(most material taken with permission from
Prof.Dr.
Jaap M. Middeldorp et al, Crit Rev Oncol Hematol. (2003) ;45(1):1-36)
Table of contents :
Epidemiology
: prevalence > 90%, first cultured in 1964ref
Genomics : like
other herpesviruses, EBV has a toroid-shaped protein core wrapped with
DNA, a nucleocapsid, a protein tegument and an outer envelope. The envelope
carries Gp spikes of which the most abundant structural glycoprotein (Gp)
is Gp350/220. The EBV genome is a linear, double stranded DNA molecule
of 172 kb and has reiterated 0.5 kb terminal repeats and a reiterated 3
kb internal direct repeat. The unique sequences also contain several repeat
elements, often encoding repeat domains in proteins. The BamHI restriction
fragments of the B95.8 laboratory strain genome have been completely sequencedref;
EBV genes are named after the BamHI restriction fragment containing
the RNA start site and their leftward or rightward transcriptional orientation.
BamHI-A
being the largest fragment, BamHI-B the second largest, with
BALF2
being the second leftward reading frame on the BamHI-A fragment
and so on. Subsequent genomic sequencing of additional EBV isolates have
revealed the existence of 2 predominant EBV-strains, named A-type
and B-type (or type-1 and -2, respectively), with significant sequence
polymorphism in the EBNA2 (BYRF1) and EBNA3A-C (BERF1-3) genesref1,
ref2
and minor ‘sub-strain’ variations in the EBNA1 (BKRF1), LMP1 (BNLF1), Zebra
(BZLF1) and various other genesref1,
ref2,
ref3.
In previous studies the A-type virus was found to prevail in most EBV-associated
diseases and showed a more efficient transformation of B-cells in vitroref.
The relative oncogenicity and biological behaviour of different EBV-strains
and variants remains a matter of debateref1,
ref2.
Most viral genomes remain in cells as episomes : rarely integrated in cell
chromosomes. 3 geographic subtypes of EBV have been identified to date
differing by their nuclear antigen EBNA2 :
-
EBNA2 AC strains predominate in Asia
-
EBNA2 AD strains predominate in the USA
-
EBNA2 B strains have all been identified in black Africa.
EBV genome contains
5 miRNAs : computer predictions of the mRNA
targets of the 5 micro RNAs include the tumor suppressor p53, retinoblastoma
binding protein 3, transcription factor E2F1, and genes involved in the
immune response, such as a BCL8 homolog and the ICOS ligand precursor,
a gene expressed in B cells that activates the immune system. The findings
could also explain why EBV is linked to cancers such as
Burkitt's
lymphoma (BL)
and
Hodgkin's
lymphoma (HL)
.
Micro RNAs are expressed quite well in latent infected cells : most EBV
genes are not expressed in latent cells—one of the reasons that the virus
becomes latent—with the exception of a few
latency-associated transcripts
(LAT). One of the micro RNAs,
miR-BART2, has the potential to
target one of the EBV genes that encodes a DNA polymerase : degradation
of that transcript could conceivably contribute to either the establishment
or maintenance of latency
ref. People have found conserved miRNA
sequences in animals and humans and plants—and now they're also in viruses.
Proteomics :
the EBV genome contains over 100 open reading frames (ORFs). These are
named after the
BamHI restriction fragment on which they are located
as indicated above. Only about 50–60% of the gene products have been characterized
to date and much remains to be learned about their pathogenic role
in
vivo. A number of technical developments, such as (multi-primed) RT-PCR
and NASBA, have allowed a sensitive and reliable documentation of EBV transcriptional
activity in most disease syndromes
ref1,
ref2.
Transcription of various subsets of these genes has been well documented
in EBV
+ cell lines and can be detected in cellular samples from
patients with a multitude of EBV-associated acute, chronic and malignant
disease syndromes.
In most EBV proliferative syndromes EBV gene expression
is rather limited and does not involve lytic genome replication or production
of new virions. In EBV-associated malignancies, the virus genome is present
in the individual tumor cells in its latent episomal (closed circular)
form and the viral genome is replicated during each cell division by host
DNA polymerase together with host chromosomes. The detection of latent
episomal EBV-DNA can be used to define tumor and virus clonality and is
taken as proof for early involvement of EBV in tumor development
ref1,
ref2.
Consequently, most EBV disease syndromes are associated with so-called
EBV latency. During such latent infections, the virus remains transcriptionally
active—albeit in a restricted fashion—and the genes expressed then are
referred to as ‘latent genes’
EBV genes and their (putative) functions
&z.cirf;, Positive; , negative; &z.cirf;, positive in some cases;
?, not determined.
-
latent gene products : comprise about 10 different gene products
that are expressed in different transcription profiles (latency types).
During latency, EBV expresses a limited number of viral genes, which are
involved in tasks such as stimulating cell proliferation, inhibiting apoptosis,
blocking viral lytic replication, and assuring accurate and equal partitioning
of the episomal viral genome to daughter cells.
Because most EBV-linked
syndromes are associated with viral latency, the functions of latent gene
products have been studied most extensively
-
EBV-encoded RNAs (EBER1 and EBER2)
are small, non-polyadenylated RNAs with an estimated abundance of 105–107
copies per cell that have a stable secondary structure with extensive intramolecular
base-pairing and some homology to tRNAsref1,
ref2,
Arrand , JR, 145–149. They are localized in the nucleus, where they
are associated with the cellular La antigenClemens, ML, 107–111
and the EBER-associated proteinref.
It has been proposed that they play a role in splicing of other viral transcripts
including primary EBNA and Latent
Membrane Protein (LMP) transcripts. Furthermore, it has been suggested
that EBER1, 2 neutralize the transcription block normally induced by eIF-2ref,
mediate prevention of apoptosisref,
suppress antiviral effects of IFN-a and -g
and induce IL-10 productionref.
These effects may be mediated by the ability of EBER1, 2 to interact with
and inhibit the function of the double stranded RNA-activated protein kinase
PKR, a crucial mediator of interferon-induced antiviral effects. Recently
an independent role in malignant transformation was suggestedref.
-
EBV nuclear antigen (EBNA)1 transcripts can
be derived from 4 different promoters. During the initial stages of primary
infection, (i.e. at the time of recircularization of the genome), transcription
is initiated in BamHI-W (Wp)ref;
once host cell transformation is established, there is a switch towards
the promoter in BamHI-C (Cp)ref.
Primary transcripts from these 2 promoters are very long (ca. 50–70 kb)
and contain coding sequences not only for EBNA1, but also for the other
EBNAsref.
It is thought that these polycistronic transcripts are spliced, resulting
in bi- or monocistronic transcripts, dependent on the needs of the virus
at a certain time point. In latency types I and
II
where EBNA1 is the only EBNA expressed, Cp and Wp are silenced by methylationref
and transcription is initiated from a promoter in BamHI-Q (Qp)ref.
Qp is considered to be the true latent promoter for EBNA1 transcription
and is regulated in a cell cycle dependent manner involving EBNA1 autoregulation
as well as by cytokine-mediated regulationref1,
ref2.
The fourth promoter is localized in BamHI-F (Fp) and is activated
upon entry in the lytic cycleref.
These individual EBNA1 promoters provide different levels of EBNA1 transcripts,
depending on the host cell type and its activation stateref.
EBNA1 is a nuclear protein consisting of a basic amino terminus, a Gly–Ala
repeat segment of variable length, another short basic domain including
a nuclear localization sequence and a long hydrophobic C-terminal domain
with DNA-binding and dimerization activityref1,
ref2.
Recently the crystal structure of the EBNA1 C-terminal dimer domain, bound
to its cognate DNA sequence, was resolved revealing unexpected strong structural
homology with the Bovine Papillomavirus E2 proteinref.
EBNA1 dimers bind to DNA by interacting with
-
a partial palindrome sequence [TAGGATAGCATA-TGCTACCCAGATCCAG] that is present
at 2 sites in the EBV genomeref.
There are 2 high-affinity sites: a set (family) of 20 tandem repeats of
the cognate sequence (FR), and a combination of 2 sequences in dyad symmetry
and 2 in tandem (DS). FR and DS elements are separated by a 1 kb intervening
sequence. Together these form oriP, the origin of plasmid replicationref1,
ref2.
Binding of EBNA1 to these sites results in looping-out of the 1 kb sequence
between FR and DS, thus enabling the replication and persistence of the
episome in cycling cellsref;
at the same time, EBNA1 is associated with host-cell chromosomes during
mitosis which is important for segregation of episomes into progeny
nucleiref1,
ref2,
ref3
-
the third EBNA1 binding site is downstream of the latency I promoter in
BamHI Q. EBNA1 binding to this site represses Qp activity, but this repression
can be overcome by the presence of E2F and thus activity of Qp is cell-cycle
dependentref1,
ref2.
-
EBNA1 furthermore enhances transcription from cellularref
and at least 2 key latency-associated EBV promoters (Cp and LMP1p), which
is correlated with its ability to link distant DNA sequencesref1,
ref2
and binding to host-cell nucleosomal transcription and splicing factors,
such as P32/Tap and EBp2ref1,
ref2,
ref3,
ref4.
In addition to its role(s) in maintenance of the viral episome and promoter
activation, an oncogenic potential has been ascribed to this protein: mice
carrying an EBNA1 transgene under control of the Ig heavy chain enhancer
develop lymphomasref.
Furthermore, the ability of EBNA1 to induce expression of the recombinase-activating
genes RAG-1 and -2 is thought to lead to genetic instabilityref1,
ref2.
These observations may be relevant in the multistep oncogenesis as discussed
above. EBNA1 is the only EBV-protein that is thought to be expressed in
all latently EBV-infected cells. Although EBNA1 is a foreign protein to
the host, EBV-infected cells expressing EBNA1 are not killed by CTLs. This
is due to a inhibitory effect of the protein's Gly/Ala repeat (GAr)
domain on proteasomal processing thereby preventing endogenous MHC
class I-restricted presentation. This is likely to prevent peptide
production from defective ribosomal products (DRiPs) of EBNA1 translation,
a key source of MHC class I presented peptidesref.
Although EBNA1 is barely recognized by CD8+ T-cells, CD4+
T-cells and antibodies reactive with EBNA1 are readily detected in most
healthy virus carriersref1,
ref2.
The latter most likely arise from cross-priming by dendritic cells loaded
with EBNA1 that is acquired via digestion of apoptosed EBV-positive cellsref1,
ref2.
-
EBNA2 is encoded in the long primary transcripts
initiated from Wp and Cpref
and is the first EBV protein to be expressed after delivery of the viral
genome to the nucleus. EBNA2 is a nuclear protein that in vitro specifically
trans-activates cellular genes, such as the B-cell activation markers CD23
and CD21ref
and the c-fgr proto-oncogeneref,
and viral genes including LMP1ref,
LMP2ref
and the cis-acting element upstream of Cpref.
The interaction of EBNA2 with its responsive elements is not a direct one
but instead occurs via the RBP J-k proteinref,
resembling the signaling exerted by the Notch protein pathwayref1,
ref2.
Together with EBNA5, EBNA2
is involved in G0-G1 transitionref.
Significant sequence diversity exists between the EBNA2 proteins of EBV
types 1 and 2, which directly relates to functional differences such as
in vitro transforming activityref.
Although EBNA2 is essential for initial growth transformation of EBV-infected
B-cells in vitroref,
its expression may be less relevant for malignant growth in vivo
because EBNA2 is not expressed in most EBV-associated tumors, except those
in immunocompromized individuals. Even there EBNA2 expression seems to
be limited and rather heterogeneous at the single cell levelref.
-
EBNA leader protein (EBNA-LP,
EBNA5) is encoded in the leader of the Wp driven EBNA mRNAsref
and is expressed simultaneously with EBNA2 in freshly infected B-cellsref.
The ORF is encoded by repeating exons in BamHI-Y and 2 short exons
in BamHI-Y leading to the synthesis of multiple protein species
ranging from 20 to 130 kDaref.
Distinct functional domains on LP were recently mappedref.
Whether the protein is actually synthesized, depends on the splicing of
the mature mRNA. EBNA-LP function and phosphorylation appear to be cell
cycle dependentref [110]. In vitro experiments have indicated
that EBNA-LP is essential for transformation and directly or indirectly
(via EBNA2) upregulates the expression of host factors required for B-cell
growthref1,
ref2
[103 and 111]. Moreover, EBNA-LP was shown to bind p53 and pRb and cellular
PKCs HA95 and HAX1, which regulate transcription by specific phosphorylation
pathways resembling B-cell receptor mediated activationref1, ref2,
ref3, ref4 [103, 112, 113 and 114], leading to G0–G1
transition of resting B-cells. This is an important step in the early stages
of EBV-mediated transformation.
-
EBNA3A (EBNA3), EBNA3B
(EBNA4) and EBNA3C (EBNA6),
like EBNA2 are encoded in the long Wp and Cp driven primary transcriptsref
[64]. Mature mRNA encoding these 3 proteins are the least abundant of all
EBNA mRNAs. EBNA3A, B and C are nuclear proteins of 140, 165 and 155 kDa,
respectively, and localize into large nuclear clusters associated with
the nuclear matrix, but excluding the nucleolusref1, ref2 [115
and 116]. The EBV type 1 and 2 strains show a sequence identity of only
84, 80 and 72%, respectively, mainly due to variations in the C-terminal
repeat regions, thus allowing strain typing. Reverse genetics have shown
that EBNA3B is dispensible for B-cell growth transformationref
[107]. On the other hand EBNA3B expression correlates with upregulation
of CD40 and downregulation of CD77
in vitroref [117].
EBNA3A–C regulate the expression of certain cellular genes and bind to
a variety of host proteins including different isoforms of the cellular
transcription factor RBP J-kref1, ref2
[118 and 119], thus modulating the EBNA2-driven upregulation of cellular
and viral promotersref [120].
-
EBNA3 / EBNA3A induces the nuclear accumulation
of F538, a novel human uridine kinase/uracil phosphoribosyltransferase
that is part of the ribonucleotide salvage pathway. Increased intranuclear
levels of UK/UPRT may contribute to the metabolic build-up that is needed
for blast transformation and rapid proliferationref
-
EBNA3C upregulates CD21 expression
in vitroref
[121], augments the EBNA2-driven upregulation of LMP1 expressionref
[122] and downregulates Cp-promoter activityref [123]. EBNA3C
resembles Adenovirus E1A and Papilloma virus E7 proteins by its ability
to disturb cell cycle checkpoints and can override the G2M checkpoint
imposed by UV irradiation or genotoxic drugs such as etoposide and cisplatin,
thus contributing to accumulation of DNA damageref1, ref2, ref3
[124, 125 and 126]. Because the expression of EBNA3C in EBV-associated
neoplasms in vivo has received little attention thusfar, its role
in supporting tumor outgrowth remains to be determined.
-
LMP1 mRNA originating from the BNLF1 ORF
is a highly abundant viral transcript in most latently infected B-cell
lines. The LMP1 protein is encoded by 3 exons and is an integral membrane
protein with its hydrophilic (small) N- and (large) C-terminus positioned
in the cytoplasm flanking 6 short hydrophobic membrane-spanning -helices
linked by three short reverse turns that are predicted to loop-out on the
extracellular side of the membraneref [127]. LMP1 forms patches
in the cell membraneref [128] and associates with various intracellular
membrane vesiclesref [129] possibly mediated via its association
with the intermediate filament protein vimentinref [130]. Part
of the LMP1 may be secreted from EBV-infected cells in the form of MHC-II
containing secretory vesicles, called exosomesref [131]. The
domains responsible for patch formation comprise the highly conserved N-terminus
and first 2 membrane spanning domainsref [132]. Newly synthesized
LMP1 remains detergent soluble for about 2 h, before it becomes phosphorylated
and relocates to detergent insoluble cytoskeleton-associated intracellular
structuresref1, ref2 [128 and 130]. > 50% of LMP1 is located
intracellularly and even in clonal and synchronized EBV+ B-cell
populations LMP1 expression is highly heterogeneous from cell to cellref
[133]. In some viral strains induction of the viral lytic cycle leads to
a boost of LMP1 expression, which, in part, may be derived from a second
start site on the mRNA, resulting in a 45 kDa truncated form, largely lacking
the transmembrane domains and devoid of most characteristic functions of
LMP1ref [134]. When expressed at low level LMP1 has clear growth
enhancing and oncogenic effectsref [135], but when expressed
at high levels LMP1 causes general cytostasisref [136]. The
cytostatic activity of LMP1 is linked to the first 2 transmembrane domains
of LMP1ref [137], which coincide with the T-cell inhibitory
domain LALLFWL located in the first transmembrane helix at AA34–40ref
[131]. The balanced expression of LMP1 and its relative positioning on
intracellular and plasma membranes in EBV transformed cells may prove to
be relevant for the cell's growth potential. The level of LMP1 expression
in different EBV-infected cell lines in vitro shows considerable
variation similar to the LMP1 expression in EBV carrying tumor cells in
vivoref1, ref2, ref3, ref4 [106, 138, 139 and 140]. The
expression of high levels of LMP1 in vivo has been linked to a worse
prognosisref [141]. LMP1 is considered to be the major EBV-encoded
transforming gene for several reasonsref [39]: transfection
of LMP1 has growth transforming effects in rodent fibroblastsref
[142] and induces many of the changes that are usually associated with
primary EBV-infection and transformation of B-cellsref [143].
Moreover, LMP1 inhibits apoptosis in B-cells by inducing bcl-2ref
[144] and A20ref [145]. In addition, LMP1 was shown to upregulate
the cytokine IL-10 that stimulates B-cell proliferation and inhibits local
immune responseref [146]. In addition, studies with LMP1 transgenic
mice have demonstrated direct oncogenic potential for LMP1. Mice carrying
an LMP1-transgene under control of the Ig heavy chain promoter and enhancer
develop lymphomasref [147]. Upregulation of A20 and Bcl-2
expression in these transgenic mice was observed, in line with the previously
found in vitro effects of LMP1. Mice carrying the LMP1 transgene
under control of the polyoma virus enhancer and promoter (which directs
specific expression in epithelia) show hyperplasia of the epidermisref
[148]. It was suggested that LMP1 expression—probably in association with
aberrant expression of other (host) genes—may predispose to carcinogenesis.
LMP1 was shown to interact with cellular proteins that are mediators of
cytoplasmic signaling from the family of TNFRref [149]. This
interaction is crucial for LMP1 to stimulate cell proliferation, both in
B-cells and epithelial cellsref1, ref2 [150 and 151]. Thus,
LMP-1 mimics the CD40
–CD40L
mediated NF-kB signal transduction pathway by
interacting with specific members of the TRAF's (esp. TRAF1 and TRAF3)
and TRADDref1, ref2 [152 and 153]. LMP1 and CD40 co-localize
in lipid rafts, but LMP1-associated TRAF-mediated signaling seems to be
more efficientref [154]. The TRAF1, 3 interaction leads to NF-kB
activation resulting in morphological changes and enhanced expression of
B-cell activation markers including CD23, CD39, CD40, CD44, MHC class II
and the cellular adhesion molecules LFA-1 and ICAM-1 [155]. LMP1 also induces
enhanced signaling through the JAK–STAT pathway via binding to JAK3 kinase
and ERK MAPK signaling, which involves TRAF2 as signal mediatorref
[156]. All these effects are mediated by at least three distinct signal
activating domains (CTAR1-3) located in the cytoplasmic C-terminal
half of LMP1ref [153]. The hydrophilic N-terminus and
first hydrophobic membrane-anchoring domain of LMP1 are essential for LMP1
functionref [132] and mediate LMP1 association with distinct
glycosphingolipid-rich raft domains in various cell membrane compartmentsref1,
ref2, ref3 [154, 157 and 158]. LMP1 has distinct effects on cytokine
and cytokine receptor synthesisref [159], that may affect angiogenesisref
[160] and inflammatory responses at the single tumor cell level in vivoref
[161], thus contributing to tumor growth and immune escaperef1, ref2,
ref3, ref4 [159, 161, 162 and 163].
-
LMP2A and LMP2B
are encoded by spliced mRNAs transcribed from the region spanning the terminal
repeats of the EBV genome. Therefore, LMP2A, B mRNA is only transcribed
once the circular viral episome is formedref [164]. They
have separate promoters resulting in unique first exons with LMP2B missing
most of the N-terminal domain; the remaining exons are common for LMP2A
and B. The LMP2 proteins both have 12 putative transmembrane domains and
a hydrophilic C-terminal domain and colocalize with LMP1 in raft domainsref
[158]. It was shown that the LMP2A 119 aa N-terminal cytoplasmic domain,
which is lacking in LMP2B, interacts via multiple phosphotyrosines arranged
in ITAM- and SH2-protein binding motifs with the tyrosine kinases Lyn and
Sykref [165]. This interaction diminishes Syk and Lyn from binding
to the cytoplasmic B-cell receptor
signalling domains, thus preventing antigen-receptor mediated activation
of EBV-infected B-cells, that would otherwise result in induction of the
lytic cycle. LMP2 is not required for B-cell transformation and has
no growth altering effects when expressed in differentiating epitheliaref1,
ref2 [39 and 166]. Studies with LMP2A transgenic mice have shown
that LMP2A—although not oncogenic in these mice—can provide an survival
signal to B-cells even when these do not express a B-cell receptorref1,
ref2 [167 and 168]. It was suggested that this signal, in combination
with the B-cell activation-preventing activity of LMP2A, is important for
persistence of EBV in the hostref [165]. This is further supported
by recent data showing a well regulated co-expression of LMP1 and LMP2,
together with EBNA1, in circulating EBV carrying memory B-cells travelling
through lymphoid follicles of the tonsil in vivo. This tightly regulated
expression of LMP1 and LMP2 appears to be important for maintaining long-term
persistence, by providing temporal growth and survival signals during passage
of these cells through lymphoid organsref1, ref2, ref3 [169,
170 and 171].
-
rightward transcripts of the BamHI-A region of the viral genome
(BARTs) can be detected in all types of EBV latency, including EBV-infected
peripheral blood B-cells and are expressed at particularly high levels
in nasopharyngeal carcinomasref1, ref2, ref3, ref4, ref5[172,
173, 174, 175 and 176]. Interestingly, these transcripts are co-terminal
and harbour the BARF0 open reading frame (ORF) at their 3?-endsref1,
ref2, ref3 [177, 178 and 179]. This ORF encodes a putative protein
of 174 amino acids. By means of alternative splicing, the BARF0 ORF may
be extended with 105 additional amino acids. The alternative ORF thus generated
is called RK-BARF0ref [180]. Whether (RK-)BARF0 protein
is expressed in vivo is at present uncertain. The presence of a
polyadenylation signal 5' to the stopcodon renders actual translation unlikelyref1,
ref2 [177 and 179], but anti-BARF0 antibody [180] and cytotoxic T
lymphocyte (CTL)ref [181] responses detected in patients
suggest that synthesis of this protein does take place. However, a publication
from the same group indicated that alternative splicing may remove the
actual BARF0 coding sequences from BART's and suggested that the original
antibody used to detect the (RK-)BARF0 proteins also detects a cellular
component of similar molecular weightref [182]. Work from our
own group, using newly developed monoclonal antibodies reactive in vitro
with various native and denatured recombinant forms of the (RK-)BARF0 protein,
did not result in any evidence for (RK-)BARF0 protein expression in
vivo in multiple EBV+ tumor and EBV-transformed cell line
specimens, despite the clear detection of abundant spliced and non-spliced
RNA transcripts encoding the putative (RK-)BARF0 protein species. In addition,
we could not reproduce the published antibody detection results using purified
(RK-)BARF0 protein or defined peptide domains thereof as antigenref
[183]. Therefore, there remains considerable dispute on a possible role
for (RK-)BARF0 protein EBV-infection in vivo. Recently, in vitro
functional
studies were initiated using heterologous expressed recombinant (RK-)BARF0
protein. One of these studies showed a nuclear localization of the proteinref
[184]. Transfected RK-BARF0 was shown to induce the expression of LMP1,
a mechanism dependent on the apparent interaction between RK-BARF0 and
Notchref [185]. It was hypothesized that this mechanism guarantees
expression of LMP1 in EBNA2-lacking latency types. The RK-BARF0 protein,
however, appears to turn over rapidly with Notchref [185], which
could be the reason why the protein could not be detected in EBV-associated
diseasesref [183]. At least 2 other putative ORFs were demonstrated
in BARTs, called RPMS1 and RPMS2 / A73ref1, ref2, ref3
[173, 186 and 187]. RPMS1 encodes a putative nuclear proteinref1,
ref2 [173 and 186] that is partially homologous to EBNA2, in particular
the WWP containing domain that interacts with RBP J-k.
It is tempting to speculate that RPMS1 acts as a substitute for EBNA2,
for example in latency types I and II. However, recent data point to a
negative influence of the putative RPMS1 protein on EBNA2 and Notch signallingref
[188]. Although transcription of RPMS1 in latently infected B-cells from
healthy donors was foundref [173], neither transcription of
RPMS1 in patient samples nor expression of RPMS1 protein have been reported.
Even less is known about potential expression and function of the A73 protein
which has been suggested to modulate integrin signalling pathwaysref
[186]. Finally, a role for BARTs in transcriptional control was suggested,
by virtue of their anti-sense orientation relative to several important
early–late leftward gene transcripts, such as BALF5 (DNA-polymerase), BALF4
(the nuclear transport membrane protein gp125) and BILF1, a putative cytokine
receptorref1, ref2, ref3, ref4, ref5 [39, 175, 176, 177 and
186]. Thus, BARTs could play a role in maintenance of latency. BARF1 is
a rightward transcript from the BamHI-A region that has its own
promoter. Originally recognized as an early lytic phase transcriptref
[189]. However, recent data indicate that BARF1 is preferentially expressed
in EBV carrying epithelial tumors but not in lymphoid tumorsref1,
ref2, ref3, ref4, ref5 [50, 54, 190, 191 and 192]. This classifies
the BARF1 gene as an interesting and useful carcinoma-specific marker.
The BARF1 protein has transforming properties both when expressed in B-cellsref
[193] and in epithelial cellsref [194]. The transforming domain
of BARF1 may be located in the N-terminus and mediates Bcl2 upregulationref
[195]. The extracellular domain of BARF1 may be selectively cleaved off
and secreted from the cell and is considered to be a functional, soluble
homolog of the human colony stimulating factor 1 (CSF-1) receptorref
[196]. As such, BARF1 is hypothesized to play a role in local immune evasion
by absorbing CSF-1 that is necessary for an effective immune response.
Thus, BARF1 constitutes a protein with pleiotropic function, involved both
in oncogenesis and immune escape with a possible significant role in EBV-associated
carcinomas, like NPC and GC.
Expression patterns of latent genes :
-
latency I (first detected in Burkitt's
lymphoma (BL)
)
: EBNA1, BARTs and EBERs (106 copies per
cells)
-
latency II (first detected in nasopharyngeal
carcinoma (NPC)
but this also proved to be the prevailing expression pattern in most other
EBV-positive tumors that occur in the immunocompetent host, such as Hodgkin's
lymphoma (HL)
and T- and B-cell NHLs) : EBERs, BARTs, EBNA1 + LMP1
+ LMP2A + LMP2B
-
latency III (lymphoblastoid cell lines
(LCLs) in vitro) : EBNA1 + EBNA2
+ EBNA3A + EBNA3C + LP + LMP1
+ LMP2. However, in vivo, expression of the
complete set of latent genes is only found in absence of a fully functional
immune response, i.e. in lymphomas of AIDS patients and of transplant recipients.
Under these circumstances individual tumor cells may display a considerable
degree of expression heterogeneity, overall resembling latency-IIIref1,
ref2 [106 and 201]
-
several other gene expression patterns are recognized that cannot be grouped
with the known latency patternsref [191]
Overview of EBV protein expression (for which suitable antibodies are available)
in EBV-associated diseases
++, Positivity in >75% of neoplastic cells in all tested tumors; +,
positivity in 10–50% of neoplastic cells in most of the cases; ±,
positivity in <5% of neoplastic cells in part of the cases. Abbreviations:
(see also Chapter 1) ePTCL, extranodal peripheral T-cell lymphoma; GC,
gastric carcinoma; nd, not determined; ±, weak positivity in part
of the cases; immunohistochemistry in these cases shows signals in small
percentage of neoplastic and reactive cells; in isolated cases, weak transcription
is found, although not confirmed by immunohistochemistry; therefore, these
signals are ascribed to EBV-infected reactive cells.
-
lytic phase : during the lytic replication
phase of the EBV life cycle, many more viral genes are expressed which
encode proteins involved in viral DNA replication and viral particle synthesis
-
kinases expressed only during the lytic form of infection
-
thymidine kinase
-
BGLF4 phosphorylates the viral early antigen
EA-Dref
-
immediate early (IE)
gene products : genes which are expressed immediately upon induction
of the lytic cycle, independently of new protein synthesis, and which encode
transcription factors that are crucial for activation (switching-on) of
lytic phase genes; essentially considered to form the switch between latent
and lytic cycle. However, IE gene expression cannot be considered synonymous
to full viral lytic replication and on the other hand some late gene products
may be expressed without concomitant IE gene expressionref1, ref2
[39 and 54].
-
BZLF1 / Zebra (Z) : the best studied IE gene
product; a powerful trans-activator of early EBV gene expressionref
[39]. The Zebra protein consists of a trans-activating domain, a basic
domain that has homology to a conserved region of the c-jun/c-fos family
of transcription factorsref [197], and a domain that enables
Zebra to interact with p53ref [198]. Overexpression of p53 and
g-irradiation
have been shown to induce Zebra expression in a NPC-derived EBV carrying
epithelial cell line leading to the (partial) expression of lytic cycle
genesref [199]. In most EBV-associated malignancies sporadic
tumor cells show detectable Zebra protein expression, probably reflecting
partial (abortive) activation of the lytic cycle, because true late gene
transcription is mostly absentref1, ref2, ref3, ref4 [54, 200,
201 and 202]. Expression of Zebra may interfere with IFN signalling, by
decreasing IFN-receptor expression and preventing IFN-induced STAT1 tyrosine
phosphorylation together, resulting in abrogation of IFN-induced MHC-II
upregulation and thereby contributing to immune escaperef [203].
BZLF1 converts the virus from the latent to the lytic form of infection
even when the viral genome is highly methylated. Methylation of CpG motifs
in Z-responsive elements of the viral BRLF1 immediate-early promoter enhances
Z binding to, and activation of, this promoter. Demethylation of the viral
genome impairs Z activation of lytic viral genes. Z is the first transcription
factor that preferentially binds to, and activates, a methylated promoterref.
-
BRLF1, BRRF1 (R)
and the BI-LF4 transactivator genesref1,
ref2 [39 and 204], have been studied only little in relation to EBV+
tumor development and will not be discussed here.
BZLF1 and BRLF1 both encode transcriptional activators, and together these
proteins induce transcription of the entire lytic viral gene program. In
latently infected B cells, the BZLF1 (Zp) and BRLF1 (Rp) promoters are
inactive. However, ligation of the B-cell receptorref,
phorbol ester treatmentref,
calcium ionophoresref,
TGF-ß1ref,
demethylating agentsref,
and agents which induce histone acetylationref1,
ref2,
ref3
are known to activate expression of the BZLF1 and BRLF1 IE promoters in
at least a portion of EBV+ B-cell lines. In the case of the
BZLF1 IE promoter (Zp), the two promoter elements which appear to be essential
for stimulation by most, if not all, of these inducing factors are termed
the ZI and ZII motifsref.
Several of the ZI motifs are bound by the MEF2D cellular transcription
factor, as well as by Sp1/Sp3ref1,
ref2,
and MEF2D has been shown to be an important regulator of EBV infection
in host cellsref1,
ref2.
The ZII motif is a CRE site which is bound by CREB, ATF-1, c-Jun, and ATF-2ref1,
ref2.
The ability of both gemcitabine and doxorubicin to activate BZLF1 transcription
in EBV-negative cells requires both the ZI and ZII binding motifs of the
BZLF1 promoter. Gemcitabine and doxorubicin also activated the BRLF1 IE
promoter in EBV-negative cells. This effect was shown to require the presence
of 2 EGR-1 (Zif268) binding sites in the promoter which were previously
shown to be important for phorbol ester-induced activation of the promoterref.
-
early gene products : genes of
which the expression is not affected by inhibition of viral DNA synthesis
including series of enzymes influencing the host cell nucleotide metabolism
and DNA synthesis; early lytic genes are barely expressed in EBV-associated
malignancies and are not considered to contribute to the oncogenic process,
with the possible exception of BHRF1. However, their expression can be
induced by chemical treatment, irradiation or membrane receptor triggering
of latently infected cells, which is mediated by prior expression of the
EBV IE transactivator protein Zebraref [39]. Enzymes included
among the early lytic genes are potential targets for antiviral drugs which
may lead to applications for future tumor therapyref1, ref2, ref3
[199, 205 and 206].
-
BHRF1 mRNAs are abundantly expressed from their
own promoter in BamHI-H (Hp) during early lytic infectionref
[39]; the protein can also be detected then and relates to the 17 kDa EA-R
antigenref [207]. However, the protein is not detectable in
most latently infected cell linesref [208] and transcripts of
BHRF1 in these cells include leader sequences found in Cp/Wp EBNA transcriptsref
[209]. In vivo, BHRF1 transcripts are predominantly found in EBV-associated
B-cell lymphomasref [210], although expression at the protein
level apparently does not occur significantlyref1, ref2 [211
and 212]. Low level BHRF1 transcription occasionally can be detected in
Hodgkin's
lymphoma (HL)
and in T-cell NHLs and even in nasopharyngeal
carcinoma (NPC)
and BHRF1 mRNA and protein expression is particularly abundant in oral
hairy leukoplakia (OHL)
ref1,
ref2 [54 and 212]. Interestingly, BHRF1 shows structural and functional
homology
to the host-encoded apoptosis inhibitor Bcl-2ref
[213] and is highly conserved among gamma herpesviruses, suggesting an
evolutionary conserved important role for BHRF1-protein in vivoref1,
ref2 [214 and 215]. It is thought that BHRF1 mediated apoptosis-inhibition
contributes to prevent early lysis of productively infected cells by cytotoxic
T-cells thus prolonging virus production time. A deregulated expression
of BHRF1 in EBV tumor cells may similarly contribute to enhanced tumor
cell survival [216].
-
late gene products : genes of which
the expression relies on new linear genomic templates and whose expression
is blocked by inhibition of lytic (linear) viral DNA synthesis, including
-
most of the virion structural proteins. Most late genes that can be identified
directly or based on their homology with other herpesvirus genes encode
structural proteins. Amongst these are :
-
VCA p18 (BFRF3) the small capsid protein
-
VCA-p40 (BdRF1) the scaffold proteinref
[217]
-
Gp125 (BALF4) the nuclear membrane protein
They are strongly immunogenic in humans and serve as targets for
serodiagnosis, because virtually all EBV carriers develop antibodies to
these proteinsref1, ref2 [92 and 218].
BLLF1 encodes the major virion
envelope glycoprotein
Gp350/220 that mediates virion binding to
CD21
/ CR2 / EBVR
and is the major target of the neutralizing antibody responseref
[219].
-
non-structural genes :
-
viral IL-10 (BCRF1)ref1,
ref2
is of particular interest because of its close homology with human IL-10
(hIL10). The structural homology consists of nearly 90% colinear amino
acid sequence identityref [220] and BCRF1 is also functionally
homologous to hIL10ref1, ref2 [221 and 222]. They share the
ability to modulate local immune responses, to inhibit the function of
macrophages and NK cells and the production of interferon-. Similar to
the BHRF1 encoded viral bcl-2 homologue, viral IL-10 probably serves to
enhance survival of virus producing cells in a hostile inflammatory environment.
Both genes were probably acquired by EBV as a consequence of close co-evolution
with its human host within cells of the immune system providing a selective
survival advantage. In vivo, vIL10 (BCRF1) expression is mainly
restricted to OHL in AIDS-patientsref [54]. OHL is a lesion
with a unique combination of latent and lytic EBV gene expression and forms
the only proliferative disorder associated with lytic EBV replicationref
[151]. In contrast, hIL10 expression seems to predominate in most EBV-associated
malignancies, probably directly linked to the expression of LMP1ref
[161]. Overall IL-10 expression is elevated in most EBV-associated diseases
and can be detected both in affected tissues and in serum and is shown
to correlate with a bad prognosisref1, ref2, ref3 [223, 224
and 225].
Transmission :
direct or indirect through oropharynx ("
kissing disease"), blood
or vehicles. Sexually active teens and adults may be exposed to a larger
dose of EBV through particularly "deep" kissing, or possibly through genital
fluids, which can carry the virus. Acquisition of EBV is enhanced by penetrative
sexual intercourse, although transmission could occur through related sexual
behaviors, such as "deep kissing." EBV type 1 infection is significantly
more likely to result in IM. A large EBV type 1 load acquired during sexual
intercourse can rapidly colonize the B cell population and induce the exaggerated
T cell response that causes IM. Thus, IM could, perhaps, be prevented with
a vaccine that reduces the viral load without necessarily inducing sterile
immunity
ref.
Under normal circumstances, EBV-infection is restricted to humans, although
some types of monkeys can be infected experimentally
ref.
EBV enters epithelial cells in the oropharyngeal mucosa through 3 CD21-independent
pathways:
-
by direct cell-to-cell contact of apical cell membranes with EBV-infected
lymphocytes
-
by entry of cell-free virions through basolateral membranes, mediated in
part through an interaction between b1or
a5b1
integrins and the EBV BMRF-2 protein
-
after initial infection, by virus spread directly across lateral membranes
to adjacent epithelial cells.
Pathogenesis :
the finding that individuals with the heritable disorder
X-linked
agammaglobulinemia
harbour no EBV in their blood or throat washings and do not have EBV specific
memory cytotoxic T lymphocyte (CTL) response, indicates that B lymphocytes,
and not oropharyngeal epithelial cells, are required for primary EBV-infection
ref
[226]. At present, it is thought that primary EBV-infection occurs in the
oropharynx via exchange of cell-free virus or productively infected cells
in saliva
ref [241]. The critical steps underlying the initial
EBV B-cell transformation are depicted below :
Schematic presentation of the initial events leading to EBV-induced
transformation of human B-cells. The first steps consist of virion CD21
binding, cell penetration and capsid transport, host cell activation and
viral gene expression following release of the linear viral genome in to
the cell nucleus. Subsequently, a well-orchestrated series of molecular
events is initiated, starting with the expression of EBNA2 and EBNA-LP
and leading to the expression of LMP1 the major viral oncogene and multiple
host genes as well and to the host-driven circularisation of the viral
genome. Finally, expression of EBNA1 is induced allowing the episomal viral
genome to replicate with dividing cells, thus completing the growth transformation
process. Additional expression of EBERs, BARTs and LMP2a will modify the
transformed state but are not essential for this process. The resulting
EBV transformed cell will continue to grow indefinitely, expressing the
latency-III program.
In vivo this program is not tolerated by the
immune system and EBV-infected cells will (be forced to) switch-off expression
of the major immunogenic proteins.
In vivo EBV persists
in transcriptionally silenced memory B-cells which only express non-coding
EBER1, 2 and BARTs and occasional low levels of LMP2 and EBNA1.
Schematic representation of EBV RNA expression profiles in different
EBV-infected B-cell populations in tonsils and peripheral blood of healthy
EBV-carriers and their proposed relationship.
Naive B-cells are
infected by EB-virions that enter the oropharyngeal lymph nodes by crossing
epithelial barriers. EBV glycoprotein gp350 binds to the receptor CD21,
present on all B-cells. Under influence of CD21-triggering and the subsequent
EBNA2-driven transcription program, naive B-cells differentiate into B
blasts that express the full set of latent EBV RNAs and which are probably
controlled by anti-EBV cytotoxic T-cell responses. The B blast further
differentiates through the germinal center via centroblast to centrocyte.
Both cell types express the latent membrane proteins LMP1 and LMP2, which
provides them with growth and survival signals in absence of antigen, and
EBNA1 which is essential for maintenance of the viral genome in the host
cell. Finally, differentiated EBV-infected cells enter the circulation
as memory B-cells, that are generally silent for viral RNA expression but
may occasionally express LMP2 RNA. Infection of B lymphocytes is mediated
through interaction of the viral Gp350/220 with
CD21
/ CR2 / EBVR
,
the physiological receptor for the complement factor C3d
ref1, ref2
[227 and 228]. After CD21 binding the viral envelope fuses with the host
cell membrane and triggers host cell activation
ref1, ref2 [227
and 229]. The nucleocapsid is then transported to the nuclear boundary
and subsequently degraded, releasing the linear viral DNA into the nucleus,
which then leads to initial EBNA2 and EBNA-LP transcription
ref
[39]. EBNA2 and EBNA-LP are essentially required for initial B-cell growth
transformation, but their activity is modulated by the subsequent expression
of EBNA3A–C. A second essential event is the recircularization of the viral
genome at the terminal repeats, for which the host cell DNA repair machinery
is used
ref1, ref2 [230 and 231]. Although episomal forms are
by far prevailing in EBV-associated malignancies
in vivo, a small
number of studies have described cell lines with integrated EBV
ref1,
ref2, ref3 [232, 233 and 234]. In the first stages after recircularization
of the genome, the full spectrum of latent proteins is expressed
ref
[39]. This complex event which leads to efficient growth transformation
of the host cell includes the well coordinated transcription of poly-cistronic
mRNAs derived from promoters in
BamHI-C and -W, encoding EBNA1 together
with one or more of the other EBNAs (EBNA2, -3a, -3b, -3c and -LP)
ref
[39] and transcription of the major EBV oncogene LMP1, encoded in BNLF1.
This transcription pattern allows
rapid polyclonal expansion of the
infected B-cells as lymhoblasts and is, therefore, referred to as the
‘growth program’
ref1, ref2, ref3 [171, 235 and 236].
In vivo,
incoming EBV predominantly infects subepithelial B-cells to become transformed
B-blasts
ref1, ref2 [39 and 237], but these are rapidly eliminated
by the host T-cell response, which is mainly directed against viral lytic
genes during early primary infection and against EBNA3a, -3b and -3c during
lifelong persistence
ref1, ref2, ref3, ref4 [237, 238, 239, 240
and 241].
The fulminant T-cell response against these freshly EBV transformed
cells in adolescents and adults is the basis of the mononucleosis syndromeref
[241]. Throughout life, outgrowth of EBV transformed B-cells is kept under
control by a persistent alert immune response to latency-associated products,
especially involving EBNA3A–C and LMP2
ref [240]. Part of the
EBV-infected lymphoblasts proliferate and differentiate through a germinal
center-type reaction and subsequently enter the peripheral B-cell pool
as resting memory cells
ref1, ref2, ref3, ref4, ref5 [171, 235,
236, 242 and 243]. It is likely that the continuous but limited expression
of latent genes associated with the growth program is responsible for maintaining
the high level of T-cell memory to these potentially dangerous cells. The
pathogenic consequences of failing T-cell surveillance is seen in immunosuppressed
individuals who are at high risk of developing lymphoproliferative disease
and malignant lymphoma, that are predominantly EBV driven
ref
[241]. Like other herpesviruses, EBV persists lifelong in its host. Total
body irradiation of EBV-seropositive individuals awaiting bone marrow transplantation
leads to eradication of the virus
ref [244]. This indicates that
the cellular compartment where the infected cells reside must be associated
with the hemopoietic tissue. More recent studies have shown that
resting
memory B-cells are the reservoir for latently present EBVref1,
ref2, ref3, ref4, ref5, ref6 [170, 171, 245, 246, 247 and 248]. In
fact, it was already known for many years that it is possible to grow EBV
transformed B-cells directly from the blood of virtually all EBV-seropositive
individuals, provided that T-cells are depleted or suppressed
in vitroref
[241].
In vivo, these infected cells can escape CTL-mediated killing
because the expression of immunogenic EBV-proteins such as EBNA3a, -3b
and -3c is silenced once latent infection has been established
ref1,
ref2, ref3, ref4, ref5 [39, 246, 247, 248 and 249]. This silencing
is accomplished by methylation of early latency promoters Cp and Wp
ref1,
ref2, ref3 [66, 250 and 251]. Because EBNA1 is indispensible for
maintenance of the viral genome in the dividing host cells
ref1, ref2
[39 and 77], its transcription is continued, but now initiated from an
autoregulated promoter in
BamHI-Q (Qp)
ref [82]. Interestingly,
Qp-driven EBNA1 transcription is found in all EBV-associated malignancies
of non-immunocompromized patients.
Although EBNA1 is a foreign protein
to the host, EBV-infected cells expressing EBNA1 are not killed by CTLs
due to the inhibitory effect of the protein's Gly–Ala repeat on proteasomal
processing and subsequent MHC class I-restricted presentationref
[90]. This is considered to be an important mechanism by which EBV
+
tumor cells escape CTL-mediated killing
ref [249].
Given the
fact that memory CTLs reactive against most EBNAs remain clearly detectable
for liferef1, ref2, ref3, ref4 [238, 239, 240 and 241], it is
generally thought that there must always be a subset of B-cells present
that express the growth program. Recently, this subset was shown to consist
of naive (IgD+) B-cells in the mantle zones of the tonsilref1,
ref2 [246 and 247]. Circulating EBV
+ B-cells may express
defined homing receptors and cytokine response receptors, that preferentially
direct them to the epithelial surfaces in the body, where these cells may
be periodically triggered into the lytic cycle in order to maintain shedding
of infectious virus in the oropharynx
ref [241]. It is thought
that this switching process is influenced by signals that normally control
B-cell behaviour, such as antigen-driven activation
ref1, ref2, ref3,
ref4 [171, 241, 242 and 243]. Indeed careful analysis of the localization
of EBV
+ B-cells in tonsils and salivary gland epithelia during
infectious mononucleosis (IM) and latent carriership have revealed that
these cells
preferentially locate in the interfollicular region
rather than in the germinal center and also accumulate around the crypts
and subepithelial layers of the tonsil
ref1, ref2, ref3 [237,
252 and 253]. The differential expression of defined cytokine receptors,
such as CCR6, CCR7, CCR10 and CXCR4 and CXCR5 may be responsible for this
phenomenon
ref [254].
Local antigen triggering and CD40 activation
may subsequently lead to virus production and transepithelial secretionref
[241]. The persistent (low level) production of infectious virus progeny
is reflected by the life-long presence of IgG antibodies to the viral capsid
antigen/membrane antigen (VCA/MA) complex
ref1, ref2 [92 and
255].
Table 2. EBV latent gene expression patterns in EBV associated disorders
Table 3. Overview of EBV transcriptiona in EBV associated diseases
Important factors during virus-induced lymphomagenesis and carcinogenesis
are growth transformation in combination with genetic instability, inhibition
of apoptosis, angiogenesis and inhibition of (or evasion from) the local
immune response. Individual EBV gene products were shown to exert such
effects in vitro, and in concerted action are considered to play an important
role in the genesis of EBV+ malignancies
in vivo. Different
EBV gene expression patterns are recognized, which are generally referred
to as the latency programs (types) I, II and III. It is conceivable that
these expression patterns are subject to the nature of the cells from which
the respective malignancies are derived, or that they are in fact subject
to the host immune response as is the case in PTLD and ARL. In addition
to these well-established gene expression patterns, differential expression
patterns were found among other EBV genes that encode homologs to human
proteins involved in proliferation, differentiation, apoptosis inhibition
and suppression of the local immune response. Some viral latency genes
may predominantly provide their function in the initial B-cell transformation,
like EBNA2 and EBNA-LP, and are subsequently downregulated or even switched
off. Other latency genes, like EBNA1 and LMP2a, may be more essential in
maintaining the long-term survival of the EBV genome in a resting cell
environment, whereas LMP1 may provide the temporary growth kick when such
cells pass through lymph nodesref [171]. The pleiotropic effects
of LMP1 expression, together with external growth or differentiation inducing
stimuli may well provide the basis for pre-malignant growth. It is fascinating
to realize that long-term evolutionary co-existence have thought the virus
to evade immune elimination by preventing the recognition of its most essential
gene products, especially EBNA1 (via Gly–Ala proteasome inhibition) and
LMP1, which is non-immunogenic and has distinct direct and indirect immunosuppressive
functionsref1, ref2, ref3 [131, 351 and 437]. As indicated before
the well regulated and well balanced, low level expression of LMP1 may
be most crucial for persistence of EBV transformed B-cells. When activated
in subepithelial layers additional viral genes can be expressed as well
providing growth or survival functions. Transcripts encoding BHRF1 (a functional
homolog of Bcl-2) were mainly found in B-cell lymphomas (both of immunocompetent
and immunocompromized patients)ref1, ref2 [210 and 211]. Weak
BHRF1 transcription signals were found in some T-cell lymphomas and HDs,
but these signals are ascribed to the presence of EBV-positive reactive
cellsref1, ref2, ref3 [54, 210 and 212]. BHRF1 transcription
and protein expression at the single cell level in most of the lymphoid
disorders has yet to be determined. In NPC the data suggest that BHRF1
expression in NPC is only limited. Preliminary data indicate that BHRF1
protein can also be detected by immunohistochemistry in this disorderref
[212]. Particularly strong expression of early BHRF1 transcripts is confined
to OHLref1, ref2 [54 and 431]. It is thought that the putative
BHRF1 protein may act in addition to the LMP1 induced Bcl-2 protein acts
in prevention of host cell apoptosis, enhancing survival of the host cells
during production of viral progeny ref1, ref2[438 and 439].
Newly synthesized virions released into the environment or by cell-cell
contact with surrounded cells EBV may be transmitted. When the virus then
is capable of entering an epithelial cell additional genes, that are not
part of the B-cell program may become expressed, one of which is the BARF1
gene. BARF1 was originally recognized as an early transcriptref
[189]. Using a reversed northern blotting technique, the transcript was
found to be preferentially expressed in epithelial but not in lymphoid
cellsref [190]. More recently, using NASBA, we found BARF1 transcription
exclusively in NPCs and in OHL but not in lymphoid disordersref1,
ref2 ( Fig. 5) [50 and 54]. In addition to these studies, BARF1 transcripts
were also detected in EBV-associated gastric carcinomasref [191].
Recently, expression of BARF1 at the protein level was detected in NPC
biopsies by immunoblottingref [192]. These findings indicate
that BARF1 expression is specific for EBV-associated epithelial malignancies.
BARF1 has clear effects on epithelial cells in vitroref
[194], in more than one aspect mimicking the role of LMP1 in B-cellsref
[440]. Transcription of BCRF1 (a functional homolog of human IL-10) occurs
almost exclusively in OHL [54], which represents a productive infection.
In this and other productive infections, the BCRF1 protein may play a role
in the inhibition of the local immune response. These observations together
suggest that multiple products encoded within the EBV genome may exert
functions that may be relevant for pathogenic events in vivo. A
number of genes with putative interesting functions, such as the cytokine
receptor encoded in BILF1, BDLF2, a protein with homology to cyclin-Bref
[54] and the BFRF1 putative virion protein, remain to be exploredref
[441]. Furthermore, the in vivo expression and functional role (if
any) of the intriguing but yet illusive proteins encoded in ORFs RPMS1,
A72 and (RK-)BARF0 located within the BARTs that are abundantly transcribed
in all EBV-associated malignancies remain to be defined. In addition, the
relative importance and interactive collaboration by the individual gene
products and the relevance of subtle mutations found in various EBV isolates
recovered from directly from tumor tissues or patients with distinct clinical
syndromes associated with EBV-infection largely remain to be established.
Further unraveling of these viral functions and their ‘well orchestrated
(inter)action(s)’ may permit the development of antiviral agents capable
of interfering with these functions with options for future therapeutic
intervention. EBV and some of its latent gene products have been demonstrated
to modulate the expression of various cellular (proto-onco-) genes in
vitro. The most obvious way to study the effect of the presence of
EBV on the expression of these genes in vivo, is the comparison
of their expression in EBV-positive and -negative cases of a single clinical
entitiy. In this context, HD is a suitable model, because both EBV-associated
HDs and EBV-negative HDs are recognized. Moreover, several findings suggest
that EBV-associated and EBV-negative HDs have a different pathogenesis.
For example, H-RS cells in EBV-associated HDs express high amounts of MHC
class I molecules, whereas MHC class I appears to be dowregulated in EBV-negative
HDsref [442]. MHC class II and a number of co-stimulatory molecules
as well as intracellular TAPs associated with peptide transport after proteasomal
degradation seems to be functionally intactref [362]. This is
also found for NPC and T/NK cell lymphomasref1, ref2 [336 and
394]. Most EBV-associated HDs express particularly abundant levels of LMP1
and LMP2, proteins which normally can evoke a CTL response. However, H-RS
cells apparently are not killed by CTLsref1, ref2 [363 and 364].
Several underlying mechanisms have been proposed for this phenomenon. It
is thought that the presence of the CTLs results in a selection of H-RS
cells that are resistant to CTL-mediated (and probably also therapy-mediated)
apoptosis. HDs in which this has occurred, are likely to express low amounts
of p53 and high amounts of bcl-2 in their H-RS cellsref [368].
However, the expression of p53 (as determined by immunohistochemistry)
is not related to the presence of EBV, and the expression of bcl-2 even
shows an inverse relation to the presence of EBVref [368]. This
indicates that induction of apoptosis resistance by modulation of p53 and
bcl-2 expression is probably not a mechanism used by EBV in HD. This may
also hold for NPC where high levels of p53 and bcl2 coexist, together with
highly expressed PCNAref1, ref2 [391 and 392]. Alternatively,
p53 mediated apoptosis may be inhibited by upregulation of A20 expression
via LMP1. We tested A20 expression in EBV-positive and -negative HD cases
using NASBA, but we could not find a relation between A20 transcription
and the presence of EBV (Brink et al., unpublished data). This seems to
be in agreement with the finding that EBV apparently does not interfere
with the normal function of p53 in HDref [204]. It would be
best to confirm these data morphologically, but at present no antibodies
are available that recognize human A20 in clinical material. To prevent
CTL-mediated killing of EBV-infected H-RS cells, inhibition of the local
CTL response by EBV is also a possible mechanism. EBV-associated HDs express
significantly higher amounts of human IL-10ref1, ref2, ref3
[161, 362 and 443], which may contribute to immune evasion. We have shown
that HDs do not express the viral homolog of Interleukin-10 (BCRF1) but
that the detected IL-10 expression is of host cell originref
[161]. Upregulation of human IL-10 upon EBV-infection in vitro has been
demonstratedref [365], and, interestingly, elevated human IL-10
expression levels have been detected in the H-RS cells of EBV-positive
HDsref1, ref2, ref3 [161, 362 and 443].
Table 6. Overview of cellular genes and their functions of which expression
and/or function is modulated by EBV
None: no differences found for EBV+ and EBV? cases.
Using RT-PCR, we have also analyzed the expression of the proto-oncogene
c-fgr. C-fgr was shown by others to be upregulated upon EBV-infection,
and EBNA2 is most likely the EBV gene to induce c-fgr expressionref1,
ref2 [96 and 444]. Moreover, alternative splicing of c-fgr transcripts
was reported in EBV-positive cell linesref [445]. Using RT-PCR,
we have performed a qualitative analysis of c-fgr transcription in EBV-positive
(n=4) and -negative HDs (n=4), in EBV-associated PTDLs (n=3) and ARLs (n=4),
and one IM case. We found no clear relation between the presence of EBV
and c-fgr transcription in HD (Brink et al., unpublished data). Moreover,
c-fgr RT-PCR signals in HD were relatively weak compared with positive
controls. In most PTLDs and ARLs we found transcription of c-fgr, including
the EBV specific alternative transcript. These data suggest that upregulation
of the c-fgr proto-oncogene is not a mechanism used by EBV in HD, but may
play a role in PTLDs, ARLs and IM. Moreover, these data strengthen the
finding that EBNA2 (which is expressed to some extent in PTLDs, ARLs and
IM but not in HD) is the EBV gene responsible for c-fgr upregulation. Future
studies should aim to investigate c-fgr protein expression at the single
cell level. In conclusion, differences in EBV gene expression patterns
exist between the various EBV-associated diseases. This is true for lymphomas
of immunocompromized versus immunocompetent patients, for lymphomas of
B-cell versus T-cell origin, and most strikingly for lymphomas on the one
hand and epithelial disorders on the other. For some of the differentially
expressed genes, such as BARF1, it remains to be investigated whether the
observed expression pattern is subject to host cell regulation factors
or contributes actively to differences in pathogenesis. Therefore, future
studies should not only aim to determine EBV gene expression in EBV-associated
diseases, but should also include more fundamental research on expression
regulation and functional interactions. Moreover, it is important to notice
that high mRNA expression does not always coincide with expression at the
protein level; we have shown this for BARF0 but it may well be that the
EBV genome gives rise to other non-translated transcripts. In the future,
it would be worthwhile to develop additional antibody reagents against
the various (putative) EBV-proteins and use these for in situ expression
analysis, in combination with mRNA profilingref.
EBV-induced
gene 3 (EBI3) is expressed in DCs and is part of the cytokine IL-27
ref
that controls Th cell development. However, its regulated expression
in DCs is poorly understood. EBI3 is expressed in splenic CD8-,
CD8+, and plasmacytoid DC subsets and is induced upon TLR signaling.
Cloning and functional analysis of the EBI3 promoter using in vivo
footprinting and mutagenesis showed that stimulation via TLR2, TLR4, and
TLR9 transactivated the promoter in primary DCs via NF-kB
and Ets binding sites at -90 and -73 bp upstream of the transcriptional
start site, respectively. Furthermore, NF-kB
p50/p65 and PU.1 were sufficient to transactivate the EBI3 promoter in
EBI3-deficient 293 cells. Finally, induced EBI3 gene expression in DCs
was reduced or abrogated in TLR-2/TLR4, TLR9, and MyD88 knockout mice,
whereas both basal and inducible EBI3 mRNA levels in DCs were strongly
suppressed in NF-kB p50-deficient mice. In summary,
EBI3 expression in DCs is transcriptionally regulated by TLR signaling
via MyD88 and NF-kB. Thus, EBI3 gene transcription
in DCs is induced rapidly by TLR signaling during innate immune responses
preceding cytokine driven Th cell developmentref.
Increased numbers of EBV-infected cells in areas of active inflammatory
bowel disease are secondary to influx or local proliferation of inflammatory
cells & do not contribute significantly to local production of EBI3ref
Release of progeny virions from polarized cells occurs from both their
apical and basolateral membranes. No CPE, no productive replication in
vivo, immortalization of B lymphocytes in vitro. Polyclonal
activation of B cells => heterophilic IgMs.
Expression of IL-7Ra
was lost from all CD8+ T cells, including EBV epitope-specific
populations, during acute infectious mononucleosis (IM). Thereafter expression
recovered quickly on total CD8+ cells but slowly and incompletely
on EBV-specific memory cells. Expression of IL-15Ra
was also lost in acute IM and remained undetectable thereafter not just
on EBV-specific CD8+ populations but on the whole peripheral
T and NK cell pool. This deficit, correlating with defective IL-15 responsiveness
in
vitro, was consistently observed in patients up to 14 years post-IM
but not in patients after CMV
-associated
mononucleosis, nor in healthy EBV carriers with no history of IM, nor in
EBV-naive individuals. By permanently scarring the immune system,
symptomatic primary EBV infection provides a unique cohort of patients
through which to study the effects of impaired IL-15 signalling on human
lymphocyte functions in vitro and in vivoref.
Symptoms &
signs :
-
largely subclinical in early childhood : latency II and III expression
patterns in tonsils of healthy asymptomatic healthy carriers
-
lymphoid diseases :
-
infectious
mononucleosis (IM) (18%; once termed Drusenfieber, i.e.
Pfeiffer's
glandular fever) after 30-60 days incubation (10-15 days in babies)
: colonization of Waldeyer's ring => fever
(66-76%, only in first 10 days), cough (14%), pharyngotonsillitis
(1.6%; "kissing tonsils" : if severe bacterial overinfection and oedema
=> cortisone therapy); anterior and posterior laterocervical, inguinal
and axillary lymphadenomegaly
(5-94%, for 2-3 weeks), mild hepatomegaly
(12%; increase in SGPT up to 100÷200 U/L at 37°C), eyelid edema
(5.3%), infantile hepatitis syndrome (0.5%), jaundice
(9%), splenomegaly
(52%) due to red pulp hyperplasia (sometimes spleen upper pole may enlarge
from the 7th÷8th intercostal space down to
the left iliac cavity : rare splenic
infarction
and splenic rupture
;
avoid splenic injuries during recovery !), maculopapular
exanthema
(8-10%), mild sinusal tachycardia (1.6%), hyperhemic tonsils with no exudate,
gross hematuria (0.5%), pericarditis
and interstitial
pneumonia
.
Genital ulcerations can be the initial manifestation of EBV in adolescents
who are neither sexually active nor immunocompromised practicing cunnilingusref.
IM can be considered the clinically manifest form of a primary EBV-infection.
Its diagnosis relies in the detection of atypical lymphoid cells in the
peripheral blood, the occurrence of so-called heterophile antibodies and
EBV-seroconversionref [276]. IM is a benign disorder with expansion
of the paracortex of lymphoid tissues. The proliferating cell populations
are EBV-infected polyclonal B blastsref [277], accompanied by
the growth of activated T-cellsref [278]. Morphologically, the
EBV-infected cells range from large immunoblasts, including H-RS like cells,
to small lymphoid cells. In addition to latently infected cells, a small
number of EBV-positive cells expressing lytic cycle antigens such as BZLF1
can be detected in tissues from IM patients. These are often found adjacent
to crypt epithelium and are frequently positive for plasma cell markersref1,
ref2, ref3 [252, 253 and 279]. It is thought that EBV can replicate
in these plasmacytic cells, thus generating a cellular source of infectious
virus in the saliva of IM patientsref1, ref2 [241 and 253].
Further EBV gene expression analysis has shown a type III EBV latency pattern
in IM [235], although the expression at the single cell level is more heterogeneous.
Most cells are EBER positive but do not express EBNA2 or LMP1 (type I latency),
whereas some large immunoblasts express LMP1 in the absence of EBNA2 (type
II latency) and many small lymphocytes express EBNA2 but not LMP1ref1,
ref2 [237 and 279].
Laboratory examinations : leukocytosis
(> 4,500÷10,000 / mL) with relative reactive
lymphocytosis due to proliferation of Downey
cells / atypical lymphocytes
(i.e. CD8+ Ts cells) :
-
type I cells : kidney-shaped or lobulated nucleus with vacuolated,
basophilic foamy cytoplasm
-
type II cells contain plasmacytoid nuclei with less vacuolated and
basophilic cytoplasm
-
type III cells have a finer chromatin pattern and 1-2 nucleoli.
..., decrease in HCT, HGB and MCV (differential diagnosis with massive
hemolysis, internal or external hemorrhages, hemoglobinopathies)
Prognosis : self-limiting within 2-4 weeks.
Complications :
-
CNS syndromes associated with primary EBV or reactivated infection : diverse
CNS syndromes can occur
-
CNS syndromes associated with chronic EBV infection
-
X-linked
lymphoproliferative syndrome (X-LPS) / fatal mononucleosis
: caused by hereditary mutations in the gene encoding the signalling lymphocyte
activation molecule (SLAM)-associated protein (SAP) on position q25 of
the X-chromosomeref1, ref2 [280 and 281]. The self-ligand SLAM
protein is present on the surface of both B and T-cells. When interactions
occur between SLAM molecules on the interface between T- and B-cells, signal
transduction pathways are initiated. The T-cell protein SAP binds to SLAM
and as such acts as a negative regulatorref1, ref2 [282 and
283]. Individuals that have inherited this trait are usually asymptomatic,
but upon primary EBV-infection their immune response becomes overreactive
because of the non-functional SAP protein. This usually results in a fulminant
IM accompanied by a EBV-associated
hemophagocytic syndrome (EBV-AHS) by which liver and bone-marrow are
destroyedref [284]. The only curative treatment for this syndrome
is allogeneic bone-marrow transplantation
ref
[285]. However, patients who do survive the infection are prone to developing
lymphomas later in liferef [286].
-
EBV-mild
acquired immune deficiency syndrome (EBV-MAIDS) in postsurgical sinusitis
Therapy : periodic intramuscular
IVIg
-
autoimmunity may cause :
-
immunoproliferative syndrome
/ lymphoproliferative disease (LPD) (latency
III expression pattern) in stem cell and organ transplant recipients
(Cohen, 2000) : the present view that lymphomas have to be considered neoplastic
counterparts of reactions normally occurring in lymphoid tissues after
antigenic stimulation, is helpful in understanding their pathogenesis.
Lymphomagenesis is considered to be a multistep processref,
during which an accumulation of genetic changes takes place. Lately, numerous
studies have identified cytogenetic abnormalities that are characteristic
of specific non-Hodgkin lymphomas (NHLs). These are frequently translocations
involving the antigen receptor loci. Since transcriptional activity of
the antigen receptor loci is inherent to the function of lymphoid cells,
translocations involving these loci usually result in overexpression of
an oncogene under the control of the antigen receptor expression regulation.
A characteristic example is the t(14; 18) in follicular lymphomas, which
results in overexpression of the apoptosis inhibitor bcl-2 under the control
of the Ig heavy chain promoterref1,
ref2.
The finding that cells containing t(14; 18) can be detected in the peripheral
blood of healthy individualsref
indicates that this translocation may be an early event in the genesis
of follicular lymphoma. Another translocation involving the Ig heavy chain
locus is t(8; 14) which occurs in many of the Burkitt's lymphomas (BL).
This translocation results in overexpression of the transcription factor
c-myc under the control of the Ig heavy chain promoterref1,
ref2.
The difference in growth rate between follicular lymphomas and BL is clearly
the consequence of the different genes involved in their respective genetic
alterations. The t(14;18) involving bcl-2 results in an extended lifespan
of the affected cells, allowing the accumulation of more genetic hits.
By contrast, genetic changes resulting in overexpression of a transcription
factor usually have a direct influence on proliferation and, therefore,
are usually associated with rapidly growing lymphomas, as is the case with
c-myc in BL. Besides cytogenetic abnormalities, chronic antigenic stimulation
is thought to play an important role in lymphomagenesis. The presence of
antigens not only stimulates proliferation (resulting in ‘fixation’ of
genetic aberrations throughout the next cell generations) but also induces
the process of Ig rearrangement, and thus increases the possibility of
more aberrant rearrangements to take placeref.
This can also be illustrated from the c-myc translocations in BL. Recent
findings show that all cases of BL harbor somatically mutated V region
genes and are as such derived from B-cells that took part in a germinal
center reactionref.
It was proposed that most, if not all, c-myc/Ig translocations in endemic
BL happen in a mutating germinal center B-cell and result from the process
of hypermutation as such. In sporadic BL, c-myc translocations are usually
targeted to the class-switch region of the Ig gene, and thus probably relate
to class switch recombination that also take place in the germinal centerref1Lenoir
GM, 173-206, ref2.
Some lymphoma subtypes are clearly related to the presence of a certain
antigen. For example, gastric lymphomas are related to infection with Helicobacter
pylori (H. pylori)ref,
and certain cutaneous B-cell lymphomas are associated with Borrelia
burgdorferi infectionref.
In AIDS-related non-Hodgkin Lymphoma (ARNHL) similar chronic antigen exposure
may be responsible for tumor outgrowth, but the triggering antigen remains
elusiveref.
For the endemic form of BL, the antigens providing chronic stimulation
are probably Epstein–Barr virus (EBV) and malariaref.
The latter provides a strong and acute B-cell activation stimulus mediated
by repetitive epitopes on the parasite surface, whereas the action of EBV
in BL may be 2-fold: it functions as antigen and as a transforming agent.
The presence of somatic hypermutations in the variable region of the Ig
heavy chain in BL cells strengthens the hypothesis that exposure to antigens
may be relevant for the pathogenesis of BLref1Lenoir GM, 173-206,
ref2.
Furthermore, the presence of Ig-gene hypermutations in the Reed–Sternberg
cells of Hodgkin's diseaseref
and the expression of functional differentiation markers such as granzyme-B
and TIA-1 in all EBV-associated T-/NK-cell NHL'sref
also suggest that these tumors are initially driven by antigen stimulation.
Similar to the situation in endemic BL, the presence of EBV genome may
provide immortalizing functions thus contributing to the multistep oncogenic
processref1,
ref2.
-
autoimmune
lymphoproliferative syndrome (ALPS)

-
Kikuchi-Fujimoto
disease (KFD) / subacute or histiocytic necrotizing lymphadenitis (HNL)
?
-
EBV-associated T/NK-cell
LPD : target cell specificity, defects in host immune responses, and
strain differences of EBV may account for ectopic EBV infections and for
the unique clinical presentations characteristic of each illnessref.
-
chronic infectious
mononucleosis (CIM) / chronic Epstein-Barr virus infection (CEBV) / chronic
active EBV infection (CAEBV) in apparently immunocompetent hosts (extraordinary
event after acute disease)
Symptoms & signs : chronic or recurrent
infectious mononucleosis-like symptoms (intermittent fever, weight loss
and liver abnormalities, rarely multiple nodular coagulation necrosis in
the liver) persisting over a long time and by an unusual pattern of anti-EBV
antibodiesRickinson AB, 13-14. Patients with this disease have
no evidence of any prior immunologic abnormalities or of any other recent
infection that might explain their conditionRickinson AB, 13-14,
ref2.
CAEBV is a disease with a high mortality and high morbidity with life-threatening
complications, such as virus-associated hemophagocytic syndrome, interstitial
pneumonia, lymphoma, coronary artery aneurysms, and central nervous system
involvementref1,
ref2,
ref3,
ref4,
ref5.
The 3 main criteria of CAEBV infection areref
:
-
(1) severe illness lasting > 6 months that began as a primary EBV infection
and that was associated with grossly abnormal EBV antibody titers, antiviral
capsid antigens (VCA) IgG > 5120, anti-early antigens (EA) IgG > 640, or
anti-EB nuclear antigens (EBNA) < 2;
-
(2) histologic evidence of major organ involvement such as interstitial
pneumonia, hypoplasia of some bone marrow elements, uveitis, lymphadenitis,
persistent hepatitisor splenomegaly; and
-
chronic hepatitis might be a manifestation of chronic EBV infection in
the lack of detectable immune deficiency; the expansion of CD28-CD27-
and increase of functional EBV-specific CD8+ T cells being the
only surrogate markers of viral activityref
-
(3) increased quantities of EBV in affected tissues.
Subsequently, Okano and his colleaguesref
proposed similar criteria to diagnose severe CAEBV infection.
Importantly, many cases have been reported that do not satisfy the
criteria described above. Some patients lack abnormal patterns of EBV-related
antibodies, whereas other patients lack major organ involvement and have
only skin symptoms, such as hypersensitivity
to mosquito bites (HMB) or hydroa
vacciniforme
-like
eruptionsref1,
ref2.
On the other hand, patients with CAEBV infection had extremely high viral
loads, as assessed by qPCRref1,
ref2.
Clonal expansion of EBV-infected T or natural killer (NK) cells could be
associated with CAEBV infectionref1,
ref2,
ref3,
ref4,
ref5,
ref,
ref7.
Detailed clinical features of some patients have been describedref1,
Morita M, 1485-1488,
ref3.
An accumulating body of evidence suggests that clonal expansion of EBV-infected
T or NK cells is associated with CAEBV infectionref1,
ref2,
ref3,
ref4,
ref5,
ref6,
ref7.
In healthy carriers, EBV exists latently in resting memory B cellsref.
It is unclear whether the invasion of blood cells other than B cells causes
CAEBV infection or the invasion is an ordinary event, but the host's immunologic
abnormalities allow the expansion of these cells. Recently, a defect in
the SAP/SH2D1A gene was implicated in XLP, a fatal lymphoproliferative
disorderref1,
ref2,
ref3.
Patients with XLP are exclusively boys in whom primary infection with EBV
causes lymphoproliferation and severe hepatitis, mimicking CAEBV infectionref1,
ref2.
Therefore, we examined the SAP/SH2D1A gene in all the male subjects enrolled
in our study, but did not find any abnormalities. However, it is possible
that a defect in another gene essential for regulating lymphocyte activation
and proliferation may be a cause of CAEBV infection. Such a defect or single
nucleotide polymorphism might influence the function of virus-specific
or nonspecific lymphocytes and thereby allow the expansion of EBV-infected
T or NK cells. It has been reported that EBV-infected T and NK cells express
a limited range of viral antigensref.
EBV-infected B cells express at least 9 antigens, some of which are antigenic
enough to be presented to CTLs. This is called latent
infection type 1. EBV-infected T and NK cells express only EBNA1 and
LMP1; this is called latent infection type 2ref.
The expression of viral antigens was examined in some CAEBV patients, and
they were type 2ref1,
ref2.
Because
these 2 proteins are less antigenic, infected cells can evade from immune
surveillance by CTLs. Therefore, they may proliferate and cause chronic
infection. Chromosomal abnormality occurred in the lymph nodes of patients
with CAEBVref
: 50% of the patients examined had chromosomal aberrations in their peripheral
blood cells and 79% of patients showed monoclonality of EBV. Because there
were no specific patterns of genetic aberration, and some patients displayed
several different aberrations, the chromosomal abnormalities seen in patients
with CAEBV infection might only reflect chromosomal fragility. However,
clonality of the EBV genome and chromosomal aberrations generally indicate
clonal expansion of EBV-infected cells. These results indicate that clonal
expansion is a common feature of CAEBV infection, and this disease might
therefore be considered lymphoproliferative rather than infectiousref.
In fact, patients with CAEBV infection frequently develop neoplasms such
as malignant lymphoma. It would therefore be better to call the cases presented
here "chronic EBV-associated lymphoproliferative disorders." Approximately
10 out of 30 patients with CAEBV infection did not meet the previously
accepted definition of CAEBV infectionref1,
ref2.
These patients had symptoms typical for CAEBV infection and had extremely
high EBV loads in their peripheral blood. High titers of EBV-related
antibody are not always necessary for the diagnosis of CAEBV infection.
On the other hand, all the patients had > 102.5 copies/µg EBV DNA
in PBMC. Furthermore, it is of note that the copy number of EBV DNA in
PBMC decreased below 102.5 copies/µg DNA in all 7 patients who underwent
hematopoietic stem cell transplantation. These results indicate that viral
loads greater than102.5 copies/µg DNA can be used not only as diagnostic
factors for CAEBV infection, but also as predictors of therapeutic efficacy.
Previous reports show that patients with CAEBV infection had cell-free
EBV DNA in plasma, although the origin of the viral DNA is unclear.37,38
Usually, the plasma of healthy individuals does not contain EBV DNAref1,
ref2.
Therefore, the presence of EBV DNA in plasma may have significance for
the diagnosis of CAEBV infection. However, it should be borne in mind that
the plasma of patients with CAEBV infection did not always test positive
for EBV DNA. Indeed, in this study, EBV DNA was not detected in plasma
from 6 patients, whereas they had large amounts of EBV DNA in their PBMC.
PBMC,
and not plasma, should be used for diagnosing and monitoring patients with
CAEBV infection. It is unclear why some patients did not have cell-free
EBV DNA. Patients with negative plasma results sometimes turned positive
at a later visit. Cell-free viral DNA may fluctuate from day to day. The
other possibility is that inhibitors in plasma might influence PCR reactions
and cause false-negative results. In conclusion, patients with CAEBV infection
were divisible into 2 subgroups: T-cell and NK-cell CAEBV infection. Each
group had different clinical features and prognosis. A high titer of EBV-related
antibodies is not always a prerequisite for the diagnosis of CAEBV infection.
Viral load, detected by quantitative PCR in PBMC, was useful for disease
diagnosis and as an indicator of therapeutic efficacy. A viral load
> 102.5 copies/µg DNA be used as a diagnostic criterion for CAEBV
infectionref.
-
NK cell-type CAEBV : HMB
and high IgE
Symptoms & signs : liver dysfunction
Therapy : vidarabine
-
T-cell type CAEBV : fever and high
titers of EBV-related antibodies. Although not statistically significant,
patients with T-cell type of CAEBV infection had higher IgG levels than
those with NK-cell type.. It is possible that EBV-infected T cells become
activated and release inflammatory cytokines such as IFN-g,
IL-6, or TNF-a, resulting in severe inflammation
and fever. In EBV-related
hemophagocytic syndrome, it has been shown that viral-infected T cells
release TNF-a and activate macrophages, resulting
in massive hemophagocytosisref1,
ref2.
These activated T cells might induce polyclonal B-cell activation through
cytokine release and thereby induce high levels of IgG and EBV-related
antibodies. On the other hand, it is unclear why HMB and high IgE were
observed in patients with NK-cell type of CAEBV infection. We should emphasize
that determining the cell type is important in predicting disease prognosis,
because the survival rates were different between the 2 groups. Differences
in survival rates are particularly important in assessing treatment choices.
T
It remains unclear whether these 2 manifestations of disease represent
different entities or simply appear different because of the nature of
the infected cells
-
severe chronic
active EBV-infection (SCAEBV) (late 1970s)
Symptoms & signs : similar to those
of chronic
fatigue syndrome (CFS)
but more severe in degree (pancytopenia, high fever, massive splenomegaly)
=> poor prognosis
Therapy : effect of in vitro-generated
autologous EBV-specific CTLs or LAK cells is limited or deteriorative
Therapy : to date, a treatment for CAEBV infection
has not yet been established. Antiviral or immunomodulating agents, such
as acyclovir, gancyclovir, vidarabine, IFN-g,
and IL-2, have been triedref1,
Wakiguchi H, 2022-2025,
ref3,
ref4.
Adoptive transfer of virus-specific CTLs to a patient with CAEBV infection
has been reportedref.
However, most of these reports were anecdotal and there are few confirmatory
reports. Immunochemotherapy consisting of etoposide, steroids, and cyclosporin
A was proposed for use in patients with advanced CAEBV infectionref,
but no evidence of its efficacy is available. Recently, successful treatment
of CAEBV infection by allogeneic bone marrow transplantation has been reportedref.
Viral loads decreased in all patients receiving transplants, some of whom
appeared to be cured. However, HSCT constitutes a considerable risk, because
4 of 7 patients died after transplantation. Prospective studies analyzing
a larger number of patients with CAEBV infection are necessary to confirm
the efficacy of transplantation and to establish safer conditioning regimens.
-
EBV-associated
hemophagocytic
syndrome (HPS)
(EBV-AHS)
-
peripheral T-cell lymphoma
-
aggressive NK-cell leukemia
-
EBV-associated extranodal T-cell and natural killer (NK) cell lymphomas
: the association of EBV with extranodal T-cell non-Hodgkin's lymphoma
(T-NHL) is surprisingref [326], because T-cells normally
express the EBV receptor CD21 at a rate 10-fold lower than B-cells doref
[327]. The association of EBV with T-NHL depends largely on siteref1,
ref2 [326 and 328]. The expression of LMP1 in T/NK cell lymphomas
is correlated with bad prognosisref [141] and a role for EBV
induced IL-10-mediated local immunosuppression in the pathogenesis of nasal
T/NK-lymphoma was recently suggestedref [336].
-
nasal type NK/T-cell lymphomas have a 100% association with EBVref1,
ref2 [329 and 330]. In NK/T-cell lymphomas in the nose, cytotoxic
proteins like TIA-1 and granzyme B are even detected in up to 100% of the
casesref [334]. Nasal (type) NK/T-cell lymphomas usually express
CD56, showing their NK cell originref [172]. These findings
have led to the hypothesis that EBV is able to infect CTLs and NK cells
during cell-cell contact with (productively) infected target cells by means
of exchange of viral (virion) genetic material and subsequent transformation
of the effector cellref [326]. EBV is actually present within
the neoplastic cytotoxic cells, demonstrating that EBV is able to persist
in these cellsref [334]. One could speculate that predominantly
mucosa-associated CTLs or NK cells become infected by EBV during the killing
of EBV-infected target cells, because most productively infected B and
epithelial cells are found in the oro/nasopharynx. In fact, EBV-infection
by means of close cell–cell contact was described by othersref
[335]. The preferential homing of the infected cytotoxic cells would explain
the presentation of EBV-associated T-NHL in the nasopharynx and the air
and food passages
-
intestinal T-NHL have a considerably lower association with EBVref
[331] : the neoplastic cells frequently express cytotoxic proteins like
TIA-1 and granzyme Bref1, ref2, ref3 [332, 333 and 334]
-
chronic granular
lymphocyte proliferative disorders (GLPD)

Double staining for EBER RNA and surface markers in T-NHLs. Double
staining for EBER RNA (red) and the B-cell marker CD20 (brown) (a) but
not for EBER (dark blue) and the T-cell marker CD3 (brown) (b) can be observed
in a nodal T-NHL. Double staining for EBER RNA (dark blue) and TIA-1 (brown)
(c) can be observed in an intestinal T-NHL.
-
nodal T-cell lymphomas :
-
nodal angioimmunoblastic lymphadenopathy with dysproteinemia (AILD)-like
lymphomas : using EBER RISH, EBV can be detected in up to 30% of casesref1,
ref2, ref3, ref4 [337, 338, 339 and 340]. These AILD-like lymphomas
were shown to contain a clonal T-cell populationref [337]. However,
EBV in these lymphomas is not present in the neoplastic T-cells, but predominantly
in B-cells which are usually polyclonal or oligoclonalref [339]
and rarely become clonal in a later stageref [340]. This indicates
that the proliferation of EBV-infected B-cells may be a secondary event
and that EBV is not causally related to the pathogenesis of these lymphomas.
The presence of EBV in B-cells in these lymphomas suggests that the expanded
meshworks of follicular dendritic cells (FDCs), which are a hallmark of
these lymphomas, stimulate proliferation of (EBV-infected) B blasts.
-
anaplastic
large cell lymphomas (ALCLs)
: using EBER RISH, EBV can be detected in the majority of CD30+
neoplastic cells of a small percentage of casesref1, ref2, ref3, ref4
[341, 342, 343 and 344]. These can, therefore, be considered as being EBV-associated.
Moreover, EBV genomes in these ALCLs are clonalref [342], suggesting
that EBV-infection is an early event and/or results in growth advantage.
Interestingly, the majority of ALCLs appear to be derived from cytotoxic
T-cellsref1, ref2 [343 and 344], suggesting a pathogenic mechanism
resembling that of extranodal T-NHL.
-
mosquito
allergy with granular lymphocyte proliferative disorder (GLPD) / severe
hypersensitivity to mosquito bites (HMB) (SHMB)
Epidemiology : mainly in Japanese patients
(at least 58 patients) in the first 2 decades of liferef
Pathogenesis : clonal expansion of EBV-infected
NK cells (NK-GLPD) aberrantly expressing CD25 (augmented in vitro
proliferative response to IL-2), Fas-L and Bcl-2, and resistance to in
vitro Fas-induced apoptotic cell death (Fas-ACD). One of the unanswered
questions is whether EBV-infected NK cell lymphoproliferative disease (LPD)
precedes the onset of mosquito allergy among these patients. At 6 months
old, a Japanese baby developed pancytopenia resulting in B-precursor
acute lymphoblastic leukemia (ALL)
without typical chromosomal abnormalities. Although she entered complete
remission following our ALL protocol, continuous chemotherapy became difficult
because of severe myelosuppression and repeated infections. Because she
had no HLA-matched related or unrelated donors, we decided to perform CD34+
cell transplantation from her HLA-haploidentical father when she was aged
1 year and 11 monthsrefYoshimoto T, 9-14. At the time of transplantation
she was negative for EBV serology and her father was positive. Preconditioning
regimen consisted of total body irradiation (TBI) (12 Gy) and melphalan
(140/m2), and 4.54 × 106/kg of CD34+
cells were infused on February 6, 1996. Tacrolimus was used for GVHD prophylaxis.
Since her posttransplantation clinical course was uneventful except for
delayed platelet recovery, tacrolimus was stopped on day 64. Two months
later, due to development of chronic GVHD of the skin, cyclosporine was
started. Although tests for sex chromatin by fluorescence in situ hybridization
(FISH) showed complete donor engraftment, poor recovery of platelet count
still continued. Therefore additional 9.64 × 106/kg of
CD34+ cells from her father were reinfused on day 148, resulting
in complete recovery of platelets. Because EBV serology was still negative
after complete engraftment, we considered that there was no contamination
of transmissible EBV-infected B cells in the purified CD34+
cells obtained from her father. 7 months after transplantation, the patient,
aged 2 years and 6 months, developed primary EBV infection with atypical
manifestations such as fever, eruptions, hypergammaglobulinemia (IgG, 3030
mg/dL; IgM, 300 mg/dL), liver dysfunction, abdominal lymph nodes swelling,
and pancytopenia. Seroconversion against EBV (VCA IgG; 320 × , EA
IgG; 10 × , EBNA; < 10 × ) was confirmed, and clinical symptoms
as well as laboratory findings were resolved by stopping cyclosporin. 2
months later she developed fever and otitis media. Laboratory findings
on this occasion revealed that T cells were 16% in the PBMC (healthy
control < 5%). She had been healthy until she developed mosquito allergy
at age 5 years and 5 months. Upon admission at this time she had multiple
skin lesions especially on the extremities and laboratory findings revealed
liver dysfunction and a reactivation pattern of antibodies against EBV.
gd
T cells gradually increased with time :
TcRg gene rearrangements were observed and
EBV DNA was detected in the sorted T cells. A skin biopsy specimen
revealed infiltrating atypical lymphocytes around the small vessels (resembling
angiocentric lymphoma) EBER+, CD3+, CD4, CD8, and
CD56, in the skin lesion, which may play an important role in the development
of mosquito allergy. The patient underwent chemotherapy and the skin lesions
have improved and the number of T cells has been reduced.
Symptoms & signs : following mosquito
bites the skin lesion at bite sites is typically a erythema or bulla that
develops into ulcer or scar, accompanied by general symptoms such as high
fever and general malaise and subsequently may experience lymphadenopathy
and hepatosplenomegaly
Prevention : vidarabine
+ foscarnet
Therapy : refractory to several conventional
chemotherapies
Prognosis : 50% of the patients reported
died of EBV-associated
hemophagocytic syndrome / malignant histiocytosisref1,
ref2,
granular lymphocyte proliferative disorder, or lymphomas. On the other
hand, nearly 33% of patients with chronic
active Epstein-Barr virus (CAEBV) infection may also have mosquito
allergy and NK-cell proliferationKawa-Ha K, 52-55, ref2,
Kawa K, 139-147, ref4.
More recently, a close relationship has been showed between mosquito allergy
and EBV-infected NK-cell lymphoproliferationref
-
hydroa vacciniforme
accompanying with EBV-infected NK-cell proliferationref1,
ref2
Therapy : reduction and elimination of EBV-infected
T/NK cells seems to be essential for the treatment of EBV-associated T/NK-cell
LPD and it is important to distinguish it from ordinary EBV-associated
B-cell LPDref1,
ref2,
ref3.
-
EBV-associated B-cell LPDs
:
-
lymphomas in immunocompromized patients :
-
post-transplant
lymphoproliferative disorder (PTLD)
due to excessive depletion of donor-derived EBV-specific CTLs during GvHD
prophylaxis. PTLDs are a feared complication in transplant recipients who
are being treated with immunosuppressive drugsref [287]. The
majority of these disorders is of B-cell origin and is associated with
EBVref [288], although exceptions existref1, ref2
[289 and 290]. Generally, the EBV present in PTLDs of solid-organ transplant
(SOT) recipients is of host origin. By contrast, in PTLDs of bone-marrow
transplant (BMT) patients the EBV generally is of donor originref1,
ref2 [241 and 291]. Most PTLDs occur in patients who are EBV-seronegative
prior to the transplantation and who, therefore, are unable to timely mount
an effective immune response to EBV transformed B-blastsref
[288]. Thus, PTLDs are frequently associated with primary infection and,
therefore, assessment of the EBV serostatus in donor and recipient is important
for risk stratification and will allow early identification of patients
at high risk for PTLD developmentref1, ref2, ref3, ref4 [259,
260, 261 and 288]. Early PTLDs are mostly polyclonal expansions of EBV-infected
B-cells that may regress spontaneously upon reduction of immunosuppresive
therapy or T-cell infusion, but which may progress to polymorphic PTLDs
and diffuse large B-cell lymphomas, both showing clonal Ig rearrangements
and EBV genomesref [292]. Immune suppression allows outgrowth
of EBV-infected B-cells, associated with the expression of immunodominant
EBV-proteins. Indeed, (using non-morphological methods such as RT-PCR and
western blotting) a latency type III-like EBV gene expression pattern can
be found in early PTLDsref1, ref2, ref3 [292, 293 and 294].
However, using immunohistochemical double stainings, we have shown the
presence of a continuous spectrum of cells expressing different amounts
of EBNA2 and LMP1 in relation to distinct morphological features: small
neoplastic cells express only EBNA1 and sometimes also EBNA2; cells of
intermediate size express EBNA1, EBNA2 and LMP1, and large blasts, sometimes
resembling H-RS cells express EBNA1 and LMP1 Fig. 3ref [106].
Whether identical biological events underly PTLD development in SOT and
BMT remains to be definedref [295]. In BMT, lymphoid tissues
are severely depleted from resident regulatory lymphoid cells that may
influence the activation and outgrowth of EBV-positive B-cells, whereas
in SOT, local regulatory (T-) cell function(s) is merely decreased by immunosuppressive
medication leaving the overall architecture of lymphoid organs intact.
The therapeutic role for EBV-reactive cytotoxic T-cell infusions in the
prevention and treatment of BMT-associated PTLD is well establishedref1,
ref2, ref3 [295, 296 and 297], but this is more complicated and controversial
in SOTref1, ref2, ref3 [298, 299 and 300]. A recent therapeutic
approach involves the use of humanized monoclonal antibodies, such as Rituximab,
directed against the pan-B-cell marker CD20, which can effectively eliminate
B-cells from the host, but is not without complicationref1, ref2
[301 and 302].
-
AIDS-related lymphomas (ARLs) are mostly of B-cell origin and contain the
patient's intrinsic EBV. Moreover, unlike PTLDs, most ARLs contain monoclonal
EBV genomes [292 and 303] and both type 1 and type 2 EBV-strains are detectable,
frequently co-infecting the hostref1, ref2 [304 and 305]. Recent
data indicate a more equal distribution of EBV types 1 and 2 in AIDS patients
and no prevalent association between EBV type 2 and ARL was foundref
[306]. 2 different types of ARL are recognized that differ amongst others
in their EBV association and EBV gene expression patterns. Most of the
cases of diffuse large B-cell lymphoma, especially those arising in the
central nervous system, are associated with EBV and have an EBV gene expression
pattern comparable to that of PTLDref1, ref2 [292 and 303].
By contrast, only 30–40% of AIDS-related BL is associated with EBV [307,
308, 309 and 310]. These lymphomas never show EBNA2 expression and only
rarely LMP1, suggesting a latency type I or II expression pattern. In addition,
AIDS-related BL, like endemic and sporadic BL, carry the t(8; 14) translocation
involving the c-myc generef [292]. Overall, AIDS-NHL represents
a spectrum of lymphomas with a somewhat different pathogenesisref
[311]. The differences between diffuse large B-cell lymphomas and Burkitt-type
lymphomas in AIDS-patients suggest that the pathogenesis of the former
resembles that of PTLDs, with an EBV driven proliferation eventually leading
to malignant lymphoma, while the pathogenesis of AIDS-related BL resembles
that of sporadic BL. In addition to chronic antigenic stimulation, it is
thought that HIV-induced B-cell proliferation plays a role in the pathogenesis
of AIDS-related BL. Interestingly, the majority of ARL contains mutations
in the Bcl-6 gene which is regarded as a marker of B-cell transition through
the germinal centerref [312]. Thus, it is conceivable that not
only AIDS-related BL but also other AIDS-related NHL are derived from germinal
center B-cells.
-
lymphomas in immunocompetent patients
-
non-Hodgkin's lymphoma (0.5%) :
-
Burkitt's lymphoma (BL)
is a B-cell lymphoma that was originally described in equatorial Africa
where it accounts for approximately 50% of all childhood cancersref
[313]. Histologically, the tumor is composed of monomorphic medium-sized
cells, often with macrophages scattered among them (‘starry sky macrophages)ref
[314]. The first indication for a viral etiology was the climate-dependent
distribution of the disease: BL had a high incidence in regions where malaria
prevailed. EBV is present in approximately 95% of these endemic BLs. However,
in Europe and the USA, the association is only 10–20%. This difference
has been ascribed to the early age of EBV-infection in Africa compared
with industrial nationsref [315]. In the endemic tumors, the
EBV latent gene expression pattern is very restricted: LMPs are not detected,
and due to Qp promoter usage for EBNA1 transcriptionref [316],
only EBNA1 but not the other EBNAs are expressed (latency
type Iref, [317]). Rarely, isolated tumor cells may express
other EBV genes like EBNA2, LMP and BZLF1ref [318]. Another
important feature of both endemic and non-endemic BL is the presence of
the t(8; 14) translocation, which juxtaposes the c-myc gene on chromosome
8 to the Ig heavy chain locus on chromosome 14, resulting in overexpression
of c-mycref [319]. It was hypothesized that EBNA1, which can
upregulate expression of RAG-1 and -2 may play a causal role for this translocationref1,
ref2 [88 and 89]. A recent study showed that RAG mRNA's could not
be detected in EBV-associated BL, nor in other EBV-associated lymphomasref
[320]. It cannot be excluded, however, and indeed it is likely, that RAG
induction is mainly important during the initial stages of BL development.
-
B-cell chronic lymphocytic
leukemia (B-CLL)

-
the rate of detection of EBV in other B-NHL is relatively low (between
2 and 10%ref1, ref2 [321 and 322]). Only in pyothorax-associated
pleural high-grade B-cell NHLs, clonal EBV genomes can often be demonstrated
in the tumor cellsref1, ref2 [323 and 324]. However, these tumors
relatively often show the presence of a presumably less transforming EBV-strain
(EBV type 2). This, and the apparent latency type
III expression pattern (EBNA2 and LMP1 positive) favor an underlying
immune defect in these casesref [325]. The EBV gene expression
pattern in B-NHL of immunocompetent patients is usually latency
type II-like, with transcription of BHRF1 but no EBNA2ref1, ref2
[210 and 322]. At present, it is not clear whether BHRF1 is expressed at
the protein level, nor whether these transcripts are expressed in the neoplastic
B-cells or in EBV-positive reactive B-cells. This, among others, illustrates
the difficulty of inferring latency patterns from RT-PCR analyses.
-
Hodgkin's lymphoma (HL)
: histologically, HD is characterized by mononuclear Hodgkin cells and
their multinucleated variant, the Reed–Sternberg cells, together abbreviated
as H-RS cells. These are embedded in a background of reactive cells, including
lymphocytes, plasma cells, histiocytes and eosinophilsref [345].
Most recent studies indicate that the H-RS cells in many (but not all)
cases of HD are derived from B-cellsref [346]. The most
characteristic epidemiological feature of HD as seen in most western populations
is the bimodal age-incidence curve. In these populations, very few cases
occur among children; the incidence then increases, peaking at about age
25; the incidence decreases to a plateau level through middle age and increases
again with age to a second peak. Males are more often affected than females,
which is most clearly observed in the older age group. There is clear evidence
that the risk for HD occurring from early childhood through middle age
is associated with factors in the childhood environment that influence
the age at which infection with EBV takes place, (i.e. social class, population
density, geographic region)ref (reviewed in [347]). Moreover,
several independent cohort studies have shown that people who have had
IM run a3-fold higher risk for getting HD. Interestingly, the majority
of HDs in patients who have had IM is not associated with EBV (summarized
inref1, ref2 [348 and 349]). It has been suggested that EBV
is an important cofactor in the pathogenesis of HD, but that in patients
with relatively intact immune systems, EBV+ neoplastic cells
are eradicated and only cells capable of virus-independent growth survive
(hit-and-run
mechanismref [350]). A role for anti-LMP1 antibody responses
was recently suggested in this process, because in a large study of juvenile
HD patients, those with highest anti-LMP1 responses had EBV-
tumorsref [351]. Detection of monoclonal EBV within the H-RS
cells of HD was first reported in 1989ref [352]. One year later,
the presence of EBER RNAs in H-RS cells of was reportedref [353].
The outcome of numerous studies shows that in most industrialized countries
EBV is associated with on average 40–60% of HD cases, depending on their
histologyref (reviewed in [347]). Depending on race and socio-economical
status of the population studied, this percentage may approach 100% as
found in some developing countriesref1, ref2 [349 and 354].
It is thought that different pathogenic mechanisms play a role in EBV-positive
and EBV-negative HDsref [355]. Although
patients with EBV-positive
HD were shown to respond better to chemotherapyref [356],
EBV genome status of HD is by itself not an independent prognosticatorref
[357]. The EBV gene expression pattern in H-RS cells resembles that in
NPC: expression of LMP1 without detectable EBNA2, characteristic of latency
type IIref1, ref2 [358 and 359]. Most intriguing is the
finding that high levels of tumor infiltrating activated CTLs are associated
with a bad prognosis of HDref [360]. This is surprising
because one would expect that CTLs present in such large number would eradicate
the neoplastic cells. However, clonotypic analysis of tumor infiltrating
T-cells revealed an oligoclonal response, indicating that these cells were
attracted
to the tumor site in an antigen independent mannerref [361].
It is possible that the CTLs present in HD may not be specific for the
tumor antigens presented, but are rather attracted by the abundance
of cytokines produced at the site of the tumorref [163].
An impairment of antigen presentation by the H-RS cells can probably be
excluded because HD cell lines transfected with immunodominant EBV-proteins
were able to process and present these endogenously synthesized proteins,
and were subsequently killed by specific CTL clonesref [362].
Alternatively, a local immune response inhibition may be present in
vivo in the tumor, as was first shown by Frisan et al.ref
[363] and subsequently confirmed by Chapman et alref [364].
An active role for LMP1-mediated enhanced local production of IL-10 in
this process is suggestedref1, ref2 [365 and 366], but quantitative
immunohistochemical analysis failed to reveal a detectable influence of
IL-10 production on local T-cell populations in EBV- cases of
HDref [161]. Another challenging possibility is the direct immunosuppression
by LMP1, secreted from H-RS cells in the form of exosomesref
[131]. On the other hand, a (long-term) process of in vivo selection
for apoptosis resistance of the H-RS cells may determine the malignant
phenotype of HD and determine the long-term prognosis, either by providing
resistance to CTL-(granzyme-B) mediated killing or by providing resistance
to therapy induced cell damage. Apoptosis resistance may be conferred by
genetic defects in the major enzymatic pathways mediating apoptosis, by
upregulation of multidrug-resistance gene-expression or by the expression
in the tumor cells of PI-9, a direct inhibitor of granzyme B functionref
[367]. Given the in vitro properties of LMP1, its expression may
result in apoptosis resistance via upregulation of host-cell encoded anti-apoptotic
factors bcl-2ref [144] and A20ref [145]. However,
several studies have shown that bcl-2 expression in the Reed–Sternberg
cells of EBV+ HD cases is not significantly higher than that
in EBV- HD casesref1, ref2 [368 and 369]. Finally,
LMP1 expressed by EBV+ H-RS cells may have additional tumor
growth enhancing effects by inducing MMPs that can affect the function
of infiltrating T-cellsref1, ref2 [370 and 371] and by inducing
the production and secretion of IL6, IL-8 or COX-2 stimulating autocrine
growth and neo-vascularizationref1, ref2, ref3 [372, 373 and
374]. Delayed exposure to infection may increase risk of EBV-positive HL
in young adults, but risk patterns differ in younger and older women for
both EBV+ and EBV- HL. Late EBV infection does not
appear relevant to risk, suggesting that other pathogens impact HL etiology
in affluent female populationsref.
Therapy : EBV proteins expressed during latency
may serve as targets for novel chemotherapeutic agents. EBV-transformed
B cells grow in tight clumps in vitro. EBV gene expression differs
among malignancies associated with the virus. EBV+ Burkitt lymphoma
tissue shows expression of EBNA-1, but not the other latency-associated
proteins (Rowe et al, 1987). Hodgkin's disease tissues show expression
of EBNA-1, LMP-1, and LMP-2 (Deacon et al, 1993) while nasopharyngeal carcinoma
(Fahraeus et al, 1988) and T-cell lymphomas (Anagnostopoulos et al, 1992)
express EBNA-1, LMP-2 and have variable expression of LMP-1. EBV lymphomas
in immunocompromised persons generally show expression of each of the 9
latency associated proteins. While the treatment for some EBV-associated
malignancies has improved in recent years, newer approaches to therapy
are needed. LMP-1 is the latency-associated protein that has been most
directly linked to oncogenesis by EBV. Expression of LMP-1 in B cells of
transgenic animals results in B-cell lymphomas (Kulwichit et al, 1998).
LMP-1 upregulates the expression of a large number of proteins on the surface
of virus-infected B cells, including ICAM-1, LFA-1, and LFA-3 (Figure 1A)
(Peng and Lundgren, 1992). Expression of LMP-1 in lymphoma cells induces
clumping of the cells (Wang et al, 1990). LMP-1 acts as a functional homologue
of a constitutively active form of CD40. LMP-1 oligomerises on the surface
of virus-infected cells and binds to TRAFs 1,2,3 and 5, TRADD, RIP, and
JAK3 (Devergne et al, 1996). The interaction of LMP-1 with these proteins
results in activation of NF-kB, c-jun N-terminal
kinase, STATs, the p38 MAP kinase pathway, and stress-activated kinases.
This results in constitutive B-cell proliferation and inhibition of apoptosis.
LMP-1 also interacts with p85 to activate the PI3K/Akt pathway and increase
cell survival (Dawson et al, 2003). LMP-1 upregulates several other antiapoptotic
proteins including A20, Mcl-1, bcl-2, and bfl-1. EBV-associated B-cell
lymphomas in humans show activation of NF-kB,
and LMP-1 colocalises with TRAF-1 and TRAF-3 (Liebowitz, 1998). In addition,
TRAFs 1, 2, and 3 and NF-kB are expressed in
post-transplantation lymphoproliferative disorders (Ramalingam et al, 2003).
Furthermore, these lesions show expression of adhesion molecules upregulated
by LMP-1 including LFA-1 (Hamilton-Dutoit et al, 1993). Thus, LMP-1 is
critical for B-cell proliferation and development of lymphomas
in vivo
-
treatment of EBV-transformed B cells with NF-kB
inhibitors (e.g. IB mutant, Bay11-7082) has been shown to induce apoptosis
of the cells (Cahir-McFarland et al, 2000, 2004)
-
treatment of EBV-transformed cells with simvastatin also inhibits
NF-kB and induces apoptosis of EBV-transformed
B cells (Katano et al, 2004). Statins inhibit 3-hydroxy-3-methylglutaryl
coenzyme A (HMG-CoA) reductase. 6 of these compounds, atorvastatin, fluvastatin,
lovastatin, pravastatin, rosuvastatin, and simvastatin, are approved by
the FDA for use in humans. Each of these compounds is used to treat elevated
serum cholesterol. HMG-CoA reductase catalyses the conversion of HMG-CoA
to mevalonate, which ultimately leads to synthesis of cholesterol. Therefore,
statins reduce the level of mevalonate with a subsequent reduction in cholesterol.
Statins have a number of other activities related to their inhibition of
HMG-CoA reductase (reviewed in Raggatt and Partridge, 2002). Mevalonate
is a precursor for isoprenoids, including geranyl pyrophosphate and farnesyl
pyrophosphate. Statins reduce the levels of these compounds. Post-translational
modification of proteins by farnesylation or geranylgeranylation results
in their association with cell membranes and activation (reviewed in Bellosta
et al, 2000). These modified proteins include members of the nuclear laminin
family, ras, inositol triphosphate 5-phosphatase, which are farnesylated,
and Rho, Rac, cdc42, Rab, Rap, and G-proteins, which are geranylgeranylated.
These changes have pleotrophic effects including inhibition of smooth muscle
proliferation (Corsini et al, 1993), inhibition of MHC class II complexes
on antigen-presenting cells (Kwak et al, 2000), increase in bone morphogenetic
protein (Mundy et al, 1999), suppression of T- and B-cell responses (Kurakata
et al, 1996), reduced NK cell activity (Hillyard et al, 2004), reduced
synthesis of chemokines (Waehre et al, 2003), and growth arrest of certain
transformed cells (Graaf et al, 2004). These effects can be reversed with
the addition of mevalonate. Certain statins have activities that are unrelated
to their inhibition of HMG-CoA reductase. Kallen et al (1999) showed that
lovastatin binds to the I-domain of LFA-1 and blocks its interaction with
ICAM-1. LFA-1 (L2 integrin) is an adhesion molecule that promotes diapedesis
of leucocytes across the endothelium and is a costimulatory molecule on
activated T cells. The I-domain of LFA-1 is separate from the site of its
binding to its ligand ICAM-1. Weitz-Schmidt et al (2001) subsequently showed
that simvastatin, mevastatin as well as lovastatin (but not pravastatin)
can bind to LFA-1 and block its binding to ICAM-1. This interaction is
independent of HMG-CoA and is not reversed by mevalonate. Simvastatin and
lovastatin concentrations of ~10 M are required to block these interactions;
this is in contrast to the nanomolar concentrations required to inhibit
HMG-CoA. A synthetic statin (LFA703) which lacks HMGCoA reductase inhibitory
activity, but which has increased LFA-1 binding activity, blocks LFA-1-induced
costimulation of T cells and suppresses the inflammatory response to thioglycollate
in a mouse model of peritonitis. Statins have been shown to affect replication
of HIV. HIV virions have ICAM-1 on their surface, which can bind to LFA-1
on target cells and enhance virus attachment (Fortin et al, 1997). Lovastatin
inhibits replication of HIV by inhibiting the interaction of ICAM-1 on
virions with LFA-1 on the surface of target cells (Giguere and Tremblay,
2004). Lovastatin inhibits HIV-induced Rho GTPase activation, which is
important for HIV infection of cells (del Real et al, 2004). Entry into
and exit from HIV-infected cells is blocked by lovastatin and this effect
can be reversed by treatment with mevalonate. Treatment of HIV-infected
severe combined immunodeficiency (SCID)-hu mice or humans with lovastatin
results in a reduction in HIV RNA loads and increased CD4+ T-cell
counts. Treatment of EBV-transformed B cells with 2 M of simvastatin, atorvastatin,
or lovastatin resulted in dissociation of cell clumps and death beginning
5 days after treatment with the drug (Katano et al, 2004). Cell death induced
by simvastatin was due to apoptosis as demonstrated by detection of fragmented
DNA. While simvastatin induced cell death in EBV-positive Burkitt lymphoma
cells (such as Akata, Mutu-1, Mutu-3, and P3HR-1 cells), EBV-negative B
cells, and EBV-negative T cells, the concentration of simvastatin required
to kill these cells (4 M) was higher than for cells transformed with EBV
in vitro. Simvastatin and lovastatin block the interaction of LFA-1
with ICAM-1, while pravastatin does not. In contrast to apoptosis of EBV-transformed
B cells induced by simvastatin, treatment of these cells with up to 16
M of pravastatin did not dissociate cell clumps or induce cell death (Katano
et al, 2004). In addition, antibody to LFA-1, which can activate lymphocytes
(Perez et al, 2003), prevented cell death induced by simvastatin. LMP-1
is present in lipid rafts on the cell membrane (Higuchi et al, 2001). Lipid
rafts are microdomains in the membrane that are rich in cholesterol and
sphingolipids and are resistant to detergent extraction. They are important
for signal transduction in B cells by CD40 or by immunoglobulin on the
surface of the cells. The carboxy terminal domain of LMP-1 is activated
when targeted to lipid rafts where it induces signalling and activation
of NF-B-mediated transcription (Kaykas et al, 2001). A mutation in one
of the transmembrane domains of EBV LMP-1 results in lack of LMP-1 localisation
to rafts, failure to bind TRAF 3, and loss of activation of NF-kB
(Yasui et al, 2004). Treatment of EBV-transformed B cells with 2 M simvastatin
for 3 days reduced the amount of LMP-1 present in lipid rafts by 85% (Katano
et al, 2004). While this effect might have been attributable to the depletion
of cholesterol by the statin, it occurred at a relatively low level of
simvastatin and treatment of cells with 8 M pravastatin did not result
in a reduced amount of LMP-1 in rafts. Treatment of EBV-transformed B cells
with 2 M simvastatin for 3 days inhibited NF-B activation. Cells treated
with 8 M pravastatin did show reduced activation of NF-kB.
Since LMP-1 also activates the phosphatidylinositol 3-kinase/Akt pathway
(Dawson et al, 2003), displacement of LMP-1 from lipid rafts may reduce
activation of this pathway and inhibit survival of EBV-transformed B cells.
While LMP-2 has also been shown to be present in lipid rafts (Dykstra et
al, 2001) and simvastatin might have an effect on LMP-2, the observation
that LMP-2 is dispensable for B-cell transformation by EBV (Speck et al,
1999), suggests that the effect of simvastatin in killing transformed B
cells is unlikely to be mediated through LMP2. The effects of simvastatin
on EBV-transformed B cells cells could be due the ability of the drug to
block adhesion molecule interactions on the surface of B cells, or to displace
LMP-1 from rafts and inhibit NF-kB (Figures
1B and 2B). Examination of multiple EBV-positive and EBV-negative cell
lines showed that induction of cell death by simvastatin correlated best
with expression of LMP-1 and activated NF-kB
in the cells prior to treatment with drug (Katano et al, 2004). Cells expressing
the highest levels of LMP-1 and NF-kB (EBV-transformed
B cells) were most susceptible to simvastatin-induced cell death; one cell
line (Mutu-3 Burkitt lymphoma cells) expressing lower, but detectable levels
of LMP-1 and NF-kB showed an intermediate sensitivity
to death by simvastatin. However, other EBV-positive cells (Akata, P3HR-1,
and Mutu-1 Burkitt lymphoma cells) that expressed low levels of NF-kB
and no detectable LMP-1 were much less sensitive to simvastatin. One of
these latter cell lines (P3HR-1) expressed levels of LFA-1 and ICAM-1 that
were similar to those in EBV-transformed B cells. Taken together, these
finding suggest that inhibition of NF-B by simvastatin may be more important
than its ability to block the interaction of LFA-1 with ICAM-1. Intraperitoneal
injection of EBV-transformed B cells into SCID mice results in development
of EBV-positive lymphomas that resemble the tumours seen in immunosuppressed
persons (Rowe et al, 1991). These tumours show a pattern of gene expression
similar to that in patients with EBV lymphoproliferative disease with EBNAs,
LMP-1, and adhesion molecules detected in the tumours. Oral treatment of
SCID mice with simvastatin beginning 3 days prior to injection with EBV-transformed
B cells resulted in a statistically significant improvement in survival
rate compared to animals not given the drug (Figure 3) (Katano et al, 2004).
While there was a trend for longer survival for mice treated with simvastatin
beginning 7 days after injection with EBV-transformed B cells, the difference
with untreated mice was not statistically significant. Some tumours from
mice that were treated with simvastatin showed downregulation of LFA-1
on their surface, compared with the EBV-transformed B cells that had been
used to inject the animals. The dose of simvastatin used to treat these
mice (250 mg/kg/day) is estimated to result in serum levels that would
be 4 to 8 times that of humans receiving the maximum dose of simvastatin
(80 mg/day) used to lower serum cholesterol. However, similar high serum
levels have been achieved in humans treated with large doses of statins
in cancer therapy trials). Statins have been shown to induce apoptosis
in several proliferating tumour cell lines, including certain leukaemia,
lymphoma, astrocytoma, pancreatic carcinoma, and neuroblastoma cell lines
(reviewed in Wong et al, 2002). This effect is due to inhibition of HMG-CoA
reductase since it can be inhibited by mevalonate. Lovastatin reduced viability
of EBV-positive or EBV-negative Burkitt lymphoma cells by >75% due to inhibition
of geranylgeranylation (van de Donk et al, 2003). Statins have been used
in animal models and have reduced the tumour burden associated with melanoma,
hepatoma, neuroblastoma, pancreatic cancer, and lung cancer (reviewed in
Wong et al, 2002). In other animal models, statins have been used as adjunctive
therapy in combination with cytotoxic agents in mouse models including
carmustine for melanoma and with doxorubicin for lung cancer. A number
of studies have tested the role of oral statin therapy in patients with
tumours (reviewed in Wong et al, 2002). Lovastatin given in doses ranging
from 2 to 45 mg/kg/day for 1 out of every 4 weeks was used to treat a variety
of tumours including astrocytoma, glioblastoma, and prostate, breast, ovarian,
and lung cancer (Thibault et al, 1996). Doses of 25 mg/kg/day for 7 days
were well tolerated. Serum levels of the drug ranged from 0.1 to 3.9 M.
One patient with an astrocytoma had a minor response to therapy. Since
mice metabolise statins more rapidly than humans, a dose of 25 mg/kg/day-1
is comparable to ~250 mg/kg/day in mice. While large doses of statins have
been used in patients with cancer, might standard doses of statins prevent
development of haematologic malignancies? Large trials of simvastatin for
the prevention of coronary events have evaluated the incidence of cancer
in study recipients. No significant decrease in the overall incidence of
cancer or of haematologic malignancies was noted in the MRC/BHF Heart Protection
Study involving 20 536 patients randomised to 40 mg of simvastatin per
day or placebo for a median of 5 years (Heart Protection Study Group, 2002).
Similarly, there was no significant decrease in the incidence or mortality
for cancer, or for lymphatic or haematopoeitic malignancies in the Scandinavian
Simvastatin Survival Study in which 4444 patients were randomised to simvastatin
(20-40 mg per day) vs placebo for a median of 5.4 years (Strandberg et
al, 2004). Thus, simvastatin at the standard doses to prevent coronary
events did not have a significant effect on the risk of haematologic cancers.
EBV lymphomas occur in immunocompromised patients such as organ or stem
cell transplant recipients, patients with HIV, or patients with congenital
immunodeficiencies. These tumours generally express each of the EBV latency
proteins and the tumours are driven by these viral proteins. Central nervous
system lymphomas account for 20% of lymphomas in AIDS patients and express
EBV LMP-1 and other latency proteins (MacMahon et al, 1991). Immunoblastic
lymphomas account for about 60% of cases of lymphoma in patients with AIDS
and usually express EBV LMP-1 and other latency proteins (Hamilton-Dutoit
et al, 1993). These tumours generally occur in AIDS patients late in the
course of disease when CD4+ T cell numbers are low. Tumours
in immunocompromised patients can occur in lymph nodes, but frequently
present at extranodal sites such as the gastrointestinal tract, central
nervous system, liver, lung, bone marrow, or transplanted organ (Cohen,
2000). The tumours can be polyclonal or monoclonal and usually lack chromosomal
translocations. Patients who lack immunity to EBV at the time of transplant
and develop primary EBV infection after transplant are more likely to develop
EBV-driven lymphomas. Transplant recipients who receive HLA-mismatched
or T-cell-depleted bone marrow or infusions of antilymphocyte antibodies
for rejection are also at increased risk for development of lymphomas.
EBV viral DNA is often elevated in peripheral blood mononuclear cells of
these patients prior to, and at the onset of lymphoma, indicative of the
EBV-driven B-cell proliferation. Patients with these tumours have impaired
T-cell immunity to EBV resulting in failure to regulate EBV-driven B-cell
proliferation. Treatment for these lymphomas includes reduction in immunosuppression
when possible. Resection of localised lesions, especially in the gastrointestinal
tract has been effective in some patients. Monoclonal anti-CD20 antibody
(rituximab) results in remissions in about 50% of patients. Interferon-
has been effective in some patients, but may increase the risk of rejection
of the transplanted organ. Lymphomas in stem cell transplant recipients
are usually of donor origin; infusions of donor T cells (which are HLA-matched)
have been effective in many cases of EBV lymphoma in these patients. Lymphomas
in organ transplant recipients are usually recipient in origin; infusions
of autologous or HLA-matched T cells have been effective. Radiation therapy,
especially for central nervous system lesions, and cytotoxic chemotherapy
are used for refractory cases. The latter two therapies are frequently
used for lymphomas in AIDS patients whose immune systems are less responsive
to immunologic-based therapies. Statins may have a role as adjunctive therapy
in some patients with EBV-driven lymphomas. These might include stem cell
transplant recipients in whom donor T cells are not available, organ transplant
recipients whose lymphomas are not responsive to reduction of immunosuppressive
therapy and in whom HLA-matched T cells are not available. In these settings,
statins might be used in combination with other therapies (e.g. rituximab
or interferon-) which by themselves result in remissions in about 50% of
patients. Statins might be tried in AIDS patients with EBV-driven lymphomas,
especially those with very low CD4T cell counts who tend to respond less
well to chemotherapy. Statins might also be considered for patients with
EBV-positive Hodgkin's disease who are refractory to chemotherapy and radiation
therapy. Tumours from these patients usually express LMP-1 as well as adhesion
molecules (Sandvej et al, 1993), and statins might reduce the viability
of the tumour cells. While nasopharyngeal carcinoma and T-cell lymphomas
frequently express LMP-1, the tumours show more variable expression of
the protein and therefore might be less susceptible to statins. Finally,
Burkitt lymphomas do not express LMP-1 and therefore would be unlikely
to respond to statins at doses that are effective for cells transformed
with EBV in vitro. Identification of signalling pathways for EBV-mediated
transformation has helped to identify new targets and potential treatments
for these tumours. Certain statins have been shown to inhibit the interactions
of adhesion molecules and block NF-B activation in EBV-transformed cells,
resulting in apoptosis. Simvastatin delays the development of EBV-lymphomas
in SCID mice inoculated with EBV-transformed B cells. The dose of statin
needed to induce apoptosis is much higher than that required for lowering
serum cholesterol, but such doses have been tolerated by patients in clinical
trials. Statins may have a role in the treatment of EBV-driven lymphomas,
most likely as part of combination therapy for these lymphomas.
-
intentional induction of the lytic form of EBV infection combined with
ganciclovir
treatmentref1,
ref2,
ref3,
ref4,
ref5.
GCV and AZT are not generally effective for treating EBV-positive tumors
because most tumor cells are infected with the latent form of EBV and therefore
do not express the kinases which activate these drugs. Virally encoded
kinases (thymidine kinase and BGLF4) which are expressed only during the
lytic form of infection convert GCV into its active, cytotoxic form. The
ability of various chemotherapy drugs to induce lytic EBV induction is
clearly cell type dependent :
-
in EBV-positive epithelial cell tumors
-
previous studies showed that certain chemotherapy agents (fluorouracil
[5-FU]
and cis-platinum
)
efficiently induce a lytic infection in vivo and in vitro and that
the addition of GCV greatly enhances the ability of both 5-FU and cis-platinum
to inhibit NPC tumor growth in nude miceref
-
in EBV-positive B-cell tumors, however, EBV infection is primarily
latent (in contrast to epithelial cells, in which the virus infection is
often lytic) and consequently it has been more difficult to identify agents
that can efficiently induce a lytic EBV infection in B-cell tumors.
-
although g-irradiation induces lytic EBV infection
and expression of the viral TK in EBV-positive lymphoblastoid cell lines
(LCLs) both in vitro and in vivoref1,
ref2
and might be useful for treating localized EBV-positive lymphomas combined
with GCV, this method cannot be used to treat widely disseminated EBV-positive
B-cell lymphomas
-
gemcitabine
and doxorubicin
(but not 5-azacytidine
,
cis-platinum
,
or fluorouracil [5-FU]
)
induce lytic EBV infection in EBV-transformed B cells in vitro and
in
vivo. Gemcitabine and doxorubicin both activated transcription from
the promoters of the 2 viral immediate-early genes, BZLF1 and BRLF1, in
EBV-negative B cells. This effect required the EGR-1 motif in the BRLF1
promoter and the CRE (ZII) and MEF-2D (ZI) binding sites in the BZLF1 promoter.
GCV enhanced cell killing by gemcitabine or doxorubicin in lymphoblastoid
cells transformed with wild-type EBV, but not in lymphoblastoid cells transformed
by a mutant virus (with a deletion in the BZLF1 immediate-early gene) that
is unable to enter the lytic form of infection. Most importantly, the combination
of gemcitabine or doxorubicin and GCV was significantly more effective
for the inhibition of EBV-driven lymphoproliferative disease in SCID mice
than chemotherapy alone. In contrast, the combination of zidovudine and
gemcitabine was no more effective than gemcitabine aloneref
-
another approach for inducing lytic EBV infection in tumors would be to
introduce the BZLF1 or BRLF1 genes under the control of a strong heterologous
promoter by gene delivery methods. It was recently shown that direct delivery
of the 2 EBV IE genes into nasopharyngeal carcinomas by using adenovirus
vectors induces lytic EBV gene expression in the tumors and inhibits tumor
growthref.
However, adenovirus delivery of the 2 EBV IE genes to B-cell lymphomas
would be limited by the fact that B cells express very little (if any)
of the major receptor (CAR) for adenovirus, although this hurdle could
potentially be overcome by the use of bispecific antibody techniquesref.
-
previous reports have shown that sodium butyrate (which induces
histone acetylation) effectively induces the lytic form of EBV infection
in at least some Burkitt's lymphoma cells either aloneref,
or associated with radiationref,
and the combination of arginine butyrate and GCV to treat EBV lymphomas
in patients is currently being studiedref
Nevertheless, drug-based strategies for inducing lytic EBV infection in
widely disseminated EBV-induced lymphoproliferative disease are more likely
to be successful clinically than gene delivery strategies
-
epithelial diseases :
-
respiratory tract infection (40%)
-
Gianotti-Crosti
syndrome (GCS) / papular acrodermatitis of childhood (PAC)

-
oral hairy
leukoplakia (OHL)
is a white epithelial lesion of the tongue which can be found in immunocompromized
individuals. EBV-DNA was first detected in this lesion in 1985, with localization
restricted to the superficial epithelial cellsref [414]. Expression
of viral lytic cycle antigens has been shown in OHL and abundant production
of virus particles has been demonstrated using TEM, indicating that epithelial
cells can support viral replicationref1, ref2 [273 and 275].
The detection of linear EBV genomes in the absence of episomal DNAref1,
ref2 [415 and 416], the exclusive localization of the virus in upper
epithelial cells, and the sensitivity of the lesion to acyclovir treatmentref
[417], support this notion. Expression of latent proteins in OHL has also
been reported but EBER RNAs are expressed at very low levelsref
[416]. Recent data indicate that LMP1 may contribute to the proliferation
and apoptosis resistance of OHL epithelial cellsref [151].
-
epithelial malignancies : as discussed above, the pathogenesis of
lymphomas is a multistep process. This also holds true for the genesis
of carcinomasref;
in fact, similar pathogenic mechanisms play a role in the genesis of various
types of carcinomas. These are among others inhibition of apoptosisref,
inactivation of tumor suppressor genesref
and genetic instabilityref.
Environmental and dietary influences are considered to play an important
role in the accumulation of genetic damages leading to disregulated control
of epithelial cell differentiation and proliferation, ultimately leading
to carcinoma formation. In addition, the presence of certain micro-organisms
is thought to contribute to carcinogenesis. Interestingly, several types
of carcinomas are specifically associated with certain micro-organisms
as is the case for lymphomas. For example, the causal relation between
cancer of the uterine cervix and high-risk human papillomavirus infection
is well established. Furthermore a (in)direct role for of hepatitis viruses
(HCV, HBV) in the genesis of hepatocellular carcinoma and H. pylori
in the outgrowth of gastric cancer is clearly indicatedParkin, DM
5-33. EBV is most clearly associated with nasopharyngeal carcinomasref
and a substantial percentage of gastric adenocarcinomasParkin, DM
5-33, ref2,
but is also found in rare salivary gland and thymic carcinomasref.
Finally, EBV-specific DNA, RNA and proteins have been detected in hepatocellular
carcinomaref
and invasive breast cancersref1,
ref2,
but the relevance of these findings remains to be determinedref1,
ref2.
How EBV acquires access to epithelial cells remains a matter of debate,
but this may involve low level CD21 (the EBV receptor) expression on IFN-activated
epithelial cells in combination with BZLF2–gp42 MHC-II mediated membrane
fusion or IgA-mediated transepithelial transportref1,
ref2,
ref3.
-
anaplastic
or undifferentiated nasopharyngeal carcinoma (NPC) / lymphoepithelioma
: latency II expression pattern. NPC usually arises from the fossa of Rosenmuller
and Eustachian cushions, or in the roof of the nasopharynx and very rarely
in its anterior and lateral wallsref [375]. Unfortunately, NPC
is often detected only after dissemination to cervical lymph nodes which
is associated with a significantly worse prognosisref [376].
The WHO histopathological classification of NPC recognizes keratinizing
squamous-cell carcinoma (well, moderately or poorly differentiated), differentiated
non-keratinizing carcinoma and undifferentiated carcinoma (including lymphoepitheliomas).
Especially undifferentiated NPCs are EBV-associated (up to 100%)ref1,
ref2 [376 and 377]. NPC shows a characteristic racial and geographical
distribution with the highest incidence among males in Southern China [378]
and natives of the Arctic regionref [379]; intermediate rates
of NPC are seen among Southeast Asian peoples of both Chinese and Malayan
backgrounds, and among Arabs and Negroid populations in Kuwait and Northern/Central
Africaref [378]. Subsequent generations of individuals from
these high-risk groups who migrate to a low-risk area have a decreased
risk for NPCref [380], and vice versaref [381]. As
a result of these distribution and migration studies, numerous factors
have been proposed to play a role in the genesis of NPC. These include
consumption of Cantonese-style salted fish and other preserved foods, exposure
to (tobacco) smoke, heavy alcohol use, use of Chinese herbal drugs and
Chinese nasal oil. Moreover, certain HLA types are associated with a higher
risk of NPC, and other hereditary factors may also be of importanceref1,
ref2 (summarized in [348 and 382]). EBV-DNA in NPC biopsies was first
detected by zur Hausen in 1970ref [25]. The epidemiological
and molecular linkage between EBV and NPC is confirmed in many studiesref1,
ref2 [241 and 383]. Clonal EBV-infection was demonstrated in biopsy
samples from NPC in China and the USA and in premalignant lesions of the
nasopharynx, suggesting involvement of EBV prior to the carcinomatous stateref
[384]. The EBV latent gene expression pattern in NPC includes BamHI-A rightward
transcripts (BARTs), EBER, EBNA1 and LMP2 and sometimes LMP1 (latency type
II)ref1, ref2, ref3, ref4, ref5 [385, 386, 387, 388 and 389].
The heterogeneous expression of LMP1 is intriguing, but remains unexplained
thusfar. In addition, also the pathogenic differences between LMP1 positive
and negative NPC tumors remain to be explored. The role of EBV variant
strains, that are frequently detected in NPC also remains unclear, because
similar variants are encountered in healthy regional controls. Like in
HD, bcl2 expression is enhanced in NPC, thus providing apoptosis resistance
to the tumor cells. This may allow for high levels of p53 and concomitant
high PCNA expression, contributing to the malignant phenotype [390, 391
and 392]. On the other hand, unlike HD, NPC cells rarely show upregulated
interleukin expressionref [374]. The detection of tumor-associated
IL-10 has been linked to bad prognosis, but a relation between LMP1 expression
and IL-10 detection at the tumor cell level was not clearly demonstratedref
[223]. In contrasts to EBV-associated lymphoid malignancies, NPC is characterized
by the abundant transcription of the BARF1 ORF ( Fig. 5)ref1, ref2,
ref3 [50, 192 and 386]. An oncogenic role for BARF1 in epithelial
cells is indicated by a number of experimental results in vitro
and in vivoref1, ref2, ref3 [190, 192 and 393]. It has
been suggested that to a certain extent BARF1 expression may compensate
for the reduced expression of LMP1 in NPC, for example by inducing apoptosis
resistance via upregulation of bcl-2ref [195]. As described
for HD, NPC is characterised by an abundant influx of CTL, but the functional
relevance of these CTL remains unknownref [394]. NPC patients
are characterised by high IgG and IgA responses to proteins of the early
and late lytic phase and to EBNA1ref1, ref2 [218 and 241]. However,
except for EBNA1 these proteins are not expressed in the tumor itself.
Therefore, these responses possibly reflect active viral replication at
distinct (sub-epithelial) sites of the body or, alternatively, are directed
against sporadic differentiating epithelial cells within the tumor, that
consequently switch-on the EBV lytic cycleref [265].
Fig. 5. BARF1 NASBA analysis on NPC and HD samples. BARF1 signals are
only found (after blotting and radioactive hybridization of NASBA products)
in the NPC material (in 6 of 7 cases). BJAB, EBV-negative BL cell line;
Louckes BARF1, EBV-negative Louckes BL cell line transfected with BARF1
expression construct.
-
gastric adenocarcinomas
: using PCR and DNA- and EBER-ISH, EBV was first detected in gastric adenocarcinomas
in the USAref [395], in 16% of the cases. Studies in other areas
including Japan, where the incidence of gastric carcinoma is very high,
showed a generally lower proportion of EBV-associated gastric adenocarcinomas
(between 2 and 8%)ref1, ref2, ref3 [396, 397 and 398]. Worldwide,
the overall incidence of EBV+ tumors in regular gastric adenocarcinomas
is 10% (6–16%). Gastric stump carcinomas show a higher frequency of EBV-association,
up to 30%ref [399]. Viral genomes in EBV-associated gastric
carcinomas are monoclonalref [400]. The EBV gene expression
pattern in gastric carcinomas is exceptional: Qp driven EBNA1 transcripts
are detected, and EBER, LMP2 but not LMP1 or EBNA2 are transcribed. Moreover,
transcription of BARF1 is detected and like in NPC it is thought that BARF1
may act as the alternative viral transforming factor [27].
-
lymphocyte-rich carcinomas / lymphoepitheliomas often harbour EBV
in their neoplastic cellsref [401]. This was shown among others
for . Furthermore,
-
oral
squamous cell carcinomas
: ref [408], and cervical
cancer
ref1,
ref2 [410 and 411]. However, using EBER RISH in oral squamous cell
carcinomasref [409] and cervical cancerref1, ref2
[412 and 413], any signals present were confined to EBV-infected reactive
cells and were not found in neoplastic cells. In addition, the absence
of other EBV transcripts in these tumors indicates that EBV does not play
a role in their pathogenesis.
-
breast carcinoma
in French womenref,
but it may have been present in infected lymphocytes in the breast (quantitative
real-time PCR analysis of EBV+ breast carcinoma tissues suggested
that < 0.1% of the cells contained viral genomes : sporadic lytic EBV
infection may contribute to PCR-based detection of EBV in traditionally
nonvirally associated epithelial malignancies), or it is possible that
EBV may enhance the action of HHMMTV, because it
is known that some retroviruses remain dormant unless activity is promoted
by other herpesvirusesref1,
ref2,
ref3.
Evidence againstref.
Recent studies reported the presence of EBV in breast cancer biopsies using
either PCR and in situ hybridizationref [31], or PCR, EBNA1
immunohistochemistry and Southern blotting analysisref [30].
We and others could not detect EBERs or any other EBV transcript in neoplastic
cells in breast cancer samples, although we did find EBV DNA in a small
percentage of cases using PCRref1, ref2 [32 and 33]. An EBER-less
gene expression pattern, however, cannot be excluded, since it was also
found in EBV-RT-PCR positive hepatocellular carcinomasref [29].
-
hepatocellular
carcinoma (HCC)
ref
-
smooth-muscle tumors
that arise with an increased frequency in transplant recipientsref
[406] and AIDS patientsref [407] are often associated with EBV.
Laboratory
examinations :
-
Paul-Bunnel hemagglutination modified by Davidsohn looks for heterophilic
IgM able to agglutinate goat and horse RBCs and to lyse bull RBCs after
pre-absorption with guinea pig kidney extract and bull RBCs (nowadays it
has been replaced by Mono-spot, in which anti-capsule Abs are looked
for). Heterophile antibody tests have similar specificity and are cheaper,
but are less sensitive in children or in adults during the early days of
the illness (SOR: C, based on validating cohort study)
-
serology is most sensitive, highly specific, and also the most expensive
for diagnosing IM (strength of recommendation [SOR]: C, based on validating
cohort study) :
-
viral capsid Ag (VCA)
-
early Ag - diffuse (EA-D)
-
early Ag - restricted (EA-R)
-
Epstein-Barr nuclear Ag (EBNA)
specific IgG avidity index (AI)ref
-
EBV-DNA
-
in the past usually detected by means of Southern blotting. The
disadvantages of this technique are its relative insensitivity (especially
when the % of EBV-infected cells in a certain sample is low) and the need
for relatively large amounts of DNA. However, Southern blotting is still
the appropriate means to determine the clonality of a population of EBV-infected
cellsref1, ref2 [52 and 256]. In addition, Southern blotting
can be used to determine whether EBV in a given sample is episomal or integrated.
-
PCRref [257] has greatly enhanced the sensitivity with
which EBV-DNA and its transcripts are detected in cell lines and clinical
samples. Thus, PCR-based techniques and not Southern blotting are recommended
for mere detection of the presence of EBV in (clinical) samples. In addition,
quantitative PCR techniques for the determination of EBV load have been
describedref1, ref2 [258 and 259] that have proven very useful
for monitoring EBV-DNA load in body fluids and lymphocytes and are currently
being incorporated in routine clinical diagnostic monitoring of high risk
patientsref1, ref2, ref3 [260, 261 and 262]. PCR is more sensitive
than the heterophile antibody test in children, is highly specific, but
is also expensive (SOR: C, based on validating cohort study).
-
% of atypical lymphocytes and total lymphocytes on a complete blood
count provide another specific and moderately sensitive, yet inexpensive,
test (SOR: C, based on validating cohort study).
-
morphological detection of EBV : the actual identification of the
EBV-infected cells is only possible using morphological techniques. First,
DNA in situ hybridization was used. However, this technique is not
very sensitive because it only detects the viral genome, which is present
in limited numbers in most EBV-linked diseases except in OHL. Alternatively,
the high abundance of the EBER1, 2 RNAs in most if not all EBV+ tumor
cells renders them a much better target for RNA in situ hybridization
(RISH) assaysref [263]. At present, EBER RISH is often performed
non-radioactivelyref [264] and it is considered to be the most
reliable technique to detect EBV in clinical materials with preservation
of morphologyref [52]. In recent year a variety of mAbs have
become available for the detection of functional EBV gene expression in
tissues and in circulating B-cells as wellref1, ref2, ref3, ref4,
ref5, ref6, ref7, ref8 [106, 140, 200, 201, 265, 266, 267, 268 and
269]. Combination of EBER RISH and mAb staining and double staining
for different EBV antigens has revealed that EBV-latency antigen expression
at the single cell level may be more heterogeneous than previously appreciatedref1,
ref2 [106 and 201]. These findings are in concordance with current
ideas on the defined and well-regulated gene expression in circulating
EBV+ B-cellsref1, ref2 [170 and 171] and may have
important consequences for understanding the genesis of lymphoproliferative
diseases.
Double staining for EBNA2 (black) and LMP1 (brown) in a post-transplantation
lymphoproliferative disorder. This staining shows a continuous spectrum
of smaller tumor cells staining strongly for EBNA2 but relatively weak
for LMP1, via intermediate phenotypes to larger, more blastic tumor cells
even resembling (mononuclear variants of) Reed–Sternberg cells, staining
positively for LMP1, but negatively for EBNA2.
-
EBV gene expression analysis : EBV transcripts were detected in
the past using northern blotting analysis; however, the RT-PCR developed
more recently allows much more sensitive detection of even low-abundant
mRNAs, and is less material-consuming. A multi-primed adaptation of the
current RT-PCR protocol allows for even more efficient use of small lymphoma
biopsies, and in addition, allows for better quality controls because control
(‘housekeeping gene’) transcripts and the transcripts of interest are reverse
transcribed simultaneouslyref [49]. However, we need additional,
less sensitive, RNA quality controls. Routine gel electrophoresis of the
RNA preparations is recommended to check for the presence of 18S/28S rRNA,
which can be used as markers for the integrity of the RNA in clinical samplesref1,
ref2 [49 and 50]. In addition, nucleic
acid sequence based amplification (NASBA
ref
[270]) proved to be especially suitable for the specific amplification
of non-spliced EBV RNAs, such as BCRF1 (vIL10) and BARF1, even in a background
of genomic DNAref1, ref2, ref3 [50, 54 and 191]. To study the
expression of mRNA at the single cell level, RNA in situ hybridization
can also be used, but its routine use is rather limited to relatively abundant
transcripts. Moreover, in order to definitively demonstrate RNA-mediated
protein expression in the tumor cells, monoclonal antibodies specific for
a variety of EBV proteins such as EBNA1ref1, ref2 [73 and 267],
EBNA2ref [271], LMP1ref1, ref2, ref3 [138, 140 and
269], LMP2ref [272], Zebraref [273], EAref1,
ref2, ref3 [151, 212 and 274] and VCAref1, ref2 [265 and
275] are available. These antibodies allow the detection of the related
proteins both by western blot analysis and by in situ immunohistochemistry.
Differential
diagnosis with other
atypical
lymphocytoses
.
A lymphocyte–white blood cell count (L/WCC) ratio > 0.35 had a specificity
of 100% and a sensitivity of 90% for the detection of mononucleosis and
against acute purulent tonsillitis
ref.
Therapy :
-
inhibitors of viral DNA polymerase : most of them act as competitive
alternative substrates, competing with GTP on the substrate-binding site,
and as DNA chain terminators, by incorporating into the growing DNA chain
and blocking its elongation due to their acyclic structureref
-
acyclic nucleoside analogues (aciclovir, ganciclovir, penciclovir, as well
as their prodrugs valaciclovir (Purifoy DJ, Beauchamp LM, de Miranda P
et al. Review of research leading to new anti-herpesvirus agents in clinical
development: valaciclovir hydrochloride (256 U, the L-valyl ester of acyclovir)
and 882C, a specific agent for varicella zoster virus. J Medical Virol
1993; Suppl. 1: 139–45), valganciclovirref
and famciclovirref,
respectively)
-
acyclovir
ref1,
ref2,
ref3,
ref4,
ref5
is a potent inhibitor of EBV replication in cell culture, but clinical
trials of aciclovir for the treatment of patients with IM have failed to
detect benefitref1,
ref2.
There are several possible explanations for these outcomes
-
the symptoms of IM are insidious in onset, which, coupled with a long incubation
period (4–6 weeks), make for late diagnosis—in contrast, for example, to
herpes labialis or chickenpox
-
EBV is shed in the saliva. The levels of aciclovir achieved in the oropharynx,
particularly after oral administration of the drug, may be inadequate as
judged in part by failure to suppress virus titres in the salivaref1,
ref2,
in contrast to the reduction in titres that are produced by aciclovir given
intravenouslyref.
Presumably, the orally administered aciclovir prodrug, valaciclovir
(Purifoy DJ, Beauchamp LM, de Miranda P et al. Review of research leading
to new anti-herpesvirus agents in clinical development: valaciclovir hydrochloride
(256 U, the L-valyl ester of acyclovir) and 882C, a specific agent for
varicella zoster virus. J Medical Virol 1993; Suppl. 1: 139–45), would
do almost as well as intravenously administered aciclovir, since the prodrug
produces high blood levels of aciclovir
-
most of the symptoms and signs of IM are due not directly to viral cytopathology
in infected tissues, but to immunopathic responses to EBV-infected cells,
particularly EBV-infected B-lymphocytes that circulate in the blood and
infiltrate tissues of affected organs. The virus is usually restricted
to infection of epithelium in the oropharynx and the cervix and of B-lymphocytes,
initially in the tonsillar region. In immunocompetent hosts, the virus
does not usually infect liver cells, neural cells or haematological cells
other than lymphocytes, although liver, neural and bone marrow tissues
and cells may be affected indirectly by immunopathic responses in complications
of IM. Indeed, the atypical lymphocytosis (up to 40% or more of total circulating
white blood cells) characteristic of IM consists of T cells, not EBV-infected
B cells, and signifies the massive cell-mediated immune response mounted
in the course of infection to the proliferating infected B cells (Pagano
JS. Epstein-Barr Virus and the Infectious Mononucleosis Syndrome. In: Humes
HD, Dupont HL, eds. Kelley's Textbook of Internal Medicine, 4th edn. Philadelphia:
Lippincott-Raven Publishers, 2000; 2181–5).
-
Ideally then it seems apparent that antiviral therapy coupled with an immunomodulatory
drug might be effective. Corticosteroids are used empirically by physicians
in treating IM, especially if there is severe pharyngitis, tonsillar swelling
or airway obstructionref,
and in fact a controlled trial of prednisolone
given with aciclovir for treatment of IM was carried out, but without apparent
benefitref.
However, the design of this trial may not have been optimal, as indicated
by the fact that prednisolone alone did not produce the expected resultsref.
Also, use of aciclovir may not have produced adequate levels of the drug,
although there was suppression of shedding of virus in the oropharynx
-
ganciclovir
.
-
acyclic nucleotide analogues (cidofovir
ref
and adefovir
ref)
-
pyrophosphate analogues [phosphonoformic
(foscarnet)
ref
and phosphonoaceticref
acids] interact directly with the pyrophosphate-binding site of the enzyme
-
4-oxo-dihydroquinolines (4-oxo-DHQs) (as represented by PNU-182171
and PNU-183792)ref1,
ref2
showed a broad spectrum of anti-herpesvirus activity. Although non-nucleosides,
the compounds seem to affect viral DNA polymerase and inhibit HSV-1 and
-2, VZV, HCMV, kaposi's sarcoma herpesvirus (KSHV)ref
and EBVref
in cell culture. The mechanism of action of these drugs has not been reported,
but it is clearly different from that of aciclovir and related drugs. Resistance
to 4-oxo-DHQs has been mapped to V823 of the viral DNA polymerase, a residue
that is conserved in the DNA polymerases of six (HSV-1 and -2, VZV, HCMV,
EBV and KSHV) of the 8 human herpesviruses, which implies that the compounds
specifically inhibit these enzymesref.
Acyclic nucleoside and nucleotide analogues require phosphorylation to
their triphosphorylated forms in order to become active. However, in the
case of the nucleoside analogues, the antiviral specificity is based in
part on the fact that viral kinases are more efficient in catalysing the
first step in the drug intracellular metabolism (monophosphorylation),
and so these compounds are more specific. EBV encodes two kinases: a thymidine
kinase (BXLF1 gene product) and a protein kinase (BGLF4 gene product).
However, in contrast to all the other herpesviruses, it is unclear which
of these enzymes is responsible for the monophosphorylation and activation
of nucleoside analoguesref1,
ref2.
Specificity is also imparted by the ultimate target of these drugs, viral
DNA polymerase, which explains why the phosphonated nucleoside analogues
like cidofovir are still specific. It is unclear if herpesvirus DNA polymerases
differ in sensitivity to these drugs since rigorous comparative determinations
of Km's and Ki's of the different herpesvirus
DNA polymerases for any given drug have not been attempted, in part because
of issues of enzyme purification (Pagano JS. Epstein-Barr virus: Therapy
of active and latent infection. In: Jeffries DJ, De Clercq E, eds. Antiviral
Chemotherapy. John Wiley & Sons Ltd. 1995; pp. 155–95). While usable,
all these compounds suffer from a number of drawbacks (i.e. toxic side
effects, poor oral bioavailability and risk for emergence of drug-resistant
virus strains).
-
compounds of mixed nature :
-
benzimidazole D-ribonucleosides inhibit HCMV
ref
but not EBV replicationref.
These compounds prevent the processing and maturation of concatameric viral
DNA to monomeric genomesref1,
ref2.
-
2,5,6-trichloro-1-ß-D-ribofuranosylbenzimidazole
(TCRB)
-
2-bromo-5,6-dichloro-1-ß-D-ribofuranosylbenzimidazole
(BDCRB)
-
maribavir
ref
is an L-ribonucleoside. Surprisingly, this novel compound has a mechanism
of action that is different from that of BDCRB and in fact not yet fully
understood. Maribavir is active against both HCMVref1,
ref2,
ref3,
ref4,
ref5
and EBVref1,
ref2,
ref3,
ref4,
and its effects on these viruses seem to involve direct or indirect inhibition
of viral protein kinasesref1,
ref2,
ref3,
ref4,
ref5
as well as possible interference with nuclear egress of virions during
viral maturationref.
Maribavir resistance in HCMV has been mapped to UL97ref
and UL27ref1,
ref2
genes. In contrast, the EBV BGLF4 gene product, a protein kinase that is
the homologue of HCMV UL97, was not inhibited directly by the drugref1,
ref2,
and EBV does not encode a homologue of UL27. However, phosphorylation of
the EBV DNA-processivity factor, EA-D, by the EBV protein kinase is inhibited
by maribavir in infected cellsref.
Maribavir is the only drug in this group for which Phase I clinical trials
have been completed, viz., for HCMV infectionref1,
ref2,
and Phase II trials were initiated in July 2004ref.
-
ß-L-5-iododioxolane uracilref
-
indolocarbazoles have emerged recently as potential inhibitors of
HCMVref
and EBVref
replication. The mechanism of action of these compounds seems to involve
the inhibition of HCMV UL97 protein kinaseref1,
ref2;
however, the same compounds (except for one, K252a) failed to directly
inhibit EBV BGLF4 protein kinaseref.
These observations suggest that although some compounds may affect different
groups of viruses, the mechanism of their action might be different for
each of these groups.
Mechanisms of action of these drugs are still under study, but they do
not involve inhibition of the viral DNA polymerase.
-
EBV-specific
CTLs
specific for have been successfully used in immunotherapy of PTLD,
Hodgkin's disease, and in HIV/AIDS patients who also have high risk of
developing EBV-associated lymphomaref
Patients given
ampicillin
or
amoxicillin
develop a rash (a.k.a.
Dave Rouse disease)
Except for OHL, which is benign and represents a purely lytic infection
with large amounts of EBV replicating freely in the lesions, all the other
EBV diseases are malignancies characterized by latent infection that relies
on cellular enzymes for EBV episomal DNA synthesis. Therefore, it is not
expected that antiviral drugs directed at viral synthetic processes would
affect EBV in the latent phase. In latent infection, the linear double-stranded
genomes characteristic of productive lytic infection and encapsidated in
virus particles are not made. Rather infection persists via controlled
replication of viral episomes that are found only in the nucleus in a nucleosomal
form. These circular supercoiled genomes are replicated once during cell
division and perpetuated in progeny cells indefinitely. Replication is
mediated by host DNA polymerase (and other enzymes of the cellular replicative
machinery). Episomal copy number is tightly regulated and remains constant
while expression of viral genes is greatly limited to several latency genes.
None of these processes is affected by, or sensitive to, antiviral drugs
to any degree, as shown by their lack of effect on latently EBV-infected
cell lines or tumours (Pagano JS. Epstein-Barr virus: Therapy of active
and latent infection. In: Jeffries DJ, De Clercq E, eds. Antiviral Chemotherapy.
John Wiley & Sons Ltd. 1995; pp. 155–95).
A possible exception might
be the initially polyclonal EBV lymphoproliferative disorders, in which
occasional cells do exhibit lytic rather than latent infection; in such
cases, theoretically antiviral therapy might prevent secondary infection
of a new population of B cells. There is only anecdotal evidence for
such an effect clinically, and it is scant. However, Chodosh et al. have
reported experiments in which treatment of latently EBV-infected cells
with
hydroxyurea
led to loss of episomes
ref,
and in a subsequent case report detailed apparently successful use of hydroxyurea
in a patient with an EBV-related CNS lymphoma
ref.
It should be noted though, that a number of groups are intensively studying
means to induce a switch from latent to cytolytic infection as a therapeutic
approach for the treatment of EBV
+ neoplasms, the rationale
being that viral replication causes cytolysis of the infected cells and
could do so in tumours
ref
:
... all of which alone or in combination induce viral reactivation. Combinations
of such inducers with ganciclovir, which is activated by EBV PK (or TK)
and thus toxic for infected and some neighbouring cells, has led to some
progress in treatment of EBV
+ malignancies in animal models
ref1,
ref2,
ref3
and in Phase I/II clinical trials
ref.
In quite another experimental approach to treatment of EBV-associated malignancies,
phosphonated nucleoside analogues have been used to cause apoptosis of
human EBV
+ anaplastic
or undifferentiated nasopharyngeal carcinoma (NPC) / lymphoepithelioma
grown in athymic mice. Since NPCs are latently infected, again the antitumour
effect must be independent of the antiviral effects of these drugs
ref1,
ref2
(Wakisaka N, Yoshizaki T, Murono S et al. Ribonucleotide reductase inhibitors
enhance cidofovir-induced apoptosis in EBV
+ nasopharyngeal carcinoma
xenografts. Int J Cancer 2005; 116: in press). Interestingly, cidofovir
has been reported to produce complete responses
in vivo in another
virus-associated neoplasm, laryngeal papillomatosis, which is caused by
human papillomavirus
ref
(Snoeck R, Andrei G & De Clercq E. Cidofovir in the treatment of HPV-associated
lesions. Verh K Acad Geneeskd Belg 2001; 63: 93–120, discussion 120–2).
Such effects of cidofovir might be linked to decreased expression of EBV
LMP1 and consequent aberrations in apoptotic mechanisms
ref,
but there is evidence that weighs against these conclusions
ref,
and so how these nucleotide analogues affect neoplastic growths is far
from clear and remains an important topic for continued study
ref
Experimental
animal models : at present it is not known to what extent the mechanisms
described
in vitro also play a role
in vivo. To gain more
insight into the
in vivo situation, several animal models have been
developed. The pathogenic aspects of separate EBV gene products have been
studied by means of transgenic mice. These studies clearly indicate putative
pathogenic roles for EBNA1
ref [87], LMP1 (both in lymphoid
ref
[47] and epithelial
ref [48] tissue), and LMP2A
ref
[168] in driving oncogenic cell transformation and inhibition of cellular
differentiation. However, the implications of these findings for the pathogenesis
of EBV-infection and malignant transformation in humans, where EBV persists
in the face of a highly responsive immune system, remain to be clarified
by further studies. In addition, animal models have been developed to study
the biology of EBV-infection. Especially cottontop tamarins are susceptible
for EBV-infection: administration of a high intravenous dose of the EBV
B85-8 strain rapidly produces gross, multiple, malignant lymphomas in 100%
of these animals
ref [418]. However, this pathogenesis is clearly
not representative for the situation in humans, where several types of
lymphomas develop only after a long period of latent EBV-infection. A more
closely related animal model was developed by Wedderburn et al. using the
common marmoset which developes a persistent infection more closely resembling
EBV-infection of humans. However, only limited experience exists in this
model
ref [419]. From lymphoma tissue arising in various monkey
species EBV-like viruses have been recovered that have subsequently been
used to develop an animal model for lymphomagenesis
ref1, ref2, ref3,
ref4 [420, 421, 422 and 423]. An excellent alternative model was
developed by Moghaddam et al.
ref [424], who did not use EBV
but a naturally endemic rhesus lymphocryptovirus. This animal model represented
several aspects of human EBV-infection, including oral transmission, atypical
lymphocytes, lymphadenopathy, latent infection in peripheral blood and
serologic responses to latent and lytic viral antigens. Interestingly,
tamarins were shown to be effectively protected from lymphomas after vaccination
with a gp340 vaccine containing the native fully glycosylated Gp
ref
[425], although in protected animals virus-carrying cells could still be
detected which showed a restricted gene expression pattern
ref
[426]. Similar studies have recently been undertaken in the common marmoset,
showing reduced shedding by EBV challenge in gp340 immunized animals
ref
[427]. In humans, during natural infection gp340 also elicits virus-neutralizing
antibodies which remain present in healthy virus carriers throughout life
ref
[428]. Indeed a small human trial with recombinant vaccinia virus expressing
gp340 showed promising results in delaying primary infection in virus-
naïve
infants
ref [429]. In addition to studies in monkeys, EBV-mediated
lymphomagenesis has also been studied in SCID mice and in nude mice
ref1,
ref2, ref3 [430, 431 and 432]. This model also proved a valuable
tool for anti-tumor therapeutic studies of EBV lymphomas
ref1, ref2
[433 and 434] and carcinomas
ref1, ref2 [435 and 436]. It is,
however, difficult to extrapolate these data to EBV-mediated lymphomagenesis
or carcinogenesis in humans, because these nude mice and SCID mice lack
(part of) the immune responses which play a role in the establishment and
control of EBV-infection in humans.
Prognosis : establishes
a life-long persistent infection in > 90% of the world's population
Web resources
:
International Association
for Research on EBV and Associated Diseases