after the initial infection has been resolved.
(d)
Inflammation is maintained by higher levels of IFN-γ and TNF-α, producing SARS-CoV-2-specific CD4+ and CD8+ T cells.
(e)
Autoantibodies produced by B cells contribute to tissue damage.
Also, monocyte-derived macrophages exposed to EBV antigen-laden exosomes express more CXCL10, which is a pro-inflammatory chemokine that attracts monocytes, eosinophils, T cells, and NK cells [70]. Therefore, it can be hypothesised that EBV antigen-laden exosomes can induce further inflammation [69].
SARS-CoV-2 infection induces a severe acute hyperinflammatory shock, termed multisystem inflammatory syndrome (MIS-C), in children and adolescents at four to eight weeks after infection [71]. Acute MIS-C is characterised by the impaired reactivation of SARS-CoV-2-reactive memory T cells, which is accompanied by the presence of TGFβ in T cells, B cells, and monocytes, along with the reduced antigen-presentation capabilities of monocytes, and can be reversed by blocking TGFβ. Goetzke C.C. et al. demonstrated that T cell receptor repertoires of patients with MIS-C exhibit an expansion of T cells expressing TCRVβ21.3, resembling EBV-reactive T cell clones; hence, serum TGFβ in patients with MIS-C can trigger EBV reactivation, which is reversible with TGFβ blockade [72].

5. EBV Reactivation Contributing to COVID-19-Associated Autoimmunity

Another common feature shared between SARS-CoV2 and EBV is the development or exacerbation of autoimmune phenomena. Although EBV has been associated with rheumatoid arthritis for the last 40 years [73], there has been a renewed interest regarding its pathogenetic role in several autoimmune diseases [74], particularly rheumatoid arthritis [75] and multiple sclerosis [76]. It has been hypothesised that EBV may increase the risk of developing autoimmunity in COVID-19 patients due to its ability to disrupt B cell tolerance [77]. Various studies have shown a high frequency of EBV reactivation in COVID-19 patients with severe illness and those suffering from Long COVID symptoms, including autoimmune diseases [16,17,19,20]. SARS-CoV-2 triggers the production of cytokine storm and causes the dysregulation of innate immune and adaptive immune responses with hyper-activation of immune system cells, and all of which lead to worsening of autoimmune diseases as well as Long COVID manifestations. The reactivation of latent EBV is favoured by COVID-19-induced immune dysregulation in the host. EBV can cause or exacerbate autoimmunity through different mechanisms, like the hyper-activation of autoreactive B cells, plasma cell differentiation, autoantibody production, tissue damage, and bystander activation. COVID-19-related immunological changes are exacerbated by EBV reactivation, which may elevate the likelihood of developing Long COVID complications [77].

6. Novel Therapeutics for EBV and Their Efficacy Against SARS-CoV-2

Various novel drugs and drugs under investigation have been used against EBV, some of which have also shown considerable potency against the SARS-CoV-2 virus, as shown in Table 4 [78,79,80,81,82,83,84,85,86,87,88,89,90,91,92].
Some of the novel drugs for EBV are as follows.

6.1. Nucleoside Analogues (Acyclovir, Valacyclovir, Ganciclovir, and Valganciclovir)

Acyclovir is an important acyclic nucleoside that is more readily phosphorylated by viral thymidine kinases than the corresponding cellular thymidine kinases, with an inherent flexibility that allows for optimised interactions in target enzyme binding sites. However, it has low bioavailability; due to this, a valine ester prodrug of acyclovir, (valacyclovir) was introduced, which had a 3-to-5-fold increase in oral bioavailability that led to an improved efficacy against herpesviruses. Gancyclovir is an acyclic guanosine mimic that retains the 3′-hydroxyl group. It also demonstrates low bioavailability, which can be increased with the addition of a valine ester to yield the prodrug valganciclovir [93].
Hocker B. et al. evaluated the efficacy of ganciclovir and valganciclovir prophylaxis on EBV viral load in EBV-naïve pediatric renal transplant recipients (R−) who had received a graft from an EBV-positive donor (D+) and found a significant decrease in the EBV primary infection [94].
Another study by Østensen A.B. et al. on pediatric liver transplant recipients showed that there was no difference in the proportion of patients with a reduction in virus load in patients treated with ganciclovir compared to untreated patients at 8 weeks [95].

6.2. Nucleotide Analogues (Cidofovir)

Cidofovir is a nucleoside analogue of deoxycytidine monophosphate primarily approved for the treatment of cytomegalovirus (CMV) retinitis in AIDS patients. It inhibits viral DNA synthesis by acting as a competitive inhibitor and alternative substrate for CMV DNA polymerase, blocking further viral DNA synthesis through its incorporation into the growing DNA strand. Cidofovir is also proven to be effective against other DNA viruses, including EBV [96]. Abdulkarim B. et al. demonstrated that cidofovir can significantly reduce LMP1 and EBNA2 mRNA and protein expression, along with the enhancement of radiation-induced apoptosis and radiosensitivity in EBV-related malignancies via the proteolytic cleavage of death effectorscaspase-9 and -3 [83].

6.3. Pyrophosphate Analogues (Foscarnet)

Foscarnet inhibits herpesvirus DNA polymerases by blocking the pyrophosphate-binding site and preventing the cleavage of pyrophosphate from deoxynucleoside triphosphates. Although approved for the treatment of CMV retinitis in AIDS patients, it has potentially shown activity against all human herpesviruses, including EBV [92], as shown in Table 4.

6.4. Fluvoxamine

Fluvoxamine is a selective serotonin reuptake inhibitor and sigma-1 receptor agonist that is primarily used to treat obsessive–compulsive disorder (OCD) but has also shown activity against SARS-CoV-2 via a reduction in platelet aggregation, decreased mast cell degranulation, the regulation of inositol-requiring enzyme 1α-driven inflammation, and interference with endolysosomal viral trafficking [97]. It also attenuates SARS-CoV-2 cell entry by inhibiting acid sphingomyelinase (ASM) activity and the formation of a ceramide-enriched membrane domain [33]. Fluvoxamine reduces XBP1 activation, which in turn modulates the ER stress response pathway and reduces cytokine storm. XBP1 plays a major role in EBV reactivation and hence its deactivation by fluvoxamine helps to prevent lytic gene expression in EBV [33].

6.5. Maribavir

Maribavir is an oral benzimidazole riboside approved by the U.S. Food and Drug Administration in 2021 for the treatment of post-transplant cytomegalovirus infection that is refractory to treatment with ganciclovir, valganciclovir, cidofovir, or foscarnet. It inhibits viral protein kinase UL97, which further inhibits the phosphorylation of the UL44 protein, thus inhibiting CMV DNA replication [98]. It has shown in vitro activity against EBV by inhibiting both viral DNA replication and viral transcription [90] along with the inhibition of BGLF4, which largely affects EBV transcript levels [99].

6.6. KAY-2-41 and KAH-39-149

KAY-2-41 (1′-methyl-substituted 4′-thiothymidine) and KAH-39-149 (4′-azido analogue of 4′-thiothymidine) are thiothymidine derivatives that have shown in vitro inhibitory activities against EBV. It has been observed that mutations in the viral thymidine kinase (TK) of KAY-2-41- and KAH-39-149-resistant herpesviruses conferred only low-level resistance to these drugs but high-level resistance to other TK-dependent antiviral agents. Coen et al. found that the antiviral efficacy of KAH-39-149 was superior to KAY-2-41 in a mouse model of gammaherpesvirus infection [79].

6.7. Brincidofovir (CMX-001)

Brincidofovir is a prodrug of cidofovir that releases cidofovir intracellularly, allowing for higher intracellular and lower plasma concentrations of cidofovir, effectively increasing its oral bioavailability and activity against dsDNA viruses. But despite its enhanced antiviral activity, it is not yet FDA-approved for adenovirus or CMV due to a lack of efficacy in clinical trials [91].

6.8. Inhibitors of EBV Nuclear Antigen 1 (EBNA1)

EBNA1 inhibitors decrease the expression of EBNA1, which is involved in the maintenance, replication, and segregation of the EBV genome. Lee E.K. et al. characterised H31 as an EBNA1 inhibitor, which inhibits EBNA1-dependent oriP sequence-specific DNA binding activity and produces the gradual loss of EBV episome, thereby delaying the growth of EBV-infected lymphoblastoid cell lines or Burkitt’s lymphoma cells [100].

6.9. Newer Drugs Used to Reduce Inflammation

6.9.1. Ibrutinib

Ibrutinib, a Bruton tyrosine kinase (BTK) inhibitor, has shown promise in treating EBV-related lymphoproliferative disorders, particularly in immunocompromised patients. Kotaki R. et al. demonstrated that Ibrutinib inhibits the survival of EBV-positive lymphoblastic cell lines (LCLs) and Aggressive NK cell leukemia (ANKL) cells in vitro [101]. This drug also reduces the ability of NLRP3 to reduce IL1β, thereby reducing inflammation [102].

6.9.2. MCC950

MCC950, a selective NLRP3 inflammasome inhibitor, plays a role in potentially treating EBV reactivation by blocking the activation of the NLRP3 inflammasome via the inhibition of its ATP hydrolysis activity and the subsequent release of inflammatory cytokines like IL-1β and IL-18. This small molecule has not yet progressed beyond the Phase I safety trial. It has good potential for preventing the development of EBV-related cancers by limiting EBV-induced B cell transformation [103].

7. Conclusions

There is sufficient evidence regarding the contribution of COVID-19 in EBV reactivation from the latent to lytic phases, leading to a worsening of Long COVID manifestations. Recent studies have hypothesised various mechanisms leading to EBV reactivation but the data is severely limited. Data on post-COVID-19 EBV reactivation in the context of autoimmune conditions is also limited, making it difficult to establish a direct role for EBV in the development or worsening of these conditions following SARS-CoV-2 infection. The boundary between the immunological effects of SARS-CoV-2 alone and the effects of latent co-infecting viruses, like EBV, still remains unclear and deserves further investigation.

Author Contributions

Each author made substantial contribution to the conception and design of the review. CRediT authorship contribution statement: S.M. (Supriya Mahajan) (Corresponding and First Author)—Formal analysis, Supervision, Validation, Writing—original draft, Writing—review & editing, Conceptualization; S.M. (Saurabh Mahajan) (Second author): Writing—review & editing, Conceptualization; S.P. (Third author): Resources, Data curation. All authors have read and agreed to the published version of the manuscript.

Funding

This research did not receive any specific grant from funding agencies in the public, commercial or profit sectors.

Conflicts of Interest

There are no conflicts of interest.

References

  1. Zanella, M.C.; Cordey, S.; Kaiser, L. Beyond Cytomegalovirus and Epstein-Barr Virus: A Review of Viruses Composing the Blood Virome of Solid Organ Transplant and Hematopoietic Stem Cell Transplant Recipients. Clin. Microbiol. Rev. 2020, 33, e00027-20. [Google Scholar] [CrossRef] [PubMed]
  2. Indari, O.; Ghosh, S.; Bal, A.S.; James, A.; Garg, M.; Mishra, A.; Karmodiya, K.; Jha, H.C. Awakening the sleeping giant: Epstein–Barr virus reactivation by biological agents. Pathog. Dis. 2024, 82, ftae002. [Google Scholar] [CrossRef]
  3. Shafiee, A.; Aghajanian, S.; Athar, M.M.T.; Gargari, O.K. Epstein-Barr virus and COVID-19. J. Med. Virol. 2022, 94, 4040–4042. [Google Scholar] [CrossRef] [PubMed]
  4. Farrell, P.J. Epstein–Barr Virus and Cancer. Annu. Rev. Pathol. Mech. Dis. 2019, 14, 29–53. [Google Scholar] [CrossRef] [PubMed]
  5. Adhikari, S.P.; Meng, S.; Wu, Y.J.; Mao, Y.P.; Ye, R.X.; Wang, Q.Z.; Sun, C.; Sylvia, S.; Rozelle, S.; Raat, H.; et al. Epidemiology, causes, clinical manifestation and diagnosis, prevention and control of coronavirus disease (COVID-19) during the early outbreak period: A scoping review. Infect. Dis. Poverty 2020, 9, 29. [Google Scholar] [CrossRef]
  6. Altmann, D.M.; Whettlock, E.M.; Liu, S.; Arachchillage, D.J.; Boyton, R.J. The immunology of long COVID. Nat. Rev. Immunol. 2023, 23, 618–634. [Google Scholar] [CrossRef]
  7. Miller, G.; El-Guindy, A.; Countryman, J.; Ye, J.; Gradoville, L. Lytic cycle switches of oncogenic human gammaherpesviruses. Adv. Cancer Res. 2007, 97, 81–109. [Google Scholar]
  8. Murata, T.; Sugimoto, A.; Inagaki, T.; Yanagi, Y.; Watanabe, T.; Sato, Y.; Kimura, H. Molecular Basis of Epstein-Barr Virus Latency Establishment and Lytic Reactivation. Viruses 2021, 13, 2344. [Google Scholar] [CrossRef]
  9. Young, L.S.; Arrand, J.R.; Murray, P.G. EBV gene expression and regulation. In Human Herpesviruses: Biology, Therapy, and Immunoprophylaxis; Arvin, A., Campadelli-Fiume, G., Mocarski, E., Moore, P.S., Roizman, B., Whitley, R., Yamanishi, K., Eds.; Cambridge University Press: Cambridge, UK, 2007; Volume 27. [Google Scholar] [PubMed]
  10. Luo, Y.; Liu, Y.; Wang, C.; Gan, R. Signaling pathways of EBV-induced oncogenesis. Cancer Cell Int. 2021, 21, 93. [Google Scholar] [CrossRef]
  11. Hashimoto, K. Detrimental effects of COVID-19 in the brain and therapeutic options for long COVID: The role of Epstein-Barr virus and the gut-brain axis. Mol. Psychiatry 2023, 28, 4968–4976. [Google Scholar] [CrossRef]
  12. Kim, J.Y.H.; Ragusa, M.; Tortosa, F.; Torres, A.; Gresh, L.; Méndez-Rico, J.A.; Alvarez-Moreno, C.A.; Lisboa, T.C.; Valderrama-Beltrán, S.L.; Aldighieri, S.; et al. Viral reactivations and co-infections in COVID-19 patients: A systematic review. BMC Infect. Dis. 2023, 23, 259. [Google Scholar] [CrossRef]
  13. Talukder, S.; Deb, P.; Parveen, M.; Zannat, K.E.; Bhuiyan, A.H.; Yeasmin, M.; Molla, M.A.; Saif-Ur-Rahman, K. Clinical features and outcomes of COVID-19 patients with concomitant herpesvirus co-infection or reactivation: A systematic review. New Microbes New Infect. 2024, 58, 101233. [Google Scholar] [CrossRef]
  14. Bernal, K.D.E.; Whitehurst, C.B. Incidence of Epstein-Barr virus reactivation is elevated in COVID-19 patients. Virus Res. 2023, 334, 199157. [Google Scholar] [CrossRef] [PubMed]
  15. Chen, T.; Song, J.; Liu, H.; Zheng, H.; Chen, C. Positive Epstein-Barr virus detection in coronavirus disease 2019 (COVID-19) patients. Sci. Rep. 2021, 11, 10902. [Google Scholar] [CrossRef]
  16. Gold, J.E.; Okyay, R.A.; Licht, W.E.; Hurley, D.J. Investigation of Long COVID Prevalence and Its Relationship to Epstein-Barr Virus Reactivation. Pathogens 2021, 10, 763. [Google Scholar] [CrossRef] [PubMed]
  17. Naendrup, J.H.; Garcia Borrega, J.; Eichenauer, D.A.; Shimabukuro-Vornhagen, A.; Kochanek, M.; Böll, B. Reactivation of EBV and CMV in Severe COVID-19-Epiphenomena or Trigger of Hyperinflammation in Need of Treatment? A Large Case Series of Critically ill Patients. J. Intensive Care Med. 2022, 37, 1152–1158. [Google Scholar] [CrossRef] [PubMed]
  18. Paolucci, S.; Cassaniti, I.; Novazzi, F.; Fiorina, L.; Piralla, A.; Comolli, G.; Bruno, R.; Maserati, R.; Gulminetti, R.; Novati, S.; et al. EBV DNA increase in COVID-19 patients with impaired lymphocyte subpopulation count. Int. J. Infect. Dis. 2021, 104, 315–319. [Google Scholar] [CrossRef]
  19. Xie, Y.; Cao, S.; Dong, H.; Lv, H.; Teng, X.; Zhang, J.; Wang, T.; Zhang, X.; Qin, Y.; Chai, Y.; et al. Clinical characteristics and outcomes of critically ill patients with acute COVID-19 with Epstein-Barr virus reactivation. BMC Infect. Dis. 2021, 21, 955. [Google Scholar] [CrossRef] [PubMed]
  20. Mattei, A.; Schiavoni, L.; Riva, E.; Ciccozzi, M.; Veralli, R.; Urselli, A.; Citriniti, V.; Nenna, A.; Pascarella, G.; Costa, F.; et al. Epstein-Barr virus, Cytomegalovirus, and Herpes Simplex-1/2 reactivations in critically ill patients with COVID-19. Intensive Care Med. Exp. 2024, 12, 40. [Google Scholar] [CrossRef]
  21. Su, Y.; Yuan, D.; Chen, D.G.; Ng, R.H.; Wang, K.; Choi, J.; Li, S.; Hong, S.; Zhang, R.; Xie, J.; et al. Multiple early factors anticipate post-acute COVID-19 sequelae. Cell 2022, 185, 881–895.e20. [Google Scholar] [CrossRef]
  22. Simonnet, A.; Engelmann, I.; Moreau, A.S.; Garcia, B.; Six, S.; El Kalioubie, A.; Robriquet, L.; Hober, D.; Jourdain, M. High incidence of Epstein-Barr virus, cytomegalovirus, and human-herpes virus-6 reactivations in critically ill patients with COVID-19. Infect. Dis. Now. 2021, 51, 296–299. [Google Scholar] [CrossRef] [PubMed]
  23. Zubchenko, S.; Kril, I.; Nadizhko, O.; Matsyura, O.; Chopyak, V. Herpesvirus infections and post-COVID-19 manifestations: A pilot observational study. Rheumatol. Int. 2022, 42, 1523–1530. [Google Scholar] [CrossRef]
  24. Gautret, P.; Lagier, J.C.; Honoré, S.; Hoang, V.T.; Colson, P.; Raoult, D. Hydroxychloroquine and azithromycin as a treatment of COVID-19: Results of an open label non-randomized clinical trial revisited. Int. J. Antimicrob. Agents 2021, 57, 106243. [Google Scholar] [CrossRef] [PubMed]
  25. Hoffmann, M.; Schroeder, S.; Kleine-Weber, H.; Müller, M.A.; Drosten, C.; Pöhlmann, S. Nafamostat Mesylate Blocks Activation of SARS-CoV-2: New Treatment Option for COVID-19. Antimicrob. Agents Chemother. 2020, 64, e00754-20. [Google Scholar] [CrossRef] [PubMed]
  26. Chen, J.; Dai, L.; Barrett, L.; Post, S.R.; Qin, Z. SARS-CoV-2 proteins and anti-COVID-19 drugs induce lytic reactivation of an oncogenic virus. Commun. Biol. 2021, 4, 682. [Google Scholar] [CrossRef] [PubMed]
  27. Chen, J.; Dai, L.; Kendrick, S.; Post, S.R.; Qin, Z. The Anti-COVID-19 Drug Remdesivir Promotes Oncogenic Herpesvirus Reactivation through Regulation of Intracellular Signaling Pathways. Antimicrob. Agents Chemother. 2022, 66, e02395-21. [Google Scholar] [CrossRef]
  28. Hill, E.R.; Koganti, S.; Zhi, J.; Megyola, C.; Freeman, A.F.; Palendira, U.; Tangye, S.G.; Farrell, P.J.; Bhaduri-McIntosh, S. Signal transducer and activator of transcription 3 limits Epstein-Barr virus lytic activation in B lymphocytes. J. Virol. 2013, 87, 11438–11446. [Google Scholar] [CrossRef]
  29. Gonnella, R.; Granato, M.; Farina, A.; Santarelli, R.; Faggioni, A.; Cirone, M. PKC theta and p38 MAPK activate the EBV lytic cycle through autophagy induction. Biochim. Biophys. Acta 2015, 1853, 1586–1595. [Google Scholar] [CrossRef]
  30. Li, X.; Burton, E.M.; Bhaduri-McIntosh, S. Chloroquine triggers Epstein-Barr virus replication through phosphorylation of KAP1/TRIM28 in Burkitt lymphoma cells. PLoS Pathog. 2017, 13, e1006249. [Google Scholar] [CrossRef]
  31. Yang, E.V.; Webster Marketon, J.I.; Chen, M.; Lo, K.W.; Kim, S.J.; Glaser, R. Glucocorticoids activate Epstein Barr virus lytic replication through the upregulation of immediate early BZLF1 gene expression. Brain Behav. Immun. 2010, 24, 1089–1096. [Google Scholar] [CrossRef]
  32. Omi, T.; Tanimukai, H.; Kanayama, D.; Sakagami, Y.; Tagami, S.; Okochi, M.; Morihara, T.; Sato, M.; Yanagida, K.; Kitasyoji, A.; et al. Fluvoxamine alleviates ER stress via induction of Sigma-1 receptor. Cell Death Dis. 2014, 5, e1332. [Google Scholar] [CrossRef]
  33. Khani, E.; Entezari-Maleki, T. Fluvoxamine and long COVID-19; a new role for sigma-1 receptor (S1R) agonists. Mol. Psychiatry 2022, 27, 3562. [Google Scholar] [CrossRef]
  34. Rosen, D.A.; Seki, S.M.; Fernández-Castañeda, A.; Beiter, R.M.; Eccles, J.D.; Woodfolk, J.A.; Gaultier, A. Modulation of the sigma-1 receptor-IRE1 pathway is beneficial in preclinical models of inflammation and sepsis. Sci. Transl. Med. 2019, 11, eaau5266. [Google Scholar] [CrossRef] [PubMed]
  35. Hsiao, J.R.; Chang, K.C.; Chen, C.W.; Wu, S.Y.; Su, I.J.; Hsu, M.C.; Jin, Y.T.; Tsai, S.T.; Takada, K.; Chang, Y. Endoplasmic reticulum stress triggers XBP-1-mediated up-regulation of an EBV oncoprotein in nasopharyngeal carcinoma. Cancer Res. 2009, 69, 4461–4467. [Google Scholar] [CrossRef]
  36. Gonzalez-Farre, B.; Rovira, J.; Martinez, D.; Valera, A.; Garcia-Herrera, A.; Marcos, M.A.; Sole, C.; Roue, G.; Colomer, D.; Gonzalvo, E.; et al. In vivo intratumoral Epstein-Barr virus replication is associated with XBP1 activation and early-onset post-transplant lymphoproliferative disorders with prognostic implications. Mod. Pathol. 2014, 27, 1599–1611. [Google Scholar] [CrossRef]
  37. Keck, K.M.; Moquin, S.A.; He, A.; Fernandez, S.G.; Somberg, J.J.; Liu, S.M.; Martinez, D.M.; Miranda, J.L. Bromodomain and extraterminal inhibitors block the Epstein-Barr virus lytic cycle at two distinct steps. J. Biol. Chem. 2017, 292, 13284–13295. [Google Scholar] [CrossRef] [PubMed]
  38. Van Gent, M.; Reich, A.; Velu, S.E.; Gack, M.U. Nonsense-mediated decay controls the reactivation of the oncogenic herpesviruses EBV and KSHV. PLoS Biol. 2021, 19, e3001097. [Google Scholar] [CrossRef] [PubMed]
  39. Fu, W.; Verma, D.; Burton, A.; Swaminathan, S. Cellular RNA Helicase DHX9 Interacts with the Essential Epstein-BarrVirus (EBV) Protein SM and Restricts EBV Lytic Replication. J. Virol. 2019, 93, e01244-18. [Google Scholar] [CrossRef] [PubMed]
  40. Tengvall, K.; Huang, J.; Hellström, C.; Kammer, P.; Biström, M.; Ayoglu, B.; Bomfim, I.L.; Stridh, P.; Butt, J.; Brenner, N.; et al. Molecular mimicry between Anoctamin 2 and Epstein-Barr virus nuclear antigen 1 associates with multiple sclerosis risk. Proc. Natl. Acad. Sci. USA 2019, 116, 16955–16960. [Google Scholar] [CrossRef]
  41. Wang, L.W.; Shen, H.; Nobre, L.; Ersing, I.; Paulo, J.A.; Trudeau, S.; Wang, Z.; Smith, N.A.; Ma, Y.; Reinstadler, B.; et al. Epstein-Barr-Virus-Induced One-Carbon Metabolism Drives B Cell Transformation. Cell Metab. 2019, 30, 539–555.e11. [Google Scholar] [CrossRef]
  42. Canaan, A.; Haviv, I.; Urban, A.E.; Schulz, V.P.; Hartman, S.; Zhang, Z.; Palejev, D.; Deisseroth, A.B.; Lacy, J.; Snyder, M.; et al. EBNA1 regulates cellular gene expression by binding cellular promoters. Proc. Natl. Acad. Sci. USA 2009, 106, 22421–22426. [Google Scholar] [CrossRef] [PubMed]
  43. Williams, M.V.; Cox, B.; Lafuse, W.P.; Ariza, M.E. Epstein-Barr Virus dUTPase Induces Neuroinflammatory Mediators: Implications for Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Clin. Ther. 2019, 41, 848–863. [Google Scholar] [CrossRef]
  44. Tsai, P.F.; Lin, S.J.; Weng, P.L.; Tsai, S.C.; Lin, J.H.; Chou, Y.C.; Tsai, C.H. Interplay between PKCδ and Sp1 on histone deacetylase inhibitor-mediated Epstein-Barr virus reactivation. J. Virol. 2011, 85, 2373–2385. [Google Scholar] [CrossRef]
  45. Gandhi, J.; Gaur, N.; Khera, L.; Kaul, R.; Robertson, E.S. COX-2 induces lytic reactivation of EBV through PGE2 by modulating the EP receptor signaling pathway. Virology 2015, 484, 1–14. [Google Scholar] [CrossRef]
  46. Ariza, M.E.; Rivailler, P.; Glaser, R.; Chen, M.; Williams, M.V.; Zhang, L. Epstein-Barr virus encoded dUTPase containing exosomes modulate innate and adaptive immune responses in human dendritic cells and peripheral blood mononuclear cells. PLoS ONE 2013, 8, e69827. [Google Scholar] [CrossRef] [PubMed]
  47. Zhao, Y.; Pang, T.Y.; Wang, Y.; Wang, S.; Kang, H.X.; Ding, W.B.; Yong, W.; Bie, Y.; Cheng, X.; Zeng, C.; et al. LMP1 stimulates the transcription of eIF4E to promote the proliferation, migration and invasion of human nasopharyngeal carcinoma. FEBS J. 2014, 281, 3004–3018. [Google Scholar] [CrossRef] [PubMed]
  48. Liu, X.; Li, Y.; Peng, S.; Yu, X.; Li, W.; Shi, F.; Luo, X.; Tang, M.; Tan, Z.; Bode, A.M.; et al. Epstein-Barr virus encoded latent membrane protein 1 suppresses necroptosis through targeting RIPK1/3 ubiquitination. Cell Death Dis. 2018, 9, 53. [Google Scholar] [CrossRef]
  49. Arvey, A.; Tempera, I.; Lieberman, P.M. Interpreting the Epstein-Barr Virus (EBV) epigenome using high-throughput data. Viruses 2013, 5, 1042–1054. [Google Scholar] [CrossRef]
  50. Chang, C.W.; Lee, C.P.; Su, M.T.; Tsai, C.H.; Chen, M.R. BGLF4 kinase modulates the structure and transport preference of the nuclear pore complex to facilitate nuclear import of Epstein-Barr virus lytic proteins. J. Virol. 2015, 89, 1703–1718. [Google Scholar] [CrossRef]
  51. Cai, W.; Yang, H. The structure and regulation of Cullin 2 based E3 ubiquitin ligases and their biological functions. Cell Div. 2016, 11, 7. [Google Scholar] [CrossRef]
  52. Lin, A.; Wang, S.; Nguyen, T.; Shire, K.; Frappier, L. The EBNA1 protein of Epstein-Barr virus functionally interacts with Brd4. J. Virol. 2008, 82, 12009–12019. [Google Scholar] [CrossRef] [PubMed]
  53. Maruyama, T.; Farina, A.; Dey, A.; Cheong, J.; Bermudez, V.P.; Tamura, T.; Sciortino, S.; Shuman, J.; Hurwitz, J.; Ozato, K. A Mammalian bromodomain protein, brd4, interacts with replication factor C and inhibits progression to S phase. Mol. Cell Biol. 2002, 22, 6509–6520. [Google Scholar] [CrossRef]
  54. Garcia, P.L.; Miller, A.L.; Gamblin, T.L.; Council, L.N.; Christein, J.D.; Arnoletti, J.P.; Heslin, M.J.; Reddy, S.; Richardson, J.H.; Cui, X.; et al. JQ1 Induces DNA Damage and Apoptosis, and Inhibits Tumor Growth in a Patient-Derived Xenograft Model of Cholangiocarcinoma. Mol. Cancer Ther. 2018, 17, 107–118. [Google Scholar] [CrossRef]
  55. Hug, N.; Longman, D.; Cáceres, J.F. Mechanism and regulation of the nonsense-mediated decay pathway. Nucleic Acids Res. 2016, 44, 1483–1495. [Google Scholar] [CrossRef] [PubMed]
  56. Li, Q.; Ali, M.A.; Cohen, J.I. Insulin degrading enzyme is a cellular receptor mediating varicella-zoster virus infection and cell-to-cell spread. Cell 2006, 127, 305–316. [Google Scholar] [CrossRef] [PubMed]
  57. Newcombe, E.A.; Camats-Perna, J.; Silva, M.L.; Valmas, N.; Huat, T.J.; Medeiros, R. Inflammation: The link between comorbidities, genetics, and Alzheimer’s disease. J. Neuroinflamm. 2018, 15, 276. [Google Scholar] [CrossRef]
  58. Herbein, G.; Wendling, D. Histone deacetylases in viral infections. Clin. Epigenet. 2010, 1, 13–24. [Google Scholar] [CrossRef]
  59. Udawatte, D.J.; Rothman, A.L. Viral Suppression of RIPK1-Mediated Signaling. mBio 2021, 12, e0172321. [Google Scholar] [CrossRef]
  60. Verma, D.; Church, T.M.; Swaminathan, S. Epstein-Barr Virus Lytic Replication Induces ACE2 Expression and Enhances SARS-CoV-2 Pseudotyped Virus Entry in Epithelial Cells. J. Virol. 2021, 95, e0019221. [Google Scholar] [CrossRef]
  61. Hamming, I.; Timens, W.; Bulthuis, M.L.C.; Lely, A.T.; Navis, G.J.; van Goor, H. Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J. Pathol. 2004, 203, 631–637. [Google Scholar] [CrossRef]
  62. Tabiasco, J.; Vercellone, A.; Meggetto, F.; Hudrisier, D.; Brousset, P.; Fournié, J.J. Acquisition of viral receptor by NK cells through immunological synapse. J. Immunol. 2003, 170, 5993–5998. [Google Scholar] [CrossRef] [PubMed]
  63. Venkatachari, N.J.; Alber, S.; Watkins, S.C.; Ayyavoo, V. HIV-1 infection of DC: Evidence for the acquisition of virus particles from infected T cells by antigen uptake mechanism. PLoS ONE 2009, 4, e7470. [Google Scholar] [CrossRef]
  64. Kongsomros, S.; Thanunchai, M.; Manopwisedjaroen, S.; Na-Ek, P.; Wang, S.F.; Taechalertpaisarn, T.; Thitithanyanont, A. Trogocytosis with monocytes associated with increased α2,3 sialic acid expression on B cells during H5N1 influenza virus infection. PLoS ONE 2020, 15, e0239488. [Google Scholar] [CrossRef]
  65. Barberis, E.; Vanella, V.V.; Falasca, M.; Caneapero, V.; Cappellano, G.; Raineri, D.; Ghirimoldi, M.; De Giorgis, V.; Puricelli, C.; Vaschetto, R.; et al. Circulating Exosomes Are Strongly Involved in SARS-CoV-2 Infection. Front. Mol. Biosci. 2021, 8, 632290. [Google Scholar] [CrossRef]
  66. Amin, S.; Aktar, S.; Rahman, M.M.; Chowdhury, M.M.H. NLRP3 inflammasome activation in COVID-19: An interlink between risk factors and disease severity. Microbes Infect. 2022, 24, 104913. [Google Scholar] [CrossRef] [PubMed]
  67. Xu, H.; Akinyemi, I.A.; Chitre, S.A.; Loeb, J.C.; Lednicky, J.A.; McIntosh, M.T.; Bhaduri-McIntosh, S. SARS-CoV-2 viroporin encoded by ORF3a triggers the NLRP3 inflammatory pathway. Virology 2022, 568, 13–22. [Google Scholar] [CrossRef] [PubMed]
  68. Burton, E.M.; Goldbach-Mansky, R.; Bhaduri-McIntosh, S. A promiscuous inflammasome sparks replication of a common tumor virus. Proc. Natl. Acad. Sci. USA 2020, 117, 1722–1730. [Google Scholar] [CrossRef]
  69. Rousseau, B.A.; Bhaduri-McIntosh, S. Inflammation and Epstein-Barr virus at the crossroads of Multiple Sclerosis and Post-Acute Sequelae of COVID-19 Infection. Viruses 2023, 15, 949. [Google Scholar] [CrossRef]
  70. Vazirinejad, R.; Ahmadi, Z.; Kazemi Arababadi, M.; Hassanshahi, G.; Kennedy, D. The biological functions, structure and sources of CXCL10 and its outstanding part in the pathophysiology of multiple sclerosis. Neuroimmunomodulation 2014, 21, 322–330. [Google Scholar] [CrossRef]
  71. Riphagen, S.; Gomez, X.; Gonzalez-Martinez, C.; Wilkinson, N.; Theocharis, P. Hyperinflammatory shock in children during COVID-19 pandemic. Lancet 2020, 395, 1607–1608. [Google Scholar] [CrossRef]
  72. Goetzke, C.C.; Massoud, M.; Frischbutter, S.; Guerra, G.M.; Ferreira-Gomes, M.; Heinrich, F.; von Stuckrad, A.S.L.; Wisniewski, S.; Licha, J.R.; Bondareva, M.; et al. TGFβ links EBV to multisystem inflammatory syndrome in children. Nature 2025, 640, 762–771. [Google Scholar] [CrossRef]
  73. Tosato, G.; Steinberg, A.D.; Blaese, R.M. Defective EBV-specific suppressor T-cell function in rheumatoid arthritis. N. Engl. J. Med. 1981, 305, 1238–1243. [Google Scholar] [CrossRef] [PubMed]
  74. Houen, G.; Trier, N.H. Epstein-Barr virus and systemic autoimmune diseases. Front. Immunol. 2021, 11, 587380. [Google Scholar] [CrossRef] [PubMed]
  75. Fanelli, I.; Rovero, P.; Hansen, P.R.; Frederiksen, J.L.; Houen, G.; Trier, N.H. Reactivity of Rheumatoid Arthritis-Associated Citrulline-Dependent Antibodies to Epstein-Barr Virus Nuclear Antigen1-3. Antibodies 2022, 11, 20. [Google Scholar] [CrossRef]
  76. Bjornevik, K.; Cortese, M.; Healy, B.C.; Kuhle, J.; Mina, M.J.; Leng, Y.; Elledge, S.J.; Niebuhr, D.W.; Scher, A.I.; Munger, K.L.; et al. Longitudinal analysis reveals high prevalence of Epstein-Barr virus associated with multiple sclerosis. Science 2022, 375, 296–301. [Google Scholar] [CrossRef]
  77. Tarasco, M.C.; Iacomino, N.; Mantegazza, R.; Cavalcante, P. COVID-19, Epstein-Barr virus reactivation and autoimmunity: Casual or causal liaisons? J. Microbiol. Immunol. Infect. 2025. ahead of print. [Google Scholar]
  78. Keith, K.A.; Hartline, C.B.; Bowlin, T.L.; Prichard, M.N. A standardized approach to the evaluation of antivirals against DNA viruses: Polyomaviruses and lymphotropic herpesviruses. Antivir. Res. 2018, 159, 122–129. [Google Scholar] [CrossRef] [PubMed]
  79. Coen, N.; Duraffour, S.; Haraguchi, K.; Balzarini, J.; van den Oord, J.J.; Snoeck, R.; Andrei, G. Antiherpesvirus activities of two novel 4′-thiothymidine derivatives, KAY-2-41 and KAH-39-149, are dependent on viral and cellular thymidine kinases. Antimicrob. Agents Chemother. 2014, 58, 4328–4340. [Google Scholar] [CrossRef]
  80. Meng, M.; Zhang, S.; Dong, X.; Sun, W.; Deng, Y.; Li, W.; Li, R.; Annane, D.; Wu, Z.; Chen, D. COVID-19 associated EBV reactivation and effects of ganciclovir treatment. Immun. Inflamm. Dis. 2022, 10, e597. [Google Scholar] [CrossRef]
  81. German, E.R.; Jairath, M.K.; Caston, J. Treatment of Long-Haul COVID Patients with Off-Label Acyclovir. Cureus 2023, 15, e37926. [Google Scholar] [CrossRef]
  82. Yoshizaki, T.; Wakisaka, N.; Kondo, S.; Murono, S.; Shimizu, Y.; Nakashima, M.; Tsuji, A.; Furukawa, M. Treatment of locally recurrent Epstein-Barr virus-associated nasopharyngeal carcinoma using the anti-viral agent cidofovir. J. Med. Virol. 2008, 80, 879–882. [Google Scholar] [CrossRef]
  83. Abdulkarim, B.; Sabri, S.; Zelenika, D.; Deutsch, E.; Frascogna, V.; Klijanienko, J.; Vainchenker, W.; Joab, I.; Bourhis, J. Antiviral agent cidofovir decreases Epstein-Barr virus (EBV) oncoproteins and enhances the radiosensitivity in EBV-related malignancies. Oncogene 2003, 22, 2260–2271. [Google Scholar] [CrossRef] [PubMed]
  84. Rahman, M.R.; Banik, A.; Chowdhury, I.M.; Sajib, E.H.; Sarkar, S. Identification of potential antivirals against SARS-CoV-2 using virtual screening method. Inform. Med. Unlocked 2021, 23, 100531. [Google Scholar] [CrossRef] [PubMed]
  85. Afshar, K.; Rao, A.P.; Patel, V.; Forrester, K.; Ganesh, S. Use of Foscarnet Therapy for EBV Infection following Control of PTLD with Enhancement of Cellular Immunity in a Lung-Transplant Recipient. J. Transplant. 2011, 2011, 919651. [Google Scholar] [CrossRef]
  86. Schneider, U.; Ruhnke, M.; Delecluse, H.J.; Stein, H.; Huhn, D. Regression of Epstein-Barr virus-associated lymphoproliferative disorders in patients with acquired immunodeficiency syndrome during therapy with foscarnet. Ann. Hematol. 2000, 79, 214–216. [Google Scholar] [CrossRef]
  87. Hashimoto, Y.; Suzuki, T.; Hashimoto, K. Comments to “Fluvoxamine and long COVID-19: A new role for sigma-1 receptor (S1R) agonists” by Khani and Entezari-Maleki. Mol Psychiatry 2022, 27, 3563–3564. [Google Scholar] [CrossRef] [PubMed]
  88. Hashimoto, K. Overview of the potential use of fluvoxamine for COVID-19 and long COVID. Discov. Ment. Health 2023, 3, 9. [Google Scholar] [CrossRef]
  89. Zacny, V.L.; Gershburg, E.; Davis, M.G.; Biron, K.K.; Pagano, J.S. Inhibition of Epstein-Barr virus replication by a benzimidazole L-riboside: Novel antiviral mechanism of 5,6-dichloro-2-(isopropylamino)-1-beta-L-ribofuranosyl-1H-benzimidazole. J. Virol. 1999, 73, 7271–7277. [Google Scholar] [CrossRef]
  90. Wang, F.Z.; Roy, D.; Gershburg, E.; Whitehurst, C.B.; Dittmer, D.P.; Pagano, J.S. Maribavir inhibits Epstein-barr virus transcription in addition to viral DNA replication. J. Virol. 2009, 83, 12108–12117. [Google Scholar] [CrossRef]
  91. Hostetler, K.Y. Alkoxyalkyl prodrugs of acyclic nucleoside phosphonates enhance oral antiviral activity and reduce toxicity: Current state of the art. Antiviral Res. 2009, 82, A84–A98. [Google Scholar] [CrossRef]
  92. Andrei, G.; Trompet, E.; Snoeck, R. Novel Therapeutics for Epstein-Barr Virus. Molecules 2019, 24, 997. [Google Scholar] [CrossRef]
  93. Yates, M.K.; Seley-Radtke, K.L. The evolution of antiviral nucleoside analogues: A review for chemists and non-chemists. Part II: Complex modifications to the nucleoside scaffold. Antiviral Res. 2019, 162, 5–21. [Google Scholar] [CrossRef]
  94. Höcker, B.; Böhm, S.; Fickenscher, H.; Küsters, U.; Schnitzler, P.; Pohl, M.; John, U.; Kemper, M.J.; Fehrenbach, H.; Wigger, M.; et al. Val-Ganciclovir prophylaxis reduces Epstein-Barr virus primary infection in pediatric renal transplantation. Transpl. Int. 2012, 25, 723–731. [Google Scholar] [CrossRef] [PubMed]
  95. Østensen, A.B.; Sanengen, T.; Holter, E.; Line, P.D.; Almaas, R. No effect of treatment with intravenous ganciclovir on Epstein-Barr virus viremia demonstrated after pediatric liver transplantation. Pediatr. Transplant. 2017, 21, 6. [Google Scholar] [CrossRef] [PubMed]
  96. Poppens, M.; Ruci, A.; Davis, J. Cutaneous Applications of the Antiviral Drug Cidofovir: A Review. J. Clin. Med. 2024, 13, 2462. [Google Scholar] [CrossRef]
  97. Sukhatme, V.P.; Reiersen, A.M.; Vayttaden, S.J.; Sukhatme, V.V. Fluvoxamine: A Review of Its Mechanism of Action and Its Role in COVID-19. Front. Pharmacol. 2021, 12, 652688. [Google Scholar] [CrossRef] [PubMed]
  98. Halpern-Cohen, V.; Blumberg, E.A. New Perspectives on Antimicrobial Agents: Maribavir. Antimicrob. Agents Chemother. 2022, 66, e0240521. [Google Scholar] [CrossRef]
  99. Whitehurst, C.B.; Sanders, M.K.; Law, M.; Wang, F.Z.; Xiong, J.; Dittmer, D.P.; Pagano, J.S. Maribavir inhibits Epstein-Barr virus transcription through the EBV protein kinase. J. Virol. 2013, 87, 5311–5315. [Google Scholar] [CrossRef] [PubMed]
  100. Lee, E.K.; Kim, S.Y.; Noh, K.W.; Joo, E.H.; Zhao, B.; Kieff, E.; Kang, M.-S. Small molecule inhibition of Epstein-Barr virus nuclear antigen-1 DNA binding activity interferes with replication and persistence of the viral genome. Antiviral Res. 2014, 104, 73–83. [Google Scholar] [CrossRef] [PubMed]
  101. Kotaki, R.; Higuchi, H.; Kotani, A. Ibrutinib As a Possible Therapeutic Strategy for Epstein-Barr Virus-Positive Lymphoma/Leukemia. Blood 2017, 130 (Suppl. S1), 1537. [Google Scholar]
  102. Weber, A.N.R. Targeting the NLRP3 Inflammasome via BTK. Front. Cell Dev. Biol. 2021, 9, 630479. [Google Scholar] [CrossRef]
  103. Xu, H.; Hutchinson, T.E.; Koganti, S.; Rousseau, B.A.; Xia, D.; McIntosh, M.T.; Bhaduri-McIntosh, S. Leveraging the interconnected unfolded protein response and NLRP3 inflammasome pathways to reactivate Epstein-Barr virus in diffuse large B-cell lymphomas. NAR Cancer 2025, 7, zcaf017. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Flowchart showing inflammatory cascade associated with EBV and SARS-CoV-2 co-infection.
Figure 1. Flowchart showing inflammatory cascade associated with EBV and SARS-CoV-2 co-infection.
Viruses 17 00903 g001
Table 1. Studies reporting EBV reactivation in patients with COVID-19.
Table 1. Studies reporting EBV reactivation in patients with COVID-19.
S. No.Type of StudyStudy PopulationStudy FindingsReference
1.Systematic
review
53 articles and 40 reactivation studies58% of COVID-19 patients presented signs of EBV reactivation[12]
2.Systematic
review
19 studies; 539 patients who were infected with both COVID-19 and HerpesviridaeEBV reactivation was frequent in
COVID-19 patients; D-dimer, C-reactive protein (CRP), length of stay in the intensive care unit (ICU), and usage of invasive mechanical ventilation were significantly associated markers
[13]
3.Retrospective
study
106 patients; 54
positive and 52
negative for COVID-19
27.1% of EBV reactivations, based on qPCR detection of EBV genomes, were from the COVID-19-positive group, while only 12.5% of reactivations belonged to the negative group[14]
4.Retrospective
study
67 COVID-19 patients with onset time within 2 weeks55.2% of patients were seropositive for EBV viral capsid antigen (VCA) IgM antibodies.
EBV/SARS-CoV-2 coinfection was associated with fever and increased inflammation
[15]
5.Retrospective
study
68 COVID-19 patients66.7% of Long COVID subjects versus 10% of control subjects were positive for EBV reactivation based on positive titers for EBV early antigen-diffuse (EA-D) IgG or EBV VCA IgM[16]
6.Retrospective
study
117 ICU patients with severe COVID-1916% of patients with severe COVID-19 developed EBV reactivation as compared to 14% in the non-COVID-19 control group[17]
7.Retrospective
study
104 COVID-19 patients, including 42
hospitalised in ICU and 62 in a sub-ICU
EBV DNA median level among ICU patients was significantly higher than that observed in SICU patients, and the B cell count was significantly increased in ICU patients[18]
8.Retrospective
study
128 COVID-19
patients
13.3% COVID-19 patients demonstrated EBV reactivation. Lymphocyte and albumin of EBV group decreased more significantly than the non-EBV group. Respiratory failure, acute respiratory distress syndrome (ARDS), and hypoproteinaemia of EBV group had more incidence than non-EBV group[19]
9.Observational retrospective cohort study120 patients with COVID-19 severe pneumonia were enrolled from ICUEBV reactivation was observed in 65% of patients, with authors claiming that viral reactivation is associated with mortality and a higher risk of developing both ventilator-associated pneumonia (VAP) and ICU bloodstream infections (BSI)[20]
10.Longitudinal study309 COVID-19 patients from initial diagnosis to convalescence (2–3 months later)EBV reactivation in COVID-19 patients at the time of COVID-19 diagnosis was positively correlated with Long COVID symptoms of fatigue and sputum production 2 to 3 months after COVID-19 onset[21]
11.Monocentric retrospective study34 COVID-19 patients from ICU were selectedEBV was detected in 82% of patients with EBV reactivation occurring early after ICU admission and was associated with longer ICU length-of-stay[22]
12.Pilot observational study88 patients with post-COVID-19 manifestations were recruitedPatients with post-COVID manifestations presented with reactivation of EBV in 42.6% of cases, with authors claiming that patients with herpes virus infections presented with more frequent fever, headache, psycho–neurological disorders, pulmonary abnormalities, myalgia, activation of liver enzymes, elevated CRP and D-dimers, and suppressed cellular immune response[23]
Table 2. Association of anti-SARS-CoV-2 drugs with EBV reactivation.
Table 2. Association of anti-SARS-CoV-2 drugs with EBV reactivation.
S. No.Anti-SARS-CoV-2 DrugsRole in EBV Reactivation
1.Remdesivir
Increases the expression of viral lytic genes—BZLF1 (immediate early gene) and BHFR1 (early gene) [27].
Reduces STAT3 but increases p38 MAPK phosphorylation from EBV+ lymphoma cells [28,29].
2.Azithromycin and nafamostat mesylate
Significantly increase viral lytic gene expression and virion production via the activation of MAPK and NF-kB signalling pathways [26].
3.Chloroquine diphosphate
Triggers EBV replication through phosphorylation of Kruppel-associated and Box-associated protein 1/tripartite motif-containing protein 28 (KAP1/TRIM28) in Burkitt lymphoma cells [30].
4.Dexamethasone
Leads to dose-dependent upregulation of immediate early gene BZLF1 mRNA expression that encodes the lytic transactivator protein ZEBRA [31].
Table 3. Role of interactive host proteins linked to their respective SARS-CoV-2 proteins in EBV reactivation.
Table 3. Role of interactive host proteins linked to their respective SARS-CoV-2 proteins in EBV reactivation.
S. No.SARS-CoV-2
Protein
Interactive Host Proteins Associated with EBV ReactivationRole in EBV Reactivation
1.EBRD4; BRD2
  • Activate EBV enhancer and promoter function to modulate gene expression.
  • JQ1 blocks BZLF1 protein production and prevents downstream transcription of lytic genes [37].
2.NUPF1; DDX21
  • UPF1 knockdown upregulates the expression of EBV lytic genes BZLF1, BMRF1, BLLF1, and BcLF1 and elevates lytic proteins ZEBRA and EA-R [38].
  • DDX21 inhibits EBV lytic replication by interacting with EBV protein SM [39].
3.MANO2; FAKD5; MPPA
  • Anoctamin 2 (ANO2) exhibits molecular mimicry with EBV nuclear antigen 1 and increases the risk of multiple sclerosis [40].
  • FAKD5 and MPPA affect the remodelling of EBV-infected B cells [41].
5.NSP2RAP1GDS1
  • Undergoes downregulation in the presence of EBNA1, which causes latent viral replication in B cells [42].
6.NSP4IDE
  • Major protein modulated by EBV protein deoxyuridine triphosphate nucleotidohydrolase (dUTPase), which is associated with neuroinflammation observed in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) [43].
7.NSP5HDAC2
  • Involved in histone deacetylase inhibitor (HDACi)-induced EBV reactivation [44].
9.NSP7PGES2
  • PGES2 produces PGE2, which is a principle COX-2-regulated downstream product. Upregulation of COX-2 modulates EBV lytic reactivation through its downstream effector PGE2 [45].
10.NSP8EXOSC3
  • Included among the major genes modulated by EBV-encoded dUTPase in human dendritic cells [46].
11.NSP9EIF4H
  • LMP1 stimulates the transcription of eIF4E via c-Myc to promote EBV-mediated nasopharyngeal carcinoma [47].
12.NSP12RIPK1; TCF12
  • LMP1 can modulate the post-translational modification of receptor-integrating protein kinase 1 (RIPK1) and LMP1-mediated K63-polyubiquitination of RIPK1, which can induce a switch from necroptotic death to survival [48].
  • LMP2 promoter binds to TCF12, blocking reactivation of EBV [49].
13.ORF3aSUN2
  • SUN2 is a nuclear envelope-associated protein that is modified by BCLF4 protein during EBV reactivation and is required for further EBV replication [50].
14.ORF10CUL2
  • CUL2 forms complexes with EBV ZEBRA proteins and induces p53 degradation for promoting EBV propagation [51].
Table 4. Novel therapeutics for EBV and their efficacy against SARS-CoV-2.
Table 4. Novel therapeutics for EBV and their efficacy against SARS-CoV-2.
S. No.Novel TherapeuticsComments
1.Nucleoside Analogues (acyclovir, valacyclovir, ganciclovir, and valganciclovir)
Studies have shown that acyclovir and ganciclovir inhibit EBV in vitro [78,79].
Meng M. et al. showed that ganciclovir-treated patients had an improved survival rate in comparison to patients with COVID-19 who did not receive ganciclovir therapy [80].
German E.R. et al. showed that acyclovir resolved the neurological symptoms of COVID-19 patients and lowered their IgG and IgM titers, supporting the use of acyclovir for COVID-19 neurologic symptoms [81].
2.Nucleotide Analogues (cidofovir)
Successful treatment of recurrent EBV-associated nasopharyngeal carcinoma with cidofovir was reported in two patients [82].
Cidofovir enhanced radiation-induced apoptosis and radiosensitivity in EBV-related malignancies (Burkitt’s lymphoma and nasopharyngeal carcinoma) [83].
In a study by Rehman et al., an ADMET (Absorption, Distribution, Metabolism, Excretion, and Toxicity) analysis revealed that cidofovir is well metabolised, distributed, and bioavailable, but has some undesirable effects [84].
3.Pyrophosphate Analogues
(foscarnet)
Foscarnet in combination with immunoglobulins was successful in controlling persistent EBV infection in a lung transplant patient that showed clinical improvement of Post-Transplant Lymphoproliferative Disorder (PTLD) following a reduction in immunosuppression intensity [85].
Schneider U. et al. reported that two AIDS patients with EBV-associated lymphoproliferative disorders who did not respond to standard chemotherapy achieved complete tumour regression after therapy with foscarnet [86].
Anti-EBV Compounds Under Investigation
a.Fluvoxamine
Considering the interaction of XBP1 and sigma-1 receptor in the ER, it is likely that sigma-1 receptor agonists (i.e., fluvoxamine), which may block EBV reactivation, would be potential therapeutic drugs to limit clinical deterioration after infection and Long COVID symptoms [87,88].
b.Maribavir (MBV)
Oral benzimidazole L-riboside with significant activity against both Human Cytomegalovirus (HCMV) and EBV but no other human herpesviruses [89].
MBV has been shown to inhibit the phosphorylation of the EBV DNA polymerase processivity factor BMRF1 [63] and has a unique dual effect against the EBV inhibition of viral DNA replication and virus transcription [90].
c.KAY-2-41 and KAH-39-149
These two thionucleoside derivatives were effective in vitro anti-EBV activity [79].
d.Brincidofovir (CMX-001)
This alkoxyalkyl ester prodrug of cidofovir has the same in vitro broad-spectrum antiviral activity as cidofovir but with an activity up to 1000-fold higher compared with cidofovir due to higher intracellular levels of cidofovir-diphosphate [91].
e.Inhibitors of EBV Nuclear Antigen 1 (EBNA1)
The EBV-encoded nuclear antigen 1 (EBNA1) is a versatile protein useful in the maintenance, replication, and segregation of EBV genome and represents an attractive therapeutic target to treat EBV-associated malignancies [92].
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Mahajan, S.; Mahajan, S.; Patgiri, S. Association and Interaction of Epstein–Barr Virus with SARS-CoV-2 Infection—A Review. Viruses 2025, 17, 903. https://doi.org/10.3390/v17070903

AMA Style

Mahajan S, Mahajan S, Patgiri S. Association and Interaction of Epstein–Barr Virus with SARS-CoV-2 Infection—A Review. Viruses. 2025; 17(7):903. https://doi.org/10.3390/v17070903

Chicago/Turabian Style

Mahajan, Supriya, Saurabh Mahajan, and Sayashree Patgiri. 2025. "Association and Interaction of Epstein–Barr Virus with SARS-CoV-2 Infection—A Review" Viruses 17, no. 7: 903. https://doi.org/10.3390/v17070903

APA Style

Mahajan, S., Mahajan, S., & Patgiri, S. (2025). Association and Interaction of Epstein–Barr Virus with SARS-CoV-2 Infection—A Review. Viruses, 17(7), 903. https://doi.org/10.3390/v17070903

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