You are on page 1of 13

Seminar

www.thelancet.com Vol 378 July 2, 2011 73


Hepatitis delta virus
Sarah A Hughes, Heiner Wedemeyer, Phillip M Harrison
Hepatitis delta virus (HDV) is a small, defective RNA virus that can infect only individuals who have hepatitis B virus
(HBV); worldwide more than 15 million people are co-infected. There are eight reported genotypes of HDV with
unexplained variations in their geographical distribution and pathogenicity. The hepatitis D virion is composed of a
coat of HBV envelope proteins surrounding the nucleocapsid, which consists of a single-stranded, circular RNA
genome complexed with delta antigen, the viral protein. HDV is clinically important because although it suppresses
HBV replication, it causes severe liver disease with rapid progression to cirrhosis and hepatic decompensation. The
range of clinical presentation is wide, varying from mild disease to fulminant liver failure. The prevalence of HDV is
declining in some endemic areas but increasing in northern and central Europe because of immigration. Treatment
of HDV is with pegylated interferon alfa; however, response rates are poor. Increased understanding of the molecular
virology of HDV will identify novel therapeutic targets for this most severe form of chronic viral hepatitis.
Introduction
Hepatitis delta virus (HDV) is a small, defective RNA
virus that is related more to plant viroids than to other
human pathogens. It can propagate only in an individual
who has coexistent hepatitis B virus (HBV), either after
simultaneous transmission of the two viruses, or via
superinfection of an established HBV carrier.
1
Clinical
expression of HDV is wide, and although it sometimes
follows a benign course, the disease is clinically
important. Studies have consistently shown that most
patients with HBV and HDV co-infection have more
severe liver disease,
2,3
more rapid progression to
cirrhosis,
4,5
and increased hepatic decompensation and
death
6,7
than do those with HBV infection alone. Advances
have improved our understanding of the transmission,
replication, and pathogenesis of the virus; however, we
do not yet fully understand the mechanisms by which it
causes such severe liver disease. In this Seminar, we will
review the biology, pathogenesis, epidemiology, natural
history, clinical presentation, and management of this
disease. We discuss how the most recent evidence might
help in the design of novel therapeutic agents for HDV,
the most severe of all the chronic viral hepatitides.
Historical perspective
Rizzetto and colleagues
8
discovered HDV in the mid-
1970s while investigating a group of patients with HBV
who had severe hepatitis. They showed a novel antigen-
antibody system, which they called delta-antigen and
delta-antibody, and noted that this system occurred only
in patients with HBV and was associated with severe
liver disease. The disease was later associated with a
particle consisting of an RNA genome of low molecular
weight that was encapsidated by HBV envelope proteins.
9

This particle was termed the delta agent, or HDV, and
was classied under the Deltavirus genus.
Viral structure
The HDV virion is a small, spherical particle of about
36 nm in diameter. It is composed of an outer coat
containing the three HBV envelope proteins termed
large, medium, and small hepatitis B surface antigen
(HBsAg),
10
and host lipids surrounding an inner
nucleocapsid, consisting of a single-stranded, circular
RNA of 1679 nucleotides and about 200 molecules of
hepatitis D antigen (HDAg) per genome.
11,12
Because of
the high GC content of the nucleotide sequence,
12
the
circular genome, which is unique to animal viruses and
more closely related to plant viroids than to human
pathogens,
13
can fold into an unbranched, rod-like
structure with 74% intra molecular base-pairing.
Virus life cycle
The HDV receptor on the human hepatocyte remains
unidentied, but is thought to be the same as that of
HBV because of the shared identity of their outer coat.
HDV infectivity is dependent upon a receptor-binding
domain in the N-terminal region of the pre-S1 moiety of
large HBsAg.
14,15
For HDV infectivity, this domain
requires modication by myristoylation.
16
Fine mapping
has identied aminoacid residues 915 as the receptor
binding site.
17
A second infectivity region in the antigenic
loop of all three envelope proteins is also required for
infectivity,
18,19
but whether the antigenic loop and the
pre-S1 determinants act synergistically or independently
in viral entry is unclear.
The virus is uncoated after entering the hepatocyte,
and a signal in HDAg translocates the nucleocapsid to
the nucleus.
20
The antigen has no RNA polymerase
Lancet 2011; 378: 7385
Published Online
April 20, 2011
DOI:10.1016/S0140-
6736(10)61931-9
Institute of Liver Studies, Kings
College Hospital, London, UK
(S A Hughes MBBCh);
Transplantation Immunology
and Mucosal Biology, Kings
College London, London,
UK (P M Harrison MD);
and Department of
Gastroenterology, Hepatology
and Endocrinology, Hannover
Medical School, Hannover,
Germany (H Wedemeyer MD)
Correspondence to:
Dr Phillip M Harrison,
Transplantation Immunology
and Mucosal Biology, Kings
College London, Denmark Hill
Campus, London SE5 9PJ, UK
phillip.harrison@kcl.ac.uk
Search strategy and selection criteria
We searched PubMed, Embase, and the Cochrane Library, with
no date or language restrictions. We used the search terms
hepatitis delta virus, hepatitis D virus, and Delta hepatitis,
in combination with the following terms: replication,
epidemiology, genotype, transmission, clinical
presentation, pathogenesis, diagnosis, and treatment. We
largely selected publications in the past 5 years. We searched the
reference lists of articles identied by this strategy, and selected
references that were most relevant. Abstracts from international
congressional proceedings in the past 3 years that were thought
to add key information were also included.
Seminar
74 www.thelancet.com Vol 378 July 2, 2011
activity; to replicate its genome, the virus hijacks the
cellular RNA polymerases of the host, which might then
treat the genome as double-stranded DNA because of its
folded, rod-like structure.
21
Three forms of RNA are made
in the host during replication: circular genomic RNA,
circular complementary antigenomic RNA, and a linear
polyadenylated antigenomic RNA of 08 kb, which is the
messenger RNA (mRNA) containing the open reading
frame of the HDAg.
Initial studies implicated RNA polymerase II in HDV
replication; however, one study has shown that RNA
polymerases I and III also interact with HDV RNA,
suggesting a more complex reliance on several host
polymerases.
22
Evidence suggests that synthesis of the
dierent RNA species occurs in dierent subcellular
locations, mediated by distinct cellular polymerases:
synthesis of antigenomic RNA occurs in the nucleolus
mediated by RNA polymerase I, whereas synthesis of
genomic RNA takes place more diusely in the
nucleoplasm by RNA polymerase II.
23
Replication of the
circular HDV RNA template occurs via a rolling
mechanism that is unique to animal viruses but analogous
to that of plant viroids. HDV RNA is rst synthesised as a
linear molecule, potentially containing many copies of
the genome, but in the genomic and antigenomic RNA, a
sequence of 85 nucleotides acts as a ribozyme, which self-
cleaves the linear HDV RNA into monomers.
24,25
These
monomers are then ligated to form circular RNA (the
involvement of a host RNA ligase remains controversial).
Ribozymes are regarded as a feature of plant viroids,
26
but
a self-cleaving RNA sequence in an intron of the CPEB3
gene is structurally and biochemically related to the HDV
ribozymes; therefore, HDV might have arisen from the
human transcriptome.
27
HDAg is the only protein that is known to be encoded
by the HDV genome. It consists of two isoforms: the
27 kDa large-HDAg with 214 aminoacids; and a 24 kDa
small-HDAg with 195 aminoacids.
28
The N-terminal
sequence of the two isoforms is the same; they dier by
only 19 aminoacids at the C-terminus of the large-HDAg.
The open reading frame of the antigenomic RNA
generates both isoforms because of heterogeneity in the
RNA at codon 196.
28
A UAG stop codon at this position
leads to translation of small-HDAg; however, RNA
editing by the cellular enzyme adenosine deaminase-1
changes the sequence to UGG and, consequently, the
longer large-HDAg is produced.
12,28,29
Small-HDAg
returns to the nucleus and supports viral replication,
30,31

whereas large-HDAg is a negative regulator of HDV
replication and is essential for virion assembly.
32

Therefore, RNA editing is central to the replication cycle
of HDV because it controls the levels of each isoform,
and consequently, the balance between viral synthesis
and particle assembly.
Post-translational modication of large-HDAg, espe-
cially prenylation of the cysteine residue at the C-terminus,
is integral to its ability to bind HBsAg and assemble the
viral particle.
33
Phosphorylation of a serine residue at
position 177 of small-HDAg increases replication of
antigenomic RNA by interacting with RNA poly-
merase II,
34,35
whereas the sumoylation of small-HDAg
enhances the synthesis of genomic RNA and mRNA, but
not of antigenomic RNAproperties that are also
ascribed to acetylation.
36
Methylation of small-HDAg by
arginine methyltransferase at arginine-13 (an RNA-
binding domain), is essential for translocation of small-
HDAg to the nucleus, and for replication of the
antigenomic RNA strand to form the genomic RNA
strand.
37
Hence, post-translational modications deter-
mine the balance of the viral life cycle, and present
attractive novel therapeutic targets for drug development.
In the nucleus, molecules of large-HDAg form complexes
with small-HDAg and new constructs of genomic RNA,
and these complexes are exported to the Golgi membranes
by a signal in the C-terminus of large-HDAg. Once there,
these complexes associate with HBV envelope proteins to
create an infectious virion.
14,38
Interaction of the C-terminus
of large-HDAg with the clathrin heavy-chain in the trans-
Golgi network is essential for viral assembly.
39
Figure 1 is
a schematic representation of the delta particle and its
life cycle.
Viral heterogeneity
The sequence of the HDV RNA genome is highly
variable, and there is divergence of up to 16% within the
same genotype, compared with 2040% between dierent
genotypes. Even in one individual the virus is a pool of
closely related quasispecies.
40
This divergence is partly
due to the scarcity of proofreading ability of RNA
polymerases. The average mutation rate for the non-
coding region of HDV is about 35210

base sub-
stitutions per genome site per year, whereas for the HDV
coding region, it is about 14910

for non-synonymous
substitutions, and 06710

for synonymous sub-


stitutions.
41
However, heterogeneity is not uniform for
the entire coding region of the genome; the self-cleaving
ribozyme sequence and the RNA-binding domain of
HDAg are highly conserved, whereas the C-terminal
region of large-HDAg is very divergent. Historically,
HDV genotyping was achieved by either immuno-
histochemical analysis of liver tissue
42
or by restriction
fragment length polymorphism of PCR products,
43
and
the virus was thought to have evolved into three major
genotypes that diered in their global distribution.
44

Genotyping by direct sequencing and analysis of
molecular phylogenetic trees has shown that HDV exists
as at least eight dierent clades, four of which seem to be
of exclusively African origin.
45,46
Epidemiology
Of the 350 million chronic carriers of HBV worldwide,
more than 15 million have serological evidence of
exposure to HDV.
47
Traditionally, the regions with high
rates of HDV carriage where the virus is endemic are
Seminar
www.thelancet.com Vol 378 July 2, 2011 75
central Africa, the Horn of Africa, the Amazon Basin,
eastern and Mediterranean Europe, the Middle East, and
parts of Asia.
48
Rates of HDV infection are generally
highest in regions where HBV is endemic, but there are
exceptionseg, HDV co-infection is uncommon in
Vietnam and Indonesia.
49,50
In Chinaa large reservoir of
HBV infectionHDV prevalence varies widely between
provinces despite a high prevalence of HBV. In one study
from Hong Kong, carriage of HDV was almost universal
in intravenous drug users who were HBsAg positive,
contrasting with low rates in non-drug users.
51
HDV genotype 1 is prevalent worldwide,
52
whereas
genotype 2 (previously termed genotype-2a) is found in
Japan, Taiwan, and the Yakutia region of Russia.
5355

Genotype 3the most divergent genotypeis common
in the Amazon Basin,
56
whereas genotype 4 (previously
2b) is found in Taiwan and Japan.
54,57
HDV genotypes 58
are found in individuals of African origin, including in
those who have migrated to northern Europe.
45,46
Figure 2
shows the worldwide prevalence of HDV and distribution
of its genotypes.
Longitudinal studies have shown a decrease in HDV
prevalence in some endemic areas, such as in Italy, where
infection in HBsAg carriers has fallen from 246% in
1983, to 14% in 1992, and 83% in 1997.
5860
Infection rates
are especially reduced in younger patients, and evidence
suggests that in Italy HDV infection is restricted to
ageing cohorts who were infected in the 1980s. In the
past three decades, reductions in HDV prevalence have
also been reported in Spain, Taiwan, and Turkey.
6163

Vaccination programmes for HBV have probably
contributed substantially to the decline in HDV in these
regions, but additional factors, including increased
awareness of the virus and its mode of transmission,
have led to better implementation of preventive measures,
such as the change to disposable needles, syringes, and
other medical equipment, and a general improvement in
socioeconomic conditions.
There are caveats to the assertion that HDV prevalence
is declining. First, developing countries have not always
had the same measures of HBV control, and have not
shown the same improvements in socioeconomic
conditions as have developed countries. Data for
prevalence in these regions are sparse, but cross-sectional
studies have shown that rates of HDV co-infection in
carriers of HBsAg remain greater than 10%, and
sometimes as high as 70% in Nigeria, Gabon, India,
Pakistan, Iran, the western Brazilian Amazon, Tajikistan,
and Mongolia.
6471
Second, HDV prevalence has not
decreased in northern Europe. Prevalence in northern
Europe and the USA was thought to be low and conned
to high-risk groups, such as intravenous drug-users;
however, in London, UK, HDV prevalence in people with
HBV increased from 26% in the 1980s to 85% in
2005.
72,73
In Germany, although the prevalence of anti-
HDV antibody in those with HBV reduced from 186% in
1992 to 68% in 1997, from 1999 onwards the rates have
increased again to between 8% and 14%.
74
HDV remains
prevalent in France.
75
The common factor in these three
countries is HDV infection in young individuals who
L-HDAg
Delta virion
Ribonucleoprotein
Hepatocyte
membrane
receptor
L-HBsAg
S-HBsAg
M-HBsAg
S-HDAg
1
2 3
9
Nucleus
Genomic RNA
Endoplasmic
reticulum
Golgi complex
Antigenomic RNA
mRNA 6
7
6
8
4 4
5
Figure 1: Schematic representation of the delta virion and its replication cycle
(1) The virion attaches to the hepatocyte via an interaction between large-HBsAg and an uncharacterised membrane receptor in the host cell; (2) the virion enters the
cell and is uncoated; (3) the RNP is targeted to the nucleus; (4) genomic RNA is transcribed in the nucleus to form antigenomic RNA, which forms the template for
replication of new transcripts of the circular genome, and mRNA, which contains the open reading frame; (5) the mRNA is exported to the cytoplasm where it is
translated at the endoplasmic reticulum to form new molecules of hepatitis D antigen; (6) the new antigen molecules return to the nucleus where the small-HDAg
isoform supports further genome replication, and where both forms of hepatitis D antigen associate with new transcripts of genomic RNA to form new RNPs;
(7) RNPs are exported to the cytoplasm where large-HDAg facilitates association with HBV envelope proteins in the ER to form new virus particles; (8) these particles
bud through an intermediate compartment; (9) they are then exported from the hepatocyte via the trans-Golgi network to re-infect further cells.
RNP=ribonucleoprotein. mRNA=messenger RNA. HBV=hepatitis B virus. HBsAg=hepatitis B surface antigen. HDAg=hepatitis D antigen. ER=endoplasmic reticulum.
Seminar
76 www.thelancet.com Vol 378 July 2, 2011
have migrated from regions of high prevalence. Even in
Italy the decline in HDV has now reached a plateau,
76
and
a report has shown a high prevalence of HDV of 17% in
non-EU citizens (mainly those from eastern Europe) who
are infected with HBV.
77
Third, clustered outbreaks of
HDV superinfection continue to be reported, notably in
Venezuela, Ecuador, Mongolia, and Greenland,
7881
which
are similar to those recorded in Samara (Russia), Okinawa
(Japan), Central Africa, and the Amazon Basin in the
1980s and 1990s.
8285
Thus, while outbreaks still occur and
population migration from endemic countries increases,
the threat of HDV infection remains.
Modes of transmission
Like HBV, HDV is transmitted via the parenteral route
through exposure to infected blood or body uids, and
tests in chimpanzees have shown that only a very small
inoculum is su cient to transmit infection.
86
Thus,
transmission rates remain high in intravenous drug
users. There is evidence for sexual transmission,
87
and
people with high-risk sexual activity are at increased risk
for infection.
88
Intrafamilial spread occurs and seems to
be common in regions of high prevalence, which is
known as inapparent parenteral transmission. Perinatal
transmission of HDV is uncommon. Because of screening
of blood products, new infections in haemophiliacs, blood
transfusion recipients, and patients receiving haemo-
dialysis are no longer seen in developed countries.
Clinical expression and natural history
With HBV and HDV co-infection, the fate of HDV is
determined by the host response to HBV, which in more
than 95% of adults results in viral clearance. Acute
co-infection can be more severe than acute mono-
infection with HBV, thereby resulting in acute liver
failure; however, disease expression is wide-ranging. By
contrast, HDV superinfection of an individual with
chronic HBV results in chronic HDV infection in most
people. In the remainder, replication of HDV stops, and
the natural history of the disease is that of the underlying
HBV; however, the residual liver disease might be
advanced. Important evidence from an Italian cohort
showed that 10% of patients with anti-HDV antibodies
cleared HBsAg after a mean follow-up of 4 years,
compared with 28% of those with HBV mono-infection.
89

The mechanism for increased rate of HBsAg loss after
clearance of HDV RNA is unknown, but an enhanced
immune response against HBV and HDV seems
plausible. Figure 3 shows the typical evolution of
serological and virological markers in co-infected patients
versus super infected patients.
Superinfection can present as acute hepatitis in a
previously undiagnosed carrier of HBsAg, and is often
misdiagnosed as acute HBV or as worsening liver
disease due to chronic HBV.
90
At initial histological
assessment, patients with HDV superinfection are
often shown to have severe hepatitis with advanced
brosis.
9193
These patients undergo more rapid
progression to cirrhosis
4,5
and increased hepatic
decompensation leading to death
6,7
than do those with
HBV infection alone. Despite the high rate of
progression to cirrhosis, not all studies show an
increased rate of hepatocellular carcinoma, perhaps
because of suppression of HBV replication by HDV.
73

High
Intermediate
Low
Very low
Insucient data
Genotype 58
Genotype 1
Genotype 1
Genotype 1/3
Genotype 1/2/4
Genotype 1
Figure 2: Worldwide prevalence of HDV and the geographic distribution of its genotypes
Two subtypes1A and 1Bhave been identied in HDV genotype 1. 1A is predominant in Asia and 1B in the USA. Both are common in the Mediterranean. HDV genotype 2 occurs in the Far East.
HDV genotype 3 occurs exclusively in the northern part of South America and is linked to HBV genotype F. Genotypes 58 have been identied primarily in patients from Africa. HDV=hepatitis D virus.
Seminar
www.thelancet.com Vol 378 July 2, 2011 77
In liver transplantation for HDV, hepatitis B
hyperimmune globulin (HBIg) leads to rapid reduction
in HBsAg concentrations, and serum HDV RNA
declines in parallel.
94
Graft re-infection is prevented by
long-term administration of HBIg, which prevents
HBV re-infection; hence, propagation of HDV cannot
be supported.
95,96
Originally, HDV was thought to persist
as an isolated or latent infection after transplantation;
however, more advanced and sensitive serological
assays have shown that this latency does not occur.
97

Hence, the outcome of liver transplantation for HDV is
very good, with 5-year survival of more than 80%, and
better than the outcome noted with transplantation for
other causes of liver disease.
95,98
Although early studies identied that HDV genotype
aects the natural history of HDV, the more recently
identied genotypes 58, from Africa, are less well
characterised. A study from Taiwan
99
showed a lower rate
of remission and more adverse outcomes in patients with
genotype 1 than in those with genotype 2. Patients with
genotype 4 often have mild liver disease,
100
but a variant
of genotype 4 on the Miyako Island in Okinawa, Japan, is
associated with greater progression to cirrhosis than
genotype 4 is in Taiwan.
101
Genotype 3 has been linked to
outbreaks of severe, orid hepatitis (Labrea fever),
culminating in acute liver failure and death in the
Amazon Basin of South America.
44
Outbreaks of severe
hepatitis due to genotype 1 have also taken place, but
genotype 1 is associated with a wide range of disease,
102

thereby making the relation between genotype and
natural history di cult to interpret.
Although HBV genotype might also determine the
natural history of HDV, any eect is di cult to study
because in many co-infected individuals the serum level
of HBV DNA is too low for genotype analysis. Nonetheless,
Su and colleagues
99
showed a signicantly lower remission
rate and more adverse outcomes in patients with HDV
superinfection with HBV genotype C than with HBV
genotype B, in a population from Taiwan who were mostly
infected with HDV genotypes 1 and 2. A study from Brazil
A
B
Time after exposure (weeks)
C
Anti-HBc IgG
Anti-HBc IgM
Anti-HD IgG
Anti-HD IgM
HBsAg
HDV RNA
ALT
Figure 3: Typical evolution of serological and virological markers in HDV infection
(A) Simultaneous co-infection with HBV and HDV, resulting in clearance of both viruses in almost all patients. (B) HDV superinfection of an HBV carrier with
self-limited outcome. The spontaneous clearance of HDV RNA might take years to occur (indicated by break on x axis) and, in few cases, can herald the loss of HBsAg
(not depicted). (C) HDV superinfection of a HBV carrier with chronic persistent viral replication; the more common outcome after superinfection. HDV=hepatitis
delta virus. ALT=alanine aminotransferase. HBV=hepatitis B virus. HBsAg=hepatitis B surface antigen.
Seminar
78 www.thelancet.com Vol 378 July 2, 2011
has suggested that HBV genotype determines the severity
of liver inammation and the HDV viral load in
individuals with co-infection; however, the consequence
of this in relation to outcome is not clear.
103
Patterns of viral dominance
Co-infection and superinfection with HDV suppresses
HBV replication in patients and in model systems.
About 7090% of patients with HDV co-infection are
HBeAg negative, and most have low serum HBV
DNA.
73,104106
Evidence has indicated that the small (p24)
and large (p27) HDV proteins downregulate HBV repli-
cation by repressing activity of the two HBV enhancer
regions, and by transactivating the interferon-inducible
MxA gene, which inhibits HBV replication by reducing
the export of viral mRNA from the nucleus.
107,108
If HDV
infection is cleared either spontaneously or after
treatment with interferon alfa, then HBV replication
can reactivate.
109
Many patients with HBV and HDV have serological
evidence of exposure to hepatitis C virus (HCV)about
30% in European cohorts.
73,105
In those with triple
infection, HDV is the dominant virus because it not only
suppresses HBV replication, but also inhibits HCV
replication.
110
Indeed, HCV RNA was detected in less
than 19% of patients who were anti-HCV antibody
positive, HBsAg positive, and anti-HDV antibody posi-
tive.
73,105,110
Most patients who are HCV RNA negative have
probably cleared HCV infection, but further long-term
follow-up studies are needed, especially because patterns
of viral dominance evolve over time in HBV and HCV
co-infection.
111
A dynamic pattern of viral dominance also
exists in HBV and HDV co-infection.
112
Pathogenesis
The mechanisms that determine whether an individual
clears HDV spontaneously or becomes chronically
infected, and the processes that cause severe hepatitis
and rapid progression of brosis, remain unclear.
HDAg is not directly cytotoxic in human hepatocytes or
in transgenic mice.
113,114
Viral load of HDV was not
associated with severity of liver injury in a cohort of
patients in a clinical trial.
106
However, evidence from
observational cohort studies suggests that in the acute
phase of HDV infection, HDV viraemia is associated
with an increased level of alanine aminotransferase and
suppressed HBV. In the chronic phase, falling HDV
RNA, reactivation of HBV, and moderate trans aminitis
culminate in a late phase that is characterised by either
the development of cirrhosis and hepatocellular
carcinoma due to replication of HDV or HBV, or
remission with clearance of both viruses.
115
Hence, viral
load of HDV and HBV uctuates according to the stage
of viral infection. Whether this uctuation represents a
direct relation with the pathogenesis of disease
progression remains unclear, and other factors should
be considered.
Host immune response
The host immune response is thought to have an
important role in viral clearance and liver injury. An early
study
116
showed that the function of natural killer cells
was active in individuals who suppressed HDV RNA
during treatment with interferon, but further studies
investigating the innate immune response in the
pathogenesis of HDV are needed. The adaptive immune
response in HDV infection is also poorly dened. In one
study,
117
weak HDAg-specic responses of CD4 T cells
were elicited in patients with inactive HDV infection
(anti-HDV antibody positive but RNA negative), but
absent in those with chronic HDV infection. Further
studies
118
have shown that chronic HDV infection is
associated with a response that is dominated by T-helper-2
(Th-2) cells, with a high frequency of T cells secreting
interleukin-10. Furthermore, after antiviral therapy,
production of interleukin-10 that was HDV-peptide
specic remained higher in patients who did not respond
to treatment than in those who did. Secretion of
interferon-inducible protein-10 was, however, more
frequent in responders.
119
These ndings suggest that
HDV subverts the adaptive immune response away from
the Th-1-biased CD4 and CD8 T-cell response that is
needed for viral clearance. Indeed, Huang and
colleagues
120
showed that the responses of HDV-specic
CD8 cytotoxic T lymphocytes were detected in those with
past, but not active, infection.
CD4 T cells are essential to the antiviral immune
response because they help CD8 T cells and B cells by
stimulating antigen presenting cells and cytokine
secretion. However, cytotoxic CD4 T cells that are
perforin-positive might also have a role in directly
killing virus-infected cells. A higher frequency of these
cells were recorded in patients who were co-infected
with HBV and HDV than with HBV or HCV
mono-infection, and the frequency of perforin-positive
cells was positively correlated with disease activity.
These ndings suggest that such perforin-positive cells
are implicated in the pathogenesis of HDV liver
disease.
121
These studies suggest an important role for
the adaptive immune response, but the precise
mechanisms are unknown.
Role of HDV genotype in pathogenesis
E ciency of RNA editing is lower with genotype 2 than
with genotype 1, but this dierence is unlikely to account
for the reduced disease activity.
122
The sequence of the
C-terminal moiety in large-HDAg varies widely between
genotypes, with up to 74% divergence between the HDV
packaging signal domains of genotype 1 and other HDV
genotypes. The C-terminal sequence of the large antigen
in genotype 1 results in better packaging ability than in
genotype 2, which interacts weakly with clathrin, leading
to less e cient assembly of particles.
123125
However, all
HDV genotypes are able to bind clathrin to some degree,
which lends support to the role of clathrin binding in the
Seminar
www.thelancet.com Vol 378 July 2, 2011 79
assembly of HDV particles.
126
Which, if any, of these
mechanisms underpin the eect of genotype on the
natural history of HDV is unclear.
Diagnosis
The development of anti-HDV antibodies is universal in
individuals with HDV; therefore, every patient who is
HBsAg positive should be tested for anti-HDV IgG
antibodies, which persist even after the patient has
cleared HDV infection. Although active HDV infection
was diagnosed historically by the presence of anti-
HDV IgM antibodies, it is now conrmed by the detection
of serum HDV RNA with sensitive real-time PCR
assay.
127
Covert HDV infection has not been reported;
therefore, testing of HDV RNA in the absence of
anti-HDV antibodies is not indicated. Because of the
variability of the genome sequence, assays of HDV RNA
might produce false-negative results, and testing of anti-
HDV IgM antibodies still has a role in patients who
test negative for HDV RNA, but have clinical features
of HDV-related liver disease. International standard-
isation of the HDV RNA assay is needed. Some
laboratories undertake quantitative assays, but serum
concentrations of HDV RNA do not correlate with
disease activity or stage of liver brosis.
106
Serial
quantication of HDV RNA is used to determine the
response to antiviral treatment.
128,129
HDV genotyping is usually only available in specialist
centres, but is gaining acceptance as a useful diagnostic
test because patients with HDV genotype 1 are at a
higher risk of developing end-stage liver disease
99
and
have a lower response to treatment with pegylated
interferon than do patients with African genotypes
(unpublished data). All patients with HDV also need
investigation for HCV and HIV because co-infection
with these viruses is common.
73,105,130
The panel shows the
relevance of serological and virological markers in HDV.
Patients who test positive for serum HDV RNA should
be further investigated with liver biopsy to assess the
severity of liver disease. Studies have shown no
association between levels of HDV RNA, HBsAg titre,
values of liver tests, and the histological staging of liver
disease, thus reinforcing the pivotal part that liver biopsy
plays in the assessment of HDV-related liver disease.
Tests for non-invasively assessing the progression of
liver brosis, such as transient elastography, can be
useful for detecting advanced hepatic brosis, but have
not been validated in chronic HDV and therefore cannot
yet be recommended. The aspartate aminotransferase to
HBsAg
Anti-HDV IgG Ab
Treat as per local HBV
guidelines
HCV/HIV Ab
Add ribavirin
+ve
-ve
>1 -ve
+ve
+ve
HCV RNA+ve HBV DNA>2000 IU/mL
Reactivation of HBV after
successful control of HDV
HDV RNA
Liver biopsy
Consider PEG- IFN- for at
least 48 weeks
Consider addition of potent
nucleos(t)ide analogue
Add potent nucleos(t)ide
analogue
Figure 4: Suggested algorithm for the investigation and management of a patient with HDV
HBsAg=hepatitis B surface antigen. HBV=hepatitis B virus. HCV=hepatitis C virus. HDV=hepatitis delta virus.
PEG-IFN-=pegylated interferon alfa. Ab=antibody.
Panel: Diagnostic markers in HDV infection and their
importance
Anti-HDV IgG antibody
Positive in all individuals exposed to HDV, and persists
long-term, even after viral clearance.
Anti-HDV IgM antibody
Positive in acute infection, negative in past infection but
persists in a large proportion of patients with chronic
infection. Sometimes used as surrogate marker for HDV
replication but not 100% sensitive or specic.
HDV RNA qualitative
Marker of HDV replication. Positive in all patients with
chronic infection. Negative in spontaneous or
treatment-induced viral clearance.
HDV RNA quantitative
Useful method to predict or monitor treatment response.
HBsAg qualitative
Must be positive for HDV infectivity.
HBsAg quantitative
Positively correlated with HDV RNA. Might be useful to
predict or monitor treatment response, since falling titre
heralds HBsAg loss, and hence HDV clearance.
HBeAg
Negative in about 85% of patients; associated with
detectable anti-HBe.
HBV DNA quantitative
Usually negative or low level because suppressed by HDV.
Might be increased, especially in patients with detectable
HBeAg. Can reactivate after spontaneous or
treatment-induced clearance of HDV.
ALT
Usually increased but does not correlate well with degree of
histological liver damage.
HDV=hepatitis delta virus. HBsAg=hepatitis B surface antigen. HBeAg=hepatitis B early
antigen. HBV=hepatitis B virus. ALT=alanine aminotransferase
Seminar
80 www.thelancet.com Vol 378 July 2, 2011
platelet ratio index was shown to have no use in
predicting the stage of brosis in patients with HDV.
106

The assessment of patients with HDV infection is
summarised in gure 4.
Treatment
The ideal endpoint of any treatment for HDV is not only
clearance of HDV, but also of the helper virus HBV.
Hence, a major challenge of dening the optimum
therapy is the added complexity of targeting two persistent
viral infections. A meta-analysis of ve studies of
treatment with recombinant interferon concluded that
such treatment was benecial for HDV in terms of serum
aminotransferase reduction, but the response was poorly
sustained after discontinuation of treatment, and was not
necessarily associated with clearance of HDV RNA.
There were better results with higher doses of
interferonat least 5 MU daily or 9 MU thrice weekly for
12 months, rather than for 6 months.
131,132
High-dose
interferon induced delayed clearance of RNA, and
sometimes, loss of HBsAg with resultant improvement
in histological inammatory activity and stage of
brosis.
132
In a single-case study,
133
extended treatment for
12 years led to clearance of HDV RNA and loss of HBsAg
associated with complete regression of brosis.
Consequently, some have advocated prolonging treatment
duration,
47
but studies have shown no additional benet
in giving treatment for 24 months rather than for
12 months.
134,135
The potential benets of this approach
should be balanced with the side-eects, cost, and
logistical implications. Interferon-induced clearance of
HDV RNA has been associated with a fall in HBsAg
concentrations, and sometimes a loss of HBsAg, with
absence of HBsAg decline in patients who do not respond
to treatment.
128
However, the decrease in HBsAg often
lags behind that of HDV RNA, and hence the mechanism
behind this nding is unclear. More data will emerge as
quantication of HBsAg becomes more routinely used.
Investigators have examined the use of pegylated
interferon for chronic HDV. Series from Italy,
136
France,
109

Summary of therapy Regimen Number of
patients
Median age
(years)
Cirrhosis
(%)
Median baseline
HDV RNA
HDV
genotype
SVR
(%)
SBR (%)
Castelnau et al (2006)
109
Open label pilot PEG monotherapy PEG-IFN2b; 15g/kg per week;
12 months
14 42 29 5* 1=86%
5=14%
43 57
Niro et al (2006)
136
Multicentre randomised trial PEG vs PEG+rbv PEG-IFN2b; 15 g/kg per week;
72 weeks
+Rbv 800 mg per day for rst
48 weeks
16
22
45
43
75
73
..
..
..
..
25
18
25
27
Erhardt et al (2006)
137
Open label pilot PEG monotherapy PEG-IFN2b; 15 g/kg per week;
48 weeks
12 34 27 7* 1 or 2 17 17
Wedemeyer et al (2011)
138
Multicentre randomised trial PEG+placebo vs
PEG+adv vs adv
PEG-IFN2a; 180 g per week;
48 weeks
+Adv 10 mg; once daily
Adv 10 mg; once daily 48 weeks
29
31
30
38
42
33
20
14
24
6*
6*
6*
1
1
1
31
26
0
45
35
10
Yurdaydin et al (2008)
139
Multicentre randomised trial LAM vs LAM+IFN vs IFN LAM 100 mg; once daily;
12 months
+IFN2a; 9 MU three times per
week for last 10 months
IFN2a; 9 MU three times per
week; 12 months
17
14
8
38
35
46
..
..
..
6*
6*
6*
..
..
..
12
36
50
24
21
50
Gunsar et al (2005)
140
Single centre randomised trial IFN vs IFN+rbv IFN2a; 9 MU three times per
week 96 weeks
+Rbv 10001200 mg/day
10
21
39
38
30
24
..
..
..
..
20
20
235
235
Farci et al (2004)
132
Single centre randomised trial High-dose IFN vs
low-dose IFN vs no Rx
IFN2a; 9 MU three times per
week 48 weeks
IFN2a; 3 MU three times per
week 48 weeks
Untreated controls
14
14
13
35
35
38
71
64
61
6
6
6
1
1
1
0
0
0
50
7
8
SVR=sustained virological response (RNA negative 6 months after end of therapy). SBR=sustained biochemical response (normal aminotransferases 6 months after end of therapy). PEG=pegylated interferon.
IFN=interferon. Rbv=ribavirin. Adv=adefovir. LAM=lamivudine. Rx=prescription drug. *Log
10
copies per mL. Log
10
genome equivalents per mL.
Table: Summary of key trials of alpha-interferon for chronic hepatitis delta virus
Seminar
www.thelancet.com Vol 378 July 2, 2011 81
and Germany,
137
have shown sustained virological
responses in 21% of 38 patients, 43% of 14 patients, and
17% of 12 patients, respectively. In the HIDIT-1 trial,
138

which randomised 90 patients from Germany, Turkey,
and Greece, pegylated interferon led to a 28% sustained
virological response; the addition of adefovir did not
improve virological response but did lead to increased
suppression of HBsAg concentrations, whereas adefovir
monotherapy was ineective.
138
Why response rates vary
between series is not clear, but might relate to baseline
clinical, demographical, and virological characteristics
that are heterogeneous. The table shows data for the
main interferon trials.
The simplicity of the delta virus, particularly the lack of
viral polymerase, limits the targets for therapeutic
compounds. Investigators have studied nucleos(t)ide
analogues, which block the HBV polymerase, but
lamivudine alone was not eective at reducing
concentrations of HDV RNA and did not increase
sustained virological response when combined with
interferon.
139,141144
Famciclovir was also ineective.
145
Riba-
virin alone,
146
or combined with interferon
140,147
or pegylated
interferon
136
did not increase the virological response.
Entecavir did not reduce HDV viraemia, but did reduce
alanine aminotransferase and HBV DNA in a small
subset of patients who had low HDV RNA and high HBV
DNA.
148
One report has shown HDV clearance associated
with seroconversion of HBsAg in a man treated with
pegylated interferon plus tenofovir and emtricitabine,
who had high levels of both HBV DNA and HDV RNA.
149

In a long-term observational study
150
of 16 patients who
were co-infected with HIV, HBV, and HDV and on highly
active antiretroviral therapy with anti-HBV activity,
including tenofovir, a signicant reduction in HDV RNA
was noted for a median of 61 years, and three patients
became RNA negative. The mechanism for this reduction
is unclear, because it was not associated with a
corresponding decrease in concentrations of HBsAg.
Hence, treatment with nucleos(t)ide analogues is not
eective at reducing HDV replication, but might be useful
in patients with high concentrations of HBV replication,
and might be of potential benet when used long term by
gradually reducing HBsAg concentrations. The use of
older analogues of nucleos(t)ides to induce HBV
mutations is controversial. Lamivudine-induced muta-
tions in the polymerase gene at rtM204V or rtM204I are
associated with changes in the overlapping envelope gene
products, such as in sW196L/S, which inhibits secretion
of HDV particles.
151
There is insu cient evidence to show
whether the failure to package and secrete HDV is
benecial, or indeed whether the retention of HDV RNA
within cells is harmful.
Baseline and treatment factors predicting the outcome
of interferon treatment are poorly dened. HDV RNA is
correlated positively with HBsAg titre,
106,128
and baseline
values of both predict response to therapy (unpublished
data). HDV genotype 1 is associated with a reduced
response to pegylated interferon (unpublished data),
but whether this response relates to a property of the
HDV sequence itself, or merely to the nding that
patients with genotype 1 have increased viral loads, is
unknown. By contrast with treatment response in
patients with chronic HCV, in which rates of sustained
virological response to pegylated interferon are
substantially reduced by the presence of underlying
cirrhosis, cirrhosis associated with HDV infection did
not obviously aect the response to pegylated inter-
feron.
136,137,152
Baseline liver biochemical tests are also not
associated with treatment outcome.
109
Although the kinetics of HDV RNA in therapy have
been used to predict long-term virological outcome, data
are scarce.
128,129
Castelnau and colleagues
109
showed that
75% of patients achieving an end-of-treatment response
were HDV RNA negative by month 6 of treatment,
compared with no patients who did not respond to
treatment; however, undetectable mid-treatment RNA
did not protect against relapse. The investigators also
noted that HDV RNA values were lower at the end of
therapy than at baseline in some patients who did not
respond, which could justify extension of therapy in this
group. Yurdaydin and colleagues
139
dened three
virological patterns of response to interferon: complete,
in which RNA negativity at 6 months predicted sustained
virological response; partial, in which incomplete
suppression of RNA at 6 months predicted rebound are
in viral replication after discontinuation of therapy; and
non-response, in which viraemia persisted without
decline throughout treatment and follow-up. Liver
transplantation is the only treatment option for patients
who have end-stage liver disease due to co-infection with
HBV and HDV, and is appropriate for those with acute
liver failure that fulls criteria for poor prognosis.
153
Re-
infection of the graft is reduced by long-term
administration of immunoglobulins against the HBsAg
(HBIg) after transplantation,
95,96
creating an HBsAg-
negative environment in which HDV cannot survive.
97

The use of potent antivirals against HBV has further
reduced the risk of HBV re-infection.
The accepted practice for treatment of chronic HDV is
subcutaneous injections of pegylated interferon every
week for at least 48 weeks. This practice should be
considered in patients with active replication of HDV
RNA, and histological evidence of disease activity, and
with no contraindications to interferon therapy. We would
advocate an individualised approach to therapy in patients
who have not achieved clearance of HDV RNA by sensitive
PCR-based assay at 48 weeks. Those who have persistent,
high-level viraemia, with detectable anti-HDV IgM
antibody, and ongoing transaminitis, are unlikely to
respond to further therapy. By contrast, patients with a
falling viral load, declining IgM antibody titre, and
resolving transaminitis, or with progressive decline in
HBsAg titre, might benet from extending therapy to
72 weeks, and perhaps beyond, if improvement is
Seminar
82 www.thelancet.com Vol 378 July 2, 2011
sustained and if tolerability is favourable. In patients with
a high concentration of HBV DNA, addition of a potent
nucleos(t)ide analogue to inhibit HBV replication is
logical; however, the long-term eectiveness has yet to be
dened. Potent nucleos(t)ide analogues are the treatment
choice for patients in whom HBV replication is reactivated
after successful control of HDV replication. Such
analogues might also be used in patients with detectable
serum HBV DNA who cannot be treated with pegylated
interferon such as those with advanced liver disease.
Novel therapiesthe future
Increased understanding of the molecular virology of
HDV, and the poor overall response to conventional
antiviral therapy, has driven the search for alternative
antiviral targets. Prenylation of large-HDAg is essential
for viral assembly and secretion,
33
and in mouse models
prenylation inhibitors inhibited the assembly and
release of HDV, leading to rapid clearance of HDV
RNA from serum.
154
These compounds are in pre-
clinical development. Other forms of post-translational
modi cation of HDAg, such as acetylation, phos-
phorylation, and methylation, might also prove fruitful
as targets for novel therapeutic compounds, although
none are yet in development. Studies showing that
myristoylated synthetic peptides specic for the
N-terminal region of the pre-S1 domain of HBsAg are
able to inhibit viral attachment and hence HDV
infectivity, draw attention to an alternative therapeutic
target.
14,17,155,156
As we learn more about cell-signalling
pathways associated with the pathogenesis of HDV,
further targets might be exposed.
Therapy for HDV deserves the resurgence of interest
it has received. Treatment eectiveness remains unsatis-
factory, and developments have lagged behind those
achieved for the treatment of hepatitis B. The renement
of conventional antiviral regimens will continue with
larger collaborative trials, and drug development will
hopefully follow the advances in HDV biology. These
developments remain necessary while worldwide
measures to control the helper virus are inconsistent and
the prevalence of HDV does not decline in many areas,
resulting in a continuing and substantial health burden.
Contributors
All authors contributed to the conception and writing of the Seminar.
Sections were divided between SAH and PMH, and each author
undertook the relevant searches and wrote the assigned sections.
SAH assembled the sections and all authors revised the nal version.
Conicts of interest
PMH has received payment for board membership from Roche;
consultancy fees from Gilead, Bristol-Myers Squibb, and Phytopharm;
and payment for participation in a speakers bureau from Gilead and
Bristol-Myers Squibb. HW has received payment for board membership
from Roche, Gilead, Bristol-Myers Squibb, and Schering Plough;
consultancy fees from Roche and Novartis; grant support from Roche
and Gilead; payment for participation in a speakers bureau from Roche,
Gilead, Bristol-Myers Squibb, Schering Plough, and Novartis; and
payment for developing educational presentations from Roche. SAH
declares that she has no conicts of interest
References
1 Rizzetto M. Hepatitis D: thirty years after. J Hepatol 2009;
50: 104350.
2 Hadler SC, De Monzon M, Ponzetto A, et al. Delta virus infection
and severe hepatitis. An epidemic in the Yucpa Indians of
Venezuela. Ann Intern Med 1984; 100: 33944.
3 Govindarajan S, Chin KP, Redeker AG, Peters RL. Fulminant B
viral hepatitis: role of delta agent. Gastroenterology 1984;
86: 141720.
4 Saracco G, Rosina F, Brunetto MR, et al. Rapidly progressive
HBsAg-positive hepatitis in Italy. The role of hepatitis delta virus
infection. J Hepatol 1987; 5: 27481.
5 Fattovich G, Boscaro S, Noventa F, et al. Inuence of hepatitis delta
virus infection on progression to cirrhosis in chronic hepatitis
type B. J Infect Dis 1987; 155: 93135.
6 Fattovich G, Giustina G, Christensen E, et al. Inuence of hepatitis
delta virus infection on morbidity and mortality in compensated
cirrhosis type B. The European Concerted Action on Viral Hepatitis
(Eurohep). Gut 2000; 46: 42026.
7 Romeo R, Del Ninno E, Rumi M, et al. A 28-year study of the course
of hepatitis Delta infection: a risk factor for cirrhosis and
hepatocellular carcinoma. Gastroenterology 2009; 136: 162938.
8 Rizzetto M, Canese MG, Arico S, et al. Immunouorescence
detection of new antigen-antibody system (delta/anti-delta)
associated to hepatitis B virus in liver and in serum of HBsAg
carriers. Gut 1977; 18: 9971003.
9 Rizzetto M, Hoyer B, Canese MG, Shih JW, Purcell RH, Gerin JL.
Delta Agent: association of delta antigen with hepatitis B surface
antigen and RNA in serum of delta-infected chimpanzees.
Proc Natl Acad Sci USA 1980; 77: 612428.
10 Sureau C. The role of the HBV envelope proteins in the HDV
replication cycle. Curr Top Microbiol Immunol 2006; 307: 11331.
11 Gudima S, Chang J, Moraleda G, Azvolinsky A, Taylor J. Parameters
of human hepatitis delta virus genome replication: the quantity,
quality, and intracellular distribution of viral proteins and RNA.
J Virol 2002; 76: 370919.
12 Wang KS, Choo QL, Weiner AJ, et al. Structure, sequence and
expression of the hepatitis delta (delta) viral genome. Nature 1986;
323: 50814.
13 Kos A, Dijkema R, Arnberg AC, van der Meide PH, Schellekens H.
The hepatitis delta (delta) virus possesses a circular RNA. Nature
1986; 323: 55860.
14 Barrera A, Guerra B, Notvall L, Lanford RE. Mapping of the
hepatitis B virus pre-S1 domain involved in receptor recognition.
J Virol 2005; 79: 978698.
15 Engelke M, Mills K, Seitz S, et al. Characterization of a hepatitis B
and hepatitis delta virus receptor binding site. Hepatology 2006;
43: 75060.
16 Abou-Jaoud G, Molina S, Maurel P, Sureau C. Myristoylation
signal transfer from the large to the middle or the small HBV
envelope protein leads to a loss of HDV particles infectivity.
Virology 2007; 365: 20409.
17 Schulze A, Schieck A, Ni Y, Mier W, Urban S. Fine mapping of
pre-S sequence requirements for hepatitis B virus large envelope
protein-mediated receptor interaction. J Virol 2010; 84: 19892000.
18 Abou-Jaoud G, Sureau C. Entry of hepatitis delta virus requires
the conserved cysteine residues of the hepatitis B virus envelope
protein antigenic loop and is blocked by inhibitors of thiol-disulde
exchange. J Virol 2007; 81: 1305766.
19 Salisse J, Sureau C. A function essential to viral entry underlies
the hepatitis B virus a determinant. J Virol 2009; 83: 932128.
20 Xia YP, Yeh CT, Ou JH, Lai MM. Characterization of nuclear
targeting signal of hepatitis delta antigen: nuclear transport as
a protein complex. J Virol 1992; 66: 91421.
21 Lai MM. RNA replication without RNA-dependent RNA
polymerase: surprises from hepatitis delta virus. J Virol 2005;
79: 795158.
22 Greco-Stewart VS, Schissel E, Pelchat M. The hepatitis delta virus
RNA genome interacts with the human RNA polymerases I and III.
Virology 2009; 386: 1215.
23 Li YJ, Macnaughton T, Gao L, Lai MM. RNA-templated replication
of hepatitis delta virus: genomic and antigenomic RNAs associate
with dierent nuclear bodies. J Virol 2006; 80: 647886.
Seminar
www.thelancet.com Vol 378 July 2, 2011 83
24 Branch AD, Benenfeld BJ, Baroudy BM, Wells FV, Gerin JL,
Robertson HD. An ultraviolet-sensitive RNA structural element
in a viroid-like domain of the hepatitis delta virus. Science 1989;
243: 64952.
25 Wu HN, Lin YJ, Lin FP, Makino S, Chang MF, Lai MM. Human
hepatitis delta virus RNA subfragments contain an autocleavage
activity. Proc Natl Acad Sci USA 1989; 86: 183135.
26 Taylor J, Pelchat M. Origin of hepatitis delta virus. Future Microbiol
2010; 5: 393402.
27 Salehi-Ashtiani K, Luptk A, Litovchick A, Szostak JW.
A genomewide search for ribozymes reveals an HDV-like sequence
in the human CPEB3 gene. Science 2006; 313: 178892.
28 Weiner AJ, Choo QL, Wang KS, et al. A single antigenomic open
reading frame of the hepatitis delta virus encodes the epitope(s) of
both hepatitis delta antigen polypeptides p24 delta and p27 delta.
J Virol 1988; 62: 59499.
29 Jayan GC, Casey JL. Inhibition of hepatitis delta virus RNA editing
by short inhibitory RNA-mediated knockdown of ADAR1 but not
ADAR2 expression. J Virol 2002; 76: 12399404.
30 Taylor JM. Structure and replication of hepatitis delta virus RNA.
Curr Top Microbiol Immunol 2006; 307: 123.
31 Yamaguchi Y, Filipovska J, Yano K, et al. Stimulation of RNA
polymerase II elongation by hepatitis delta antigen. Science 2001;
293: 12427.
32 Chang MF, Chen CJ, Chang SC. Mutational analysis of delta
antigen: eect on assembly and replication of hepatitis delta virus.
J Virol 1994; 68: 64653.
33 Glenn JS, Watson JA, Havel CM, White JM. Identication
of a prenylation site in delta virus large antigen. Science 1992;
256: 133133.
34 Chen YS, Huang WH, Hong SY, Tsay YG, Chen PJ.
ERK1/2-mediated phosphorylation of small hepatitis delta antigen
at serine 177 enhances hepatitis delta virus antigenomic RNA
replication. J Virol 2008; 82: 934558.
35 Hong SY, Chen PJ. Phosphorylation of serine 177 of the small
hepatitis delta antigen regulates viral antigenomic RNA replication
by interacting with the processive RNA polymerase II. J Virol 2010;
84: 143038.
36 Tseng CH, Cheng TS, Shu CY, Jeng KS, Lai MM. Modication of
small hepatitis delta virus antigen by SUMO protein. J Virol 2010;
84: 91827.
37 Li YJ, Stallcup MR, Lai MM. Hepatitis delta virus antigen is
methylated at arginine residues, and methylation regulates
subcellular localization and RNA replication. J Virol 2004;
78: 1332534.
38 Wang CJ, Chen PJ, Wu JC, Patel D, Chen DS. Small-form hepatitis
B surface antigen is su cient to help in the assembly of hepatitis
delta virus-like particles. J Virol 1991; 65: 663036.
39 Huang C, Chang SC, Yu IC, Tsay YG, Chang MF. Large hepatitis
delta antigen is a novel clathrin adaptor-like protein. J Virol 2007;
81: 598594.
40 Dny P. Hepatitis delta virus genetic variability: from genotypes I,
II, III to eight major clades? Curr Top Microbiol Immunol 2006;
307: 15171.
41 Krushkal J, Li WH. Substitution rates in hepatitis delta virus.
J Mol Evol 1995; 41: 72126.
42 Hsu SC, Syu WJ, Ting LT, Wu JC. Immunohistochemical
dierentiation of hepatitis D virus genotypes. Hepatology 2000;
32: 111116.
43 Wu JC, Choo KB, Chen CM, Chen TZ, Huo TI, Lee SD. Genotyping
of hepatitis D virus by restriction-fragment length polymorphism
and relation to outcome of hepatitis D. Lancet 1995; 346: 93941.
44 Casey JL, Brown TL, Colan EJ, Wignall FS, Gerin JL. A genotype
of hepatitis D virus that occurs in northern South America.
Proc Natl Acad Sci USA 1993; 90: 901620.
45 Radjef N, Gordien E, Ivaniushina V, et al. Molecular phylogenetic
analyses indicate a wide and ancient radiation of African hepatitis
delta virus, suggesting a deltavirus genus of at least seven major
clades. J Virol 2004; 78: 253744.
46 Le Gal F, Gault E, Ripault MP, et al. Eighth major clade for hepatitis
delta virus. Emerg Infect Dis 2006; 12: 144750.
47 Farci P. Delta hepatitis: an update. J Hepatol 2003;
39 (suppl 1): S21219.
48 Rizzetto M, Ponzetto A, Forzani I. Hepatitis delta virus as a global
health problem. Vaccine 1990; 8 (suppl): S1014.
49 Nguyen VT, McLaws ML, Dore GJ. Highly endemic hepatitis B
infection in rural Vietnam. J Gastroenterol Hepatol 2007;
22: 2093100.
50 Mulyanto, Depamede SN, Surayah K, et al. A nationwide molecular
epidemiological study on hepatitis B virus in Indonesia:
identication of two novel subgenotypes, B8 and C7. Arch Virol
2009; 154: 104759.
51 Abbas Z, Jafri W, Raza S. Hepatitis D: scenario in the Asia-Pacic
region. World J Gastroenterol 2010; 16: 55462.
52 Shakil AO, Hadziyannis S, Hoofnagle JH, Di Bisceglie AM,
Gerin JL, Casey JL. Geographic distribution and genetic variability
of hepatitis delta virus genotype I. Virology 1997; 234: 16067.
53 Imazeki F, Omata M, Ohto M. Heterogeneity and evolution rates
of delta virus RNA sequences. J Virol 1990; 64: 559499.
54 Wu JC, Chiang TY, Sheen IJ. Characterization and phylogenetic
analysis of a novel hepatitis D virus strain discovered by restriction
fragment length polymorphism analysis. J Gen Virol 1998;
79: 110513.
55 Ivaniushina V, Radjef N, Alexeeva M, et al. Hepatitis delta virus
genotypes I and II cocirculate in an endemic area of Yakutia,
Russia. J Gen Virol 2001; 82: 270918.
56 Paran R, Kay A, Molinet F, et al. HDV genotypes in the Western
Brazilian Amazon region: a preliminary report. Am J Trop Med Hyg
2006; 75: 47579.
57 Sakugawa H, Nakasone H, Nakayoshi T, et al. Hepatitis delta virus
genotype IIb predominates in an endemic area, Okinawa, Japan.
J Med Virol 1999; 58: 36672.
58 Smedile A, Lavarini C, Farci P, et al. Epidemiologic patterns of
infection with the hepatitis B virus-associated delta agent in Italy.
Am J Epidemiol 1983; 117: 22329.
59 Sagnelli E, Stroolini T, Ascione A, et al. Decrease in HDV
endemicity in Italy. J Hepatol 1997; 26: 2024.
60 Gaeta GB, Stroolini T, Chiaramonte M, et al. Chronic hepatitis D:
a vanishing Disease? An Italian multicenter study. Hepatology 2000;
32: 82427.
61 Navascus CA, Rodrguez M, Sotorro NG, et al. Epidemiology
of hepatitis D virus infection: changes in the last 14 years.
Am J Gastroenterol 1995; 90: 198184.
62 Huo TI, Wu JC, Lin RY, Sheng WY, Chang FY, Lee SD. Decreasing
hepatitis D virus infection in Taiwan: an analysis of contributory
factors. J Gastroenterol Hepatol 1997; 12: 74751.
63 Deertekin H, Yalin K, Yakut M, Yurdaydin C. Seropositivity
for delta hepatitis in patients with chronic hepatitis B and liver
cirrhosis in Turkey: a meta-analysis. Liver Int 2008; 28: 49498.
64 Nwokediuko SC, Ijeoma U. Seroprevalence of antibody to HDV
in Nigerians with hepatitis B virus-related liver diseases.
Niger J Clin Pract 2009; 12: 43942.
65 Makuwa M, Mintsa-Ndong A, Souquire S, Nkogh D, Leroy EM,
Kazanji M. Prevalence and molecular diversity of hepatitis B virus
and hepatitis delta virus in urban and rural populations in
northern Gabon in central Africa. J Clin Microbiol 2009;
47: 226568.
66 Chakraborty P, Kailash U, Jain A, et al. Seroprevalence of
hepatitis D virus in patients with hepatitis B virus-related liver
diseases. Indian J Med Res 2005; 122: 25457.
67 Seetlani NK, Abbas Z, Raza S, Yakoob J, Jafris W. Prevalence
of hepatitis D in HBsAg positive patients visiting liver clinics.
J Pak Med Assoc 2009; 59: 43437.
68 Somi MH, Farhang S, Miri SM, Pouri AA, Mjidi G, Alavian SM.
The frequency of hepatitis D virus in patients with hepatitis B in
Iran: an increasing rate? Trop Doct 2009; 39: 15456.
69 Viana S, Paran R, Moreira RC, Compri AP, Macedo V. High
prevalence of hepatitis B virus and hepatitis D virus in the western
Brazilian Amazon. Am J Trop Med Hyg 2005; 73: 80814.
70 Khan A, Kurbanov F, Tanaka Y, et al. Epidemiological and clinical
evaluation of hepatitis B, hepatitis C, and delta hepatitis viruses in
Tajikistan. J Med Virol 2008; 80: 26876.
71 Tsatsralt-Od B, Takahashi M, Nishizawa T, Endo K, Inoue J,
Okamoto H. High prevalence of dual or triple infection of
hepatitis B, C, and delta viruses among patients with chronic liver
disease in Mongolia. J Med Virol 2005; 77: 49199.
Seminar
84 www.thelancet.com Vol 378 July 2, 2011
72 Smith HM, Alexander GJ, Webb G, McManus T, McFarlane IG,
Williams R. Hepatitis B and delta virus infection among at risk
populations in south east London. J Epidemiol Community Health
1992; 46: 14447.
73 Cross TJ, Rizzi P, Horner M, et al. The increasing prevalence of
hepatitis delta virus (HDV) infection in South London. J Med Virol
2008; 80: 27782.
74 Wedemeyer H, Heidrich B, Manns MP. Hepatitis D virus
infectionnot a vanishing disease in Europe! Hepatology 2007;
45: 133132.
75 Le Gal F, Castelneau C, Gault E, et al. Hepatitis D virus
infectionnot a vanishing disease in Europe! Hepatology 2007;
45: 133233.
76 Gaeta GB, Stroolini T, Smedile A, Niro G, Mele A. Hepatitis delta
in Europe: vanishing or refreshing? Hepatology 2007; 46: 131213.
77 Piccolo P, Lenci I, Telesca C, et al, for the Help B Free Network
Investigators. Patterns of chronic hepatitis B in Central Italy:
a cross-sectional study. Eur J Public Health 2009; published online
Nov 2. DOI:10.1093/eurpub/ckp168.
78 Nakano T, Shapiro CN, Hadler SC, et al. Characterization of
hepatitis D virus genotype III among Yucpa Indians in Venezuela.
J Gen Virol 2001; 82: 218389.
79 Manock SR, Kelley PM, Hyams KC, et al. An outbreak of fulminant
hepatitis delta in the Waorani, an indigenous people of the Amazon
basin of Ecuador. Am J Trop Med Hyg 2000; 63: 20913.
80 Tsatsralt-Od B, Takahashi M, Endo K, et al. Infection with
hepatitis A, B, C, and delta viruses among patients with acute
hepatitis in Mongolia. J Med Virol 2006; 78: 54250.
81 Borresen ML, Olsen OR, Ladefoged K, et al. Hepatitis D outbreak
among children in a hepatitis B hyper-endemic settlement in
Greenland. J Viral Hepat 2010; 17: 16270.
82 Flodgren E, Bengtsson S, Knutsson M, et al. Recent high incidence
of fulminant hepatitis in Samara, Russia: molecular analysis of
prevailing hepatitis B and D virus strains. J Clin Microbiol 2000;
38: 331116.
83 Sakugawa H, Nakasone H, Shokita H, et al. Seroepidemiological
study of hepatitis delta virus infection in Okinawa, Japan.
J Med Virol 1995; 45: 31215.
84 Lesbordes JL, Ravisse P, Georges AJ, et al. Studies on the role
of HDV in an outbreak of fulminant hepatitis in Bangui (Central
African Republic). Prog Clin Biol Res 1987; 234: 45159.
85 Casey JL, Niro GA, Engle RE, et al. Hepatitis B virus
(HBV)/hepatitis D virus (HDV) coinfection in outbreaks of acute
hepatitis in the Peruvian Amazon basin: the roles of HDV
genotype III and HBV genotype F. J Infect Dis 1996; 174: 92026.
86 Ponzetto A, Hoyer BH, Popper H, Engle R, Purcell RH, Gerin JL.
Titration of the infectivity of hepatitis D virus in chimpanzees.
J Infect Dis 1987; 155: 7278.
87 Wu JC, Chen CM, Sheen IJ, Lee SD, Tzeng HM, Choo KB. Evidence
of transmission of hepatitis D virus to spouses from sequence
analysis of the viral genome. Hepatology 1995; 22: 165660.
88 Rosenblum L, Darrow W, Witte J, et al. Sexual practices in the
transmission of hepatitis B virus and prevalence of hepatitis delta
virus infection in female prostitutes in the United States. JAMA
1992; 267: 247781.
89 Niro GA, Gravinese E, Martini E, et al. Clearance of hepatitis B
surface antigen in chronic carriers of hepatitis delta antibodies.
Liver 2001; 21: 25459.
90 Farci P, Smedile A, Lavarini C, et al. Delta hepatitis in inapparent
carriers of hepatitis B surface antigen. A disease simulating acute
hepatitis B progressive to chronicity. Gastroenterology 1983; 85: 66973.
91 Rizzetto M, Verme G, Recchia S, et al. Chronic hepatitis in
carriers of hepatitis B surface antigen, with intrahepatic
expression of the delta antigen. An active and progressive disease
unresponsive to immunosuppressive treatment. Ann Intern Med
1983; 98: 43741.
92 Colombo M, Cambieri R, Rumi MG, Ronchi G, Del Ninno E,
De Franchis R. Long-term delta superinfection in hepatitis B
surface antigen carriers and its relationship to the course of chronic
hepatitis. Gastroenterology 1983; 85: 23539.
93 Govindarajan S, Kanel GC, Peters RL. Prevalence of delta-antibody
among chronic hepatitis B virus infected patients in the
Los Angeles area: its correlation with liver biopsy diagnosis.
Gastroenterology 1983; 85: 16062.
94 Mederacke I, Filmann N, Bremer B, et al. Early HDV-RNA kinetics
after liver transplantation. Hepatology 2009;
50 (suppl 4): 547A (abstr).
95 Samuel D, Zignego AL, Reynes M, et al. Long-term clinical and
virological outcome after liver transplantation for cirrhosis caused
by chronic delta hepatitis. Hepatology 1995; 21: 33339.
96 Rosenau J, Kreutz T, Kujawa M, et al. HBsAg level at time of liver
transplantation determines HBsAg decrease and anti-HBs increase
and aects HBV DNA decrease during early immunoglobulin
administration. J Hepatol 2007; 46: 63544.
97 Smedile A, Casey JL, Cote PJ, et al. Hepatitis D viremia following
orthotopic liver transplantation involves a typical HDV virion with
a hepatitis B surface antigen envelope. Hepatology 1998; 27: 172329.
98 Lerut JP, Donataccio M, Ciccarelli O, et al. Liver transplantation and
HBsAg-positive postnecrotic cirrhosis: adequate
immunoprophylaxis and delta virus co-infection as the signicant
determinants of long-term prognosis. J Hepatol 1999; 30: 70614.
99 Su CW, Huang YH, Huo TI, et al. Genotypes and viremia of
hepatitis B and D viruses are associated with outcomes of chronic
hepatitis D patients. Gastroenterology 2006; 130: 162535.
100 Wu JC. Functional and clinical signicance of hepatitis D virus
genotype II infection. Curr Top Microbiol Immunol 2006; 307: 17386.
101 Watanabe H, Nagayama K, Enomoto N, et al. Chronic hepatitis
delta virus infection with genotype IIb variant is correlated with
progressive liver disease. J Gen Virol 2003; 84: 327589.
102 Niro GA, Smedile A, Andriulli A, Rizzetto M, Gerin JL, Casey JL.
The predominance of hepatitis delta virus genotype I among
chronically infected Italian patients. Hepatology 1997; 25: 72834.
103 Kiesslich D, Crispim MA, Santos C, et al. Inuence of hepatitis B
virus (HBV) genotype on the clinical course of disease in patients
coinfected with HBV and hepatitis delta virus. J Infect Dis 2009;
199: 160811.
104 Sagnelli E, Coppola N, Scolastico C, et al. Virologic and clinical
expressions of reciprocal inhibitory eect of hepatitis B, C, and
delta viruses in patients with chronic hepatitis. Hepatology 2000;
32: 110610.
105 Heidrich B, Deterding K, Tillmann HL, Raupach R, Manns MP,
Wedemeyer H. Virological and clinical characteristics of delta
hepatitis in Central Europe. J Viral Hepat 2009; 16: 88394.
106 Zachou K, Yurdaydin C, Drebber U, et al, for the HIDT-1 Study
Group. Quantitative HBsAg and HDV-RNA levels in chronic delta
hepatitis. Liver Int 2010; 30: 43037.
107 Williams V, Brichler S, Radjef N, et al. Hepatitis delta virus proteins
repress hepatitis B virus enhancers and activate the alpha/beta
interferon-inducible MxA gene. J Gen Virol 2009; 90: 275967.
108 Gordien E, Rosmorduc O, Peltekian C, Garreau F, Brchot C,
Kremsdorf D. Inhibition of hepatitis B virus replication by the
interferon-inducible MxA protein. J Virol 2001; 75: 268491.
109 Castelnau C, Le Gal F, Ripault MP, et al. E cacy of peginterferon
alpha-2b in chronic hepatitis delta: relevance of quantitative RT-PCR
for follow-up. Hepatology 2006; 44: 72835.
110 Mathurin P, Thibault V, Kadidja K, et al. Replication status and
histological features of patients with triple (B, C, D) and dual (B, C)
hepatic infections. J Viral Hepat 2000; 7: 1522.
111 Raimondo G, Brunetto MR, Pontisso P, et al. Longitudinal
evaluation reveals a complex spectrum of virological proles in
hepatitis B virus/hepatitis C virus-coinfected patients. Hepatology
2006; 43: 10007.
112 Schaper M, Rodriguez-Frias F, Jardi R, et al. Quantitative
longitudinal evaluations of hepatitis delta virus RNA and hepatitis B
virus DNA shows a dynamic, complex replicative prole in chronic
hepatitis B and D. J Hepatol 2010; 52: 65864.
113 Verme G, Amoroso P, Lettieri G, et al. A histological study of
hepatitis delta virus liver disease. Hepatology 1986; 6: 130307.
114 Guilhot S, Huang SN, Xia YP, La Monica N, Lai MM, Chisari FV.
Expression of the hepatitis delta virus large and small antigens in
transgenic mice. J Virol 1994; 68: 105258.
115 Wu JC, Chen TZ, Huang YS, et al. Natural history of hepatitis D
viral superinfection: signicance of viremia detected by polymerase
chain reaction. Gastroenterology 1995; 108: 796802.
116 Actis GC, Maran E, Rosina F, et al. Natural killer response to
exogenous interferon in delta hepatitis: boost or depression dened
within the rst week of therapy. Digestion 1987; 37: 5158.
Seminar
www.thelancet.com Vol 378 July 2, 2011 85
117 Nisini R, Paroli M, Accapezzato D, et al. Human CD4+ T-cell
response to hepatitis delta virus: identication of multiple epitopes
and characterization of T-helper cytokine proles. J Virol 1997;
71: 224151.
118 Aslan N, Yurdaydin C, Bozkaya H, et al. Analysis and function of
delta hepatitis virus-specic cellular immune responses. J Hepatol
2003; 38: 1516.
119 Wedemeyer H, Ciner A, Yurdaydin C, et al. Dierential cytokine
pattern of HDV-specic cellular immune responses distinguishes
treatment responder and nonresponder to PEG-IFN-2a treatment:
results from the HEP-NET/International HIDIT-1 study. J Hepatol
2007; 46 (suppl 1): S1314.
120 Huang YH, Tao MH, Hu CP, Syu WJ, Wu JC. Identication of
novel HLA-A*0201-restricted CD8+ T-cell epitopes on hepatitis delta
virus. J Gen Virol 2004; 85: 308998.
121 Aslan N, Yurdaydin C, Wiegand J, et al. Cytotoxic CD4 T cells
in viral hepatitis. J Viral Hepat 2006; 13: 50514.
122 Hsu SC, Syu WJ, Sheen IJ, Liu HT, Jeng KS, Wu JC. Varied
assembly and RNA editing e ciencies between genotypes I and II
hepatitis D virus and their implications. Hepatology 2002;
35: 66572.
123 Shih HH, Shih C, Wang HW, Su CW, Sheen IJ, Wu JC. Pro-205 of
large hepatitis delta antigen and Pro-62 of major hepatitis B surface
antigen inuence the assembly of dierent genotypes of hepatitis D
virus. J Gen Virol 2010; 91: 100412.
124 Shih HH, Jeng KS, Syu WJ, et al. Hepatitis B surface antigen levels
and sequences of natural hepatitis B virus variants inuence the
assembly and secretion of hepatitis D virus. J Virol 2008;
82: 225064.
125 Huang C, Chang SC, Yang HC, Chien CL, Chang MF.
Clathrin-mediated post-Golgi membrane tra cking in the
morphogenesis of hepatitis delta virus. J Virol 2009; 83: 1231424.
126 Wang YC, Huang CR, Chao M, Lo SJ. The C-terminal sequence of
the large hepatitis delta antigen is variable but retains the ability to
bind clathrin. Virol J 2009; 6: 31.
127 Mederacke I, Bremer B, Heidrich B, et al. Establishment of a novel
quantitative hepatitis D virus (HDV) RNA assay using the Cobas
TaqMan platform to study HDV RNA kinetics. J Clin Microbiol 2010;
48: 202229.
128 Manesis EK, Schina M, Le Gal F, et al. Quantitative analysis of
hepatitis D virus RNA and hepatitis B surface antigen serum levels
in chronic delta hepatitis improves treatment monitoring.
Antivir Ther 2007; 12: 38188.
129 Le Gal F, Gordien E, Aolabi D, et al. Quantication of hepatitis
delta virus RNA in serum by consensus real-time PCR indicates
dierent patterns of virological response to interferon therapy in
chronically infected patients. J Clin Microbiol 2005; 43: 236369.
130 Boyd A, Lacombe K, Miailhes P, et al. Longitudinal evaluation of
viral interactions in treated HIV-hepatitis B co-infected patients
with additional hepatitis C and D virus. J Viral Hepat 2010;
17: 6576.
131 Hadziyannis SJ. Use of alpha-interferon in the treatment of chronic
delta hepatitis. J Hepatol 1991; 13 (suppl 1): S2126.
132 Farci P, Roskams T, Chessa L, et al. Long-term benet of interferon
alpha therapy of chronic hepatitis D: regression of advanced hepatic
brosis. Gastroenterology 2004; 126: 174049.
133 Lau DT, Kleiner DE, Park Y, Di Bisceglie AM, Hoofnagle JH.
Resolution of chronic delta hepatitis after 12 years of interferon alfa
therapy. Gastroenterology 1999; 117: 122933.
134 Di Marco V, Giacchino R, Timitilli A, et al. Long-term
interferon-alpha treatment of children with chronic hepatitis delta:
a multicentre study. J Viral Hepat 1996; 3: 12328.
135 Yurdaydin C, Bozkaya H, Karaaslan H, et al. A pilot study of 2 years
of interferon treatment in patients with chronic delta hepatitis.
J Viral Hepat 2007; 14: 81216.
136 Niro GA, Ciancio A, Gaeta GB, et al. Pegylated interferon alpha-2b
as monotherapy or in combination with ribavirin in chronic
hepatitis delta. Hepatology 2006; 44: 71320.
137 Erhardt A, Gerlich W, Starke C, et al. Treatment of chronic hepatitis
delta with pegylated interferon-alpha2b. Liver Int 2006; 26: 80510.
138 Wedemeyer H, Yurdaydin C, Dalekos GN, et al. Peginterferon plus
adefovir versus either drug alone for hepatitus delta. N Engl J Med
2011; 364: 32231.
139 Yurdaydin C, Bozkaya H, Onder FO, et al. Treatment of chronic
delta hepatitis with lamivudine vs lamivudine + interferon vs
interferon. J Viral Hepat 2008; 15: 31421.
140 Gunsar F, Akarca US, Ersoz G, et al. Two-year interferon therapy
with or without ribavirin in chronic delta hepatitis. Antivir Ther
2005; 10: 72126.
141 Lau DT, Doo E, Park Y, et al. Lamivudine for chronic delta hepatitis.
Hepatology 1999; 30: 54649.
142 Niro GA, Ciancio A, Tillman HL, et al. Lamivudine therapy in
chronic delta hepatitis: a multicentre randomized-controlled pilot
study. Aliment Pharmacol Ther 2005; 22: 22732.
143 Wolters LM, van Nunen AB, Honkoop P, et al. Lamivudine-high
dose interferon combination therapy for chronic hepatitis B
patients co-infected with the hepatitis D virus. J Viral Hepat 2000;
7: 42834.
144 Canbakan B, Senturk H, Tabak F, et al. E cacy of interferon
alpha-2b and lamivudine combination treatment in comparison to
interferon alpha-2b alone in chronic delta hepatitis: a randomized
trial. J Gastroenterol Hepatol 2006; 21: 65763.
145 Yurdaydin C, Bozkaya H, Gurel S, et al. Famciclovir treatment
of chronic delta hepatitis. J Hepatol 2002; 37: 26671.
146 Garripoli A, Di Marco V, Cozzolongo R, et al. Ribavirin treatment
for chronic hepatitis D: a pilot study. Liver 1994; 14: 15457.
147 Kaymakoglu S, Karaca C, Demir K, et al. Alpha interferon and
ribavirin combination therapy of chronic hepatitis D.
Antimicrob Agents Chemother 2005; 49: 113538.
148 Onder F, Yakut M, Idilman R, et al. Entecavir may be benecial in
a subset of patients with chronic delta hepatitis. Hepatology 2009;
50 (suppl 4): 735A (abstr).
149 Mansour W, Ducancelle A, Le Gal F, et al. Resolution of chronic
hepatitis Delta after 1 year of combined therapy with pegylated
interferon, tenofovir and emtricitabine. J Clin Virol 2010; 47: 9799.
150 Sheldon J, Ramos B, Toro C, et al. Does treatment of hepatitis B
virus (HBV) infection reduce hepatitis delta virus (HDV) replication
in HIV-HBV-HDV-coinfected patients? Antivir Ther 2008;
13: 97102.
151 Vietheer PT, Netter HJ, Sozzi T, Bartholomeusz A. Failure of the
lamivudine-resistant rtM204I hepatitis B virus mutants to e ciently
support hepatitis delta virus secretion. J Virol 2005; 79: 657073.
152 Yurdaydin C, Kabacam G, Cakaloglu Y, et al. E cacy of pegylated
interferon-based treatment in patients with cirrhosis due to chronic
delta hepatitis: comparison with non-cirrhotic patients. Hepatology
2009; 50 (suppl 4): 736A (abstr).
153 OGrady JG, Alexander GJ, Hayllar KM, Williams R. Early indicators
of prognosis in fulminant hepatic failure. Gastroenterology 1989;
97: 43945.
154 Bordier BB, Ohkanda J, Liu P, et al. In vivo antiviral e cacy of
prenylation inhibitors against hepatitis delta virus. J Clin Invest
2003; 112: 40714.
155 Glebe D, Urban S, Knoop EV, et al. Mapping of the hepatitis B virus
attachment site by use of infection-inhibiting preS1 lipopeptides
and tupaia hepatocytes. Gastroenterology 2005; 129: 23445.
156 Gripon P, Cannie I, Urban S. E cient inhibition of hepatitis B
virus infection by acylated peptides derived from the large viral
surface protein. J Virol 2005; 79: 161322.

You might also like