You are on page 1of 9

Journal of Clinical Virology 102 (2018) 84–92

Contents lists available at ScienceDirect

Journal of Clinical Virology


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

Review

Primary Epstein-Barr virus infection T


a b a,b,⁎
Samantha K. Dunmire , Priya S. Verghese , Henry H. Balfour Jr.
a
Department of Laboratory Medicine and Pathology, USA
b
Department of Pediatrics, University of Minnesota Medical Center, Minneapolis, MN 55455, USA

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

Keywords: Epstein-Barr virus (EBV) infects about 90% of adults worldwide. It is the main cause of infectious mononucleosis,
Epstein-Barr virus which is observed most frequently in adolescents. The disease can last several weeks and is characterized by
Primary infection lymphocytosis, sore throat, lymphadenopathy, and fatigue. Exposure to oral secretions during deep kissing has
Infectious mononucleosis been identified as the major source for primary EBV infection in adolescents. Oral secretions are also thought to
Mono
be the source for younger children through intimate intact or sharing food and eating utensils, although this has
Vaccine
not been confirmed. Unlike most acute viral illnesses such as influenza, the incubation period of symptomatic
Immune responses
primary EBV infection is unusually long, lasting about six weeks. Diagnosis is typically made by heterophile
antibody tests and/or EBV-specific antibody tests. Long-term consequences may result from acquisition of the
virus, including nasopharyngeal carcinoma and lymphomas. Nevertheless, there remains a surprising dearth of
knowledge regarding the establishment of an immune response to persistent EBV infection, especially during the
incubation period. This lack of knowledge has impaired our ability to develop an effective prophylactic EBV
vaccine, despite various attempts. Our greatest challenges in EBV research are to develop a prophylactic vaccine
and devise treatment strategies for persons already infected with EBV.

1. Introduction of the world's population has been infected by the time they reach
adulthood [1,12,13]. The prevalence of EBV infection in children varies
Epstein-Barr virus (EBV) is arguably the most ubiquitous of human from 10 to 90%, depending on their age, geographic location, and race/
viruses, infecting at least 90% of adults worldwide [1]. It is also the first ethnicity [1,13–15]. For example, the age-specific prevalence of EBV
human cancer virus, having been recognized as the cause of endemic antibodies among white, non-Hispanic children tested in 2011 in the
Burkitt lymphoma in the 1960s [2–4]. In addition to Burkitt lymphoma, Minneapolis-St. Paul metropolitan area did not reach 50% until age 18,
EBV is implicated in the pathogenesis of Hodgkin lymphoma [5], na- whereas 50% of non-white children were antibody positive by the age
sopharyngeal carcinoma [6,7], gastric cancer [8], and a myriad of of 10 [15].
malignancies in individuals with inherited or acquired im- Children may be acquiring primary EBV infection at a later age in
munodeficiency [9]. EBV is also considered to be a major environ- developed countries. Among entering freshmen at the University of
mental risk factor for multiple sclerosis [10]. Despite the enormous Minnesota, EBV antibody prevalence declined from 64% in 2006 to
disease burden attributed to EBV infection, we do not yet have a li- 52% in 2012 [16,17]. This is consistent with data from the National
censed vaccine to prevent it, or an approved treatment for it. Health and Nutrition Examination Surveys (NHANES). In NHANES,
EBV antibody prevalence among US children 6–19 years old declined
2. Epidemiology from an average of 72% during 2003–2004 to 65% during 2009–2010
[13]. The decline in antibody prevalence has averaged approximately
2.1. Prevalence of EBV infection 2% annually. A decrease in primary EBV infection especially among
young children has also been observed in England and Wales [18] and
The discovery of EBV was reported in 1964 [2,3]. Shortly after that, Japan [19]. The explanation for this could be improvement in socio-
methods to detect EBV-specific antibodies were devised [6,11] and economic status, since higher household income and education level
applied to cross-sectional studies, which showed that the vast majority were significantly associated with a lower prevalence of EBV antibody

Abbreviations: CMV, cytomegalovirus; EBV, Epstein-Barr virus; EBNA, Epstein-Barr virus nuclear antigen; LMP, latent membrane protein; NHANES, National Health and Nutrition
Examination Surveys; VCA, viral capsid antigen

Corresponding author at: MMC 609, 420 Delaware St SE, University of Minnesota, Minneapolis, MN, 55455, USA.
E-mail address: balfo001@umn.edu (H.H. Balfour).

https://doi.org/10.1016/j.jcv.2018.03.001
Received 3 January 2018; Received in revised form 2 March 2018; Accepted 3 March 2018
1386-6532/ © 2018 Elsevier B.V. All rights reserved.
S.K. Dunmire et al. Journal of Clinical Virology 102 (2018) 84–92

in the NHANES study [13]. This trend is important to monitor because period of continuous oral shedding was 28 days and the longest was 3.1
if the trend continues, the number of adolescents and young adults years.
susceptible to infectious mononucleosis in developed countries will
increase. 3.2. Sexual intercourse

2.2. Incidence of primary EBV infection and infectious mononucleosis Sexual intercourse has been reported to enhance transmission [35].
among adolescents and young adults However, our University of Minnesota prospective studies found that
subjects who engaged in deep kissing with or without penetrative
The development of EBV-specific antibody tests enabled serosurveys sexual intercourse had the same higher risk of primary EBV infection
to be conducted in college students and military recruits, which es- throughout their undergraduate years as compared with subjects who
tablished unequivocally that EBV was the cause of infectious mono- reported no kissing and no sex [16,17].
nucleosis [20–25]. The annual incidence of primary EBV infection and
the proportion of those subjects diagnosed with infectious mono- 3.3. Blood transfusion
nucleosis varied widely, most likely because the study protocols and
populations recruited were quite different. Of interest, the investigation Blood products have been reported to transmit EBV infection, which
of the Yale class of 1973 found that during their freshman year, 17 in some instances results in an infectious mononucleosis syndrome
(74%) of 23 students who acquired primary EBV infections experienced [36–38]. The risk appears to be low, but the exact incidence is not
infectious mononucleosis [23]. This is exactly the same proportion of known, likely because most adults who receive blood products remain
infectious mononucleosis we found among our University of Minnesota asymptomatic if they are superinfected due to residual immunity from a
students studied some 3 decades later (described in Section 4.2 below). previous EBV infection.

2.3. Relevance of age of acquisition of primary EBV infection 3.4. Allograft transplantation

There is a complex relationship between age of acquisition of pri- Transmission of EBV from donor to recipient has been documented
mary EBV, severity of the acute illness, and subsequent risk of EBV- in hematopoietic cell transplantation [39] and solid organ transplan-
spurred cancers or autoimmune diseases. Infectious mononucleosis tation [40]. Shapiro et al. demonstrated donor to recipient transmission
occurs in preadolescents, although it is less common than in adolescents of EBV during hematopoietic cell transplantation by comparing re-
and young adults. Interestingly, younger age at the time of primary EBV striction fragment length polymorphisms of EBV strains in blood, bone
infection among Kenyan infants was associated with elevated levels of marrow, and tissue [39]. Verghese and colleagues used sequence var-
EBV viremia throughout infancy, leading the investigators to postulate iation in the EBV latent membrane protein 1 (LMP-1) gene to document
that these infants were at higher risk for endemic Burkitt lymphoma transmission of EBV from a living, unrelated kidney donor to an EBV-
[26,27]. Melbye and colleagues found that Greenland Eskimo children naïve, 16-year-old boy, who subsequently developed posttransplant
acquired primary EBV infection at an earlier age and had higher titers lymphoproliferative disorder [40].
of IgG antibody against EBV viral capsid antigen (VCA) than age-mat-
ched Danish children [28]. The authors speculated that early infection 3.5. Close contact with household members or caregivers?
with “a large inoculum of EBV” explained why Eskimos were at high
risk for nasopharyngeal carcinoma versus Danes who were not. Finally, How preadolescent children contract EBV is unknown. A reasonable
early acquisition of EBV is a significant risk for MS in childhood [29]. supposition is that they are infected by their parents, siblings, other
On the other hand, late acquisition of primary EBV infection by household members, or caregivers who are “carriers” of the virus and
adolescents and young adults is thought to result in infectious mono- shed EBV periodically into their oral secretions [41,42]. An example of
nucleosis more often than it does in children [30], although there are no this is the very early acquisition of EBV among three distinct Melane-
hard data to support this. Studies performed on cohorts of children in sian populations whose infants have multiple caregivers that pre-
Uganda and The Gambia suggest that young children seroconvert masticate the baby’s food [43]. Another paper studying child care in
asymptomatically [31,32]. Jayasooriya et al. showed that infants under South Africa also mentions premastication of food as a way by which
two years old who acquire the virus were observed to have activated children may be exposed to saliva, in addition to cleaning the child’s
EBV-specific T cells and viral loads comparable to adolescents pre- face or mouth with saliva [44]. Sharing items such as eating utensils,
senting with infectious mononucleosis, but lacked simultaneous lym- drinking glasses, toothbrushes, or toys with an infected person has been
phocytosis at the time periods examined [31]. It is important to note, implicated but not proven to be a route of transmission.
however, that in patients with infectious mononucleosis EBV-specific T
cell activation persists for a number of months. Indeed, VCA IgM titers 4. Clinical features
are highest in the so-called “subacute” phase, after bulk CD8 T cells
have already largely contracted [16,33]. 4.1. Incubation period

3. Routes of transmission The incubation period for asymptomatic primary EBV infections is
nearly impossible to define. For infectious mononucleosis, Hoagland’s
3.1. Deep kissing clinical records suggested an incubation period of 32–49 days, based on
the dates of kissing episodes until the onset of symptoms [34]. A well-
EBV infection among adolescents and young adults is acquired documented Swedish case of infectious mononucleosis occurred 38 days
primarily by deep kissing as proposed by Hoagland's careful clinical after a kissing event [45]. Behavioral data from our prospectively col-
observations [34] and confirmed decades later by prospective studies lected medical history questionnaires are consistent with an incubation
among undergraduate university students [16,17]. The amount of EBV period of 42 days [16,17].
DNA in washes after primary EBV infection can be extremely high and
may persist for months, which is supportive evidence for this route of 4.2. Clinical findings during the acute infection
transmission. Median EBV oral loads peaked at 4.8 log10 copies/mL
(63,000 copies/mL) a median of 2 months after onset of infection and Three prospective studies of college students have been published
persisted for a median of 5.2 months [16,17]. The shortest documented since the turn of the century [16,17,35]. Crawford and colleagues found

85
S.K. Dunmire et al. Journal of Clinical Virology 102 (2018) 84–92

that 101 (46%) of 241 Edinburgh University students (17–24 years old Table 1
on enrollment) contracted a primary EBV infection during their first 3 Symptoms of infectious mononucleosis in 60 undergraduate students studied pro-
spectively (41 women, 19 men; median age at onset, 19; range, 18–22 years).
undergraduate years, which was an annual infection rate of 15.2% [35].
Twenty-seven subjects (25%) developed infectious mononucleosis, Symptom Number of Median duration
while the remainder were reported to have been asymptomatic. subjects (percent) (days)a
The main difference between the Scottish study and ours was the
Sore throat 59 (98%) 7.5
incidence of symptomatic infection. All laboratory-documented pri-
Swollen or tender cervical 53 (88%) 15.0
mary EBV infections in our prospective studies were assigned to one of lymphadenopathy
three clinical categories according to the following criteria. Infectious Fatigue 47 (78%) 15.5
mononucleosis was an acute illness with at least two of the following Decreased appetite 39 (65%) 10.0
findings: sore throat, cervical lymphadenopathy, fever, fatigue. Headache 38 (63%) 8.0
Felt febrile 32 (53%) 5.5
Symptomatic meant having some symptoms but not fulfilling the cri-
Body aches (myalgia) 30 (50%) 8.0
teria for infectious mononucleosis. Asymptomatic subjects were entirely Upper respiratory symptoms (cough, 29 (48%) 8.0
well during the 3 months after the laboratory diagnosis of primary EBV runny nose, nasal stuffiness)
infection. Of the 81 subjects (18–22 years old) who had primary EBV Abdominal pain 9 (15%) 16.0
infections while under surveillance in our two prospective studies, 60 a
Of symptomatic subjects.
(74%) experienced infectious mononucleosis, 11 (14%) were sympto-
matic, and 10 (12%) were entirely asymptomatic. Why the rate of
and is most likely due to transient penicillin hypersensitivity induced by
symptomatic primary EBV infection was so much higher in the
the immune dysregulation accompanying the acute stage of infection
Minnesota students (74% versus 25%) is not clear. Explanations could
[47]. Subclinical hepatitis documented by elevated levels of alanine
be that the study designs were quite different, the geographic locations
aminotransferase was documented in approximately 75% of our sub-
were different, and years of surveillance were different.
jects, and overt hepatitis with tender hepatomegaly and jaundice de-
The severity of illness (SOI) of the Minnesota students was graded
veloped in 5% of cases.
on a scale from 0 to 6 according to the subjects' reports of the degree of
The assumption has been that the majority of primary EBV infec-
pain and limitation of activities [46]. A score of 0 meant the subject was
tions before puberty are asymptomatic or mild but that is not ne-
entirely asymptomatic, while a score of 6 meant that the subject was
cessarily so. One consideration is that young children, especially those
bedridden. The distribution of maximum SOI scores (Fig. 1) emphasizes
under the age of 4 years, may not develop a positive heterophile anti-
that primary EBV infection in adolescents and young adults is not tri-
body response during primary EBV infection [48]. Unless EBV-specific
vial: 33 subjects (41%) had a maximum SOI score above the modal
assays are performed, the diagnosis will be missed.
score of 3.
A noteworthy aspect of infectious mononucleosis is its long dura-
tion. The median duration of the acute illness was 18 days (mean, 4.3. Complications of the acute illness
19 days; range, 3–54 days) in the 60 subjects with infectious mono-
nucleosis. Table 1 shows the frequency of clinical findings in these Serious complications during the acute phase of primary EBV in-
subjects. Most findings had a median duration of 8 days or less, but fection are uncommon. Complications estimated to occur in at least 1%
fatigue and cervical lymphadenopathy persisted longer. Other findings, of patients with infectious mononucleosis are: airway obstruction due
seen in fewer than 20% of cases, included hepatomegaly, splenomegaly, to oropharyngeal inflammation, meningoencephalitis, streptococcal
nausea, vomiting, palatal petechiae, periorbital and eyelid edema, and pharyngitis, hemolytic anemia, and thrombocytopenia [49–51]. Splenic
rash. Rash occurs more often in patients given penicillin derivatives, rupture is a rare but feared complication [52] that may keep athletes
out of competition for weeks. A reasonable recommendation is that
athletes with infectious mononucleosis may resume contact sports after
3 weeks of illness as long as their acute signs and symptoms have
subsided [53].
We recommend that students gradually return to their normal ex-
ercise level. If strenuous activities are resumed too quickly, clinical
symptoms, especially fatigue, may recur.
Gall bladder involvement during infectious mononucleosis is un-
derappreciated, and usually resolves as the period of generalized im-
mune upregulation subsides [54]. We are aware of patients with in-
fectious mononucleosis who underwent an unnecessary
cholecystectomy. Abdominal pain accompanying the acute illness could
be due to mesenteric lymphadenitis, pressure on the liver or splenic
capsule due to organomegaly, or acalculous cholecystitis.

4.4. Chronic infectious mononucleosis

Primary EBV infection may result in a chronic multisystem disease


in a yet to be determined percentage of patients who do not have a
defined genetic reason for poor control of the virus. Their persistent
antibody profile is: negative VCA IgM indices, very high VCA IgG in-
dices, and modestly elevated EBNA-1 IgG levels. There appear to be two
Fig. 1. Distribution of severity of illness scores in 81 subjects who contracted primary clinical patterns of chronic infectious mononucleosis. The first and
EBV infection during prospective surveillance. Subjects with a score of 0 were entirely more common is recovery from the initial disease but persistent lin-
asymptomatic, whereas subjects with a score of 6 were bedridden. The figure illustrates gering or recurring symptoms that develop months to years later. These
that primary EBV infection in adolescents and young adults is not trivial: 33 subjects patients may report that they have had infectious mononucleosis more
(41%) had a maximum SOI score above the modal score of 3.
than once, but we have not been able to detect acquisition of a different

86
S.K. Dunmire et al. Journal of Clinical Virology 102 (2018) 84–92

Fig. 2. A model of primary EBV infection in the or-


opharynx and periphery. The days relative to
symptom onset were calculated from our 81 pro-
spectively followed students experiencing primary
EBV infection. Epithelial cell derived EBV (purple
hexagons) is transmitted to an EBV naïve host via
infected saliva about six weeks prior to symptom
onset. Viral particles may either infect susceptible B
cells in the crypt of the tonsil or epithelial cells which
later infect B cells. Infected B cells become trans-
formed, resulting in LatencyIII cells. These cells re-
plicate in the germinal center, creating a reservoir of
virus. Some become mature infected B cells, with the
latent membrane proteins augmenting survival sig-
nals to the cell. Latency0 cells are memory-like,
changing their trafficking patterns so they recirculate
between the periphery and oropharynx, with the
median for first detection being five days prior to
symptom onset. Around the same time, EBV specific
CD8 T cells also begin circulating, first becoming
detectable about five days post symptom onset.
Other LatencyIII cells undergo reactivation, produ-
cing virus which infects epithelial cells. Virus is then
shed into the oral cavity.

strain of EBV using LMP-1 sequence variation analysis. We suspect that symptomatic primary EBV infection.
the originally acquired viral strain is the culprit. The second pattern is a
continuous “mono-like” illness that lasts indefinitely. There is no bona 6. Virologic events
vide treatment regimen for these patients, but we have had some an-
ecdotal success with a combination of antiviral drugs and an anti-in- 6.1. Exposure and acquisition
flammatory diet, which are posted on our website (http://z.umn.edu/
ebvdiseases). What conditions are ideal for transmission of EBV are not known. A
higher inoculum of virus would presumably be more likely to result in
4.5. EBV-spurred malignancies and autoimmune disease transmission, but the amount likely varies based on genetic factors of
both the host and the virus itself. Once transmitted, EBV infects oral
More than 50 years after the discovery of EBV, the chronic con- epithelial cells, most likely squamous epithelial cells in the tonsil [58].
sequences of EBV infection are still incompletely understood. EBV is EBV also infects B cells in the lymphatic tissues of Waldeyer’s Ring,
clearly implicated in the etiology of lymphomas, nasopharyngeal car- which include the tonsils.
cinoma, and gastric cancer [2–8]. Also, “There is an undeniable asso- Although it is unknown which cell type is infected first, evidence
ciation between EBV and multiple sclerosis” [55]. exists for a ‘switch tropism’ for host cell types depending on which cells
EBV replicates in. Surface markers including gp42 on salivary virus
5. Genetic influence on acquisition and severity of primary EBV suggest it is derived from oral epithelial cells, making B cells the pri-
infection mary targets during acquisition [59]. These B cells then become
transformed, which may result in malignancies as observed during the
Genetics likely influence the age at which primary EBV is acquired, discovery of EBV [2].
and its severity. Two separate cross-sectional surveys of US children Four possible types of viral latency may occur in B cells after in-
found that early acquisition of EBV infection, as evidenced by having fection. These states are characterized by expression of specific EBV
IgG antibodies against EBV VCA, was a racial disparity that could not be encoded genes. Cells in latency III are the most replicative, expressing
explained solely by socioeconomic factors [13,15]. The enormous dif- all EBV nuclear antigens (EBNAs), LMPs, and EBV encoded small RNAs
ference in overall age- and sex-adjusted EBV antibody prevalence be- (EBER) [60]. In contrast, latency II cells express EBNA-1 and LMP-1 and
tween non-Hispanic white (32%) and non-Hispanic black children −2. These cells are thought to be important precursors to some EBV
(62%) in the study by Condon et al. [15] is consistent with a genetic related malignancies [61]. Latency I involves expression of only
basis for the racial clustering of EBV infection in younger black chil- EBNA1, which serves to help replicate and segregate the viral genome
dren. during cell divisions [62]. A final latency 0 is thought to exist wherein
In terms of the severity of primary EBV infection, Hwang and col- the virally encoded gene expression is completely silenced, allowing for
leagues found that concordance for infectious mononucleosis in infection to persist without detection by the immune system. A model of
monozygotic twins was twice that of dizygotic twins [56]. They stated primary infection is shown in Fig. 2.
that their results were “compatible with a heritable contribution to the
risk of infectious mononucleosis." Rostgaard et al. [57] extended these 6.2. Incubation period
findings by tracking familial clustering of hospitalized cases of in-
fectious mononucleosis. Using large Danish national databases, these Relatively little is known about the incubation period due to the
investigators reported that same-sex twins had a rate ratio of 9.3 for difficulty of obtaining samples from this stage of infection. A few pro-
infectious mononucleosis as compared with 2.3 for first-degree relatives spective studies, however, have allowed some insight into the viral
(opposite-sex twins, siblings and parents), 1.4 for second degree re- progression during acute infectious mononucleosis. Following the in-
latives (half-siblings, grandparents, uncles and aunts) and 1.0 for third- oculating event, virus remains undetectable for the first four to five
degree relatives (first cousins). The 95% confidence intervals for those weeks of the incubation period. We detected EBV in the oral cavity
four classes of relationships did not overlap, supporting the conclusion about a week before onset of symptoms and the peripheral blood ten to
that degree of relatedness increased the likelihood of having a seven days prior to symptom onset [63].

87
S.K. Dunmire et al. Journal of Clinical Virology 102 (2018) 84–92

This result fits with a model whereby latently infected B cells re- bulk CD8 T cells, however, did appear to be affected. In the ten days
plicated slowly, eventually entering germinal centers within the tonsil. prior to symptom onset, bulk CD8 T cells showed significant upregu-
The B cells are then reactivated through B cell receptor signaling aug- lation of activation marker CD38. By contrast, EBV tetramer specific
mented by EBV encoded proteins such as LMP [60]. The terminal dif- CD8 T cells were not activated until symptom onset, indicating the
ferentiation of these cells into mature plasmablasts results in robust response during the incubation period is probably induced by inter-
shedding of virus back into the basal layer of epithelial cells in the oral ferons in the circulation [63]. The lack of circulating tetramer reactive
cavity [64]. cells until symptom onset suggests any EBV-specific cells activated in
the oropharynx must stay within local lymphatic tissue.
6.3. Acute and convalescent phases
7.2. Acute and convalescent phases
The acute phase of infectious mononucleosis is characterized by
explosive viral replication in both the periphery and oral cavity. In the Until recently the innate immune system was an understudied as-
peripheral blood, virus is primarily contained within infected B cells, pect of the overall immune response during infectious mononucleosis.
and only small amounts are released into the plasma [46]. During acute In patients presenting with infectious mononucleosis, a decrease in the
infectious mononucleosis as many as 50% of the circulating resting number of circulating CD56bright immature NK cells was observed and
memory B cells may be infected with EBV, but as the patient con- the proportion of NK cells CD56bright remained significantly depressed
valesces this number declines to between 1 and 50 per 106 B cells [65]. up to 50 days post symptom onset. By contrast, the proportion of NK
A B cell with actively replicating virus may have thousands of copies of cells that were CD56dim NKG2A+ KIR− increases and remained ele-
the EBV genome [66]. Persistent EBV is maintained in latently infected vated well into convalescence [63,74,75]. It has been posited from
resting memory-like B cells at about 5–500 copies per cell [65,67]. work in humanized mice that NK cells are important for early control of
Blood viral loads are highest within two weeks of symptom onset. EBV, particularly in killing lytically infected B cells [76].
The median peak viremia of 7700 copies/mL was reached 8 days after Bridging the gap from innate to adaptive immunity, γd T cells have
onset of symptoms. Blood viral loads > 100,000 copies/mL were ob- also been implicated as having an important role in controlling EBV
served in some of our study participants. Virus can be found in oral during primary infection. γδ T cells exist primarily in the lymphatics of
epithelial cells and in the supernatant from the oral wash, showing that the gut and other mucosal tissues and bear highly conserved γδ T cell
in addition to cells being infected, virus is actively being shed into the receptors [77,78]. The prevalence of EBV related nasopharyngeal car-
oral cavity. Over the following weeks, the amount of virus in the per- cinomas and gastric cancers exist as a potent reminder to the im-
ipheral blood contracts, but elevated viral shedding in the oral cavity portance of controlling EBV-infected epithelial cells at these sites. Re-
can persist for over a year [16,63]. cent findings show that about 50% of the donors tested had γδ T cells
that responded robustly to EBV infected cells in an NKG2D dependent
6.4. Prevalence and pathogenicity of EBNA types manner. Interestingly, only cell lines known to maintain type I latency
such as Daudi elicited activation, which suggests the innate immune
Two EBNA types of EBV exist, differentiated from one another pri- system mounts a layered response to EBV with NK cells killing lytically
marily through differences in the sequence of the EBNA-3 genes [68]. infected cells and γδ targeting latently infected cells [79].
Type 1 is the most common type worldwide, but type 2 has about equal During acute infectious mononucleosis, CD8 T cells undergo ex-
prevalence with type 1 in Africa. No difference has been described to plosive expansion and comprise both EBV-specific and non-EBV-specific
date regarding the pathogenicity of the two types [69]. Seventy-nine of cells [80]. Tetramer positive EBV specific CD8 T cells increase rapidly
our subjects with primary infections had samples available for EBNA in number at symptom onset and bear elevated levels of CD38+ for
typing. EBNA type 1 was present in 69 subjects (87%), EBNA type 2 in 8 several weeks. Similarly, bulk CD8 T cells are also activated and pro-
subjects (10%) and mixtures of the 2 types in 2 subjects (2.5%). There duce Granzyme B, though these cells show an increased activation state
was no correlation between disease severity and EBNA type. for up to six months after symptom onset [63]. The number of CD4 T
cells is not significantly affected, but the cells do become activated and
7. Immune responses respond to EBV MHC class II tetramers [81].
CD4 T cell responses are important for subsequent generation of
7.1. Incubation period antibodies to EBV, as demonstrated by the parallel between im-
munodominant CD4 T cell epitopes and subsequent humoral response.
The initiation of the immune response to EBV occurs around the Work in patients with infectious mononucleosis has further suggested
time the viral load begins to be measurable in the oropharynx and there is a correlation between the kinetics and strength of the antibody
peripheral blood. Starting about two weeks prior to symptom onset, a response to the patient’s overall severity of illness. Those study parti-
systemic type I interferon gene signature was observed in peripheral cipants that had more rapid generation of anti-gp350 antibody had
blood mononuclear cells [63]. By the time patients begin to experience lower severity and duration of illness [17].
symptoms related to the acute illness, this response had waned sig-
nificantly, transitioning to a signature more closely related to hemo- 8. Diagnosis
phagocytic syndromes due to rapid CD8 T cell expansion [70].
Interestingly, the number of circulating plasmacytoid dendritic cells The diagnosis of primary EBV infection relies on laboratory tests.
were drastically reduced coincident with loss of the type I interferon Point-of-care tests are almost always based on detection of heterophile
signature, although whether these cells are killed or sequestered in antibodies. These are IgM class antibodies directed against mammalian
tissue sites remains unknown [63,71]. Furthermore, the type I inter- erythrocytes, which are raised during the generalized immune upre-
feron signature observed lacked certain typical genes such as OAS1 and gulation that accompanies acute primary EBV infection [82]. The het-
MX1. EBV is known to encode miRNAs that interfere with the host's erophile test has been the standard clinical diagnostic tool ever since it
ability to mount certain immune responses [72]. In particular, BART16 was developed in 1932, 32 years before the discovery of EBV. Hetero-
has been shown to have a dampening effect in type I interferon sig- phile tests are a practical method for confirming the clinical diagnosis.
naling in vitro, which may account for the loss of this subset of type I However, they have several limitations. Approximately 40% of children
interferon genes [73]. 4 years of age or younger do not develop heterophile antibodies fol-
No significant difference in proportions of CD8 T cells, CD4 T cells, lowing a primary EBV infection [48]. If the heterophile is the only test
or NK cells was observed prior to symptom onset. The activation state of ordered, the diagnosis may be missed. Second, heterophile antibodies

88
S.K. Dunmire et al. Journal of Clinical Virology 102 (2018) 84–92

by definition are not specific and may be present in infections caused by 9.2. Antiviral prophylaxis
other pathogens, malignancies, or autoimmune diseases [83,84]. Fi-
nally, heterophile antibodies can persist for a year or more and there- Acyclovir, valacyclovir, ganciclovir, and valganciclovir are fre-
fore are not always diagnostic of an acute EBV infection [85]. quently prescribed for 3–6 months after transplantation to prevent or
The most useful EBV-specific antibody tests are VCA IgM, VCA IgG suppress herpesvirus infections, with the major focus being on pre-
and EBNA-1 IgG usually measured by an immunoassay platform. In our vention of CMV disease. Whereas these drugs clearly reduce the in-
prospective studies, 79 of 81 subjects who experienced primary EBV cidence and severity of posttransplant CMV disease [88,89], their role
infections had adequate samples for EBV-specific antibody testing. VCA in the prevention of posttransplant EBV disease is uncertain [90,91]. A
IgM antibodies were detected in 68 (86%) of the 79 subjects. These unique approach is to give antiviral drugs to the donor pretransplant to
antibodies declined over the next 3 months, so this is a good test to prevent donor to recipient transmission of herpesviruses. This has
confirm a primary EBV infection. A caveat is that false-positive EBV shown some promise in a small double-blind, placebo-controlled trial
VCA IgM results have been reported especially with cytomegalovirus [40].
(CMV) infection [86].
The heterophile test is almost as sensitive as the VCA IgM, albeit 9.3. Vaccines
nonspecific. It was positive in 63 (80%) of the 79 subjects. All 79 of our
subjects with primary EBV infection who were followed for at least 3 Three prophylactic EBV vaccine candidates have been tested in
months after onset of infection developed IgG antibodies to VCA. EBV humans. Two are based on gp350, which is the major surface glyco-
VCA IgG antibodies persist indefinitely, so this is an excellent labora- protein of EBV that allows the virus to attach to the CD21 receptor of
tory test to document a previous EBV infection. Antibodies against human B lymphocytes and subsequently infect them.
EBNA-1, which are typically used in commercial diagnostic tests, de- The first candidate was a vaccinia virus construct expressing the
velop slowly and usually are not detectable until 3 months or longer EBV membrane glycoprotein gp220-350 [92]. This trial included 19
after onset of illness [16]. EBV-naïve Chinese children 1–3 years of age. Nine subjects received the
Subjects do not generally present with both VCA IgM and EBNA-1 vaccine as a single dose by scarification and 10 subjects served as
antibodies due to the kinetics of these responses. If both are present, controls. The vaccine was immunogenic and during 16 months of
then the person has likely been infected at least three months. follow-up, 3 of 9 vaccinees versus 10 of 10 subjects in the control group
Therefore, the presence of EBNA-1 antibodies during an acute illness became infected with EBV evidenced by development of antibodies
rules out a recent primary EBV infection. It is important to note, against EBV VCA (not contained in the vaccine). No further develop-
however, that some individuals may not produce EBNA-1 antibodies at ment of this vaccine has been reported, possibly because it contains live
all. About 8% of students who participated in our prospective study did vaccinia, which is associated with potentially serious adverse events
not develop EBNA-1 antibodies [16]. Thus, the absence of EBNA-1 [93].
antibodies is not a definitive indicator that the patient has never been The second candidate was a subunit EBV gp350 vaccine originally
infected before, in which case VCA IgG may be a more reliable surro- developed in Chinese hamster ovary (CHO) cells by Jackman and col-
gate. leagues [94]. This vaccine has been given to humans in four clinical
Other assays are sometimes used. Immunoblotting may include trials and is still under development. Parenthetically, it was recently
immediate early, early, and late EBV antigens on one test strip, which demonstrated that the gp350 vaccine construct must contain a con-
permits evaluation of the stage of EBV infection using a single serum or formationally correct 72A1 epitope to effectively neutralize EBV [95].
plasma sample [87]. Our virology research laboratory uses the Mik- The first human trial with the subunit gp350 vaccine was a phase 1
rogen line blot immunoassay system, which provides a choice of re- study evaluating the safety and immunogenicity of a 3-dose regimen of
agents for detection of IgA, IgG, and IgM class antibodies and IgG an- vaccine given intramuscularly [96]. EBV antibody-negative and anti-
tibody avidity. While valuable from a research perspective, body-positive subjects 18–25 years of age were randomized to receive
immunoblotting is not practical for routine diagnosis of primary EBV the vaccine adjuvanted with 3-O-desacyl-40-monophosphoryl lipid A
infection because of cost and low throughput. Serial quantitative and aluminum salt (AS04) or aluminum salt alone. The second trial was
measurements of EBV DNA in the blood have been used to track post- a phase 1/2 study that randomized EBV-naïve subjects 18–37 years old
transplant EBV infections, but are not necessary to document primary to unadjuvanted vaccine, vaccine adjuvanted with AS04, or vaccine
EBV infections in the otherwise normal host. adjuvanted with aluminum salt only [96]. The combined data from 138
subjects in these two trials showed that the vaccine was safe with one
exception. Ten days after receiving the second dose of vaccine ad-
9. Prevention juvanted with AS04, an EBV antibody-positive subject was hospitalized
for an apparent autoimmune reaction involving the central nervous
9.1. Limiting exposure to EBV system and multiple joints. The immunogenicity data, which included
measurement of gp350 and neutralizing antibodies, indicated that
Avoiding exposure to EBV is nearly impossible, as reflected by the vaccine adjuvanted with AS04 was superior to both non-adjuvanted
very high prevalence of EBV antibodies in adults worldwide. More vaccine and vaccine adjuvanted with aluminum salt.
needs to be learned about how children acquire EBV before appropriate The third trial was a phase 2, placebo-controlled, double-blind
measures of prevention can be recommended for them. Frequent evaluation of the safety, immunogenicity, and efficacy of gp350 vaccine
handwashing and not sharing items such as eating utensils, drinking in EBV-naïve Belgian students ages 16–25 [97]. There were no sig-
glasses, or toothbrushes is good hygiene in general, but there are no nificant adverse events and 76 (98.7%) of 77 vaccine recipients who
solid data that EBV can be transmitted by fomites. were not subsequently infected by wildtype EBV developed gp350 an-
Acquisition of primary EBV infection after transplantation of solid tibodies. The vaccine did not prevent infection: 13 (14%) of 90 vaccine
organs or hematopoietic cells could be prevented, at least in part, by recipients became infected versus 18 (20%) of 91 placebo subjects.
selecting EBV-naïve donors for EBV-naïve recipients. Because at least However, it had a significant effect on clinical disease. In the intent-to-
90% of adults are antibody-positive, identifying a suitably matched treat population, infectious mononucleosis developed in 2 (2%) of 90
antibody-negative donor is difficult. Even if an EBV-naïve donor could vaccinees as compared with 9 (10%) of 91 placebo recipients (P = 0.03,
be found, the virus still might be acquired by the natural route after Fisher exact test, 1-sided).
transplantation. The fourth trial of the gp350 subunit vaccine was a phase 1 study of
gp350 with an aluminum hydroxide adjuvant conducted in 16 pediatric

89
S.K. Dunmire et al. Journal of Clinical Virology 102 (2018) 84–92

renal transplant candidates [98]. All 13 evaluable subjects mounted an prove that active donor EBV replication at time of organ donation in-
antibody response to gp350 but only four made neutralizing antibody. creased risk of PTLD [104].
Because there was no control group, vaccine efficacy could not be as- The clinical features of PTLD can be non-specific and usually in-
sessed. However, this small phase 1 trial did show that immunization of clude fever, weight loss, fatigue, and malaise. PTLD may also present as
children awaiting transplantation for chronic renal disease is safe and lymph node enlargement anywhere in the body including in the gas-
feasible. trointestinal tract, brain, liver, kidney or in SOT recipients, it may be a
The third prophylactic EBV vaccine candidate given to humans was mass in the graft itself. Extra-nodal PTLD is also common. Computed
an adjuvanted EBNA-3A peptide restricted by HLA B8, which was de- tomography and positron emission tomography imaging can reveal
signed to control expansion of EBV-infected B cells by generating CD8 masses in the abdomen, thorax, and central nervous system, but a tissue
T-cell immunity to EBNAs [99]. EBV-naïve individuals were immunized diagnosis is required for confirmation of PTLD. Therapy is determined
on a two-month interval schedule. Of the 14 enrolled subjects, four by World Health Organization classification: early lesions; polymorphic
received placebo, two were immunized with 50 μg dose of peptide and PTLD; monomorphic PTLD (which includes B and T cell non-Hodgkin
the remaining 8 individuals were immunized with a 5 μg dose of pep- lymphoma); and classical Hodgkin lymphoma [105]. Treatment stra-
tide. This strategy was effective at generating a peptide-specific CD8+ tegies can include reduction of immunosuppression, conversion of im-
T-cell response in most individuals as measured by ex vivo peptide- munosuppression to sirolimus inclusive regimens, rituximab, che-
specific interferon gamma production. Among subjects who subse- motherapy, surgery or radiation, or combinations thereof. There is no
quently acquired wild-type EBV, infectious mononucleosis occurred in proven role for antiviral drugs in PTLD, although some centers still use
1 of 2 subjects in the placebo group versus 0 of 4 in the vaccinated them as additional therapy.
cohort, a trend suggesting that this vaccine might prevent symptomatic Re-transplantation in PTLD survivors is successful with only one
EBV infection. The general utility of epitope vaccines is limited by the reported case of PTLD recurrence [106]. Screening for PTLD has fo-
fact that they only target specific HLA types. Nonetheless epitope vac- cused on measuring quantitative peripheral blood loads of EBV. In
cines might be employed to prevent posttransplant lymphoproliferative general, higher EBV loads are present in PTLD versus non-PTLD cases
disorder (PTLD), because the HLA type of transplant patients is known. [107], although we reported a case without any detectable EBV [40].
Unfortunately, quantitative PCR assays for EBV cannot be used to de-
10. Treatment velop threshold values or patterns for intervention because results vary
widely between centers and assay platforms. Rituximab and cytotoxic T
10.1. Otherwise normal host cell infusions have been attempted as preventive measures in HCT but
their role in PTLD prevention remains debatable. Administration of
The use of corticosteroids in acute infectious mononucleosis is antiviral drugs to prevent PTLD is also of questionable value [108].
somewhat controversial [100]. Most clinicians would prescribe a short
course of corticosteroids for patients with any degree of airway ob- 11. Future challenges
struction, or autoimmune phenomena such as anemia and/or throm-
bocytopenia [51]. Despite being discovered more than 50 years ago, there remain gaps
Several nucleoside analogs have in vitro activity against EBV [101] in our knowledge base about EBV. These gaps urgently need to be filled,
but a clinical benefit has not yet been proven for any of them. Vala- especially regarding the epidemiology and pathogenesis of the virus
cyclovir is worth mentioning because it is generic and has very few side since more than 90% of the population eventually becomes infected. Of
effects. We evaluated valacyclovir (3 g/day for 14 days) in 20 Uni- particular significance is learning more about EBV infection in children.
versity of Minnesota undergraduates with infectious mononucleosis due Relatively little is known regarding transmission of the virus in children
to laboratory-confirmed primary EBV infection [102]. Ten subjects re- under twelve years of age. While it is purported in the medical com-
ceived valacyclovir and 10 received no antiviral drugs. There was a munity that primary infection in this age group is usually asympto-
significant reduction in viral load in the oral compartment but not in matic, more research is needed to fully address this issue. Likewise,
the blood. The number of reported symptoms and the severity of illness further investigation into asymptomatic primary infection in adoles-
were reduced significantly among the valacyclovir recipients as com- cents and young adults warrants attention. Finally, a perennial sticking
pared with the control subjects. Limitations of our study were the small point is the lack of a licensed prophylactic vaccine for EBV or, indeed,
number of subjects enrolled and the lack of a placebo control. These an approved specific treatment following infection. Although we have
results are promising but need to be confirmed in a larger, double-blind, described both vaccine development and antiviral regimens in this re-
placebo-controlled trial. view, the importance of forging ahead in both areas cannot be over-
stated.
10.2. Posttransplant lymphoproliferative disorder
Conflicts of interest
PTLD, a heterogeneous spectrum of disorders including lymphomas,
is a well-recognized complication of both solid organ transplantation The authors, Henry H Balfour, Priya S. Verghese, and Samantha K.
(SOT) and hematopoietic cell transplantation (HCT). PTLD is char- Dunmire, have no potential conflicts of interest or conflicts of interest to
acterized by the abnormal proliferation of lymphoid immune cells in declare.
extrinsically immunosuppressed transplant recipients due to impaired
immune surveillance. EBV is a key pathogenic driver in many cases of Disclosure
PTLD, through known and unknown mechanisms.
After initial infection and lytic cycling, EBV transitions to a latency The authors of this manuscript have no conflicts of interest to de-
gene program in the germinal center to avoid host recognition. Some of clare.
these genes co-stimulate host B cells to proliferate and differentiate. In
immunocompetent hosts, EBV-specific CD8+ effector and memory T Funding
cells control the abnormal proliferation of EBV- infected B cells, but this
ability is lost in immunosuppressed transplant recipients with resulting This article was supported by grants from the University of
B cell proliferation and PTLD [103]. In HCT, the PTLD is most often of Minnesota International Center for Antiviral Research and
donor cell origin and is usually associated with EBV of recipient cell Epidemiology, the Richard M. Schulze Family Foundation, the Randy
origin. The opposite is usually the case in SOT and we were unable to Shaver Cancer Research and Community Fund, and the University of

90
S.K. Dunmire et al. Journal of Clinical Virology 102 (2018) 84–92

Minnesota Foundation. [32] S. Gantt, J. Orem, E.M. Krantz, et al., Prospective characterization of the risk
factors for transmission and symptoms of primary human herpesvirus infections
among Ugandan infants, J. Infect. Dis. 214 (2016) 36–44.
References [33] H.H. Balfour Jr., P. Verghese, Primary Epstein-Barr virus infection: impact of age
at acquisition, coinfection, and viral load, J. Infect. Dis. 207 (2013) 1787–1789.
[1] G. de-The, N.E. Day, A. Geser, et al., Sero-epidemiology of the Epstein-Barr virus: [34] R.J. Hoagland, The transmission of infectious mononucleosis, Am. J. Med. Sci. 229
preliminary analysis of an international study – a review, IARC Sci. Publ. (1975) (1955) 262–272.
3–16. [35] D.H. Crawford, K.F. Macsween, C.D. Higgins, et al., A cohort study among uni-
[2] M.A. Epstein, Y.M. Barr, Cultivation in vitro of human lymphoblasts from Burkitt's versity students: identification of risk factors for Epstein-Barr virus seroconversion
malignant lymphoma, Lancet 1 (1964) 252–253. and infectious mononucleosis, Clin. Infect. Dis. 43 (2006) 276–282.
[3] M.A. Epstein, B.G. Achong, Y.M. Barr, Virus particles in cultured lymphoblasts [36] P. Gerber, J.H. Walsh, E.N. Rosenblum, et al., Association of EB-virus infection
from Burkitt's lymphoma, Lancet 1 (1964) 702–703. with the post-perfusion syndrome, Lancet 1 (1969) 593–595.
[4] M.A. Epstein, G. Henle, B.G. Achong, et al., Morphological and biological studies [37] C. Alfieri, J. Tanner, L. Carpentier, et al., Epstein-Barr virus transmission from a
on a virus in cultured lymphoblasts from Burkitt's lymphoma, J. Exp. Med. 121 blood donor to an organ transplant recipient with recovery of the same virus strain
(1965) 761–770. from the recipient's blood and oropharynx, Blood 87 (1996) 812–817.
[5] H. Hjalgrim, J. Askling, K. Rostgaard, et al., Characteristics of Hodgkin's lym- [38] H. Trottier, C. Buteau, N. Robitaille, et al., Transfusion-related Epstein-Barr virus
phoma after infectious mononucleosis, N. Engl. J. Med. 349 (2003) 1324–1332. infection among stem cell transplant recipients: a retrospective cohort study in
[6] L.J. Old, E.A. Boyse, H.F. Oettgen, et al., Precipitating antibody in human serum to children, Transfusion (Paris) 52 (2012) 2653–2663.
an antigen present in cultured burkitt's lymphoma cells, Proc. Natl. Acad. Sci. [39] R.S. Shapiro, K. McClain, G. Frizzera, et al., Epstein-Barr virus associated B cell
U.S.A. 56 (1966) 1699–1704. lymphoproliferative disorders following bone marrow transplantation, Blood 71
[7] H. zur Hausen, H. Schulte-Holthausen, G. Klein, et al., EBV DNA in biopsies of (1988) 1234–1243.
Burkitt tumours and anaplastic carcinomas of the nasopharynx, Nature 228 (1970) [40] P.S. Verghese, D.O. Schmeling, J.A. Knight, et al., Valganciclovir administration to
1056–1058. kidney donors to reduce the burden of cytomegalovirus and Epstein-Barr virus
[8] D. Shibata, M. Tokunaga, Y. Uemura, et al., Association of Epstein-Barr virus with transmission during transplantation, Transplantation 99 (2015) 1186–1191.
undifferentiated gastric carcinomas with intense lymphoid infiltration. [41] C.V. Sumaya, Y. Ench, Epstein-Barr virus infections in families: the role of children
Lymphoepithelioma-like carcinoma, Am. J. Pathol. 139 (1991) 469–474. with infectious mononucleosis, J. Infect. Dis. 154 (1986) 842–850.
[9] D.A. Thorley-Lawson, A. Gross, Persistence of the Epstein-Barr virus and the ori- [42] K.H. Johnson, C.H. Webb, D.O. Schmeling, et al., Epstein-Barr virus dynamics in
gins of associated lymphomas, N. Engl. J. Med. 350 (2004) 1328–1337. asymptomatic immunocompetent adults: an intensive 6-month study, Clin. Transl.
[10] A. Ascherio, K.L. Munger, J.D. Lunemann, The initiation and prevention of mul- Immunol. 5 (2016) e81.
tiple sclerosis, Nat. Rev. Neurol. 8 (2012) 602–612. [43] D.J. Lang, R.M. Garruto, D.C. Gajdusek, Early acquisition of cytomegalovirus and
[11] G. Henle, W. Henle, Immunofluorescence, interference, and complement fixation Epstein-Barr virus antibody in several isolated Melanesian populations, Am. J.
technics in the detection of the herpes-type virus in Burkitt tumor cell lines, Cancer Epidemiol. 105 (1977) 480–487.
Res. 27 (1967) 2442–2446. [44] L.M. Butler, T.B. Neilands, A. Mosam, et al., A population-based study of how
[12] L.I. Levin, K.L. Munger, E.J. O'Reilly, et al., Primary infection with the Epstein- children are exposed to saliva in KwaZulu-Natal Province, South Africa: implica-
Barr virus and risk of multiple sclerosis, Ann. Neurol. 67 (2010) 824–830. tions for the spread of saliva-borne pathogens to children, Trop. Med. Int. Health.
[13] H.H. Balfour Jr., F. Sifakis, J.A. Sliman, et al., Age-specific prevalence of Epstein- 15 (2010) 442–453.
Barr virus infection among individuals aged 6–19 years in the United States and [45] E. Svedmyr, I. Ernberg, J. Seeley, et al., Virologic, immunologic, and clinical ob-
factors affecting its acquisition, J. Infect. Dis. 208 (2013) 1286–1293. servations on a patient during the incubation, acute, and convalescent phases of
[14] A.R. Venkitaraman, G.M. Lenoir, T.J. John, The seroepidemiology of infection due infectious mononucleosis, Clin. Immunol. Immunopathol. 30 (1984) 437–450.
to Epstein-Barr virus in southern India, J. Med. Virol. 15 (1985) 11–16. [46] H.H. Balfour Jr., C.J. Holman, K.M. Hokanson, et al., A prospective clinical study
[15] L.M. Condon, L.E. Cederberg, M.D. Rabinovitch, et al., Age-specific prevalence of of Epstein-Barr virus and host interactions during acute infectious mononucleosis,
Epstein-Barr virus infection among Minnesota children: effects of race/ethnicity J. Infect. Dis. 192 (2005) 1505–1512.
and family environment, Clin. Infect. Dis. 59 (2014) 501–508. [47] H.H. Balfour Jr., F.A. Forte, R.B. Simpson, et al., Penicillin-related exanthems in
[16] H.H. Balfour Jr., O.A. Odumade, D.O. Schmeling, et al., Behavioral, virologic, and infectious mononucleosis identical to those associated with ampicillin, Clin.
immunologic factors associated with acquisition and severity of primary Epstein- Pediatr. (Phila.) 11 (1972) 417–421.
Barr virus infection in university students, J. Infect. Dis. 207 (2013) 80–88. [48] C.A. Horwitz, W. Henle, G. Henle, et al., Clinical and laboratory evaluation of
[17] J.M. Grimm, D.O. Schmeling, S.K. Dunmire, et al., Prospective studies of infectious infants and children with Epstein-Barr virus-induced infectious mononucleosis:
mononucleosis in university students, Clin. Transl. Immunol. 5 (2016) e94. report of 32 patients (aged 10–48 months), Blood 57 (1981) 933–938.
[18] M.C. Morris, W.J. Edmunds, The changing epidemiology of infectious mono- [49] L.R. White, P.S. Karofsky, Review of the clinical manifestations, laboratory find-
nucleosis, J. Infect. 45 (2002) 107–109. ings, and complications of infectious mononucleosis, Wis. Med. J. 84 (1985)
[19] K. Takeuchi, K. Tanaka-Taya, Y. Kazuyama, et al., Prevalence of Epstein-Barr virus 19–25.
in Japan: trends and future prediction, Pathol. Int. 56 (2006) 112–116. [50] K.P. Connelly, L.D. DeWitt, Neurologic complications of infectious mononucleosis,
[20] A.S. Evans, J.C. Niederman, R.W. McCollum, Seroepidemiologic studies of in- Pediatr. Neurol. 10 (1994) 181–184.
fectious mononucleosis with EB virus, N. Engl. J. Med. 279 (1968) 1121–1127. [51] H.B. Jenson, Acute complications of Epstein-Barr virus infectious mononucleosis,
[21] D.E. Lehane, A seroepidemiologic study of infectious mononucleosis: the devel- Curr. Opin. Pediatr. 12 (2000) 263–268.
opment of EB virus antibody in a military population, JAMA 212 (1970) [52] R.J. Hoagland, H.M. Henson, Splenic rupture in infectious mononucleosis, Ann.
2240–2242. Intern. Med. 46 (1957) 1184–1191.
[22] A Infectious mononucleosis and its relationship to EB virus antibody. A joint in- [53] M. Putukian, F.G. O'Connor, P. Stricker, et al., Mononucleosis and athletic parti-
vestigation by university health physicians and P.H.L.S. laboratories, Br. Med. J. 4 cipation: an evidence-based subject review, Clin. J. Sport Med. 18 (2008)
(1971) 643–646. 309–315.
[23] R.N. Sawyer, A.S. Evans, J.C. Niederman, et al., Prospective studies of a group of [54] C. Iaria, L. Arena, G. Di Maio, et al., Acute acalculous cholecystitis during the
Yale University freshmen: I. Occurrence of infectious mononucleosis, J. Infect. Dis. course of primary Epstein-Barr virus infection: a new case and a review of the
123 (1971) 263–270. literature, Int. J. Infect. Dis. 12 (2008) 391–395.
[24] T.J. Hallee, A.S. Evans, J.C. Niederman, et al., Infectious mononucleosis at the [55] S. Burnard, J. Lechner-Scott, R.J. Scott, EBV and MS: Major cause, minor con-
United States Military Academy: a prospective study of a single class over four tribution or red-herring? Mult. Scler. Relat. Disord. 16 (2017) 24–30.
years, Yale J. Biol. Med. 47 (1974) 182–195. [56] A.E. Hwang, A.S. Hamilton, M.G. Cockburn, et al., Evidence of genetic suscept-
[25] R. Dan, R.S. Chang, A prospective study of primary Epstein-Barr virus infections ibility to infectious mononucleosis: a twin study, Epidemiol. Infect. 140 (2012)
among university students in Hong Kong, Am. J. Trop. Med. Hyg. 42 (1990) 2089–2095.
380–385. [57] K. Rostgaard, J. Wohlfahrt, H. Hjalgrim, A genetic basis for infectious mono-
[26] E. Piriou, A.S. Asito, P.O. Sumba, et al., Early age at time of primary Epstein-Barr nucleosis: evidence from a family study of hospitalized cases in Denmark, Clin.
virus infection results in poorly controlled viral infection in infants from Western Infect. Dis. 58 (2014) 1684–1689.
Kenya: clues to the etiology of endemic Burkitt lymphoma, J. Infect. Dis. 205 [58] B. Chandran, L. Hutt-Fletcher, et al., Gammaherpesviruses entry and early events
(2012) 906–913. during infection, in: A. Arvin, G. Campadelli-Fiume, E. Mocarski (Eds.), Human
[27] J.A. Slyker, C. Casper, K. Tapia, et al., Clinical and virologic manifestations of Herpesviruses: Biology, Therapy, and Immunoprophylaxis, 2007 Cambridge.
primary Epstein-Barr virus (EBV) infection in Kenyan infants born to HIV-infected [59] L.M. Hutt-Fletcher, Epstein-Barr virus entry, J. Virol. 81 (2007) 7825–7832.
women, J. Infect. Dis. 207 (2013) 1798–1806. [60] D.A. Thorley-Lawson, Epstein-Barr virus: exploiting the immune system, Nat. Rev.
[28] M. Melbye, P. Ebbesen, T. Bennike, Infectious mononucleosis in Greenland: a Immunol. 1 (2001) 75–82.
disease of the non-indigenous population, Scand. J. Infect. Dis. 16 (1984) 9–15. [61] L.S. Young, J.R. Arrand, P.G. Murray, et al., EBV gene expression and regulation,
[29] B. Banwell, L. Krupp, J. Kennedy, et al., Clinical features and viral serologies in in: A. Arvin, G. Campadelli-Fiume, E. Mocarski (Eds.), Human Herpesviruses:
children with multiple sclerosis: a multinational observational study, Lancet Biology, Therapy, and Immunoprophylaxis, 2007 Cambridge.
Neurol. 6 (2007) 773–781. [62] M. Rowe, D.T. Rowe, C.D. Gregory, et al., Differences in B cell growth phenotype
[30] S. Krabbe, J. Hesse, P. Uldall, Primary Epstein-Barr virus infection in early reflect novel patterns of Epstein-Barr virus latent gene expression in Burkitt's
childhood, Arch. Dis. Child. 56 (1981) 49–52. lymphoma cells, EMBO J. 6 (1987) 2743–2751.
[31] S. Jayasooriya, T.I. de Silva, J. Njie-jobe, et al., Early virological and im- [63] S.K. Dunmire, J.M. Grimm, D.O. Schmeling, et al., The incubation period of pri-
munological events in asymptomatic Epstein-Barr virus infection in African chil- mary Epstein-Barr virus infection: viral dynamics and immunologic events, PLoS
dren, PLoS Pathog. 11 (2015) e1004746. Pathog. 11 (2015) e1005286.

91
S.K. Dunmire et al. Journal of Clinical Virology 102 (2018) 84–92

[64] L.M. Hutt-Fletcher, The long and complicated relationship between Epstein-Barr cytomegalovirus disease and early death in recipients of solid-organ transplants: a
virus and epithelial cells, J. Virol. 91 (2017) 7825–7832. systematic review of randomised controlled trials, Lancet 365 (2005) 2105–2115.
[65] D. Hochberg, T. Souza, M. Catalina, et al., Acute infection with Epstein-Barr virus [89] A.C. Kalil, J. Levitsky, E. Lyden, et al., Meta-analysis: the efficacy of strategies to
targets and overwhelms the peripheral memory B-cell compartment with resting, prevent organ disease by cytomegalovirus in solid organ transplant recipients,
latently infected cells, J. Virol. 78 (2004) 5194–5204. Ann. Intern. Med. 143 (2005) 870–880.
[66] W.C. Summers, G. Klein, Inhibition of Epstein-Barr virus DNA synthesis and late [90] A. Humar, D. Hebert, H.D. Davies, et al., A randomized trial of ganciclovir versus
gene expression by phosphonoacetic acid, J. Virol. 18 (1976) 151–155. ganciclovir plus immune globulin for prophylaxis against Epstein-Barr virus re-
[67] B. Sugden, M. Phelps, J. Domoradzki, Epstein-Barr virus DNA is amplified in lated posttransplant lymphoproliferative disorder, Transplantation 81 (2006)
transformed lymphocytes, J. Virol. 31 (1979) 590–595. 856–861.
[68] J. Sample, L. Young, B. Martin, et al., Epstein-Barr virus types 1 and 2 differ in [91] J.M. Smith, L. Corey, P.J. Healey, et al., Adolescents are more likely to develop
their EBNA-3A, EBNA-3B, and EBNA-3C genes, J. Virol. 64 (1990) 4084–4092. posttransplant lymphoproliferative disorder after primary Epstein-Barr virus in-
[69] E. Puchhammer-Stockl, I. Gorzer, Cytomegalovirus and Epstein-Barr virus sub- fection than younger renal transplant recipients, Transplantation 83 (2007)
types–the search for clinical significance, J. Clin. Virol. 36 (2006) 239–248. 1423–1428.
[70] S.K. Dunmire, O.A. Odumade, J.L. Porter, et al., Primary EBV infection induces an [92] S.Y. Gu, T.M. Huang, L. Ruan, et al., First EBV vaccine trial in humans using re-
expression profile distinct from other viruses but similar to hemophagocytic syn- combinant vaccinia virus expressing the major membrane antigen, Dev. Biol.
dromes, PLoS One 9 (2014) e85422. Stand. 84 (1995) 171–177.
[71] A. Panikkar, C. Smith, A. Hislop, et al., Cytokine-mediated loss of blood dendritic [93] D.M. Maurer, B. Harrington, J.M. Lane, Smallpox vaccine: contraindications, ad-
cells during Epstein-Barr virus-associated acute infectious mononucleosis: im- ministration, and adverse reactions, Am. Fam. Physician 68 (2003) 889–896.
plication for immune dysregulation, J. Infect. Dis. 212 (2015) 1957–1961. [94] W.T. Jackman, K.A. Mann, H.J. Hoffmann, et al., Expression of Epstein-Barr virus
[72] M. Albanese, T. Tagawa, A. Buschle, et al., MicroRNAs of Epstein-Barr virus gp350 as a single chain glycoprotein for an EBV subunit vaccine, Vaccine 17
control innate and adaptive antiviral immunity, J. Virol. 91 (2017) e01667–16. (1999) 660–668.
[73] M.J.G. Hooykaas, M. van Gent, J.A. Soppe, et al., EBV MicroRNA BART16 sup- [95] E. Servat, B.W. Ro, C. Cayatte, et al., Identification of the critical attribute(s) of
presses type I IFN signaling, J. Immunol. 198 (2017) 4062–4073. EBV gp350 antigen required for elicitation of a neutralizing antibody response in
[74] D.W. Hendricks, H.H. Balfour Jr., S.K. Dunmire, et al., Cutting edge: vivo, Vaccine 33 (2015) 6771–6777.
NKG2C(hi)CD57+ NK cells respond specifically to acute infection with cytome- [96] M. Moutschen, P. Leonard, E.M. Sokal, et al., Phase I/II studies to evaluate safety
galovirus and not Epstein-Barr virus, J. Immunol. 192 (2014) 4492–4496. and immunogenicity of a recombinant gp350 Epstein-Barr virus vaccine in healthy
[75] T. Azzi, A. Lunemann, A. Murer, et al., Role for early-differentiated natural killer adults, Vaccine 25 (2007) 4697–4705.
cells in infectious mononucleosis, Blood 124 (2014) 2533–2543. [97] E.M. Sokal, K. Hoppenbrouwers, C. Vandermeulen, et al., Recombinant gp350
[76] O. Chijioke, A. Muller, R. Feederle, et al., Human natural killer cells prevent in- vaccine for infectious mononucleosis: a phase 2, randomized, double-blind, pla-
fectious mononucleosis features by targeting lytic Epstein-Barr virus infection, cebo-controlled trial to evaluate the safety, immunogenicity, and efficacy of an
Cell. Rep. 5 (2013) 1489–1498. Epstein-Barr virus vaccine in healthy young adults, J. Infect. Dis. 196 (2007)
[77] A.C. Hayday, S. Roberts, E. Ramsburg, Gammadelta cells and the regulation of 1749–1753.
mucosal immune responses, Am. J. Respir. Crit. Care Med. 162 (2000) S161–163. [98] L. Rees, E.J. Tizard, A.J. Morgan, et al., A phase I trial of Epstein-Barr virus gp350
[78] W.K. Born, C.L. Reardon, R.L. O'Brien, The function of gammadelta T cells in in- vaccine for children with chronic kidney disease awaiting transplantation,
nate immunity, Curr. Opin. Immunol. 18 (2006) 31–38. Transplantation 88 (2009) 1025–1029.
[79] Z. Djaoud, L.A. Guethlein, A. Horowitz, et al., Two alternate strategies for innate [99] S.L. Elliott, A. Suhrbier, J.J. Miles, et al., Phase I trial of a CD8+ T-cell peptide
immunity to Epstein-Barr virus: one using NK cells and the other NK cells and epitope-based vaccine for infectious mononucleosis, J. Virol. 82 (2008)
gammadelta T cells, J. Exp. Med. 214 (2017) 1827–1841. 1448–1457.
[80] A.D. Hislop, G.S. Taylor, D. Sauce, et al., Cellular responses to viral infection in [100] B. Candy, M. Hotopf, Steroids for symptom control in infectious mononucleosis
humans: lessons from Epstein-Barr virus, Annu. Rev. Immunol. 25 (2007) (review), Cochrane Database Syst. Rev. 3 (2006) CD004402.
587–617. [101] C.A. Romain, H.H. Balfour Jr., H.E. Vezina, et al., A method for evaluating anti-
[81] H.M. Long, O.L. Chagoury, A.M. Leese, et al., MHC II tetramers visualize human viral drug susceptibility of Epstein-Barr virus, Virus Adapt. Treat. 2 (2010) 1–7.
CD4+ T cell responses to Epstein-Barr virus infection and demonstrate atypical [102] H.H. Balfour Jr., K.M. Hokanson, R.M. Schacherer, et al., A virologic pilot study of
kinetics of the nuclear antigen EBNA1 response, J. Exp. Med. 210 (2013) 933–949. valacyclovir for infectious mononucleosis, J. Clin. Virol. 39 (2007) 16–21.
[82] J.R. Paul, W.W. Bunnell, The presence of heterophile antibodies in infectious [103] V.R. Dharnidharka, Comprehensive review of post-organ transplant hematologic
mononucleosis, Am. J. Med. Sci. 183 (1932) 90–104. cancers, Am. J. Transplant. 18 (2018) 537–549.
[83] C.A. Horwitz, W. Henle, G. Henle, et al., Persistent falsely positive rapid tests for [104] P.S. Verghese, D.O. Schmeling, J.A. Knight, et al., The impact of donor viral re-
infectious mononucleosis. Report of five cases with four–six-year follow-up data, plication at transplant on recipient infections posttransplant: a prospective study,
Am. J. Clin. Pathol. 72 (1979) 807–811. Transplantation 99 (2015) 602–608.
[84] B.A. Fisher, S. Bhalara, False-positive result provided by rapid heterophile anti- [105] N.L. Harris, E.S. Jaffe, J. Diebold, et al., The world health organization classifi-
body test in a case of acute infection with hepatitis E virus, J. Clin. Microbiol. 42 cation of hematological malignancies report of the Clinical Advisory Committee
(2004) 4411. Meeting, Airlie House, Virginia, November 1997, Mod. Pathol. 13 (2000)
[85] J.M. Blake, J.M. Edwards, W. Fletcher, et al., Measurement of heterophil antibody 193–207.
and antibodies to EB viral capsid antigen IgG and IgM in suspected cases of in- [106] S.R. Johnson, W.S. Cherikh, H.M. Kauffman, et al., Retransplantation after post-
fectious mononucleosis, J. Clin. Pathol. 29 (1976) 841–847. transplant lymphoproliferative disorders: an OPTN/UNOS database analysis, Am.
[86] A. Guerrero-Ramos, M. Patel, K. Kadakia, et al., Performance of the architect EBV J. Transplant. 6 (2006) 2743–2749.
antibody panel for determination of Epstein-Barr virus infection stage in im- [107] C.J. Holman, A.B. Karger, B.D. Mullan, et al., Quantitative Epstein-Barr virus
munocompetent adolescents and young adults with clinical suspicion of infectious shedding and its correlation with the risk of post-transplant lymphoproliferative
mononucleosis, Clin. Vaccine Immunol. 21 (2014) 817–823. disorder, Clin. Transplant. 26 (2012) 741–747.
[87] G. Bauer, Simplicity through complexity: immunoblot with recombinant antigens [108] M.A. AlDabbagh, M.R. Gitman, D. Kumar, et al., The role of antiviral prophylaxis
as the new gold standard in Epstein-Barr virus serology, Clin. Lab. 47 (2001) for the prevention of Epstein-Barr virus-associated posttransplant lymphoproli-
223–230. ferative disease in solid organ transplant recipients: a systematic review, Am. J.
[88] E.M. Hodson, C.A. Jones, A.C. Webster, et al., Antiviral medications to prevent Transplant. 17 (2017) 770–781.

92

You might also like