Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Infection, Genetics and Evolution 112 (2023) 105443

Contents lists available at ScienceDirect

Infection, Genetics and Evolution


journal homepage: www.elsevier.com/locate/meegid

T cell-mediated immunity during Epstein–Barr virus infections in children


Mengjia Liu a, b, Ran Wang a, b, *, Zhengde Xie a, b, *
a
Beijing Key Laboratory of Pediatric Respiratory Infectious Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research
Center for Respiratory Diseases, Laboratory of Infection and Virology, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University,
National Center for Children’s Health, Beijing 100045, China
b
Research Unit of Critical Infection in Children, 2019RU016, Chinese Academy of Medical Sciences, Beijing 100045, China

A R T I C L E I N F O A B S T R A C T

Keywords: Epstein–Barr virus (EBV) infection is extremely common worldwide, with approximately 90% of adults testing
Epstein–Barr virus (EBV) positive for EBV antibodies. Human are susceptible to EBV infection, and primary EBV infection typically occurs
Cellular immunity early in life. EBV infection can cause infectious mononucleosis (IM) as well as some severe non-neoplastic dis­
Infectious mononucleosis (IM)
eases, such as chronic active EBV infection (CAEBV) and EBV-associated hemophagocytic lymphohistiocytosis
Chronic active EBV infection (CAEBV)
EBV-associated hemophagocytic
(EBV-HLH), which can have a heavy disease burden. After primary EBV infection, individuals develop robust
lymphohistiocytosis (EBV-HLH) EBV-specific T cell immune responses, with EBV-specific CD8+ and part of CD4+ T cells functioning as cytotoxic
Vaccine T cells, defending against virus. Different proteins expressed during EBV’s lytic replication and latent prolifer­
ation can cause varying degrees of cellular immune responses. Strong T cell immunity plays a key role in con­
trolling infection by decreasing viral load and eliminating infected cells. However, the virus persists as latent
infection in EBV healthy carriers even with robust T cell immune response. When reactivated, it undergoes lytic
replication and then transmits virions to a new host. Currently, the relationship between the pathogenesis of
lymphoproliferative diseases and the adaptive immune system is still not fully clarified and needs to be explored
in the future. Investigating the T cell immune responses evoked by EBV and utilizing this knowledge to design
promising prophylactic vaccines are urgent issues for future research due to the importance of T cell immunity.

1. Introduction lymphoproliferative disorders (PTLD) (Singavi et al., 2015; Dharnid­


harka et al., 2016; Subspecialty Group of Infectious Diseases and Na­
Epstein–Barr virus (EBV) is a ubiquitous and successful virus that tional Children’s Epstein-Barr Virus Infection Cooperative, 2021). EBV
infects a large portion of the population. It is also recognized as the first infection triggers a strong T cell-mediated immunity that plays a crucial
virus associated with tumors. The primary mode of transmission for EBV role in the control of infection and maintenance of virus-host balance.
is through saliva, although it can also spread through blood transfusion The disease burden caused by EBV infection is significant, as it can
and organ transplantations. Following initial infection, individuals may impact both physical and mental health of children. However, prophy­
experience either asymptomatic infection or infectious mononucleosis lactic and therapeutic T cell-mediated vaccines against EBV are still
(IM) (Dunmire et al., 2015; Dunmire et al., 2018). After recovering from being researched. By exploring the role of T cell immunity in infection
primary EBV infection, most individuals enter into a host-virus balance control and its relationship with EBV protein, we can better understand
state and become healthy carriers. However, a small minority of patients the progression of EBV infection and develop more effective immuno­
may experience chronic active EBV infection (CAEBV), EBV-associated logical methods. In this review, we summarize T cell immunity in
hemophagocytic lymphohistiocytosis (EBV-HLH), diffuse large B cell common EBV -associated diseases in children, including IM, CAEBV and
lymphoma, Hodgkin’s lymphoma (HL), and other EBV-related NK/T EBV-HLH.
lymphoproliferation-related diseases (Kimura et al., 2012; Fujiwara and
Nakamura, 2020). Additionally, EBV-seronegative recipients who 1.1. Epstein–Barr virus genome
receive organs from EBV- seropositive donors are at a high risk of
developing post-transplant lymphoma and related post-transplant EBV was discovered in 1964 by Michael A. Epstein and Yvonne Barr

* Corresponding authors at: Corresponding authors at: Beijing Children’s Hospital, Capital Medical University, Nan Li Shi Rd 56#, Xi Cheng District, Beijing
100045, China.
E-mail addresses: randall@mail.ccmu.edu.cn (R. Wang), xiezhengde@bch.com.cn (Z. Xie).

https://doi.org/10.1016/j.meegid.2023.105443
Received 31 January 2023; Received in revised form 25 April 2023; Accepted 15 May 2023
Available online 16 May 2023
1567-1348/© 2023 Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
M. Liu et al. Infection, Genetics and Evolution 112 (2023) 105443

in a cultured lymphoblast line derived from an African Burkitt lym­ into a latent infection state. Infected B cells then move back to the
phoma patient through electron microscopy (Epstein et al., 1964; Liu oropharynx, occur lytic replication, and circulate in the oropharynx (Bu
et al., 2020). EBV belongs to the family Herpesviridae, and is one of the et al., 2019). The process of EBV entering host cells requires the syner­
eight known herpesviruses. It is a human lymphotropic herpesvirus that gistic action of numerous glycoproteins (Fig. 1) (Connolly et al., 2021).
has been named human γ herpesvirus and is classified under the genus Viral attachment occurs by binding the viral envelope glycoprotein
Lymphocryptovirus (Dunmire et al., 2015). EBV particles have a charac­ gp350 to the C3d complement receptor CR2 (CD21), which expresses on
teristic three-layer configuration like other herpes viruses and are the surface of naïve B lymphocytes. On the surface of epithelial cells
around 120 to 180 nm in diameter (Cohen, 2000; Odumade et al., 2011; without CD21 expression, viral BMRF2 protein binds to integrin and
Li et al., 2020). The viral double-stranded DNA genome encodes various triggers endocytosis of particles (Cohen, 2000; Molesworth et al., 2000;
proteins necessary for viral DNA replication and the formation of viral Borza and Hutt-Fletcher, 2002; Chen et al., 2018). The penetration and
structure (Cruchley et al., 1997; Cohen, 2000; Farrell, 2019). It’s fusion of the virus particles with the host cell membrane require
approximately 172 kb in length and enclosed by the pseudo-icosahedral glycoprotein heterodimer complex gH-gL, gp42, and viral fusion pro­
nucleocapsid. The material outside the nucleocapsid is proteinaceous teins gB (Wang et al., 1998). During the process of EBV infecting
tegument, which contains various viral proteins. The outermost layer of epithelial cells, the heterodimer gH-gL binds to integrin αVβ5, αVβ6, or
the proteinaceous tegument is the lipid bilayer membrane containing αVβ8 (Chesnokova and Hutt-Fletcher, 2011) and then subsequently
glycoproteins like gp350, gB, gH, gL, and gp42. These glycoproteins are binds to Ephrin type A receptor 2 (EphA2) receptors, resulting in the
responsible for host recognition and the fusion process between the virus activation of the fusion protein gB and promoting the process of virus-
and host membrane (Odumade et al., 2011; Li et al., 2020). Currently, cell membrane fusion (Chen et al., 2018; Bu et al., 2019; Connolly
there are 130 published EBV genome sequence using EBV genome et al., 2021). During EBV infection of B cells, the heterodimer gH-gL
sequencing technology (Correia et al., 2017). Based on the differences in forms a complex with gp42 glycoprotein. Human Leukocyte Antigen
the EBV nuclear antigen (EBNA) loci between EBNA2 and EBNA3, EBV is (HLA)-II molecules, as cofactors for infection of B lymphocytes, bind to
isolated into type 1 and type 2. EBV-1 is the dominant type worldwide, gp42 glycoprotein, and subsequently induce fusion protein gB to
while EBV-2 has a high prevalence in the New Guinea population and mediate the process by which the virion enters the host cell (Li et al.,
sub-Saharan Africa (Odumade et al., 2011; Farrell, 2015; Correia et al., 1997; Molesworth et al., 2000; Borza and Hutt-Fletcher, 2002).
2017).
1.3. Life cycle of EBV
1.2. The process of Epstein–Barr virus entering host cells
Infected B cells undergo continuous latent proliferation in the
Primary EBV infection generally occurs by coming into contact with germinal centers of the tonsils (Dunmire et al., 2018). The EBV releases
oral secretions from EBV seropositive individuals. The primary target the nucleocapsid into the cytoplasm via vesicles. After the dissolution of
cells for EBV are epithelial cells and B lymphocytes (Cruchley et al., the nucleocapsid, the viral genome is transported to the nucleus through
1997; Balfour Jr. et al., 2015). In the oropharyngeal epithelial cells, the nuclear pore complexes (Lee and Chen, 2021). The expression products
viruses amplified through lytic replication and then infects B cells. EBV of EBV’s latency genes play an essential role in the maintaining latent
can directly infects resting B cells in tonsil crypts, transforming them infections of EBV-infected B cells. These gene products include EBV-

Fig. 1. The process of EBV entering host cells. MHC, major histocompatibility complex; EphA2, Ephrin type A receptor 2.

2
M. Liu et al. Infection, Genetics and Evolution 112 (2023) 105443

encoded microRNAs (miRNAs), BamHI A-encoded transcripts (BARTs), activation, the immediate early trans-activator Zta (BZLF1) and Rta
EBV-encoded small RNAs (EBER1 and EBER2), six Epstein–Barr nuclear (BRLF1) genes are preferentially expressed. The genes are transcribed
antigen proteins (EBNA1, EBNA2, EBNA3A, EBNA3B, EBNA3C, and and cohesively bind to the origin of lytic replication to form the core
EBNA-LP) and three latent membrane proteins (LMP-1, LMP-2A, and replication complex (Murata, 2014; Zhang et al., 2017). Consequently,
LMP-2B) (Li et al., 1997; Wang and Hutt-Fletcher, 1998; Tugizov et al., lytic virus genes are orderly expressed, producing proteins that are
2003; Tsurumi et al., 2005; Tsao et al., 2012; Dunmire et al., 2018). EBV involved in the formation of daughter virus particles. Early genes are
latency patterns can be classified into four stages (Latency stage 0-III) expressed to produce over 30 early proteins, such as BMLF1, BHRF1,
(Fig. 2) (Bailey, 1994; Tsuchiya, 2002; Odumade et al., 2011). In la­ BALF1, and DNase BGLF5, all of which are essential in the replication of
tency stage III, all latency genes are expressed, including EBNA1, the EBV genome (McKenzie and El-Guindy, 2015). The episomal form
EBNA2, EBNA3A, EBNA3B, EBNA3C, LMP-1, LMP-2, EBER, miRNAs, genome is linearly activated as viral genome replication takes place.
and BARTs. These genes can activate naïve B cells, promote prolifera­ Finally, the late genes are expressed, producing late viral protein capsid
tion, and contribute to the virus transformation. Because EBNA3s are protein VCA/gp350/ gp 220, and among others, which take part in the
dominate targets of CD8+ T cell response, a part of the infected cells is formation of daughter viral structural proteins. Once complete viral
eliminated by cytotoxic T lymphocytes (CTLs) in latency stage III. This particles are formed, they are excreted through exocytosis (Tsurumi
pattern is associated with IM and lymphoproliferative diseases (Cohen, et al., 2005; Ressing et al., 2015; Lee and Chen, 2021).
2000; Speck and Ganem, 2010). In latency stage II, I, and 0, restricted
latency genes are expressed in EBV-infected cells, allowing them to 1.4. Pathogenicity of EBV
establish latency infections and evade CD8+ CTLs (Kimura et al., 2008).
In latency stage II, the programmed expression of EBNA1, LMP-1, LMP- After primary infection with EBV, individuals may develop asymp­
2, EBER, and BARTs can induce the B cells differentiation and establish tomatic infections or present with clinical manifestations of IM. In the
the phenotype of memory B cells (Speck and Ganem, 2010; Odumade developed country, primary EBV infection usually causes IM in adoles­
et al., 2011). This pattern is characteristic of HL, epithelial tumor cents and young adults, while in infants and younger children, it is
nasopharyngeal carcinoma (NPC), and CAEBV. In latency stage I, usually asymptomatic or presents with non-specific symptoms (Dunmire
EBNA1/EBER and BARTs are expressed, which can facilitate B cell et al., 2018). Conversely, in developing countries such as China, peak
survival and can be found in Burkitt lymphoma (Yin et al., 2003; Herbert incidence of IM was found in preschoolers, particularly in the 4–6 years
and Pimienta, 2016). In latency stage 0, only EBER and BARTs are old age group (Hu et al., 2021; Liu et al., 2022). IM is characterized by a
expressed, and during this phase, EBV memory B cells are thought to be long incubation period before the onset of clinical manifestations
silent. Individual in this stage is healthy carrier in a virus-host balance (Dunmire et al., 2015). In children, typical clinical manifestations of IM
state (Yin et al., 2003; Minarovits, 2006). include pharyngitis, fever, and lymphadenopathy, sometimes accom­
Under the inducement of reactivation signals, some infected resting panied by liver dysfunction, hepatosplenomegaly, and edema of eyelids.
B cells and memory B cells undergo viral reactivation. This results in the Children with IM usually have IgM and low-affinity IgG antibodies
lytic replication of infected cells and the generation of progeny virus against viral capsid antigen (VCA), or only low-affinity IgG antibodies
particles, as shown in Fig. 2 (Price and Luftig, 2014). Following virus against VCA. Several months later, IgG antibodies against EBNA and

Fig. 2. Life cycle of EBV and latency types in EBV-associated diseases. EBV, Epstein–Barr virus; EBNA, Epstein–Barr nuclear antigen; miRNAs,EBV-encoded
microRNAs; BARTs, BamHI A-encoded transcripts; LMP, latent membrane proteins; IM, infectious mononucleosis; PTLD, post-transplant lymphoproliferative dis­
eases; CAEBV, chronic active EBV infection.

3
M. Liu et al. Infection, Genetics and Evolution 112 (2023) 105443

high-affinity IgG antibodies against VCA become detectable, which is infection. EBV-specific CD4+ T cells identified as helper T cells partici­
typical of serological tests in healthy carriers of EBV (Subspecialty pate in high-affinity antibody production, initiate and maintain CTL
Group of Infectious Diseases and National Children’s Epstein-Barr Virus function and are highly activated, producing cytotoxic substance such as
Infection Cooperative, 2021). There is no specific standard for the Granzyme B and perforin, to control EBV infection (Landais et al., 2004;
treatment of IM, which is mainly symptomatic and supportive (Womack Martorelli et al., 2010; Meckiff et al., 2019; Dowell et al., 2021).
and Jimenez, 2015). Most cases of IM have a good prognosis, while some EBV-specific memory T cells act as immune surveillance to maintain the
patients may develop complications such as encephalitis, myocarditis, virus-host balance in the infected host. Understanding T cell-mediated
or even EBV-HLH (Jenson, 2000; Allen and McClain, 2015; Yao et al., cellular immunity during EBV infection will contribute to providing
2021; Liu et al., 2022). EBV-HLH can occur during different states of data support for the development of EBV vaccine.
EBV infection and is essentially organ damage caused by excessive
production of inflammatory cytokines (Imashuku et al., 2021). Patients 2.1. Asymptomatic infection
with EBV-HLH may present with persistent high fever, hep­
atosplenomegaly, lymph node enlargement, anemia, and jaundice It is well-known that primary EBV infection in infants and toddlers is
(Subspecialty Group of Infectious Diseases and National Children’s almost always asymptomatic (Silins et al., 2001), though asymptomatic
Epstein-Barr Virus Infection Cooperative, 2021; El-Mallawany et al., individuals are occasionally discovered through EBV-specific antibody
2022). The main treatments for EBV-HLH are symptomatic treatment, spectrum contemporary assays. This makes it challenging for clinicians
chemotherapy, and hematopoietic stem cell transplantation (HSCT); to identify asymptomatic EBV infections in a timely manner (Subspe­
however, the prognosis of EBV-HLH is poor, and mortality rates are high cialty Group of Infectious Diseases and National Children’s Epstein-Barr
(Yanagaisawa et al., 2019). Virus Infection Cooperative, 2021). Consequently, there are a limited
In addition, some patients exhibit persistent or recurring IM-like number of studies on asymptomatic infection. Silins et al. reported that
symptoms for over three months (Kimura and Cohen, 2017; Shafiee individuals with asymptomatic infection maintain a diverse T cell re­
et al., 2022), which is referred as a lymphoproliferative disorder called ceptor (TCR) repertoire. Interestingly, the TCR repertoires remain stable
CAEBV. The clinical manifestations of CAEBV are diverse including throughout the process of disease recovery without amplification or
fever, hepatosplenomegaly, liver dysfunction, lymphadenopathy, narrowing (Silins et al., 2001). Jayasooriya et al. found that asymp­
thrombocytopenia, and hypersensitivity to mosquito bites, excluding tomatic African children aged 14–18 months had higher levels of plasma
known immunodeficiency diseases (Arai, 2021). Depending on the EBV-DNA load, similar to IM, which decreased over time. Moreover,
infected cell types of clonal proliferation, CAEBV can be divided into EBV-specific CD8+ T cells were activated, and the response to one in­
NK/T cell type and B cell type. The T cell type CAEBV can be further dividual epitope accounted for roughly 15% of the total CD8+ T cells.
divided into CD4+ T cell type, CD8+ T cell type, γδ T cell type, and other Highly activated EBV-specific CD8+ T cells express activation markers
T cell types (Kimura and Cohen, 2017). CAEBV characterized by EBV- HLA-DR and CD38. Strikingly, despite observing such changes, the study
infected NK/T cell clone proliferation is more common in children in found no significant expansion of CD8+ T lymphocytes and CD8+ T
Asia, particularly in East Asia, while EBV-infected B-cell-related CAEBV compartment (Fig. 3A), which is different from what is usually seen in
is more prevalent in western country (Kimura et al., 2012; Bollard and children with primary EBV infection with clinical manifestations
Cohen, 2018). The etiology of CAEBV is not clearly understood (Fuji­ (Jayasooriya et al., 2015). Similarly, Abbott et al. reported that
wara and Nakamura, 2020). Additionally, there is still no satisfactory asymptomatic adolescents displayed the same level of EBV-DNA as IM
therapy for CAEBV, and the most effective treatment is HSCT (Subspe­ patients but a lower magnitude cell-mediated immune response (Abbott
cialty Group of Infectious Diseases and National Children’s Epstein-Barr et al., 2017).Tamaki et al. found that CTL lines from children aged
Virus Infection Cooperative, 2021). 20–35 months recognized at least one EBV latent gene expression
products, with almost all the children showing CTLs response against
2. T cell-mediated immune responses during EBV infection EBNA3s (EBNA3A, 3B, 3C), of which EBNA3C was observed most
frequently (Tamaki et al., 1995). These results suggest that asymptom­
After primary infection with EBV, both humoral and cellular immune atic EBV infection in children causes high viral load with a decreasing
responses are triggered in the infected individual. B cells produce anti­ process while inducing an EBV-specific CD8+ T cells response that can
bodies to participate in controlling EBV infection. IgG antibody titers control EBV infection without excessive amplification of CD8+ T cells.
against lytic cycle antigens such as the VCA complex peak after primary This provides support for the notion that IM is an immunopathological
EBV infection and subsequently decline to stable levels. The production disease. Lam et al. observed an increase in PBMC and plasma viral load at
of antibodies by B cells requires T helper cells. Antigen-specific helper T the same level in asymptomatic and IM children. During the convales­
cells interact with antigen-specific B cells via the “antigen-bridge” and cent phase, both PBMC and plasma viral loads continued to decrease.
eventually produce specific antibodies that are involved in controlling Furthermore, both latent and lytic antigen-specific CD4+ and CD8+ T
infection (Lanzavecchia, 1985; Tomita et al., 1998; Zhong et al., 2022). cells displayed polyfunctionality (Lam et al., 2018). Detection of EBV-
During primary EBV infection, EBV usually infects naïve B cells and specific CD8+ T cells markers found that the expression of the cell
leads to latent infection (Shafiee et al., 2022). These latent infected B proliferation marker Ki-67 was increased and anti-apoptotic protein Bcl-
cells induce T cell immunity, which plays a critical role in maintaining 2 was decreased, indicating that the cells were in the sensitive phase of
the balance between the host immune system and virus. After initial proliferation and apoptosis (Jayasooriya et al., 2015; Abbott et al.,
exposure to EBV, robust EBV-specific T cell immune responses are 2017).
induced in the host, and these responses decrease in magnitude over
time, forming memory immunity that persists throughout life of the 2.2. Infectious mononucleosis
host. CD8+ and CD4+ T cells specifically target infected cells that express
a wide range of EBV proteins during the latent and lytic stages of viral 2.2.1. Relation between viral load and T cell-mediated immune response
infection. CD8+ T cells exert effector function of cytotoxicity and control Primary EBV infection typically presents as IM in older children,
virus infection by recognizing the short peptide derived from the including elementary and adolescent students (Subspecialty Group of
endocellular breakdown of EBV protein and combining with major Infectious Diseases and National Children’s Epstein-Barr Virus Infection
histocompatibility complex (MHC)-I molecular on the infected cell Cooperative, 2021). To investigate the cellular immune response to
surface. CD4+ T cells that bind to MHC-II molecules expressed on the cell primary EBV infection, researchers typically examine the peripheral
surface can be categorized as helper T lymphocytes (Th), CTLs, and blood of IM patients. The period between viral infection and the onset of
regulatory T cells (Treg), which function in the immune response to viral clinical symptoms is approximately 42 days, and infection may have

4
M. Liu et al. Infection, Genetics and Evolution 112 (2023) 105443

Fig. 3. T cell-mediated immune response in EBV-associated diseases. T cell-mediated immune response in the asymptomatic, IM and healthy carriers (A), CAEBV (B),
EBV-HLH (C). IM, infectious mononucleosis; CAEBV, chronic active EBV infection; EBV-HLH, EBV-associated hemophagocytic lymphohistiocytosis.

developed 3–6 weeks prior to sample collection (Dunmire et al., 2018). Both CD4+ and CD8+ T cells are involved in the control of viral load in
It is difficult to capture immune response samples during the incubation vivo, and T cell immune responses play an important function in the
period. control of infection.
During the acute phase of IM, there is a high level of EBV-DNA load The oropharyngeal viral load is higher during the acute period of IM,
which decreased rapidly during the convalescent phase. This decrease is after while it decreases slightly after this phase, it remains high for many
consistent with the decrease in the ratio of EBV-specific CD8+ T cells months following IM (Hislop et al., 2005). The viral load is inversely
participating in the elimination of EBV (Fig. 3A) (Hoshino et al., 2011). correlated with the number of EBV-specific CD8+ T cells expressing T-
CD8+ T cells in children with IM expressed inhibitory receptors and bet, which perhaps participate in the control of infection (Barros et al.,
differentiation markers, and an increased EBV viral load appears to be 2019).
related to increased frequencies of PD-1 and KLRG1 positive CD8+ T
cells (Chatterjee et al., 2019). 2.2.2. Immunophenotype of T cell subsets
Antigen-driven EBV-specific CD4+ T cell responses decrease in par­
allel with viral load (Precopio et al., 2003), and the ratio of circulating 2.2.2.1. CD8+ T cells. IM is an immunopathological disease in which an
follicular help T cells (cTfh) to circulating follicular regulatory T cells increased proportion and number of lymphocytes and atypical lym­
(cTfr) is positively correlated with EBV-DNA load (Qian et al., 2020). phocytes are detected in peripheral blood. Further studies have shown

5
M. Liu et al. Infection, Genetics and Evolution 112 (2023) 105443

that the increased lymphocytes are confirmed to be high expansion of stage of EBV infection for an extended period (Amyes et al., 2003).
CD8+ T lymphocytes, and the atypical lymphocytes are activated CD8+ Primary EBV infection results in oligoclonal populations of cytotoxic
T lymphocytes (Dunmire et al., 2015). The largely amplified CD8+ T EBV-specific CD4+ T cells, that secrete Granzyme B and perforin in
cells are associated with the clinical manifestations of IM (Jayasooriya response to direct in vitro to challenge with EBV-infected B cells (Meckiff
et al., 2015). Additionally, the high expansion of CD8+ T cells is due to et al., 2019; Tamura et al., 2022). During convalescence, the number of
antigen-driven responses, and these CD8+ T cells are mostly EBV- cytotoxic EBV-specific CD4+ T cells gradually decreases to undetectable
specific (Callan et al., 1998; Hoshino et al., 1999). Highly expanded levels (Appay et al., 2002; Takeuchi and Saito, 2017; Meckiff et al.,
CD8+ T cells can serve as an important indicator of assisted diagnosis of 2019). Activated EBV-specific CD4+ T cells produce one or more of IL-2,
IM based on the above characteristics. Flow cytometry with HLA-I TNF-α, IFN-γ, CD107a, and cytokine polyfunctionality, contributing to
tetramer staining makes it possible to study the phenotype of highly the decrease of EBV-DNA load in the blood to control infection (Lam
expanded CD8+ T cells (Silins et al., 2001). In children with IM, the et al., 2018). Meckiff et al. reported that T-bet expression was common
percentages of naïve CD8+ T cells (CCR7+CD45RA+) and effector CD8+ among EBV-specific CD4+ T cells in IM patients and significantly higher
T cells (CCR7− CD45RA+) are decreased in peripheral blood lymphocyte than in EBV-specific memory CD4+ T cells in healthy EBV carriers
subsets, accompanied by an increase in the percentage of CD8+ T (Meckiff et al., 2019). In the tonsil microenvironment, Th2-like
effector memory cells (CCR7− CD45RA− ) and CD8+ T central memory (CD4+CMAF+) CD4+ T cells constitute the most significant subset, fol­
cells (CCR7+CD45RA− ) (Wang et al., 2021). Chatterjee et al. detected lowed by Tregs (CD4+FoxP3+) and Th1-like (CD4+Tbet+) cells (Chai
expansion of PD-1 positive CD8+ T cells and high-frequency expression et al., 2011; Barros et al., 2019). The proportion and number of Tregs
of TIM-3, 2B4, and KLRG1 in children with IM. Blocking the PD-1 axis (CD4+FoxP3+) remain stable, and Th1-like (CD4+Tbet+) cells retain
could compromise EBV-specific immune control (Chatterjee et al., cytotoxicity for a lifetime following primary EBV infection (Wingate
2019), suggesting that PD-1 expression marks functional CD8+ T cells et al., 2009; Barros et al., 2019).
from the IM phase to convalescence.
The co-expression of CD38 and HLA-DR on the surface of EBV- 2.2.3. Lytic and latent antigens targeted by T cell immune responses
specific CD8+ T cells indicates an activated state, with an increased
ratio of TCR-αβ and TCR-γδ suggesting their function as CTLs. The 2.2.3.1. CD8+ T cells. During the acute phase of IM, EBV-specific CD8+
activated cells are primarily of a classical CD8+CD27+HLA-DR+ T cell responses are directed toward lytic proteins (Callan et al., 1998;
immunophenotype (Callan et al., 1998; Fedyanina et al., 2021; Wang Adem et al., 2002). Steven et al. screened antigenic epitopes from pe­
et al., 2021). While some EBV-specific CD8+ T cells express the CD45RO ripheral blood mononuclear cells (PBMCs) of IM patients and found that
isoform, markers for homing molecule CCR7 and CD62L are rare (Cat­ individual responses to the lytic antigens mainly focus on the epitopes of
alina et al., 2002). Furthermore, EBV-specific CD8+ T cells demonstrate the BZLF1/BRLF1, as well as BMLF1 and BMRF1 induced by BZLF1/
a lower expression of the anti-apoptotic protein Bcl-2, indicating high BRLF1 (Steven et al., 1997). This conclusion was consistent with other
apoptotic sensitivity in vivo (Soares et al., 2004). The presence of the cell studies which found that EBV-specific CD8+ T cells are more responsive
surface degranulation marker CD107a, as a surrogate for cytotoxicity, is to BZLF1 and BMLF1 epitopes (Precopio et al., 2003). CD8+ T cell re­
predominantly observed in CD8+ T cells during IM and involved in sponses to the late lytic antigens are subdominant and at low levels, and
infection control (Lam et al., 2018; Chatterjee et al., 2019). Interest­ they are rarely detected during primary EBV infection (Abbott et al.,
ingly, while CD8+ T cells undergo substantial expansion of in peripheral 2013). However, BMLF1-specific CD8+ T cell clones could recognize and
blood during EBV infection, a predomination of expended CD8+ T cell is eliminate EBV-lytically replicating autologous lymphoblastoid cell lines
not observed in tonsil, probably due to their inability to enter B-cell (LCLs) in vitro, demonstrating the effector function of cytotoxicity of
lymphoid follicles and germinal centers to eliminate infected B cells via specific CD8+ T cell in vivo (Antsiferova et al., 2014). Furthermore,
a lack of lymphocyte homing receptors (Chen et al., 2001; Montesoro BMLF1-specific CD8+ T cells expressing PD-1 and Ki-67 were observed,
et al., 2006; MacArthur et al., 2007). In tonsil, EBV-specific CD8+ T cells suggesting that despite the presence of inhibitory receptors, CD8+ T cells
show less differentiation and a higher expression level of co-stimulatory can still proliferate vigorously (Chatterjee et al., 2019). Brooks et al.
molecule CD28 than those in peripheral blood (Soares et al., 2004). reported that BHRF1, as the “first wave antigens” expressed after EBV
Additionally, the number of CD8+ T cells expressing cytotoxic markers infection of B cells, induced strong T cell imunity in some individuals,
TIA1 and Granzyme B is higher than that of CD8+ T cells expressing making it a promising candidate in the devolpoment of prophylactic
FoxP, indicating a domination of the cytotoxic signature rather than vaccines (Brooks et al., 2016). Additionally, Rühl et al. found that lytic
regulatory T cells phenotype in tonsil (Barros et al., 2019). antigen-specific CD8+ T cells have specific
PD1+TIM− 3+KLRG1+CXCR5+TCF-1+ and BATF3+ phenotypes that may
2.2.2.2. CD4+ T cells. Contrary to the extensive proliferation observed relate to their ability to control EBV-infected B cells within the germinal
in CD8+ T cells, no expansion of CD4+ T cells nor an increase in their center (Ruhl et al., 2020). To prevent CD8+ T cells from targeting EBV-
absolute numbers are observed in children with IM (Balfour Jr. et al., infected cells, the lytic genes of EBV downregulate the expression of
2013). Analysis of peripheral blood lymphocyte subsets in IM children HLA-I molecules, inhibiting antigen recognition and presentation. Early
indicates a decreased proportion of naïve CD4+ T cells protein product DNA exonuclease BGLF5 plays a crucial role in inhib­
(CCR7+CD45RA+) accompanied by an increase in the percentage of iting antigen recognition and presentation by downregulating HLA-I
CD4+ T effector memory T cells (CCR7− CD45RA− ). In addition, a molecules mRNA expression (van Gent et al., 2015).
reduced percentage of Tregs (CD4+CD25+CD127low) correlates with the Compared to the lytic-specific CD8+ T cell response, CD8+ T cells
lower cTfh (FoxP3+CXCR5+PD-1+CD4+)/cTfr (FoxP3− CXCR5+PD- exhibit a lower frequency of responding to latent infected cells (Munz,
1+CD4+) ratio in primary EBV infections than in healthy carrier 2020). CTL responses mainly target the major latent antigen epitopes
resulting from a remarkable increase in cTfr, a marker of ongoing hu­ that are derived from the EBNA3 proteins, and such epitopes show much
moral activity. This imbalance in cTfr and cTfh cells is involved in the promise in the design of an EBV prophylactic vaccine (Steven et al.,
immune control of children with IM (Qian et al., 2020). 1996). Subdominant epitopes like EBNA2 and LMP2 can also elicit a
CD38 and HLA-DR co-expression is detected on the surface of EBV- strong CD8+ T cell immune response in some populations (Chen et al.,
specific CD4+ T cells, indicating the activation. In contrast to the ratio 2009). Brooks et al. reported that EBNA2-specific CD8+ T cells can
of TCR-αβ, that of TCR-γδ is higher among EBV-specific CD4+ T cells recognize infected B cells within the first day of infection and effectively
(Wang et al., 2021). Similarly, a proportion of EBV-specific CD4+ T cells inhibit the growth of EBV-transformed B cell lines in vitro. They sug­
function as effector CD4+ T cells and can survive until the persistent gested that the EBNA2 protein may serve as a new target antigen for

6
M. Liu et al. Infection, Genetics and Evolution 112 (2023) 105443

prophylactic vaccines (Brooks et al., 2016). The EBNA1 protein has a T cell-mediated immune response (Chen et al., 2009).
glycine-alanine (Gly-Ala) repeat domain consisting solely of repeated
glycine and alanine residues, which impede the proteasome degradation
process that breaks down viral proteins into short peptides that can be 2.3. Healthy carrier
recognized by antigen-presenting cells (APC). This interference in the
presentation of the virus antigen to CD8+ T cells leads to the failure of 2.3.1. T cell-mediated immune response in EBV-host homeostasis
the EBNA1 epitope to be recognized by such T cells (Levitskaya et al., After recovering from primary EBV infection, the patients develop a
1997; Apcher et al., 2009; Farrell, 2019). The presence of this repeat virus-host balance and become long-term healthy carriers. The level of
domain guarantees a sufficient supply of EBNA1 to maintain the EBV-DNA load in plasma drops sharply during the convalescence stage
episomal form of the EBV genome in infected cells, thereby preventing and is undetectable during the healthy carrier stage. However, the level
attacks by cytotoxic cells (Valentine et al., 2010). of EBV-DNA load in PBMCs may plateau 4–6 months after infection,
declines sharply to a lower level from 6 to 12 months after infection, and
2.2.3.2. CD4+ T cells. The CD4+ T cell responses of individuals epitopes then reaches equilibrium. This phenomenon may be related to the in­
are less frequent than CD8+ T responses (Landais et al., 2004). T cell crease in the proportion of polyfunctional T cells, which can simulta­
clone analysis suggests that CD4+ T cells responses to immediate early neously secrete two or more types of cytokines to participate in infection
protein (IE), early protein (E), and late protein (L) are at the same level. control (Lam et al., 2018). Additionally, the level of EBV-DNA load in
The results of peptide stimulation in vitro indicate that frequency of the tonsil is much lower than during the acute phase (Hislop et al.,
BZLF1-specific CD4+ T cell responses increase rapidly in the acute phase 2005). In long-term healthy carriers, infected B cells are often charac­
of IM, peak, and decline in the convalescent phase (Precopio et al., 2003; terized by latent infection status in a dormant state, and activated to
Calarota et al., 2013). Dowell et al. found that structural protein BORF1- undergo lytic replication under the stimulation of certain factors. EBV is
specific CD4+ T cells express Granzyme B and perforin in IM patients, reactivated in vivo and viral load is temporarily elevated, but most in­
and the expression levels persist in healthy carriers. (Dowell et al., dividuals do not develop clinical symptoms, which is related to the
2021). During the acute stage of primary EBV infection, the response involvement of EBV-specific memory T cells in controlling the infection.
frequency of CD4+ T cells to the lytic epitope is high, and gradually
decreases in the convalescent phase. Since most of the lytic proteins are 2.3.2. Immunophenotype of T cell subsets
involved in the formation of EBV genome replication and structural
proteins into complete EBV particles, it is possible that CD4+ T cells 2.3.2.1. CD8+ T cells. HLA-I tetramer staining of memory CD8+ T cells
participate in the control of viral infection in vivo by interfering with the has shown that the resting T cells are present and can activate again
replication of virus particles. BZLF2 binds to HLA-II-peptide complexes, upon stimulation by antigen, leading to the development of CTLs that
resulting in a novel complex that contributes to the binding process of secret cytokines to control infection (Fig. 3A) (Hislop et al., 2001). EBV-
MHC-II and TCR on the surface of CD4+ Th cells. This interferes with the specific CD8+ T cells exhibit stable phenotype, functional characteris­
antigen recognition ability of Th cells and constrains cellular immunity tics, and TCR clone composition over time (Lelic et al., 2012). A subset of
(Ressing et al., 2003). latent antigen-specific CD8+ T cells that express high levels of CCR7 and
Unlike CD8+ T cells, CD4+ T cells have a lower response frequency to CD62L circulate in lymphatic tissue and upregulate expression of anti-
an individual epitope and the target epitopes that are widely distributed apoptotic protein Bcl-2, a feature of central memory T cells (Dunne
in EBNAs (Leen et al., 2001; Hislop et al., 2007). Precopio et al. noted et al., 2002).
that the response of EBV latency-specific CD4+ T cells was the highest
within three weeks of initial exposure to EBV antigen, but then rapidly 2.3.2.2. CD4+ T cells. Analysis of CD4+ T cell phenotypes suggests an
decreased (Precopio et al., 2003). During the acute stage of IM, CD4+ T equitable distribution between central memory T cells and effector
cells have a strong response of to the EBNA2 epitope, but the response to memory T cells (Long et al., 2013). Central memory T cells express
the EBNA1 epitope is substantially delayed, which parallels the late CD45RO and CD27simultaneously, while effector memory T cells ex­
emergence of EBNA1 IgG antibody (Yang et al., 2011; Long et al., 2013; press CD45RO. In patients with IM, responsive CD4+ T cells secrete IFN-
Meckiff et al., 2019). Thus, the delayed appearance of EBNA1 IgG γ, which is the dominant cytokine (Meckiff et al., 2019). However, in
antibody may be related to the immune response of CD4+ T cells. healthy carriers, EBV-specific CD4+ memory T cells secrete TNF-α, with
Meckiff et al. found that under EBNA2 peptide pool stimulation, CD4+ T decreased secretion of IFN-γ and CD107a (Lam et al., 2018), indicating
cells expressing Granzyme B and perforin were closely associated with an increase in cytokine polyfunctionality but a decrease in cytotoxic
cellular activation, and these responsive CD4+ T cells usually overex­ activity (Meckiff et al., 2019).
pressed CD38 (Meckiff et al., 2019). Lam et al. observed a dramatic in­
crease in CD4+ T cell immune response against EBNA3B from primary 2.3.3. Lytic and latent antigens targeted by T cell immune responses
EBV infection to the period of long-term carrier (Lam et al., 2018). T cell immune responses can target both lytic and latent EBV anti­
Intriguingly, the EBNA3 protein family epitope is located in a 992- gens. The distinction between the two types of antigens is significant, as
amino-acids (aa) fragment that can induce maximal proliferation of lytic antigens are expressed during active viral replication and can be
CD4+ T cells in vitro. Furthermore, 13-aa repeat region of EBNA3C targeted, while latent antigens are expressed in the absence of replica­
(aa741 to 779) was previously reported to an immunodominant target tion and are less accessible to the immune system. The interplay be­
for antibody response and contained numerous overlapping and mixed tween CD4+ and CD8+ T cells in targeting different types of EBV
CD4+ T antigen epitopes (Rajnavolgyi et al., 2000), making it a prom­ antigens remains an area of active research.
ising vaccine target (Cirac et al., 2018; Cirac et al., 2021). Regarding the
response of CD4+ T cell to latent membrane proteins, LMP1 epitopes 2.3.3.1. CD8+ T cells. As long as individuals undergo primary infection,
appear to induce a stronger immune response than LMP2 epitopes. whether they develop IM or not, EBV-specific T cell responses targeting
Marshall et al. reported that CD4+ T cells were involved in controlling lytic and latent epitopes can be detected in healthy carriers, albeit with
CTLs activities and responded to LMP1 during the acute phase of pri­ reduced frequency and specificity. The hierarchy of immunodominance
mary EBV infection, resembling a Th-1-like role; however, over time, for EBV-specific CD8+ T cells specific to EBV lytic cycle proteins is well-
they seem to switch to a regulatory IL-10-like role during convalescence established, with IE>E>L rank system maintained, which is possibly
(Marshall et al., 2007; Morales et al., 2012). Chen et al. demonstrated determined during primary EBV infection and matched in efficiency for
that Th-1 like cells predominantly dominated the LMP2A-specific CD4+ epitope recognition with CD8+ T cells (de Bleser and Poeck, 1985; Long
et al., 2011; Forrest et al., 2018). For EBV-specific CD8+ T cells specific

7
M. Liu et al. Infection, Genetics and Evolution 112 (2023) 105443

to EBV latency antigen, a certain degree of immunodominance hierarchy perforin genes that impairs the function of CTLs. As a result, these in­
is maintained, with the immune response size to latent antigens, rep­ dividuals may be unable to control chronic infections, which can prog­
resented by EBNA3A/EBNA3B/EBNA3C > LMP2 > EBNA1 > EBNA2/ ress to CAEBV (Katano et al., 2004; Collins et al., 2021). Kimura et al.
EBNA-LP > LMP1 hierarchy (Ning et al., 2011; Munz, 2020). The im­ reported that transformed lymphocytes from patients with CAEBV tested
mune response of EBV-specific CD8+ memory T cell is mainly focused on positive for EBER but negative for Granzyme B, a potent killer factor in
the latent epitopes EBNA3s, whereas the response to the lytic epitope is the immune effect of CTL-mediated granulocyte exocytosis (Kimura
slightly decreased compared with the acute IM phase (Wang et al., et al., 2012). In addition, myeloid-derived suppressor cells (MDSCs) that
2014a). develop during chronic infections can suppress host cellular immunity
and cause defective antiviral T cell function, leading to persistent
2.3.3.2. CD4+ T cells. The hierarchy of immunodominance for EBV- chronic infection (Gabrilovich and Nagaraj, 2009). Collins et al. found
specific CD4+ T cells responses to lytic epitopes is not well- that patients with CAEBV had a large number of MDSCs that suppressed
established, unlike the EBV-specific CD8+ T cells responses (Long the function of effector T cells, resulting in persistent uncontrolled EBV
et al., 2011; Munz, 2020). Dowell et al. reported that structural antigen- infection (Collins et al., 2021).
specific CD4+ T cells in primary EBV infection exhibit cytotoxic poten­ On possible reason for immune suppression is host gene mutation,
tial, an ability maintained in structural antigen-specific memory CD4+ T with CD27 or CD70 deficiency being common (Guan et al., 2021). In
cells in healthy carriers (Dowell et al., 2021). Similarly, Adhikary et al. CAEBV patients, the interaction between CD27 and CD70 is crucial as it
observed the detection of cytotoxic potential of BNRF1-specific CD4+ T induces protective EBV-specific T cell immunity and immune surveil­
cells in healthy carriers and surmise that BNRF1 may be a common lance of infected-B cells, which is due to the expansion of EBV- specific T
target of responsive CD4+ T cells (Adhikary et al., 2020). These findings cells (Izawa et al., 2017; Kruger et al., 2020). CD27 is especially vital in
imply that structural antigens may serve as target antigens in inducing the expansion of lytic EBV antigen-specific CD8+ T cells, and blocking
CD4+ T cells to rapidly eliminate virus-infected cells, thereby presenting the CD27-CD70 pathway compromises EBV-specific immune control
novel possible antigen targets and supporting data for rophylactic vac­ (Deng et al., 2021). Izawa et al. pointed out that the deficiency of CD27
cine devolopment. and CD70 leads to impaired proliferation and insufficient accumulation
Furthermore, the hierarchy of immunodominance for EBV-specific of activated T cells during EBV infection. CD70, which is expressed on
CD4+ T memory cells response differs from the EBV-specific CD8+ T the surface of B cells, provides the necessary co-stimulatory signal for
memory cells’ targeting of latent epitopes; the EBV-specific CD4+ T TCR-mediated proliferation. CD70-dependent proliferating T cells ex­
memory cells’ response presents an EBNA1 > EBNA3A/EBNA3B/ press high levels of CD27, CD25, and CD45RO but not CD70 and CD62L,
EBNA3C > LMP2 > LMP1 hierarchy (Calarota et al., 2013). EBNA1 and are able to secrete IFN-γ and TNF-α. This indicates that the preser­
epitope could elicit a strong CD4+ T cell immune response in some vation of memory T cell immunity shows that CD70-CD27 interactions
healthy carriers (Munz et al., 2000). Mullen et al. reported that CD4+ affect the proliferation of activated T cells without affecting cytotoxicity,
memory T lymphocytes targeting EBNA1 or EBNA3C epitopes appear to production of IFN-γ, and differentiation (Izawa et al., 2017).
secrete different cytokines upon in vitro antigen stimulation. EBNA1- In patients with CAEBV, pro-inflammatory cytokines are secreted by
specific CD4+ memory T cells are considered Th0-like effector T cells activated T cells (Fig. 3B), which can induce hemophagocytic syndrome.
that could simultaneously produce type I cytokines (IFN-γ) and type II TNF-α plays a dominate role in this process. Murakami et al. found that
cytokines (IL-5 and IL-13), whereas EBNA3-specific CD4+ memory T the serum TNF-α concentration was ten times higher in CAEBV patients
cells appeared to produce only type I cytokines (Steigerwald-Mullen than in the healthy carrier group. CAEBV patients also exhibited IFNGR1
et al., 2000). gene overexpression, with expression levels of IFN-γ at 3.8 times higher
than the control group. In addition, CAEBV patients showed up-
regulated IL-10, IL-2, and INHBA gene expression by about 10 times
2.4. Chronic active EBV infection higher than the control group (Murakami et al., 2014). Ohga et al.
investigated the relationship between viral load and cytokine gene
CAEBV is a lymphoproliferative disease characterized by signifi­ expression in activated T cells in CAEBV patients. Results showed that
cantly elevated levels of EBV-DNA load in the blood and invasion of activated T cells with the surface molecular marker CD3+HLA-DR+ had
organs by EBV-positive lymphocytes (Kimura and Cohen, 2017). Typi­ higher EBV-DNA copies and increased levels of IL-10, IL-2, and INF- γ
cally, the EBV-DNA load of PBMCs in CAEBV patients is higher than in than those with CD3+HLA-DR− (Ohga et al., 2001). Th1 cells secrete IL-
IM patients, and this characteristic is used for the diagnosis of CAEBV. 2, TNF-α, and INF-γ, while Th2 cells secrete IL-10. These cytokines may
According to diagnostic criteria, the diagnostic cut-off EBV-DNA load for be produced by activated T cells in CAEBV, contributing to the induction
CAEBV approximately is 102.5 copies/μg DNA (Okano et al., 2005; of clinical symptoms (Ohga et al., 2004; Solomon et al., 2014).
Subspecialty Group of Infectious Diseases and National Children’s
Epstein-Barr Virus Infection Cooperative, 2021). Decreased numbers of 2.5. EBV-associated hemophagocytic lymphohistiocytosis
EBV-specific T cells and increased viral loads in PBMCs are observed in
CAEBV patients, which may be associated with the impaired ability of EBV-induced HLH is the most common subtype of secondary virus-
EBV-specific T cells to control infection (Tsuge et al., 2001). Addition­ associated HLH during childhood, and is characterized by uncon­
ally, the latent type II pattern in CAEBV patients contributes to the trolled activation of T lymphocytes and macrophages. This leads to
emergence of high viral loads. This is because infected cells maintain excessive production of inflammatory cytokines, ultimately causing
latent infection by expressing a limited number of latent proteins that extensive tissue damage (Ishii, 2016). Patients with EBV-HLH typically
are not easily recognized by the immune system. have a high viral load of EBV and a high number of cells containing
The pathogenesis of CAEBV is not yet fully understood, but potential EBERs in their peripheral blood or tissues (Wang et al., 2014b; Ai and
reasons include dysfunction of EBV-specific CD8+ T cells, which are Xie, 2018). Active EBV-HLH develops rapidly and is associated with a
unable to eliminate infected cells (Kimura et al., 2005; Xing et al., 2013). poor prognosis, with a high fatality rate of over 50%. Children with IM
In chronic infections, antigen-specific CD8+ T cells may display complicated by HLH is most likely to have a primary immunodeficiency
decreased effector function, cytotoxicity, proliferation, and upregula­ disease (PID), such as X-linked lymphoproliferative disease (XLP) and
tion of coinhibitory receptors, known as “T cell exhaustion”. This phe­ CD27 deficiency (Qiang et al., 2012). In patients with EBV-HLH and
nomenon is not beneficial in disease control and may lead to recurrent XLP, a typical mutation in SH2D1A, which encodes the signaling
infection (Chatterjee et al., 2019; Busselaar et al., 2020). Several studies lymphocyte activator molecule (SLAM) associated protein (SAP), in­
have reported that some patients with CAEBV have mutation in their activates an important cytolytic activating receptor, NKG2D. This

8
M. Liu et al. Infection, Genetics and Evolution 112 (2023) 105443

receptor is expressed on the surface of CD8+ T cells, and its inactivation et al., 2008). Chen et al. reported that gB-specific CD8+ T cells obstruct
results in impaired cytotoxic function of CD8+ T cells and disabled B-cell transformation in vitro, which facilitates immune surveillance and
function of controlling infection (Latour and Fischer, 2019). the development of the gB subunit vaccine. gB is considered to be a
The study of T cell response in patients with EBV-HLH is limited. valuable T cell epitope due to its precise localization and epitopes con­
Analysis of PBMCs in some EBV-HLH patients showed that activated T servation. Therefore, it may be a promising epitope for the development
cell subsets are usually hyperactivated CD8+ T cells that expresses CD38, of subunit vaccines (Chen et al., 2021).
whereas the number of CD4+ T cells are decrease (Kasahara et al., 2001;
Yang et al., 2017). The imbalance of immune regulation of CTLs leads to 3.2. Antigen-armed antibodies
the production of large amount of inflammatory cytokines, (Fig. 3C),
resulting in severe tissue damage (Kasahara et al., 2001). In EBV-HLH Another candidate for a vaccine is based on AgAbs, which stimulate
patients, the secretion of many pro-inflammatory cytokines, such as immune response by fusing the antigen to endocytic receptor DEC-205, a
IFN-γ and TNF-α, is elevated, while IL-10, which plays a role in immu­ monoclonal antibody against dendritic cells, to deliver the antigen
nosuppressive to maintain the balance of inflammation in the immune specifically to antigen-presenting cells (Iberg and Hawiger, 2019;
system, is also significantly increased. A previous report suggests that Padilla-Quirarte et al., 2019). However, laboratory experiments have
the high level of IL-10 in EBV-HLH patients might be related to HLH shown that DEC-205 tends to process antigens to MHC-II molecules.
itself (Wada et al., 2013). Interestingly, CD4+ T cells spontaneously Furthermore, presenting DEC-205-targeted antigens to MHC-II mole­
function and complete differentiation process in EBV-HLH patients; cules elicit an amplified T cell immune response (Leung et al., 2013; Yu
therefore, the function of CD8+ CTLs may be impaired during molecular et al., 2015). Recombinant AgAbs vaccines of αDEC-205 equipped with
immune control processes in patients with EBV-HLH, while the function EBNA1 or LMP1 induce T cell responses, primarily EBNA1-specific CD4+
of CD4+ T cells is not affected. T cell response and less frequently EBNA1-specific CD8+ T cell immune
responses, resulting in the amplification of specific memory CTLs (Leung
3. Prophylactic vaccine and biologics et al., 2013). Further studies have demonstrated that the combination of
recombinant AgAbs vaccines targeting DEC-205 and adenovirus
Prophylactic vaccination can make the population less prone to EBV possibly protect mice from T cell lymphoma challenge. This suggests
infection and contribute to a decrease in severe cases, thus providing that heterologous prime-boost and antigen-antibody conjugates ap­
some level of protection in host-virus interaction. However, there are proaches should be considered to elicit T cell-mediated immune control
ongoing investigations into the effectiveness of EBV-related prophylac­ against EBV (Gurer et al., 2008; Ruhl et al., 2019). The AgAbs of αDEC-
tic vaccines in mitigating the burden of EBV-associated diseases in 205 equipped with BZLF1 elicit strong EBV-specific cellular responses in
general population. As such, the development of a prophylactic vaccine humanized mice and lead to a remarkable expansion of EBV-specific
against EBV remains an urgent concern. memory CTLs, which is a promising vaccine category (Ahmed et al.,
Previous research on prophylactic EBV vaccines has focused on 2021).
blocking EBV infection in target cells, primarily through the induction of
neutralizing antibodies. The first clinical trial of a prophylactic vaccine 3.3. Virus-like particles
involved the use of anti-gp350 prophylactic vaccines. Gu et al. observed
high levels of neutralizing antibody against EBV in vaccinated children, VLP have been utilized in the development of EBV vaccines. These
with only three individuals being infected (Gu et al., 1995). Other VLPs are structurally similar to EBV particles but lack EBV DNA due to
promising prophylactic vaccine targets include the proteins gp350, gH, knockout of certain genes. A cell line for EBV-derived VLPs was created
gL, and gp42, which have shown potential in eliciting higher neutral­ by deleting terminal repeat (TR) involved in DNA packaging and po­
izing antibody titers, although there are currently no clinical trials that tential EBV oncogenes BZLF1, EBNA2, EBNA3A, 3B, 3C, and LMP1.
can confirm complete protection against EBV infection (Reynolds, 2013; Inoculated BALB/c mice showed EBV-specific humoral and cellular
Cui et al., 2016; Cohen, 2018; Bu et al., 2019). Recent studies have immune responses (Ruiss et al., 2011). Subsequent research discovered
focused on the development of novel prophylactic vaccines against EBV. that latent antigen fused with envelope protein could enhance the
Kanekiyo et al. developed a ferritin nanoparticle vaccine (ferritin-gp350) antigenic spectrum of EBV VLP. These VLPs containing latent antigens
with the target antigen of gp350 and found a 10- to 100-fold increase in could stimulate CD8+and CD4+ T cell responses specific to latent pro­
neutralization in a mouse model (Kanekiyo et al., 2015). Heeke et al. teins, thereby contributing to immune protection against EBV infection
developed a glucopyranosyl lipid A incorporated into the stable emul­ in humanized mice (van Zyl et al., 2018). Therefore, the antigenic cargo
sion (GLA/ SE)-gp350 vaccine targeting gp350 with GLA/SE as an in EBV VLP can be fine-tuned to induce an immune response by
adjuvant, and detected high neutralizing antibody titers and robust immunodominant latent antigen. A new vaccine platform was devel­
polyfunctional CD4+ T cell response against gp350 in mice and rabbits oped by Perez et al. to incorporating gH/gL-EBNA1 or gB-LMP2 with
(Heeke et al., 2016). Currently, phase I clinical trials of the Modena VLPs. This vaccine candidate was stably produced in CHO cells and
mRNA vaccine (mRNA-1189) containing four antigens, gH, gL, gp42, induced neutralizing antibodies in inoculated BALB/c mice, as well as
and gp220, are underway (Zhong et al., 2022). Epitope vaccines, EBNA1- or LMP2-specific T cell responses (Perez et al., 2017). This
antigen-armed antibodies (AgAbs), and virus-like particles (VLP) are approach provides a viable and effective vaccine system.
popular vaccine categories that elicit cellular immunity. It is hoped that
these advancements will aid in the development of more effective pro­ 4. Conclusions
phylactic vaccines against EBV.
EBV is a prevalent virus that can cause a significant disease burden.
3.1. Epitope-based vaccine Understanding T cell immune function controls infection and how it
contributes to disease is essential. However, the mechanisms by which T
The simplest vaccine system is EBNA3A epitope peptide-based vac­ cells responses control infection are still not fully understood, and
cine. This vaccine is made by mixing EBNA3A epitope peptides with further investigation is needed, including the immune response in
tetanus toxin, which serve as a source of CD4+ T cell, in a water-in-oil asymptomatic individuals versus IM patients, the role of cellular im­
adjuvant. Clinical trial results have shown that inoculation with munity in CAEBV, and which antigen epitope triggers the strongest EBV-
EBNA3A epitope peptide successfully induces CD8+ T cell response in specific T cell responses. Ultimately, designing an effective prophylactic
most of subjects. However, although subjects are mostly asymptomatic vaccine is crucial for mitigating clinical manifestations even if it cannot
after EBV infection, the vaccine fails to prevent EBV infection (Elliott prevent primary EBV infection.

9
M. Liu et al. Infection, Genetics and Evolution 112 (2023) 105443

Credit author statement Barros, M.H.M., Vera-Lozada, G., Segges, P., Hassan, R., Niedobitek, G., 2019. Revisiting
the tissue microenvironment of infectious mononucleosis: identification of EBV
infection in T cells and deep characterization of immune profiles. Front. Immunol.
Mengjia Liu wrote the first draft. Ran Wang and Zhengde Xie revised 10, 146.
the manuscript. Bollard, C.M., Cohen, J.I., 2018. How I treat T-cell chronic active Epstein-Barr virus
disease. Blood 131, 2899–2905.
Borza, C.M., Hutt-Fletcher, L.M., 2002. Alternate replication in B cells and epithelial cells
Data statement switches tropism of Epstein-Barr virus. Nat. Med. 8, 594–599.
Brooks, J.M., Long, H.M., Tierney, R.J., Shannon-Lowe, C., Leese, A.M., Fitzpatrick, M.,
Taylor, G.S., Rickinson, A.B., 2016. Early T cell recognition of B cells following
None.
Epstein-Barr virus infection: identifying potential targets for prophylactic
vaccination. PLoS Pathog. 12, e1005549.
Ethical statement Bu, W., Joyce, M.G., Nguyen, H., Banh, D.V., Aguilar, F., Tariq, Z., Yap, M.L.,
Tsujimura, Y., Gillespie, R.A., Tsybovsky, Y., Andrews, S.F., Narpala, S.R.,
McDermott, A.B., Rossmann, M.G., Yasutomi, Y., Nabel, G.J., Kanekiyo, M.,
This review does not involve the ethical issues. Cohen, J.I., 2019. Immunization with components of the viral fusion apparatus
elicits antibodies that neutralize Epstein-Barr virus in B cells and epithelial cells.
Immunity 50, 1305–1316 e1306.
Declaration of Competing Interest Busselaar, J., Tian, S., van Eenennaam, H., Borst, J., 2020. Helpless priming sends CD8(
+) T cells on the road to exhaustion. Front. Immunol. 11, 592569.
Calarota, S.A., Chiesa, A., Zelini, P., Comolli, G., Minoli, L., Baldanti, F., 2013. Detection
The authors have declared no conflict of interest. of Epstein-Barr virus-specific memory CD4+ T cells using a peptide-based cultured
enzyme-linked immunospot assay. Immunology 139, 533–544.
Callan, M.F., Tan, L., Annels, N., Ogg, G.S., Wilson, J.D., O’Callaghan, C.A., Steven, N.,
Data availability
McMichael, A.J., Rickinson, A.B., 1998. Direct visualization of antigen-specific CD8
+ T cells during the primary immune response to Epstein-Barr virus in vivo. J. Exp.
No data was used for the research described in the article. Med. 187, 1395–1402.
Catalina, M.D., Sullivan, J.L., Brody, R.M., Luzuriaga, K., 2002. Phenotypic and
functional heterogeneity of EBV epitope-specific CD8+ T cells. J. Immunol. 168,
Acknowledgments 4184–4191.
Chai, L., Ge, X., Biswas, M.K., Deng, X., 2011. Molecular analysis and expression of a
floral organ-relative F-box gene isolated from ‘Zigui shatian’ pummelo (Citrus
R.W. was supported by the Beijing Natural Science Foundation
grandis Osbeck). Mol. Biol. Rep. 38, 4429–4436.
(7222059), National Natural Science Foundation of China (82002130). Chatterjee, B., Deng, Y., Holler, A., Nunez, N., Azzi, T., Vanoaica, L.D., Muller, A.,
ZD.X. was supported by the CAMS Innovation Fund for Medical Sciences Zdimerova, H., Antsiferova, O., Zbinden, A., Capaul, R., Dreyer, J.H., Nadal, D.,
Becher, B., Robinson, M.D., Stauss, H., Munz, C., 2019. CD8+ T cells retain
(2019-I2M-5-026).
protective functions despite sustained inhibitory receptor expression during Epstein-
Barr virus infection in vivo. PLoS Pathog. 15, e1007748.
References Chen, G., Shankar, P., Lange, C., Valdez, H., Skolnik, P.R., Wu, L., Manjunath, N.,
Lieberman, J., 2001. CD8 T cells specific for human immunodeficiency virus,
Epstein-Barr virus, and cytomegalovirus lack molecules for homing to lymphoid sites
Abbott, R.J., Quinn, L.L., Leese, A.M., Scholes, H.M., Pachnio, A., Rickinson, A.B., 2013.
of infection. Blood 98, 156–164.
CD8+ T cell responses to lytic EBV infection: late antigen specificities as
Chen, Y., Sun, H., Liu, G., Wang, B., Wang, F., Sun, B., Yao, K., 2009. EBV LMP2A-specific
subdominant components of the total response. J. Immunol. 191, 5398–5409.
T cell immune responses elicited by dendritic cells loaded with LMP2A protein. Cell.
Abbott, R.J., Pachnio, A., Pedroza-Pacheco, I., Leese, A.M., Begum, J., Long, H.M.,
Mol. Immunol. 6, 269–276.
Croom-Carter, D., Stacey, A., Moss, P.A.H., Hislop, A.D., Borrow, P., Rickinson, A.B.,
Chen, J., Sathiyamoorthy, K., Zhang, X., Schaller, S., Perez White, B.E., Jardetzky, T.S.,
Bell, A.I., 2017. Asymptomatic primary infection with Epstein-Barr virus:
Longnecker, R., 2018. Ephrin receptor A2 is a functional entry receptor for Epstein-
observations on young adult cases. J. Virol. 91.
Barr virus. Nat. Microbiol. 3, 172–180.
Adem, C., Sebo, T.J., Riehle, D.L., Lohse, C.M., Nascimento, A.G., 2002. Recurrent and
Chen, H., Zhang, X., Zhang, S., Duan, X., Xiang, T., Zhou, X., Zhang, W., Zhang, X.,
non-recurrent pigmented villonodular synovitis 1. Ann. Pathol. 22, 448–452.
Feng, Q., Kang, Y., Li, J., Deng, L., Wang, L., Lv, X., Zeng, M., Zeng, Y.X., Xu, M.,
Adhikary, D., Damaschke, J., Mautner, J., Behrends, U., 2020. The Epstein-Barr virus
2021. T cell epitope screening of Epstein-Barr virus fusion protein gB. J. Virol.
major tegument protein BNRF1 is a common target of cytotoxic CD4(+) T cells.
https://doi.org/10.1128/JVI.00081-21.
J. Virol. 94.
Chesnokova, L.S., Hutt-Fletcher, L.M., 2011. Fusion of Epstein-Barr virus with epithelial
Ahmed, E.H., Brooks, E., Sloan, S., Schlotter, S., Jeney, F., Hale, C., Mao, C., Zhang, X.,
cells can be triggered by alphavbeta5 in addition to alphavbeta6 and alphavbeta8,
McLaughlin, E., Shindiapina, P., Shire, S., Das, M., Prouty, A., Lozanski, G.,
and integrin binding triggers a conformational change in glycoproteins gHgL.
Mamuye, A.T., Abebe, T., Alinari, L., Caligiuri, M.A., Baiocchi, R.A., 2021. Targeted
J. Virol. 85, 13214–13223.
delivery of BZLF1 to DEC205 drives EBV-protective immunity in a spontaneous
Cirac, A., Stutzle, S., Dieckmeyer, M., Adhikary, D., Moosmann, A., Korber, N., Bauer, T.,
model of EBV-driven lymphoproliferative disease. Vaccines (Basel) 9.
Witter, K., Delecluse, H.J., Behrends, U., Mautner, J., 2018. Epstein-Barr virus strain
Ai, J., Xie, Z., 2018. Epstein-Barr virus-positive T/NK-cell lymphoproliferative diseases in
heterogeneity impairs human T-cell immunity. Cancer Immunol. Immunother. 67,
Chinese Mainland. Front. Pediatr. 6, 289.
663–674.
Allen, C.E., McClain, K.L., 2015. Pathophysiology and epidemiology of hemophagocytic
Cirac, A., Poirey, R., Dieckmeyer, M., Witter, K., Delecluse, H.J., Behrends, U.,
lymphohistiocytosis. Hematol. Am. Soc. Hematol. Educ. Program. 2015, 177–182.
Mautner, J., 2021. Immunoinformatic analysis reveals antigenic heterogeneity of
Amyes, E., Hatton, C., Montamat-Sicotte, D., Gudgeon, N., Rickinson, A.B.,
Epstein-Barr virus is immune-driven. Front. Immunol. 12, 796379.
McMichael, A.J., Callan, M.F., 2003. Characterization of the CD4+ T cell response to
Cohen, J.I., 2000. Epstein-Barr virus infection. N. Engl. J. Med. 343, 481–492.
Epstein-Barr virus during primary and persistent infection. J. Exp. Med. 198,
Cohen, J.I., 2018. Vaccine development for Epstein-Barr virus. Adv. Exp. Med. Biol.
903–911.
1045, 477–493.
Antsiferova, O., Muller, A., Ramer, P.C., Chijioke, O., Chatterjee, B., Raykova, A.,
Collins, P.J., Fox, C.P., George, L., Pearce, H., Ryan, G., De Santo, C., Mussai, F.,
Planas, R., Sospedra, M., Shumilov, A., Tsai, M.H., Delecluse, H.J., Munz, C., 2014.
Lewis, D., Long, H., Shannon-Lowe, C., 2021. Characterizing EBV-associated
Adoptive transfer of EBV specific CD8+ T cell clones can transiently control EBV
lymphoproliferative diseases and the role of myeloid-derived suppressor cells. Blood
infection in humanized mice. PLoS Pathog. 10, e1004333.
137, 203–215.
Apcher, S., Komarova, A., Daskalogianni, C., Yin, Y., Malbert-Colas, L., Fahraeus, R.,
Connolly, S.A., Jardetzky, T.S., Longnecker, R., 2021. The structural basis of herpesvirus
2009. mRNA translation regulation by the Gly-Ala repeat of Epstein-Barr virus
entry. Nat. Rev. Microbiol. 19, 110–121.
nuclear antigen 1. J. Virol. 83, 1289–1298.
Correia, S., Palser, A., Elgueta Karstegl, C., Middeldorp, J.M., Ramayanti, O., Cohen, J.I.,
Appay, V., Zaunders, J.J., Papagno, L., Sutton, J., Jaramillo, A., Waters, A.,
Hildesheim, A., Fellner, M.D., Wiels, J., White, R.E., Kellam, P., Farrell, P.J., 2017.
Easterbrook, P., Grey, P., Smith, D., McMichael, A.J., Cooper, D.A., Rowland-
Natural variation of Epstein-Barr virus genes, proteins, and primary MicroRNA.
Jones, S.L., Kelleher, A.D., 2002. Characterization of CD4(+) CTLs ex vivo.
J. Virol. 91.
J. Immunol. 168, 5954–5958.
Cruchley, A.T., Williams, D.M., Niedobitek, G., Young, L.S., 1997. Epstein-Barr virus:
Arai, A., 2021. Chronic active Epstein-Barr virus infection: the elucidation of the
biology and disease. Oral Dis. 3 (Suppl. 1), S156–S163.
pathophysiology and the development of therapeutic methods. Microorganisms 9.
Cui, X., Cao, Z., Chen, Q., Arjunaraja, S., Snow, A.L., Snapper, C.M., 2016. Rabbits
Bailey, R.E., 1994. Diagnosis and treatment of infectious mononucleosis. Am. Fam.
immunized with Epstein-Barr virus gH/gL or gB recombinant proteins elicit higher
Physician 49, 879–888.
serum virus neutralizing activity than gp350. Vaccine 34, 4050–4055.
Balfour Jr., H.H., Odumade, O.A., Schmeling, D.O., Mullan, B.D., Ed, J.A., Knight, J.A.,
de Bleser, R., Poeck, K., 1985. Analysis of prosody in the spontaneous speech of patients
Vezina, H.E., Thomas, W., Hogquist, K.A., 2013. Behavioral, virologic, and
with CV-recurring utterances. Cortex 21, 405–415.
immunologic factors associated with acquisition and severity of primary Epstein-
Deng, Y., Chatterjee, B., Zens, K., Zdimerova, H., Muller, A., Schuhmachers, P., Ligeon, L.
Barr virus infection in university students. J. Infect. Dis. 207, 80–88.
A., Bongiovanni, A., Capaul, R., Zbinden, A., Holler, A., Stauss, H.,
Balfour Jr., H.H., Dunmire, S.K., Hogquist, K.A., 2015. Infectious mononucleosis. Clin.
Transl. Immunol. 4, e33.

10
M. Liu et al. Infection, Genetics and Evolution 112 (2023) 105443

Hammerschmidt, W., Munz, C., 2021. CD27 is required for protective lytic EBV Izawa, K., Martin, E., Soudais, C., Bruneau, J., Boutboul, D., Rodriguez, R., Lenoir, C.,
antigen-specific CD8+ T-cell expansion. Blood 137, 3225–3236. Hislop, A.D., Besson, C., Touzot, F., Picard, C., Callebaut, I., de Villartay, J.P.,
Dharnidharka, V.R., Webster, A.C., Martinez, O.M., Preiksaitis, J.K., Leblond, V., Moshous, D., Fischer, A., Latour, S., 2017. Inherited CD70 deficiency in humans
Choquet, S., 2016. Post-transplant lymphoproliferative disorders. Nat. Rev. Dis. reveals a critical role for the CD70-CD27 pathway in immunity to Epstein-Barr virus
Primers 2, 15088. infection. J. Exp. Med. 214, 73–89.
Dowell, A.C., Haigh, T.A., Ryan, G.B., Turner, J.E., Long, H.M., Taylor, G.S., 2021. Jayasooriya, S., de Silva, T.I., Njie-jobe, J., Sanyang, C., Leese, A.M., Bell, A.I.,
Cytotoxic CD4+ T-cells specific for EBV capsid antigen BORF1 are maintained in McAulay, K.A., Yanchun, P., Long, H.M., Dong, T., Whittle, H.C., Rickinson, A.B.,
long-term latently infected healthy donors. PLoS Pathog. 17, e1010137. Rowland-Jones, S.L., Hislop, A.D., Flanagan, K.L., 2015. Early virological and
Dunmire, S.K., Hogquist, K.A., Balfour, H.H., 2015. Infectious mononucleosis. Curr. Top. immunological events in asymptomatic Epstein-Barr virus infection in African
Microbiol. Immunol. 390, 211–240. children. PLoS Pathog. 11, e1004746.
Dunmire, S.K., Verghese, P.S., Balfour Jr., H.H., 2018. Primary Epstein-Barr virus Jenson, H.B., 2000. Acute complications of Epstein-Barr virus infectious mononucleosis.
infection. J. Clin. Virol. 102, 84–92. Curr. Opin. Pediatr. 12, 263–268.
Dunne, P.J., Faint, J.M., Gudgeon, N.H., Fletcher, J.M., Plunkett, F.J., Soares, M.V., Kanekiyo, M., Bu, W., Joyce, M.G., Meng, G., Whittle, J.R., Baxa, U., Yamamoto, T.,
Hislop, A.D., Annels, N.E., Rickinson, A.B., Salmon, M., Akbar, A.N., 2002. Epstein- Narpala, S., Todd, J.P., Rao, S.S., McDermott, A.B., Koup, R.A., Rossmann, M.G.,
Barr virus-specific CD8(+) T cells that re-express CD45RA are apoptosis-resistant Mascola, J.R., Graham, B.S., Cohen, J.I., Nabel, G.J., 2015. Rational design of an
memory cells that retain replicative potential. Blood 100, 933–940. Epstein-Barr virus vaccine targeting the receptor-binding site. Cell 162, 1090–1100.
Elliott, S.L., Suhrbier, A., Miles, J.J., Lawrence, G., Pye, S.J., Le, T.T., Rosenstengel, A., Kasahara, Y., Yachie, A., Takei, K., Kanegane, C., Okada, K., Ohta, K., Seki, H.,
Nguyen, T., Allworth, A., Burrows, S.R., Cox, J., Pye, D., Moss, D.J., Bharadwaj, M., Igarashi, N., Maruhashi, K., Katayama, K., Katoh, E., Terao, G., Sakiyama, Y.,
2008. Phase I trial of a CD8+ T-cell peptide epitope-based vaccine for infectious Koizumi, S., 2001. Differential cellular targets of Epstein-Barr virus (EBV) infection
mononucleosis. J. Virol. 82, 1448–1457. between acute EBV-associated hemophagocytic lymphohistiocytosis and chronic
El-Mallawany, N.K., Curry, C.V., Allen, C.E., 2022. Haemophagocytic active EBV infection. Blood 98, 1882–1888.
lymphohistiocytosis and Epstein-Barr virus: a complex relationship with diverse Katano, H., Ali, M.A., Patera, A.C., Catalfamo, M., Jaffe, E.S., Kimura, H., Dale, J.K.,
origins, expression and outcomes. Br. J. Haematol. 196, 31–44. Straus, S.E., Cohen, J.I., 2004. Chronic active Epstein-Barr virus infection associated
Epstein, M.A., Achong, B.G., Barr, Y.M., 1964. Virus particles in cultured lymphoblasts with mutations in perforin that impair its maturation. Blood 103, 1244–1252.
from Burkitt’s lymphoma. Lancet 1, 702–703. Kimura, H., Cohen, J.I., 2017. Chronic active Epstein-Barr virus disease. Front. Immunol.
Farrell, P.J., 2015. Epstein-Barr Virus Strain Variation. Curr. Top. Microbiol. Immunol. 8, 1867.
390, 45–69. Kimura, H., Hoshino, Y., Hara, S., Sugaya, N., Kawada, J., Shibata, Y., Kojima, S.,
Farrell, P.J., 2019. Epstein-Barr virus and cancer. Annu. Rev. Pathol. 14, 29–53. Nagasaka, T., Kuzushima, K., Morishima, T., 2005. Differences between T cell-type
Fedyanina, O.S., Filippova, A.E., Demina, O.I., Zhuliabina, O.A., Tikhomirov, D.S., and natural killer cell-type chronic active Epstein-Barr virus infection. J. Infect. Dis.
Filatov, A.V., Chebotareva, T.A., Kuznetsova, S.A., 2021. The nature and clinical 191, 531–539.
significance of atypical mononuclear cells in infectious mononucleosis caused by the Kimura, H., Ito, Y., Suzuki, R., Nishiyama, Y., 2008. Measuring Epstein-Barr virus (EBV)
Epstein-Barr virus in children. J. Infect. Dis. 223, 1699–1706. load: the significance and application for each EBV-associated disease. Rev. Med.
Forrest, C., Hislop, A.D., Rickinson, A.B., Zuo, J., 2018. Proteome-wide analysis of CD8+ Virol. 18, 305–319.
T cell responses to EBV reveals differences between primary and persistent infection. Kimura, H., Ito, Y., Kawabe, S., Gotoh, K., Takahashi, Y., Kojima, S., Naoe, T., Esaki, S.,
PLoS Pathog. 14, e1007110. Kikuta, A., Sawada, A., Kawa, K., Ohshima, K., Nakamura, S., 2012. EBV-associated
Fujiwara, S., Nakamura, H., 2020. Chronic active Epstein-Barr virus infection: is it T/NK-cell lymphoproliferative diseases in nonimmunocompromised hosts:
immunodeficiency, malignancy, or both? Cancers (Basel) 12. prospective analysis of 108 cases. Blood 119, 673–686.
Gabrilovich, D.I., Nagaraj, S., 2009. Myeloid-derived suppressor cells as regulators of the Kruger, R., Martin, E., Dmytrus, J., Feiterna-Sperling, C., Meisel, C., Unterwalder, N.,
immune system. Nat. Rev. Immunol. 9, 162–174. Kolsch, U., Wahn, V., Hofmann, J., Korn, P., Latour, S., Boztug, K., von Bernuth, H.,
Gu, S.Y., Huang, T.M., Ruan, L., Miao, Y.H., Lu, H., Chu, C.M., Motz, M., Wolf, H., 1995. 2020. CD70 deficiency associated with chronic Epstein-Barr virus infection,
First EBV vaccine trial in humans using recombinant vaccinia virus expressing the recurrent airway infections and severe gingivitis in a 24-year-old woman. Front.
major membrane antigen. Dev. Biol. Stand. 84, 171–177. Immunol. 11, 1593.
Guan, Y.Q., Shen, K.F., Yang, L., Cai, H.D., Zhang, M.L., Wang, J.C., Long, X.L., Xiong, J., Lam, J.K.P., Hui, K.F., Ning, R.J., Xu, X.Q., Chan, K.H., Chiang, A.K.S., 2018. Emergence
Gu, J., Zhang, P.L., Xiao, M., Zhang, W., Zhou, J.F., 2021. Inherited genetic of CD4+ and CD8+ polyfunctional T cell responses against immunodominant lytic
susceptibility to nonimmunosuppressed Epstein-Barr virus-associated T/NK-cell and latent EBV antigens in children with primary EBV infection. Front. Microbiol. 9,
lymphoproliferative diseases in Chinese patients. Curr. Med. Sci. 41, 482–490. 416.
Gurer, C., Strowig, T., Brilot, F., Pack, M., Trumpfheller, C., Arrey, F., Park, C.G., Landais, E., Saulquin, X., Scotet, E., Trautmann, L., Peyrat, M.A., Yates, J.L., Kwok, W.
Steinman, R.M., Munz, C., 2008. Targeting the nuclear antigen 1 of Epstein-Barr W., Bonneville, M., Houssaint, E., 2004. Direct killing of Epstein-Barr virus (EBV)-
virus to the human endocytic receptor DEC-205 stimulates protective T-cell infected B cells by CD4 T cells directed against the EBV lytic protein BHRF1. Blood
responses. Blood 112, 1231–1239. 103, 1408–1416.
Heeke, D.S., Lin, R., Rao, E., Woo, J.C., McCarthy, M.P., Marshall, J.D., 2016. Lanzavecchia, A., 1985 Apr 11-17. Antigen-specific interaction between T and B cells.
Identification of GLA/SE as an effective adjuvant for the induction of robust humoral Nature 314 (6011), 537–539. https://doi.org/10.1038/314537a0.
and cell-mediated immune responses to EBV-gp350 in mice and rabbits. Vaccine 34, Latour, S., Fischer, A., 2019. Signaling pathways involved in the T-cell-mediated
2562–2569. immunity against Epstein-Barr virus: lessons from genetic diseases. Immunol. Rev.
Herbert, K.M., Pimienta, G., 2016. Consideration of Epstein-Barr virus-encoded 291, 174–189.
noncoding RNAs EBER1 and EBER2 as a functional backup of viral oncoprotein Lee, C.P., Chen, M.R., 2021. Conquering the nuclear envelope barriers by EBV lytic
latent membrane protein 1. mBio 7 e01926–01915. replication. Viruses 13.
Hislop, A.D., Gudgeon, N.H., Callan, M.F., Fazou, C., Hasegawa, H., Salmon, M., Leen, A., Meij, P., Redchenko, I., Middeldorp, J., Bloemena, E., Rickinson, A., Blake, N.,
Rickinson, A.B., 2001. EBV-specific CD8+ T cell memory: relationships between 2001. Differential immunogenicity of Epstein-Barr virus latent-cycle proteins for
epitope specificity, cell phenotype, and immediate effector function. J. Immunol. human CD4(+) T-helper 1 responses. J. Virol. 75, 8649–8659.
167, 2019–2029. Lelic, A., Verschoor, C.P., Ventresca, M., Parsons, R., Evelegh, C., Bowdish, D., Betts, M.
Hislop, A.D., Kuo, M., Drake-Lee, A.B., Akbar, A.N., Bergler, W., Hammerschmitt, N., R., Loeb, M.B., Bramson, J.L., 2012. The polyfunctionality of human memory CD8+
Khan, N., Palendira, U., Leese, A.M., Timms, J.M., Bell, A.I., Buckley, C.D., T cells elicited by acute and chronic virus infections is not influenced by age. PLoS
Rickinson, A.B., 2005. Tonsillar homing of Epstein-Barr virus-specific CD8+ T cells Pathog. 8, e1003076.
and the virus-host balance. J. Clin. Invest. 115, 2546–2555. Leung, C.S., Maurer, M.A., Meixlsperger, S., Lippmann, A., Cheong, C., Zuo, J., Haigh, T.
Hislop, A.D., Taylor, G.S., Sauce, D., Rickinson, A.B., 2007. Cellular responses to viral A., Taylor, G.S., Munz, C., 2013. Robust T-cell stimulation by Epstein-Barr virus-
infection in humans: lessons from Epstein-Barr virus. Annu. Rev. Immunol. 25, transformed B cells after antigen targeting to DEC-205. Blood 121, 1584–1594.
587–617. Levitskaya, J., Sharipo, A., Leonchiks, A., Ciechanover, A., Masucci, M.G., 1997.
Hoshino, Y., Morishima, T., Kimura, H., Nishikawa, K., Tsurumi, T., Kuzushima, K., Inhibition of ubiquitin/proteasome-dependent protein degradation by the Gly-Ala
1999. Antigen-driven expansion and contraction of CD8+-activated T cells in repeat domain of the Epstein-Barr virus nuclear antigen 1. Proc. Natl. Acad. Sci. U. S.
primary EBV infection. J. Immunol. 163, 5735–5740. A. 94, 12616–12621.
Hoshino, Y., Nishikawa, K., Ito, Y., Kuzushima, K., Kimura, H., 2011. Kinetics of Epstein- Li, Q., Spriggs, M.K., Kovats, S., Turk, S.M., Comeau, M.R., Nepom, B., Hutt-Fletcher, L.
Barr virus load and virus-specific CD8+ T cells in acute infectious mononucleosis. M., 1997. Epstein-Barr virus uses HLA class II as a cofactor for infection of B
J. Clin. Virol. 50, 244–246. lymphocytes. J. Virol. 71, 4657–4662.
Hu, H., Deng, H., Bi, J., Xu, Y., Li, S., Xie, Y., Sun, X., Wang, D., Li, X., Ouyang, W., Li, Z., Zhang, X., Dong, L., Pang, J., Xu, M., Zhong, Q., Zeng, M.S., Yu, X., 2020. CryoEM
Hu, B., Zhang, Y., Tang, H., Fang, C., Zhang, H., Guo, L., Wang, C., Wang, T., structure of the tegumented capsid of Epstein-Barr virus. Cell Res. 30, 873–884.
Yang, F., Jiang, T., Xie, Z., Liu, G., 2021. Clinical characteristics and effectiveness of Liu, W., Cui, Y., Wang, C., Li, Z., Gong, D., Dai, X., Bi, G.Q., Sun, R., Zhou, Z.H., 2020.
antiviral agents in hospitalized children with infectious mononucleosis in China: a Structures of capsid and capsid-associated tegument complex inside the Epstein-Barr
multicenter retrospective study. Pediatr. Investig. 5, 188–194. virus. Nat. Microbiol. 5, 1285–1298.
Iberg, C.A., Hawiger, D., 2019. Advancing immunomodulation by in vivo antigen Liu, M., Wang, X., Zhang, L., Feng, G., Zeng, Y., Wang, R., Xie, Z., 2022. Epidemiological
delivery to DEC-205 and other cell surface molecules using recombinant chimeric characteristics and disease burden of infectious mononucleosis in hospitalized
antibodies. Int. Immunopharmacol. 73, 575–580. children in China: a nationwide retrospective study. Virol. Sin. https://doi.org/
Imashuku, S., Morimoto, A., Ishii, E., 2021. Virus-triggered secondary hemophagocytic 10.1016/j.virs.2022.07.007.
lymphohistiocytosis. Acta Paediatr. 110, 2729–2736. Long, H.M., Leese, A.M., Chagoury, O.L., Connerty, S.R., Quarcoopome, J., Quinn, L.L.,
Ishii, E., 2016. Hemophagocytic lymphohistiocytosis in children: pathogenesis and Shannon-Lowe, C., Rickinson, A.B., 2011. Cytotoxic CD4+ T cell responses to EBV
treatment. Front. Pediatr. 4, 47.

11
M. Liu et al. Infection, Genetics and Evolution 112 (2023) 105443

contrast with CD8 responses in breadth of lytic cycle antigen choice and in lytic cycle nuclear antigen 6 comprises overlapping T cell epitopes which induce HLA-DR-
recognition. J. Immunol. 187, 92–101. restricted CD4(+) T lymphocytes. Int. Immunol. 12, 281–293.
Long, H.M., Chagoury, O.L., Leese, A.M., Ryan, G.B., James, E., Morton, L.T., Abbott, R. Ressing, M.E., van Leeuwen, D., Verreck, F.A., Gomez, R., Heemskerk, B., Toebes, M.,
J., Sabbah, S., Kwok, W., Rickinson, A.B., 2013. MHC II tetramers visualize human Mullen, M.M., Jardetzky, T.S., Longnecker, R., Schilham, M.W., Ottenhoff, T.H.,
CD4+ T cell responses to Epstein-Barr virus infection and demonstrate atypical Neefjes, J., Schumacher, T.N., Hutt-Fletcher, L.M., Wiertz, E.J., 2003. Interference
kinetics of the nuclear antigen EBNA1 response. J. Exp. Med. 210, 933–949. with T cell receptor-HLA-DR interactions by Epstein-Barr virus gp42 results in
MacArthur, G.J., Wilson, A.D., Birchall, M.A., Morgan, A.J., 2007. Primary CD4+ T-cell reduced T helper cell recognition. Proc. Natl. Acad. Sci. U. S. A. 100, 11583–11588.
responses provide both helper and cytotoxic functions during Epstein-Barr virus Ressing, M.E., van Gent, M., Gram, A.M., Hooykaas, M.J., Piersma, S.J., Wiertz, E.J.,
infection and transformation of fetal cord blood B cells. J. Virol. 81, 4766–4775. 2015. Immune evasion by Epstein-Barr virus. Curr. Top. Microbiol. Immunol. 391,
Marshall, N.A., Culligan, D.J., Johnston, P.W., Millar, C., Barker, R.N., Vickers, M.A., 355–381.
2007. CD4(+) T-cell responses to Epstein-Barr virus (EBV) latent membrane protein Reynolds, A.M., 2013. Selection pressures give composite correlated random walks Levy
1 in infectious mononucleosis and EBV-associated non-Hodgkin lymphoma: Th1 in walk characteristics. J. Theor. Biol. 332, 117–122.
active disease but Tr1 in remission. Br. J. Haematol. 139, 81–89. Ruhl, J., Citterio, C., Engelmann, C., Haigh, T., Dzionek, A., Dreyer, J., Khanna, R.,
Martorelli, D., Muraro, E., Merlo, A., Turrini, R., Rosato, A., Dolcetti, R., 2010. Role of Taylor, G.S., Wilson, J.B., Leung, C.S., Munz, C., 2019. Heterologous prime-boost
CD4+ cytotoxic T lymphocytes in the control of viral diseases and cancer. Int. Rev. vaccination protects against EBV antigen-expressing lymphomas. J. Clin. Invest. 129,
Immunol. 29, 371–402. 2071–2087.
McKenzie, J., El-Guindy, A., 2015. Epstein-Barr virus lytic cycle reactivation. Curr. Top. Ruhl, J., Leung, C.S., Munz, C., 2020. Vaccination against the Epstein-Barr virus. Cell.
Microbiol. Immunol. 391, 237–261. Mol. Life Sci. 77, 4315–4324.
Meckiff, B.J., Ladell, K., McLaren, J.E., Ryan, G.B., Leese, A.M., James, E.A., Price, D.A., Ruiss, R., Jochum, S., Wanner, G., Reisbach, G., Hammerschmidt, W., Zeidler, R., 2011.
Long, H.M., 2019. Primary EBV infection induces an acute wave of activated A virus-like particle-based Epstein-Barr virus vaccine. J. Virol. 85, 13105–13113.
antigen-specific cytotoxic CD4(+) T cells. J. Immunol. 203, 1276–1287. Shafiee, A., Shamsi, S., Kohandel Gargari, O., Beiky, M., Allahkarami, M.M., Miyanaji, A.
Minarovits, J., 2006. Epigenotypes of latent herpesvirus genomes. Curr. Top. Microbiol. B., Aghajanian, S., Mozhgani, S.H., 2022. EBV associated T- and NK-cell
Immunol. 310, 61–80. lymphoproliferative diseases: a comprehensive overview of clinical manifestations
Molesworth, S.J., Lake, C.M., Borza, C.M., Turk, S.M., Hutt-Fletcher, L.M., 2000. Epstein- and novel therapeutic insights. Rev. Med. Virol. 32, e2328.
Barr virus gH is essential for penetration of B cells but also plays a role in attachment Silins, S.L., Sherritt, M.A., Silleri, J.M., Cross, S.M., Elliott, S.L., Bharadwaj, M., Le, T.T.,
of virus to epithelial cells. J. Virol. 74, 6324–6332. Morrison, L.E., Khanna, R., Moss, D.J., Suhrbier, A., Misko, I.S., 2001. Asymptomatic
Montesoro, E., Castelli, G., Morsilli, O., Nisini, R., Stafsnes, M.H., Care, A., Peschle, C., primary Epstein-Barr virus infection occurs in the absence of blood T-cell repertoire
Chelucci, C., 2006. Unilineage monocytopoiesis in hematopoietic progenitor culture: perturbations despite high levels of systemic viral load. Blood 98, 3739–3744.
switching cytokine treatment at all Mo developmental stages induces differentiation Singavi, A.K., Harrington, A.M., Fenske, T.S., 2015. Post-transplant lymphoproliferative
into dendritic cells. Cell Death Differ. 13, 250–259. disorders. Cancer Treat. Res. 165, 305–327.
Morales, O., Depil, S., Mrizak, D., Martin, N., Ndour, P.A., Dufosse, F., Miroux, C., Soares, M.V., Plunkett, F.J., Verbeke, C.S., Cook, J.E., Faint, J.M., Belaramani, L.L.,
Coll, J., de Launoit, Y., Auriault, C., Pancre, V., Delhem, N., 2012. EBV latency II- Fletcher, J.M., Hammerschmitt, N., Rustin, M., Bergler, W., Beverley, P.C.,
derived peptides induce a specific CD4+ cytotoxic T-cell activity and not a CD4+ Salmon, M., Akbar, A.N., 2004. Integration of apoptosis and telomere erosion in
regulatory T-cell response. J. Immunother. 35, 254–266. virus-specific CD8+ T cells from blood and tonsils during primary infection. Blood
Munz, C., 2020. Redirecting T cells against Epstein-Barr virus infection and associated 103, 162–167.
oncogenesis. Cells 9. Solomon, S.R., Sizemore, C.A., Zhang, X., Brown, S., Holland, H.K., Morris, L.E.,
Munz, C., Bickham, K.L., Subklewe, M., Tsang, M.L., Chahroudi, A., Kurilla, M.G., Bashey, A., 2014. Preemptive DLI without withdrawal of immunosuppression to
Zhang, D., O’Donnell, M., Steinman, R.M., 2000. Human CD4(+) T lymphocytes promote complete donor T-cell chimerism results in favorable outcomes for high-risk
consistently respond to the latent Epstein-Barr virus nuclear antigen EBNA1. J. Exp. older recipients of alemtuzumab-containing reduced-intensity unrelated donor
Med. 191, 1649–1660. allogeneic transplant: a prospective phase II trial. Bone Marrow Transplant. 49,
Murakami, M., Hashida, Y., Imajoh, M., Maeda, A., Kamioka, M., Senda, Y., Sato, T., 616–621.
Fujieda, M., Wakiguchi, H., Daibata, M., 2014. PCR array analysis of gene expression Speck, S.H., Ganem, D., 2010. Viral latency and its regulation: lessons from the gamma-
profiles in chronic active Epstein-Barr virus infection. Microbes Infect. 16, 581–586. herpesviruses. Cell Host Microbe 8, 100–115.
Murata, T., 2014. Regulation of Epstein-Barr virus reactivation from latency. Microbiol. Steigerwald-Mullen, P., Kurilla, M.G., Braciale, T.J., 2000. Type 2 cytokines predominate
Immunol. 58, 307–317. in the human CD4(+) T-lymphocyte response to Epstein-Barr virus nuclear antigen
Ning, R.J., Xu, X.Q., Chan, K.H., Chiang, A.K., 2011. Long-term carriers generate Epstein- 1. J. Virol. 74, 6748–6759.
Barr virus (EBV)-specific CD4(+) and CD8(+) polyfunctional T-cell responses which Steven, N.M., Leese, A.M., Annels, N.E., Lee, S.P., Rickinson, A.B., 1996. Epitope focusing
show immunodominance hierarchies of EBV proteins. Immunology 134, 161–171. in the primary cytotoxic T cell response to Epstein-Barr virus and its relationship to T
Odumade, O.A., Hogquist, K.A., Balfour Jr., H.H., 2011. Progress and problems in cell memory. J. Exp. Med. 184, 1801–1813.
understanding and managing primary Epstein-Barr virus infections. Clin. Microbiol. Steven, N.M., Annels, N.E., Kumar, A., Leese, A.M., Kurilla, M.G., Rickinson, A.B., 1997.
Rev. 24, 193–209. Immediate early and early lytic cycle proteins are frequent targets of the Epstein-
Ohga, S., Nomura, A., Takada, H., Ihara, K., Kawakami, K., Yanai, F., Takahata, Y., Barr virus-induced cytotoxic T cell response. J. Exp. Med. 185, 1605–1617.
Tanaka, T., Kasuga, N., Hara, T., 2001. Epstein-Barr virus (EBV) load and cytokine Subspecialty Group of Infectious Diseases and National Children’s Epstein-Barr Virus
gene expression in activated T cells of chronic active EBV infection. J. Infect. Dis. Infection Cooperative G, 2021. Experts consensus on diagnosis and treatment of
183, 1–7. Epstein-Barr virus infection-related diseases in children. Zhonghua Er Ke Za Zhi 59,
Ohga, S., Nomura, A., Takada, H., Tanaka, T., Furuno, K., Takahata, Y., Kinukawa, N., 905–911.
Fukushima, N., Imai, S., Hara, T., 2004. Dominant expression of interleukin-10 and Takeuchi, A., Saito, T., 2017. CD4 CTL, a cytotoxic subset of CD4(+) T cells, their
transforming growth factor-beta genes in activated T-cells of chronic active Epstein- differentiation and function. Front. Immunol. 8, 194.
Barr virus infection. J. Med. Virol. 74, 449–458. Tamaki, H., Beaulieu, B.L., Somasundaran, M., Sullivan, J.L., 1995. Major
Okano, M., Kawa, K., Kimura, H., Yachie, A., Wakiguchi, H., Maeda, A., Imai, S., histocompatibility complex class I-restricted cytotoxic T lymphocyte responses to
Ohga, S., Kanegane, H., Tsuchiya, S., Morio, T., Mori, M., Yokota, S., Imashuku, S., Epstein-Barr virus in children. J. Infect. Dis. 172, 739–746.
2005. Proposed guidelines for diagnosing chronic active Epstein-Barr virus infection. Tamura, Y., Yamane, K., Kawano, Y., Bullinger, L., Wirtz, T., Weber, T., Sander, S.,
Am. J. Hematol. 80, 64–69. Ohki, S., Kitajima, Y., Okada, S., Rajewsky, K., Yasuda, T., 2022. Concomitant
Padilla-Quirarte, H.O., Badillo-Godinez, O., Gutierrez-Xicotencatl, L., Acevedo- cytotoxic effector differentiation of CD4(+) and CD8(+) T cells in response to EBV-
Betancur, Y., Luna-Andon, J.D., Montiel-Hernandez, J.L., Lopez-Guerrero, D.V., infected B cells. Cancers (Basel) 14.
Esquivel-Guadarrama, F., 2019. Targeting M2e to DEC-205 induces an enhanced Tomita, Y., Avila-Cariño, J., Yamamoto, K., Mellstedt, H., Klein, E., 1998. Recognition of
serum antibody-dependent heterosubtypic protection against influenza A virus B-CLL cells experimentally infected with EBV by autologous T lymphocytes.
infection. Vaccine 37, 2624–2633. Immunology. letters 60 (2–3), 73–79. https://doi.org/10.1016/s0165-2478(97)
Perez, E.M., Foley, J., Tison, T., Silva, R., Ogembo, J.G., 2017. Novel Epstein-Barr virus- 00142-9.
like particles incorporating gH/gL-EBNA1 or gB-LMP2 induce high neutralizing Tsao, S.W., Tsang, C.M., Pang, P.S., Zhang, G., Chen, H., Lo, K.W., 2012. The biology of
antibody titers and EBV-specific T-cell responses in immunized mice. Oncotarget 8, EBV infection in human epithelial cells. Semin. Cancer Biol. 22, 137–143.
19255–19273. Tsuchiya, S., 2002. Diagnosis of Epstein-Barr virus-associated diseases. Crit. Rev. Oncol.
Precopio, M.L., Sullivan, J.L., Willard, C., Somasundaran, M., Luzuriaga, K., 2003. Hematol. 44, 227–238.
Differential kinetics and specificity of EBV-specific CD4+ and CD8+ T cells during Tsuge, I., Morishima, T., Kimura, H., Kuzushima, K., Matsuoka, H., 2001. Impaired
primary infection. J. Immunol. 170, 2590–2598. cytotoxic T lymphocyte response to Epstein-Barr virus-infected NK cells in patients
Price, A.M., Luftig, M.A., 2014. Dynamic Epstein-Barr virus gene expression on the path with severe chronic active EBV infection. J. Med. Virol. 64, 141–148.
to B-cell transformation. Adv. Virus Res. 88, 279–313. Tsurumi, T., Fujita, M., Kudoh, A., 2005. Latent and lytic Epstein-Barr virus replication
Qian, J., Yu, Q., Chen, G., Wang, M., Zhao, Z., Zhang, Y., Qiu, L., 2020. Altered ratio of strategies. Rev. Med. Virol. 15, 3–15.
circulating follicular regulatory T cells and follicular helper T cells during primary Tugizov, S.M., Berline, J.W., Palefsky, J.M., 2003. Epstein-Barr virus infection of
EBV infection. Clin. Exp. Med. 20, 373–380. polarized tongue and nasopharyngeal epithelial cells. Nat. Med. 9, 307–314.
Qiang, Q., Zhengde, X., Shuang, Y., Kunling, S., 2012. Prevalence of coinfection in Valentine, R., Dawson, C.W., Hu, C., Shah, K.M., Owen, T.J., Date, K.L., Maia, S.P.,
children with Epstein-Barr virus-associated hemophagocytic lymphohistiocytosis. Shao, J., Arrand, J.R., Young, L.S., O’Neil, J.D., 2010. Epstein-Barr virus-encoded
J. Pediatr. Hematol. Oncol. 34, e45–e48. EBNA1 inhibits the canonical NF-kappaB pathway in carcinoma cells by inhibiting
Rajnavolgyi, E., Nagy, N., Thuresson, B., Dosztanyi, Z., Simon, A., Simon, I., Karr, R.W., IKK phosphorylation. Mol. Cancer 9, 1.
Ernberg, I., Klein, E., Falk, K.I., 2000. A repetitive sequence of Epstein-Barr virus van Gent, M., Gram, A.M., Boer, I.G.J., Geerdink, R.J., Lindenbergh, M.F.S., Lebbink, R.
J., Wiertz, E.J., Ressing, M.E., 2015. Silencing the shutoff protein of Epstein-Barr

12
M. Liu et al. Infection, Genetics and Evolution 112 (2023) 105443

virus in productively infected B cells points to (innate) targets for immune evasion. Xing, Y., Song, H.M., Wei, M., Liu, Y., Zhang, Y.H., Gao, L., 2013. Clinical significance of
J. Gen. Virol. 96, 858–865. variations in levels of Epstein-Barr virus (EBV) antigen and adaptive immune
van Zyl, D.G., Tsai, M.H., Shumilov, A., Schneidt, V., Poirey, R., Schlehe, B., Fluhr, H., response during chronic active EBV infection in children. J. Immunotoxicol. 10,
Mautner, J., Delecluse, H.J., 2018. Immunogenic particles with a broad antigenic 387–392.
spectrum stimulate cytolytic T cells and offer increased protection against EBV Yanagaisawa, R., Matsuda, K., Ohga, S., Kanegane, H., Morimoto, A., Okamoto, Y.,
infection ex vivo and in mice. PLoS Pathog. 14, e1007464. Ohara, A., Fukushima, K., Sotomatsu, M., Nomura, K., Saito, A.M., Horibe, K.,
Wada, T., Muraoka, M., Yokoyama, T., Toma, T., Kanegane, H., Yachie, A., 2013. Ishii, E., Nakazawa, Y., 2019. Factors predicting the recurrence of Epstein-Barr virus-
Cytokine profiles in children with primary Epstein-Barr virus infection. Pediatr. associated hemophagocytic lymphohistiocytosis in children after treatment using the
Blood Cancer 60, E46–E48. HLH-2004 protocol. Int. J. Hematol. 109, 612–617.
Wang, X., Hutt-Fletcher, L.M., 1998. Epstein-Barr virus lacking glycoprotein gp42 can Yang, D., Shao, Q., Sun, H., Mu, X., Gao, Y., Jiang, R., Hou, J., Yao, K., Chen, Y., Sun, B.,
bind to B cells but is not able to infect. J. Virol. 72, 158–163. 2011. Evaluation of Epstein-Barr virus latent membrane protein 2 specific T-cell
Wang, X., Kenyon, W.J., Li, Q., Mullberg, J., Hutt-Fletcher, L.M., 1998. Epstein-Barr virus receptors driven by T-cell specific promoters using lentiviral vector. Clin. Dev.
uses different complexes of glycoproteins gH and gL to infect B lymphocytes and Immunol. 2011, 716926.
epithelial cells. J. Virol. 72, 5552–5558. Yang, C., Zhu, X., Zhang, T., Ye, Q., 2017. EBV-HLH children with reductions in CD4+ T
Wang, Y., Aissi-Rothe, L., Virion, J.M., De Carvalho, Bittencourt M., Ulas, N., cells and excessive activation of CD8+ T cells. Pediatr. Res. 82, 952–957.
Audonnet, S., Salmon, A., Clement, L., Venard, V., Jeulin, H., Stoltz, J.F., Decot, V., Yao, S., Wang, Y., Sun, Y., Liu, L., Zhang, R., Fang, J., Jin, R., Yu, J., Li, F., Bai, J.,
Bensoussan, D., 2014a. Combination of Epstein-Barr virus nuclear antigen 1, 3 and Zeng, Y., Zhang, C., Tan, H., Zhou, F., Chen, Y., Zhang, Q., Wang, Z., 2021.
lytic antigen BZLF1 peptide pools allows fast and efficient stimulation of Epstein- Epidemiological investigation of hemophagocytic lymphohistiocytosis in China.
Barr virus-specific T cells for adoptive immunotherapy. Cytotherapy 16, 122–134. Orphanet J. Rare Dis. 16, 342.
Wang, R.C., Chang, S.T., Hsieh, Y.C., Huang, W.T., Hsu, J.D., Tseng, C.E., Wang, M.C., Yin, Y., Manoury, B., Fahraeus, R., 2003. Self-inhibition of synthesis and antigen
Hwang, W.S., Wang, J., Chuang, S.S., 2014b. Spectrum of Epstein-Barr virus- presentation by Epstein-Barr virus-encoded EBNA1. Science 301, 1371–1374.
associated T-cell lymphoproliferative disorder in adolescents and young adults in Yu, X., Ilecka, M., Bartlett, E.J., Schneidt, V., Bhat, R., Mautner, J., Feederle, R.,
Taiwan. Int. J. Clin. Exp. Pathol. 7, 2430–2437. Delecluse, H.J., 2015. Antigen-armed antibodies targeting B lymphoma cells
Wang, Y., Luo, Y., Tang, G., Ouyang, R., Zhang, M., Jiang, Y., Wang, T., Zhang, X., effectively activate antigen-specific CD4+ T cells. Blood 125, 1601–1610.
Yin, B., Huang, J., Wei, W., Huang, M., Wang, F., Wu, S., Hou, H., 2021. HLA-DR Zhang, S., Yin, J., Zhong, J., 2017. Chaetocin reactivates the lytic replication of Epstein-
expression level in CD8(+) T cells correlates with the severity of children with acute Barr virus from latency via reactive oxygen species. Sci. China Life Sci. 60, 66–71.
infectious mononucleosis. Front. Immunol. 12, 753290. Zhong, L., Krummenacher, C., Zhang, W., Hong, J., Feng, Q., Chen, Y., Zhao, Q., Zeng, M.
Wingate, P.J., McAulay, K.A., Anthony, I.C., Crawford, D.H., 2009. Regulatory T cell S., Zeng, Y.X., Xu, M., Zhang, X., 2022. Urgency and necessity of Epstein-Barr virus
activity in primary and persistent Epstein-Barr virus infection. J. Med. Virol. 81, prophylactic vaccines. NPJ Vaccines 7, 159. https://doi.org/10.1038/s41541-022-
870–877. 00587-6.
Womack, J., Jimenez, M., 2015. Common questions about infectious mononucleosis. Am.
Fam. Physician 91, 372–376.

13

You might also like