Hepatitis D review from the Lancet

download Hepatitis D review from the Lancet

of 13

Transcript of Hepatitis D review from the Lancet

  • 8/12/2019 Hepatitis D review from the Lancet

    1/13

    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 withunexplained variations in their geographical distribution and pathogenicity. The hepatitis D virion is composed of acoat of HBV envelope proteins surrounding the nucleocapsid, which consists of a single-stranded, circular RNAgenome complexed with delta antigen, the viral protein. HDV is clinically important because although it suppressesHBV replication, it causes severe liver disease with rapid progression to cirrhosis and hepatic decompensation. Therange of clinical presentation is wide, varying from mild disease to fulminant liver failure. The prevalence of HDV isdeclining in some endemic areas but increasing in northern and central Europe because of immigration. Treatmentof HDV is with pegylated interferon alfa; however, response rates are poor. Increased understanding of the molecularvirology of HDV will identify novel therapeutic targets for this most severe form of chronic viral hepatitis.

    IntroductionHepatitis delta virus (HDV) is a small, defective RNAvirus that is related more to plant viroids than to otherhuman pathogens. It can propagate only in an individualwho has coexistent hepatitis B virus (HBV), either aftersimultaneous transmission of the two viruses, or viasuperinfection of an established HBV carrier.1 Clinicalexpression of HDV is wide, and although it sometimesfollows a benign course, the disease is clinicallyimportant. Studies have consistently shown that mostpatients with HBV and HDV co-infection have moresevere liver disease,2,3 more rapid progression tocirrhosis,4,5 and increased hepatic decompensation and

    death

    6,7

    than do those with HBV infection alone. Advanceshave improved our understanding of the transmission,replication, and pathogenesis of the virus; however, wedo not yet fully understand the mechanisms by which itcauses such severe liver disease. In this Seminar, we willreview the biology, pathogenesis, epidemiology, naturalhistory, clinical presentation, and management of thisdisease. We discuss how the most recent evidence mighthelp in the design of novel therapeutic agents for HDV,the most severe of all the chronic viral hepatitides.

    Historical perspectiveRizzetto and colleagues8 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 anddelta-antibody, and noted that this system occurred onlyin patients with HBV and was associated with severeliver disease. The disease was later associated with aparticle consisting of an RNA genome of low molecularweight that was encapsidated by HBV envelope proteins.9This particle was termed the delta agent, or HDV, andwas classified under the Deltavirus genus.

    Viral structureThe HDV virion is a small, spherical particle of about36 nm in diameter. It is composed of an outer coatcontaining the three HBV envelope proteins termedlarge, medium, and small hepatitis B surface antigen

    (HBsAg),10 and host lipids surrounding an innernucleocapsid, consisting of a single-stranded, circularRNA of 1679 nucleotides and about 200 molecules ofhepatitis D antigen (HDAg) per genome.11,12 Because ofthe high GC content of the nucleotide sequence, 12 thecircular genome, which is unique to animal viruses andmore closely related to plant viroids than to humanpathogens,13 can fold into an unbranched, rod-likestructure with 74% intramolecular base-pairing.

    Virus life cycleThe HDV receptor on the human hepatocyte remainsunidentified, 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-bindingdomain in the N-terminal region of the pre-S1 moiety oflarge HBsAg.14,15 For HDV infectivity, this domainrequires modification by myristoylation.16Fine mappinghas identified aminoacid residues 915 as the receptorbinding site.17A second infectivity region in the antigenicloop of all three envelope proteins is also required forinfectivity,18,19 but whether the antigenic loop and thepre-S1 determinants act synergistically or independentlyin viral entry is unclear.

    The virus is uncoated after entering the hepatocyte,and a signal in HDAg translocates the nucleocapsid tothe nucleus.20 The antigen has no RNA polymerase

    Lancet2011; 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);

    andDepartment of

    Gastroenterology, Hepatologyand 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

    [email protected]

    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 identified 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.

  • 8/12/2019 Hepatitis D review from the Lancet

    2/13

    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 thentreat the genome as double-stranded DNA because of itsfolded, rod-like structure.21Three forms of RNA are madein the host during replication: circular genomic RNA,circular complementary antigenomic RNA, and a linearpolyadenylated antigenomic RNA of 08 kb, which is themessenger RNA (mRNA) containing the open readingframe of the HDAg.

    Initial studies implicated RNA polymerase II in HDVreplication; however, one study has shown that RNApolymerases I and III also interact with HDV RNA,suggesting a more complex reliance on several hostpolymerases.22 Evidence suggests that synthesis of thedifferent RNA species occurs in different subcellular

    locations, mediated by distinct cellular polymerases:synthesis of antigenomic RNA occurs in the nucleolusmediated by RNA polymerase I, whereas synthesis ofgenomic RNA takes place more diffusely in thenucleoplasm by RNA polymerase II.23Replication of thecircular HDV RNA template occurs via a rollingmechanism that is unique to animal viruses but analogousto that of plant viroids. HDV RNA is first synthesised as alinear molecule, potentially containing many copies ofthe genome, but in the genomic and antigenomic RNA, asequence of 85 nucleotides acts as a ribozyme, which self-cleaves the linear HDV RNA into monomers.24,25 Thesemonomers are then ligated to form circular RNA (theinvolvement of a host RNA ligase remains controversial).Ribozymes are regarded as a feature of plant viroids,26buta self-cleaving RNA sequence in an intron of the CPEB3gene is structurally and biochemically related to the HDVribozymes; therefore, HDV might have arisen from thehuman transcriptome.27

    HDAg is the only protein that is known to be encodedby the HDV genome. It consists of two isoforms: the27 kDa large-HDAg with 214 aminoacids; and a 24 kDasmall-HDAg with 195 aminoacids.28 The N-terminalsequence of the two isoforms is the same; they differ byonly 19 aminoacids at the C-terminus of the large-HDAg.The open reading frame of the antigenomic RNAgenerates both isoforms because of heterogeneity in the

    RNA at codon 196.28A UAG stop codon at this positionleads to translation of small-HDAg; however, RNAediting by the cellular enzyme adenosine deaminase-1changes the sequence to UGG and, consequently, thelonger large-HDAg is produced.12,28,29 Small-HDAgreturns to the nucleus and supports viral replication,30,31whereas large-HDAg is a negative regulator of HDVreplication and is essential for virion assembly.32Therefore, RNA editing is central to the replication cycleof HDV because it controls the levels of each isoform,and consequently, the balance between viral synthesisand particle assembly.

    Post-translational modification 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 ofantigenomic RNA by interacting with RNA poly-merase II,34,35 whereas the sumoylation of small-HDAgenhances the synthesis of genomic RNA and mRNA, butnot of antigenomic RNAproperties that are alsoascribed to acetylation.36Methylation of small-HDAg byarginine methyltransferase at arginine-13 (an RNA-binding domain), is essential for translocation of small-HDAg to the nucleus, and for replication of theantigenomic RNA strand to form the genomic RNAstrand.37 Hence, post-translational modifications deter-mine the balance of the viral life cycle, and presentattractive 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 membranesby a signal in the C-terminus of large-HDAg. Once there,these complexes associate with HBV envelope proteins tocreate an infectious virion.14,38Interaction of the C-terminusof large-HDAg with the clathrin heavy-chain in the trans-Golgi network is essential for viral assembly.39Figure 1 isa schematic representation of the delta particle and itslife cycle.

    Viral heterogeneityThe sequence of the HDV RNA genome is highlyvariable, and there is divergence of up to 16% within thesame genotype, compared with 2040% between differentgenotypes. Even in one individual the virus is a pool ofclosely related quasispecies.40 This divergence is partlydue to the scarcity of proofreading ability of RNApolymerases. 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 HDVcoding region, it is about 14910 for non-synonymoussubstitutions, and 06710 for synonymous sub-stitutions.41 However, heterogeneity is not uniform forthe entire coding region of the genome; the self-cleavingribozyme sequence and the RNA-binding domain ofHDAg are highly conserved, whereas the C-terminalregion of large-HDAg is very divergent. Historically,

    HDV genotyping was achieved by either immuno-histochemical analysis of liver tissue42 or by restrictionfragment length polymorphism of PCR products,43 andthe virus was thought to have evolved into three majorgenotypes that differed in their global distribution.44Genotyping by direct sequencing and analysis ofmolecular phylogenetic trees has shown that HDV existsas at least eight different clades, four of which seem to beof exclusively African origin.45,46

    EpidemiologyOf the 350 million chronic carriers of HBV worldwide,more than 15 million have serological evidence ofexposure to HDV.47Traditionally, the regions with highrates of HDV carriage where the virus is endemic are

  • 8/12/2019 Hepatitis D review from the Lancet

    3/13

    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, andparts of Asia.48 Rates of HDV infection are generallyhighest in regions where HBV is endemic, but there areexceptionseg, HDV co-infection is uncommon inVietnam and Indonesia.49,50In Chinaa large reservoir ofHBV infectionHDV prevalence varies widely betweenprovinces despite a high prevalence of HBV. In one studyfrom Hong Kong, carriage of HDV was almost universalin intravenous drug users who were HBsAg positive,contrasting with low rates in non-drug users.51

    HDV genotype 1 is prevalent worldwide,52 whereasgenotype 2 (previously termed genotype-2a) is found inJapan, Taiwan, and the Yakutia region of Russia.5355Genotype 3the most divergent genotypeis common

    in the Amazon Basin,56whereas genotype 4 (previously2b) is found in Taiwan and Japan.54,57HDV genotypes 58are found in individuals of African origin, including inthose who have migrated to northern Europe.45,46Figure 2shows the worldwide prevalence of HDV and distributionof its genotypes.

    Longitudinal studies have shown a decrease in HDVprevalence in some endemic areas, such as in Italy, whereinfection in HBsAg carriers has fallen from 246% in1983, to 14% in 1992, and 83% in 1997.5860Infection ratesare especially reduced in younger patients, and evidencesuggests that in Italy HDV infection is restricted toageing cohorts who were infected in the 1980s. In thepast three decades, reductions in HDV prevalence havealso been reported in Spain, Taiwan, and Turkey.6163

    Vaccination programmes for HBV have probablycontributed substantially to the decline in HDV in theseregions, but additional factors, including increasedawareness of the virus and its mode of transmission,have led to better implementation of preventive measures,such as the change to disposable needles, syringes, andother medical equipment, and a general improvement insocioeconomic conditions.

    There are caveats to the assertion that HDV prevalenceis declining. First, developing countries have not alwayshad the same measures of HBV control, and have notshown the same improvements in socioeconomicconditions as have developed countries. Data forprevalence in these regions are sparse, but cross-sectionalstudies have shown that rates of HDV co-infection in

    carriers of HBsAg remain greater than 10%, andsometimes as high as 70% in Nigeria, Gabon, India,Pakistan, Iran, the western Brazilian Amazon, Tajikistan,and Mongolia.6471 Second, HDV prevalence has notdecreased in northern Europe. Prevalence in northernEurope and the USA was thought to be low and confinedto high-risk groups, such as intravenous drug-users;however, in London, UK, HDV prevalence in people withHBV increased from 26% in the 1980s to 85% in2005.72,73 In Germany, although the prevalence of anti-HDV antibody in those with HBV reduced from 186% in1992 to 68% in 1997, from 1999 onwards the rates haveincreased again to between 8% and 14%. 74HDV remainsprevalent in France.75The common factor in these threecountries is HDV infection in young individuals who

    L-HDAg

    Delta virion

    Ribonucleoprotein

    Hepatocytemembrane

    receptor

    L-HBsAg

    S-HBsAg

    M-HBsAg

    S-HDAg

    12 3

    9

    Nucleus

    Genomic RNA

    Endoplasmicreticulum

    Golgi complex

    Antigenomic RNAmRNA 6

    7

    68

    4 4

    5

    Figure : 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.

  • 8/12/2019 Hepatitis D review from the Lancet

    4/13

    Seminar

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

    have migrated from regions of high prevalence. Even inItaly the decline in HDV has now reached a plateau,76anda report has shown a high prevalence of HDV of 17% innon-EU citizens (mainly those from eastern Europe) whoare infected with HBV.77 Third, clustered outbreaks ofHDV superinfection continue to be reported, notably inVenezuela, Ecuador, Mongolia, and Greenland,7881whichare similar to those recorded in Samara (Russia), Okinawa(Japan), Central Africa, and the Amazon Basin in the1980s and 1990s.8285Thus, while outbreaks still occur andpopulation migration from endemic countries increases,the threat of HDV infection remains.

    Modes of transmissionLike HBV, HDV is transmitted via the parenteral routethrough exposure to infected blood or body fluids, andtests in chimpanzees have shown that only a very small

    inoculum is suffi cient to transmit infection.86 Thus,transmission rates remain high in intravenous drugusers. There is evidence for sexual transmission,87 andpeople with high-risk sexual activity are at increased riskfor infection.88Intrafamilial spread occurs and seems tobe common in regions of high prevalence, which isknown as inapparent parenteral transmission. Perinataltransmission of HDV is uncommon. Because of screeningof blood products, new infections in haemophiliacs, bloodtransfusion recipients, and patients receiving haemo-dialysis are no longer seen in developed countries.

    Clinical expression and natural historyWith HBV and HDV co-infection, the fate of HDV isdetermined by the host response to HBV, which in more

    than 95% of adults results in viral clearance. Acuteco-infection can be more severe than acute mono-infection with HBV, thereby resulting in acute liverfailure; however, disease expression is wide-ranging. Bycontrast, HDV superinfection of an individual withchronic HBV results in chronic HDV infection in mostpeople. In the remainder, replication of HDV stops, andthe natural history of the disease is that of the underlyingHBV; however, the residual liver disease might beadvanced. Important evidence from an Italian cohortshowed that 10% of patients with anti-HDV antibodiescleared HBsAg after a mean follow-up of 4 years,compared with 28% of those with HBV mono-infection.89The mechanism for increased rate of HBsAg loss afterclearance of HDV RNA is unknown, but an enhancedimmune response against HBV and HDV seemsplausible. Figure 3 shows the typical evolution of

    serological and virological markers in co-infected patientsversus superinfected patients.

    Superinfection can present as acute hepatitis in apreviously undiagnosed carrier of HBsAg, and is oftenmisdiagnosed as acute HBV or as worsening liverdisease due to chronic HBV.90 At initial histologicalassessment, patients with HDV superinfection areoften shown to have severe hepatitis with advancedfibrosis.9193 These patients undergo more rapidprogression to cirrhosis4,5 and increased hepaticdecompensation leading to death6,7 than do those withHBV infection alone. Despite the high rate ofprogression to cirrhosis, not all studies show anincreased rate of hepatocellular carcinoma, perhapsbecause of suppression of HBV replication by HDV.73

    HighIntermediateLowVery low

    Insufficient data

    Genotype 58

    Genotype 1

    Genotype 1

    Genotype 1/3

    Genotype 1/2/4

    Genotype 1

    Figure : Worldwide prevalence of HDV and the geographic distribution of i ts genotypes

    Two subtypes1A and 1Bhave been identified 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 identified primarily in patients from Africa. HDV=hepatitis D virus.

  • 8/12/2019 Hepatitis D review from the Lancet

    5/13

    Seminar

    www.thelancet.com Vol 378 July 2, 2011 77

    In liver transplantation for HDV, hepatitis Bhyperimmune globulin (HBIg) leads to rapid reductionin HBsAg concentrations, and serum HDV RNAdeclines in parallel.94Graft re-infection is prevented bylong-term administration of HBIg, which prevents

    HBV re-infection; hence, propagation of HDV cannotbe supported.95,96Originally, HDV was thought to persistas an isolated or latent infection after transplantation;however, more advanced and sensitive serologicalassays have shown that this latency does not occur.97Hence, the outcome of liver transplantation for HDV isvery good, with 5-year survival of more than 80%, andbetter than the outcome noted with transplantation forother causes of liver disease.95,98

    Although early studies identified that HDV genotypeaffects the natural history of HDV, the more recentlyidentified genotypes 58, from Africa, are less wellcharacterised. A study from Taiwan99showed a lower rateof remission and more adverse outcomes in patients withgenotype 1 than in those with genotype 2. Patients with

    genotype 4 often have mild liver disease,100but a variantof genotype 4 on the Miyako Island in Okinawa, Japan, isassociated with greater progression to cirrhosis thangenotype 4 is in Taiwan.101Genotype 3 has been linked tooutbreaks of severe, florid hepatitis (Labrea fever),

    culminating in acute liver failure and death in theAmazon Basin of South America.44Outbreaks of severehepatitis due to genotype 1 have also taken place, butgenotype 1 is associated with a wide range of disease, 102thereby making the relation between genotype andnatural history diffi cult to interpret.

    Although HBV genotype might also determine thenatural history of HDV, any effect is diffi cult to studybecause in many co-infected individuals the serum levelof HBV DNA is too low for genotype analysis. Nonetheless,Su and colleagues99showed a significantly lower remissionrate and more adverse outcomes in patients with HDVsuperinfection with HBV genotype C than with HBVgenotype B, in a population from Taiwan who were mostlyinfected with HDV genotypes 1 and 2. A study from Brazil

    A

    B

    Time after exposure (weeks)

    C

    Anti-HBc IgGAnti-HBc IgMAnti-HD IgGAnti-HD IgM

    HBsAgHDV RNAALT

    Figure : 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.

  • 8/12/2019 Hepatitis D review from the Lancet

    6/13

    Seminar

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

    has suggested that HBV genotype determines the severity

    of liver inflammation and the HDV viral load inindividuals with co-infection; however, the consequenceof this in relation to outcome is not clear.103

    Patterns of viral dominanceCo-infection and superinfection with HDV suppressesHBV replication in patients and in model systems.About 7090% of patients with HDV co-infection areHBeAg negative, and most have low serum HBVDNA.73,104106Evidence has indicated that the small (p24)and large (p27) HDV proteins downregulate HBV repli-cation by repressing activity of the two HBV enhancerregions, and by transactivating the interferon-inducibleMxA gene, which inhibits HBV replication by reducing

    the export of viral mRNA from the nucleus.107,108If HDVinfection is cleared either spontaneously or aftertreatment with interferon alfa, then HBV replicationcan reactivate.109

    Many patients with HBV and HDV have serologicalevidence of exposure to hepatitis C virus (HCV)about30% in European cohorts.73,105 In those with tripleinfection, HDV is the dominant virus because it not onlysuppresses HBV replication, but also inhibits HCVreplication.110 Indeed, HCV RNA was detected in lessthan 19% of patients who were anti-HCV antibodypositive, HBsAg positive, and anti-HDV antibody posi-tive.73,105,110Most patients who are HCV RNA negative haveprobably cleared HCV infection, but further long-termfollow-up studies are needed, especially because patternsof viral dominance evolve over time in HBV and HCVco-infection.111A dynamic pattern of viral dominance alsoexists in HBV and HDV co-infection.112

    PathogenesisThe mechanisms that determine whether an individualclears HDV spontaneously or becomes chronicallyinfected, and the processes that cause severe hepatitisand rapid progression of fibrosis, remain unclear.HDAg is not directly cytotoxic in human hepatocytes orin transgenic mice.113,114 Viral load of HDV was notassociated with severity of liver injury in a cohort of

    patients in a clinical trial.106 However, evidence fromobservational cohort studies suggests that in the acutephase of HDV infection, HDV viraemia is associatedwith an increased level of alanine aminotransferase andsuppressed HBV. In the chronic phase, falling HDVRNA, reactivation of HBV, and moderate transaminitisculminate in a late phase that is characterised by eitherthe development of cirrhosis and hepatocellularcarcinoma due to replication of HDV or HBV, orremission with clearance of both viruses.115Hence, viralload of HDV and HBV fluctuates according to the stageof viral infection. Whether this fluctuation represents adirect relation with the pathogenesis of diseaseprogression remains unclear, and other factors shouldbe considered.

    Host immune response

    The host immune response is thought to have animportant role in viral clearance and liver injury. An earlystudy116 showed that the function of natural killer cellswas active in individuals who suppressed HDV RNAduring treatment with interferon, but further studiesinvestigating the innate immune response in thepathogenesis of HDV are needed. The adaptive immuneresponse in HDV infection is also poorly defined. In onestudy,117 weak HDAg-specific responses of CD4 T cellswere elicited in patients with inactive HDV infection(anti-HDV antibody positive but RNA negative), butabsent in those with chronic HDV infection. Furtherstudies118 have shown that chronic HDV infection isassociated with a response that is dominated by T-helper-2

    (Th-2) cells, with a high frequency of T cells secretinginterleukin-10. Furthermore, after antiviral therapy,production of interleukin-10 that was HDV-peptidespecific remained higher in patients who did not respondto treatment than in those who did. Secretion ofinterferon-inducible protein-10 was, however, morefrequent in responders.119 These findings suggest thatHDV subverts the adaptive immune response away fromthe Th-1-biased CD4 and CD8 T-cell response that isneeded for viral clearance. Indeed, Huang andcolleagues120showed that the responses of HDV-specificCD8 cytotoxic T lymphocytes were detected in those withpast, but not active, infection.

    CD4 T cells are essential to the antiviral immuneresponse because they help CD8 T cells and B cells bystimulating antigen presenting cells and cytokinesecretion. However, cytotoxic CD4 T cells that areperforin-positive might also have a role in directlykilling virus-infected cells. A higher frequency of thesecells were recorded in patients who were co-infectedwith HBV and HDV than with HBV or HCVmono-infection, and the frequency of perforin-positivecells was positively correlated with disease activity.These findings suggest that such perforin-positive cellsare implicated in the pathogenesis of HDV liverdisease.121These studies suggest an important role forthe adaptive immune response, but the precise

    mechanisms are unknown.

    Role of HDV genotype in pathogenesisEffi ciency of RNA editing is lower with genotype 2 thanwith genotype 1, but this difference is unlikely to accountfor the reduced disease activity.122 The sequence of theC-terminal moiety in large-HDAg varies widely betweengenotypes, with up to 74% divergence between the HDVpackaging signal domains of genotype 1 and other HDVgenotypes. The C-terminal sequence of the large antigenin genotype 1 results in better packaging ability than ingenotype 2, which interacts weakly with clathrin, leadingto less effi cient assembly of particles.123125 However, allHDV genotypes are able to bind clathrin to some degree,which lends support to the role of clathrin binding in the

  • 8/12/2019 Hepatitis D review from the Lancet

    7/13

    Seminar

    www.thelancet.com Vol 378 July 2, 2011 79

    assembly of HDV particles.126 Which, if any, of thesemechanisms underpin the effect of genotype on thenatural history of HDV is unclear.

    DiagnosisThe development of anti-HDV antibodies is universal inindividuals with HDV; therefore, every patient who isHBsAg positive should be tested for anti-HDV IgGantibodies, which persist even after the patient hascleared HDV infection. Although active HDV infectionwas diagnosed historically by the presence of anti-HDV IgM antibodies, it is now confirmed by the detectionof serum HDV RNA with sensitive real-time PCRassay.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 thevariability of the genome sequence, assays of HDV RNAmight produce false-negative results, and testing of anti-HDV IgM antibodies still has a role in patients whotest negative for HDV RNA, but have clinical featuresof HDV-related liver disease. International standard-isation of the HDV RNA assay is needed. Somelaboratories undertake quantitative assays, but serumconcentrations of HDV RNA do not correlate withdisease activity or stage of liver fibrosis.106 Serialquantification of HDV RNA is used to determine theresponse to antiviral treatment.128,129

    HDV genotyping is usually only available in specialistcentres, but is gaining acceptance as a useful diagnostictest because patients with HDV genotype 1 are at ahigher risk of developing end-stage liver disease99 andhave a lower response to treatment with pegylatedinterferon than do patients with African genotypes(unpublished data). All patients with HDV also need

    investigation for HCV and HIV because co-infectionwith these viruses is common.73,105,130The panel shows therelevance of serological and virological markers in HDV.Patients who test positive for serum HDV RNA shouldbe further investigated with liver biopsy to assess theseverity of liver disease. Studies have shown noassociation between levels of HDV RNA, HBsAg titre,values of liver tests, and the histological staging of liverdisease, thus reinforcing the pivotal part that liver biopsyplays in the assessment of HDV-related liver disease.Tests for non-invasively assessing the progression ofliver fibrosis, such as transient elastography, can beuseful for detecting advanced hepatic fibrosis, but havenot been validated in chronic HDV and therefore cannotyet be recommended. The aspartate aminotransferase to

    HBsAg

    Anti-HDV IgG AbTreat as per local HBV

    guidelinesHCV/HIV Ab

    Add ribavirin

    +ve

    -ve

    >1 -ve

    +ve

    +ve

    HCV RNA+ve HBV DNA>2000 IU/mL

    Reactivation of HBV aftersuccessful control of HDV

    HDV RNA

    Liver biopsy

    Consider PEG- IFN- for atleast 48 weeks

    Consider addition of potentnucleos(t)ide analogue

    Add potent nucleos(t)ideanalogue

    Figure :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 specific.

    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

  • 8/12/2019 Hepatitis D review from the Lancet

    8/13

    Seminar

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

    platelet ratio index was shown to have no use inpredicting the stage of fibrosis in patients with HDV.106The assessment of patients with HDV infection issummarised in figure 4.

    TreatmentThe ideal endpoint of any treatment for HDV is not onlyclearance of HDV, but also of the helper virus HBV.Hence, a major challenge of defining the optimumtherapy is the added complexity of targeting two persistentviral infections. A meta-analysis of five studies oftreatment with recombinant interferon concluded thatsuch treatment was beneficial for HDV in terms of serumaminotransferase reduction, but the response was poorlysustained after discontinuation of treatment, and was notnecessarily associated with clearance of HDV RNA.There were better results with higher doses ofinterferonat least 5 MU daily or 9 MU thrice weekly for12 months, rather than for 6 months.131,132 High-doseinterferon induced delayed clearance of RNA, and

    sometimes, loss of HBsAg with resultant improvementin histological inflammatory activity and stage offibrosis.132In a single-case study,133extended treatment for12 years led to clearance of HDV RNA and loss of HBsAgassociated with complete regression of fibrosis.

    Consequently, some have advocated prolonging treatmentduration,47but studies have shown no additional benefitin giving treatment for 24 months rather than for12 months.134,135 The potential benefits of this approachshould be balanced with the side-effects, cost, andlogistical implications. Interferon-induced clearance ofHDV RNA has been associated with a fall in HBsAgconcentrations, and sometimes a loss of HBsAg, withabsence of HBsAg decline in patients who do not respondto treatment.128 However, the decrease in HBsAg oftenlags behind that of HDV RNA, and hence the mechanismbehind this finding is unclear. More data will emerge asquantification of HBsAg becomes more routinely used.

    Investigators have examined the use of pegylatedinterferon for chronic HDV. Series from Italy,136France,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 vsPEG+rbv PEG-IFN2b; 15 g/kg per week;

    72 weeks

    +Rbv 800 mg per day for first

    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 vsadv

    PEG-IFN2a; 180 g per week;

    48 weeks

    +Adv 10 mg; once dailyAdv 10 mg; once daily 48 weeks

    29

    3130

    38

    4233

    20

    1424

    6*

    6*6*

    1

    11

    31

    260

    45

    3510

    Yurdaydin et al (2008)139

    Multicentre randomised trial LAM vsLAM+IFN vsIFN 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 vsIFN+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 vsno Rx

    IFN2a; 9 MU three times per

    week 48 weeksIFN2a; 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. *Log10copies per mL. Log10genome equivalents per mL.

    Table:Summary of key trials of alpha-interferon for chronic hepatitis delta virus

  • 8/12/2019 Hepatitis D review from the Lancet

    9/13

    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, and17% of 12 patients, respectively. In the HIDIT-1 trial, 138which randomised 90 patients from Germany, Turkey,and Greece, pegylated interferon led to a 28% sustainedvirological response; the addition of adefovir did notimprove virological response but did lead to increasedsuppression of HBsAg concentrations, whereas adefovirmonotherapy was ineffective.138Why response rates varybetween series is not clear, but might relate to baselineclinical, demographical, and virological characteristicsthat are heterogeneous. The table shows data for themain interferon trials.

    The simplicity of the delta virus, particularly the lack ofviral polymerase, limits the targets for therapeutic

    compounds. Investigators have studied nucleos(t)ideanalogues, which block the HBV polymerase, butlamivudine alone was not effective at reducingconcentrations of HDV RNA and did not increasesustained virological response when combined withinterferon.139,141144Famciclovir was also ineffective.145Riba-virin alone,146or combined with interferon140,147or pegylatedinterferon136 did not increase the virological response.Entecavir did not reduce HDV viraemia, but did reducealanine aminotransferase and HBV DNA in a smallsubset of patients who had low HDV RNA and high HBVDNA.148One report has shown HDV clearance associatedwith seroconversion of HBsAg in a man treated withpegylated interferon plus tenofovir and emtricitabine,who had high levels of both HBV DNA and HDV RNA.149In a long-term observational study150of 16 patients whowere co-infected with HIV, HBV, and HDV and on highlyactive antiretroviral therapy with anti-HBV activity,including tenofovir, a significant reduction in HDV RNAwas noted for a median of 61 years, and three patientsbecame RNA negative. The mechanism for this reductionis unclear, because it was not associated with acorresponding decrease in concentrations of HBsAg.Hence, treatment with nucleos(t)ide analogues is noteffective at reducing HDV replication, but might be usefulin patients with high concentrations of HBV replication,and might be of potential benefit when used long term by

    gradually reducing HBsAg concentrations. The use ofolder analogues of nucleos(t)ides to induce HBVmutations is controversial. Lamivudine-induced muta-tions in the polymerase gene at rtM204V or rtM204I areassociated with changes in the overlapping envelope geneproducts, such as in sW196L/S, which inhibits secretionof HDV particles.151There is insuffi cient evidence to showwhether the failure to package and secrete HDV isbeneficial, or indeed whether the retention of HDV RNAwithin cells is harmful.

    Baseline and treatment factors predicting the outcomeof interferon treatment are poorly defined. HDV RNA iscorrelated positively with HBsAg titre,106,128and baselinevalues of both predict response to therapy (unpublisheddata). HDV genotype 1 is associated with a reduced

    response to pegylated interferon (unpublished data),

    but whether this response relates to a property of theHDV sequence itself, or merely to the finding thatpatients with genotype 1 have increased viral loads, isunknown. By contrast with treatment response inpatients with chronic HCV, in which rates of sustainedvirological response to pegylated interferon aresubstantially reduced by the presence of underlyingcirrhosis, cirrhosis associated with HDV infection didnot obviously affect the response to pegylated inter-feron.136,137,152Baseline liver biochemical tests are also notassociated with treatment outcome.109

    Although the kinetics of HDV RNA in therapy havebeen used to predict long-term virological outcome, dataare scarce.128,129 Castelnau and colleagues109 showed that

    75% of patients achieving an end-of-treatment responsewere HDV RNA negative by month 6 of treatment,compared with no patients who did not respond totreatment; however, undetectable mid-treatment RNAdid not protect against relapse. The investigators alsonoted that HDV RNA values were lower at the end oftherapy than at baseline in some patients who did notrespond, which could justify extension of therapy in thisgroup. Yurdaydin and colleagues139 defined threevirological patterns of response to interferon: complete,in which RNA negativity at 6 months predicted sustainedvirological response; partial, in which incompletesuppression of RNA at 6 months predicted rebound flarein viral replication after discontinuation of therapy; andnon-response, in which viraemia persisted withoutdecline throughout treatment and follow-up. Livertransplantation is the only treatment option for patientswho have end-stage liver disease due to co-infection withHBV and HDV, and is appropriate for those with acuteliver failure that fulfils criteria for poor prognosis. 153Re-infection of the graft is reduced by long-termadministration of immunoglobulins against the HBsAg(HBIg) after transplantation,95,96 creating an HBsAg-negative environment in which HDV cannot survive.97The use of potent antivirals against HBV has furtherreduced the risk of HBV re-infection.

    The accepted practice for treatment of chronic HDV is

    subcutaneous injections of pegylated interferon everyweek for at least 48 weeks. This practice should beconsidered in patients with active replication of HDVRNA, and histological evidence of disease activity, andwith no contraindications to interferon therapy. We wouldadvocate an individualised approach to therapy in patientswho have not achieved clearance of HDV RNA by sensitivePCR-based assay at 48 weeks. Those who have persistent,high-level viraemia, with detectable anti-HDV IgMantibody, and ongoing transaminitis, are unlikely torespond to further therapy. By contrast, patients with afalling viral load, declining IgM antibody titre, andresolving transaminitis, or with progressive decline inHBsAg titre, might benefit from extending therapy to72 weeks, and perhaps beyond, if improvement is

  • 8/12/2019 Hepatitis D review from the Lancet

    10/13

    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 potentnucleos(t)ide analogue to inhibit HBV replication islogical; however, the long-term effectiveness has yet to bedefined. Potent nucleos(t)ide analogues are the treatmentchoice for patients in whom HBV replication is reactivatedafter successful control of HDV replication. Suchanalogues might also be used in patients with detectableserum HBV DNA who cannot be treated with pegylatedinterferon such as those with advanced liver disease.

    Novel therapiesthe futureIncreased understanding of the molecular virology ofHDV, and the poor overall response to conventionalantiviral therapy, has driven the search for alternative

    antiviral targets. Prenylation of large-HDAg is essentialfor viral assembly and secretion,33and in mouse modelsprenylation inhibitors inhibited the assembly andrelease of HDV, leading to rapid clearance of HDVRNA from serum.154 These compounds are in pre-clinical development. Other forms of post-translationalmodification of HDAg, such as acetylation, phos-phorylation, and methylation, might also prove fruitfulas targets for novel therapeutic compounds, althoughnone are yet in development. Studies showing thatmyristoylated synthetic peptides specific for theN-terminal region of the pre-S1 domain of HBsAg areable to inhibit viral attachment and hence HDVinfectivity, draw attention to an alternative therapeutictarget.14,17,155,156 As we learn more about cell-signallingpathways associated with the pathogenesis of HDV,further targets might be exposed.

    Therapy for HDV deserves the resurgence of interestit has received. Treatment effectiveness remains unsatis-factory, and developments have lagged behind thoseachieved for the treatment of hepatitis B. The refinementof conventional antiviral regimens will continue withlarger collaborative trials, and drug development willhopefully follow the advances in HDV biology. Thesedevelopments remain necessary while worldwidemeasures to control the helper virus are inconsistent andthe 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 authorundertook the relevant searches and wrote the assigned sections.SAH assembled the sections and all authors revised the final version.

    Conflicts 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 andBristol-Myers Squibb. HW has received payment for board membershipfrom Roche, Gilead, Bristol-Myers Squibb, and Schering Plough;consultancy fees from Roche and Novartis; grant support from Rocheand Gilead; payment for participation in a speakers bureau from Roche,Gilead, Bristol-Myers Squibb, Schering Plough, and Novartis; andpayment for developing educational presentations from Roche. SAH

    declares that she has no conflicts of interest

    References1 Rizzetto M. Hepatitis D: thirty years after.J Hepatol2009;

    50:104350.2 Hadler SC, De Monzon M, Ponzetto A, et al. Delta virus infection

    and severe hepatitis. An epidemic in the Yucpa Indians ofVenezuela. Ann Intern Med1984; 100:33944.

    3 Govindarajan S, Chin KP, Redeker AG, Peters RL. Fulminant Bviral hepatitis: role of delta agent. Gastroenterology1984;86:141720.

    4 Saracco G, Rosina F, Brunetto MR, et al. Rapidly progressiveHBsAg-positive hepatitis in Italy. The role of hepatitis delta virusinfection.J Hepatol1987; 5:27481.

    5 Fattovich G, Boscaro S, Noventa F, et al. Influence of hepatitis deltavirus infection on progression to cirrhosis in chronic hepatitistype B.J Infect Dis1987; 155:93135.

    6 Fattovich G, Giustina G, Christensen E, et al. Influence of hepatitisdelta virus infection on morbidity and mortality in compensatedcirrhosis type B. The European Concerted Action on Viral Hepatitis(Eurohep). Gut2000; 46:42026.

    7 Romeo R, Del Ninno E, Rumi M, et al. A 28-year study of the courseof hepatitis Delta infection: a risk factor for cirrhosis andhepatocellular carcinoma. Gastroenterology2009; 136:162938.

    8 Rizzetto M, Canese MG, Arico S, et al. Immunofluorescencedetection of new antigen-antibody system (delta/anti-delta)associated to hepatitis B virus in liver and in serum of HBsAgcarriers. Gut1977; 18:9971003.

    9 Rizzetto M, Hoyer B, Canese MG, Shih JW, Purcell RH, Gerin JL.Delta Agent: association of delta antigen with hepatitis B surfaceantigen and RNA in serum of delta-infected chimpanzees.Proc Natl Acad Sci USA1980; 77:612428.

    10 Sureau C. The role of the HBV envelope proteins in the HDVreplication cycle. Curr Top Microbiol Immunol2006; 307:11331.

    11 Gudima S, Chang J, Moraleda G, Azvolinsky A, Taylor J. Parametersof human hepatitis delta virus genome replication: the quantity,quality, and intracellular distribution of viral proteins and RNA.J Virol2002; 76:370919.

    12 Wang KS, Choo QL, Weiner AJ, et al. Structure, sequence and

    expression of the hepatitis delta (delta) viral genome. Nature1986;323:50814.

    13 Kos A, Dijkema R, Arnberg AC, van der Meide PH, Schellekens H.The hepatitis delta (delta) virus possesses a circular RNA. Nature1986; 323:55860.

    14 Barrera A, Guerra B, Notvall L, Lanford RE. Mapping of thehepatitis B virus pre-S1 domain involved in receptor recognition.J Virol2005; 79:978698.

    15 Engelke M, Mills K, Seitz S, et al. Characterization of a hepatitis Band hepatitis delta virus receptor binding site. Hepatology2006;43:75060.

    16 Abou-Jaoud G, Molina S, Maurel P, Sureau C. Myristoylationsignal transfer from the large to the middle or the small HBVenvelope protein leads to a loss of HDV particles infectivity.Virology2007; 365:20409.

    17 Schulze A, Schieck A, Ni Y, Mier W, Urban S. Fine mapping ofpre-S sequence requirements for hepatitis B virus large envelopeprotein-mediated receptor interaction.J Virol2010; 84:19892000.

    18 Abou-Jaoud G, Sureau C. Entry of hepatitis delta virus requiresthe conserved cysteine residues of the hepatitis B virus envelopeprotein antigenic loop and is blocked by inhibitors of thiol-disulfideexchange.J Virol2007; 81:1305766.

    19 Salisse J, Sureau C. A function essential to viral entry underliesthe hepatitis B virus a determinant.J Virol2009; 83:932128.

    20 Xia YP, Yeh CT, Ou JH, Lai MM. Characterization of nucleartargeting signal of hepatitis delta antigen: nuclear transport asa protein complex.J Virol1992; 66:91421.

    21 Lai MM. RNA replication without RNA-dependent RNApolymerase: surprises from hepatitis delta virus.J Virol2005;79:795158.

    22 Greco-Stewart VS, Schissel E, Pelchat M. The hepatitis delta virusRNA genome interacts with the human RNA polymerases I and III.Virology2009; 386:1215.

    23 Li YJ, Macnaughton T, Gao L, Lai MM. RNA-templated replicationof hepatitis delta virus: genomic and antigenomic RNAs associate

    with different nuclear bodies.J Virol2006; 80:647886.

  • 8/12/2019 Hepatitis D review from the Lancet

    11/13

    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. Science1989;243:64952.

    25 Wu HN, Lin YJ, Lin FP, Makino S, Chang MF, Lai MM. Humanhepatitis delta virus RNA subfragments contain an autocleavageactivity. Proc Natl Acad Sci USA1989; 86:183135.

    26 Taylor J, Pelchat M. Origin of hepatitis delta virus. Future Microbiol2010; 5:393402.

    27 Salehi-Ashtiani K, Luptk A, Litovchick A, Szostak JW.A genomewide search for ribozymes reveals an HDV-like sequencein the human CPEB3gene. Science2006; 313:178892.

    28 Weiner AJ, Choo QL, Wang KS, et al. A single antigenomic openreading frame of the hepatitis delta virus encodes the epitope(s) ofboth hepatitis delta antigen polypeptides p24 delta and p27 delta.J Virol1988; 62:59499.

    29 Jayan GC, Casey JL. Inhibition of hepatitis delta virus RNA editingby short inhibitory RNA-mediated knockdown of ADAR1but notADAR2expression.J Virol2002; 76:12399404.

    30 Taylor JM. Structure and replication of hepatitis delta virus RNA.Curr Top Microbiol Immunol2006; 307:123.

    31 Yamaguchi Y, Filipovska J, Yano K, et al. Stimulation of RNApolymerase II elongation by hepatitis delta antigen. Science2001;293:12427.

    32 Chang MF, Chen CJ, Chang SC. Mutational analysis of deltaantigen: effect on assembly and replication of hepatitis delta virus.J Virol1994; 68:64653.

    33 Glenn JS, Watson JA, Havel CM, White JM. Identificationof a prenylation site in delta virus large antigen. Science1992;256:133133.

    34 Chen YS, Huang WH, Hong SY, Tsay YG, Chen PJ.ERK1/2-mediated phosphorylation of small hepatitis delta antigenat serine 177 enhances hepatitis delta virus antigenomic RNAreplication.J Virol2008; 82:934558.

    35 Hong SY, Chen PJ. Phosphorylation of serine 177 of the smallhepatitis delta antigen regulates viral antigenomic RNA replicationby interacting with the processive RNA polymerase II.J Virol2010;

    84:143038.36 Tseng CH, Cheng TS, Shu CY, Jeng KS, Lai MM. Modification of

    small hepatitis delta virus antigen by SUMO protein.J Virol2010;84:91827.

    37 Li YJ, Stallcup MR, Lai MM. Hepatitis delta virus antigen ismethylated at arginine residues, and methylation regulatessubcellular localization and RNA replication.J Virol2004;78:1332534.

    38 Wang CJ, Chen PJ, Wu JC, Patel D, Chen DS. Small-form hepatitisB surface antigen is suffi cient to help in the assembly of hepatitisdelta virus-like particles.J Virol1991; 65:663036.

    39 Huang C, Chang SC, Yu IC, Tsay YG, Chang MF. Large hepatitisdelta antigen is a novel clathrin adaptor-like protein.J Virol2007;81:598594.

    40 Dny P. Hepatitis delta virus genetic variability: from genotypes I,II, III to eight major clades? Curr Top Microbiol Immunol2006;307:15171.

    41 Krushkal J, Li WH. Substitution rates in hepatitis delta virus.J Mol Evol1995; 41:72126.

    42 Hsu SC, Syu WJ, Ting LT, Wu JC. Immunohistochemicaldifferentiation of hepatitis D virus genotypes. Hepatology2000;32:111116.

    43 Wu JC, Choo KB, Chen CM, Chen TZ, Huo TI, Lee SD. Genotypingof hepatitis D virus by restriction-fragment length polymorphismand relation to outcome of hepatitis D. Lancet1995; 346:93941.

    44 Casey JL, Brown TL, Colan EJ, Wignall FS, Gerin JL. A genotypeof hepatitis D virus that occurs in northern South America.Proc Natl Acad Sci USA1993; 90:901620.

    45 Radjef N, Gordien E, Ivaniushina V, et al. Molecular phylogeneticanalyses indicate a wide and ancient radiation of African hepatitisdelta virus, suggesting a deltavirus genus of at least seven majorclades.J Virol2004; 78:253744.

    46 Le Gal F, Gault E, Ripault MP, et al. Eighth major clade for hepatitisdelta virus. Emerg Infect Dis2006; 12:144750.

    47 Farci P. Delta hepatitis: an update.J Hepatol2003;

    39(suppl 1):S21219.

    48 Rizzetto M, Ponzetto A, Forzani I. Hepatitis delta virus as a globalhealth problem. Vaccine1990; 8(suppl):S1014.

    49 Nguyen VT, McLaws ML, Dore GJ. Highly endemic hepatitis Binfection in rural Vietnam.J Gastroenterol Hepatol2007;22:2093100.

    50 Mulyanto, Depamede SN, Surayah K, et al. A nationwide molecularepidemiological study on hepatitis B virus in Indonesia:identification of two novel subgenotypes, B8 and C7. Arch Virol2009; 154:104759.

    51 Abbas Z, Jafri W, Raza S. Hepatitis D: scenario in the Asia-Pacificregion. World J Gastroenterol2010; 16:55462.

    52 Shakil AO, Hadziyannis S, Hoofnagle JH, Di Bisceglie AM,Gerin JL, Casey JL. Geographic distribution and genetic variabilityof hepatitis delta virus genotype I. Virology1997; 234:16067.

    53 Imazeki F, Omata M, Ohto M. Heterogeneity and evolution ratesof delta virus RNA sequences.J Virol1990; 64:559499.

    54 Wu JC, Chiang TY, Sheen IJ. Characterization and phylogeneticanalysis of a novel hepatitis D virus strain discovered by restrictionfragment length polymorphism analysis.J Gen Virol1998;

    79:110513.55 Ivaniushina V, Radjef N, Alexeeva M, et al. Hepatitis delta virusgenotypes I and II cocirculate in an endemic area of Yakutia,Russia.J Gen Virol2001; 82:270918.

    56 Paran R, Kay A, Molinet F, et al. HDV genotypes in the WesternBrazilian Amazon region: a preliminary report. Am J Trop Med Hyg2006; 75:47579.

    57 Sakugawa H, Nakasone H, Nakayoshi T, et al. Hepatitis delta virusgenotype IIb predominates in an endemic area, Okinawa, Japan.J Med Virol1999; 58:36672.

    58 Smedile A, Lavarini C, Farci P, et al. Epidemiologic patterns ofinfection with the hepatitis B virus-associated delta agent in Italy.Am J Epidemiol1983; 117:22329.

    59 Sagnelli E, Stroffolini T, Ascione A, et al. Decrease in HDVendemicity in Italy.J Hepatol1997; 26:2024.

    60 Gaeta GB, Stroffolini T, Chiaramonte M, et al. Chronic hepatitis D:a vanishing Disease? An Italian multicenter study. Hepatology2000;32:82427.

    61 Navascus CA, Rodrguez M, Sotorro NG, et al. Epidemiologyof hepatitis D virus infection: changes in the last 14 years.Am J Gastroenterol1995; 90:198184.

    62 Huo TI, Wu JC, Lin RY, Sheng WY, Chang FY, Lee SD. Decreasinghepatitis D virus infection in Taiwan: an analysis of contributoryfactors.J Gastroenterol Hepatol1997; 12:74751.

    63 Deertekin H, Yalin K, Yakut M, Yurdaydin C. Seropositivityfor delta hepatitis in patients with chronic hepatitis B and livercirrhosis in Turkey: a meta-analysis. Liver Int2008; 28:49498.

    64 Nwokediuko SC, Ijeoma U. Seroprevalence of antibody to HDVin Nigerians with hepatitis B virus-related liver diseases.Niger J Clin Pract2009; 12:43942.

    65 Makuwa M, Mintsa-Ndong A, Souquire S, Nkogh D, Leroy EM,Kazanji M. Prevalence and molecular diversity of hepatitis B virusand hepatitis delta virus in urban and rural populations innorthern Gabon in central Africa.J Clin Microbiol2009;47:226568.

    66 Chakraborty P, Kailash U, Jain A, et al. Seroprevalence ofhepatitis D virus in patients with hepatitis B virus-related liverdiseases. Indian J Med Res2005; 122:25457.

    67 Seetlani NK, Abbas Z, Raza S, Yakoob J, Jafris W. Prevalenceof hepatitis D in HBsAg positive patients visiting liver clinics.J Pak Med Assoc2009; 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 inIran: an increasing rate? Trop Doct2009; 39:15456.

    69 Viana S, Paran R, Moreira RC, Compri AP, Macedo V. Highprevalence of hepatitis B virus and hepatitis D virus in the westernBrazilian Amazon. Am J Trop MedHyg2005; 73:80814.

    70 Khan A, Kurbanov F, Tanaka Y, et al. Epidemiological and clinicalevaluation of hepatitis B, hepatitis C, and delta hepatitis viruses inTajikistan.J Med Virol2008; 80:26876.

    71 Tsatsralt-Od B, Takahashi M, Nishizawa T, Endo K, Inoue J,Okamoto H. High prevalence of dual or triple infection ofhepatitis B, C, and delta viruses among patients with chronic liver

    disease in Mongolia.J Med Virol2005; 77:49199.

  • 8/12/2019 Hepatitis D review from the Lancet

    12/13

    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 Health1992; 46:14447.

    73 Cross TJ, Rizzi P, Horner M, et al. The increasing prevalence ofhepatitis delta virus (HDV) infection in South London.J Med Virol2008; 80:27782.

    74 Wedemeyer H, Heidrich B, Manns MP. Hepatitis D virusinfectionnot a vanishing disease in Europe! Hepatology2007;45:133132.

    75 Le Gal F, Castelneau C, Gault E, et al. Hepatitis D virusinfectionnot a vanishing disease in Europe! Hepatology2007;45:133233.

    76 Gaeta GB, Stroffolini T, Smedile A, Niro G, Mele A. Hepatitis deltain Europe: vanishing or refreshing? Hepatology2007; 46:131213.

    77 Piccolo P, Lenci I, Telesca C, et al, for the Help B Free NetworkInvestigators. Patterns of chronic hepatitis B in Central Italy:a cross-sectional study. Eur J PublicHealth2009; published onlineNov 2. DOI:10.1093/eurpub/ckp168.

    78 Nakano T, Shapiro CN, Hadler SC, et al. Characterization ofhepatitis D virus genotype III among Yucpa Indians in Venezuela.J Gen Virol2001; 82:218389.

    79 Manock SR, Kelley PM, Hyams KC, et al. An outbreak of fulminanthepatitis delta in the Waorani, an indigenous people of the Amazonbasin of Ecuador. Am J Trop MedHyg2000; 63:20913.

    80 Tsatsralt-Od B, Takahashi M, Endo K, et al. Infection withhepatitis A, B, C, and delta viruses among patients with acutehepatitis in Mongolia.J Med Virol2006; 78:54250.

    81 Borresen ML, Olsen OR, Ladefoged K, et al. Hepatitis D outbreakamong children in a hepatitis B hyper-endemic settlement inGreenland.J Viral Hepat2010; 17:16270.

    82 Flodgren E, Bengtsson S, Knutsson M, et al. Recent high incidenceof fulminant hepatitis in Samara, Russia: molecular analysis ofprevailing hepatitis B and D virus strains.J Clin Microbiol2000;38:331116.

    83 Sakugawa H, Nakasone H, Shokita H, et al. Seroepidemiologicalstudy of hepatitis delta virus infection in Okinawa, Japan.

    J Med Virol1995; 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 (CentralAfrican Republic). Prog Clin Biol Res1987; 234:45159.

    85 Casey JL, Niro GA, Engle RE, et al. Hepatitis B virus(HBV)/hepatitis D virus (HDV) coinfection in outbreaks of acutehepatitis in the Peruvian Amazon basin: the roles of HDVgenotype III and HBV genotype F.J Infect Dis1996; 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 Dis1987; 155:7278.

    87 Wu JC, Chen CM, Sheen IJ, Lee SD, Tzeng HM, Choo KB. Evidenceof transmission of hepatitis D virus to spouses from sequenceanalysis of the viral genome. Hepatology1995; 22:165660.

    88 Rosenblum L, Darrow W, Witte J, et al. Sexual practices in thetransmission of hepatitis B virus and prevalence of hepatitis deltavirus infection in female prostitutes in the United States.JAMA1992; 267:247781.

    89 Niro GA, Gravinese E, Martini E, et al. Clearance of hepatitis Bsurface antigen in chronic carriers of hepatitis delta antibodies.Liver2001; 21:25459.

    90 Farci P, Smedile A, Lavarini C, et al. Delta hepatitis in inapparentcarriers of hepatitis B surface antigen. A disease simulating acutehepatitis B progressive to chronicity. Gastroenterology1983; 85:66973.

    91 Rizzetto M, Verme G, Recchia S, et al. Chronic hepatitis incarriers of hepatitis B surface antigen, with intrahepaticexpression of the delta antigen. An active and progressive diseaseunresponsive to immunosuppressive treatment. Ann Intern Med1983; 98:43741.

    92 Colombo M, Cambieri R, Rumi MG, Ronchi G, Del Ninno E,De Franchis R. Long-term delta superinfection in hepatitis Bsurface antigen carriers and its relationship to the course of chronichepatitis. Gastroenterology1983; 85:23539.

    93 Govindarajan S, Kanel GC, Peters RL. Prevalence of delta-antibodyamong chronic hepatitis B virus infected patients in theLos Angeles area: its correlation with liver biopsy diagnosis.Gastroenterology1983; 85:16062.

    94 Mederacke I, Filmann N, Bremer B, et al. Early HDV-RNA kineticsafter liver transplantation. Hepatology2009;

    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 causedby chronic delta hepatitis. Hepatology1995; 21:33339.

    96 Rosenau J, Kreutz T, Kujawa M, et al. HBsAg level at time of livertransplantation determines HBsAg decrease and anti-HBs increaseand affects HBV DNA decrease during early immunoglobulinadministration.J Hepatol2007; 46:63544.

    97 Smedile A, Casey JL, Cote PJ, et al. Hepatitis D viremia followingorthotopic liver transplantation involves a typical HDV virion witha hepatitis B surface antigen envelope. Hepatology1998; 27:172329.

    98 Lerut JP, Donataccio M, Ciccarelli O, et al. Liver transplantation andHBsAg-positive postnecrotic cirrhosis: adequateimmunoprophylaxis and delta virus co-infection as the significantdeterminants of long-term prognosis.J Hepatol1999; 30:70614.

    99 Su CW, Huang YH, Huo TI, et al. Genotypes and viremia ofhepatitis B and D viruses are associated with outcomes of chronichepatitis D patients. Gastroenterology2006; 130:162535.

    100 Wu JC. Functional and clinical significance of hepatitis D virusgenotype II infection. Curr Top Microbiol Immunol2006; 307:17386.

    101 Watanabe H, Nagayama K, Enomoto N, et al. Chronic hepatitisdelta virus infection with genotype IIb variant is correlated withprogressive liver disease.J Gen Virol2003; 84:327589.

    102 Niro GA, Smedile A, Andriulli A, Rizzetto M, Gerin JL, Casey JL.The predominance of hepatitis delta virus genotype I amongchronically infected Italian patients. Hepatology1997; 25:72834.

    103 Kiesslich D, Crispim MA, Santos C, et al. Influence of hepatitis Bvirus (HBV) genotype on the clinical course of disease in patientscoinfected with HBV and hepatitis delta virus.J Infect Dis2009;199:160811.

    104 Sagnelli E, Coppola N, Scolastico C, et al. Virologic and clinicalexpressions of reciprocal inhibitory effect of hepatitis B, C, anddelta viruses in patients with chronic hepatitis. Hepatology2000;32:110610.

    105 Heidrich B, Deterding K, Tillmann HL, Raupach R, Manns MP,

    Wedemeyer H. Virological and clinical characteristics of deltahepatitis in Central Europe.J Viral Hepat2009; 16:88394.

    106 Zachou K, Yurdaydin C, Drebber U, et al, for the HIDT-1 StudyGroup. Quantitative HBsAg and HDV-RNA levels in chronic deltahepatitis. Liver Int2010; 30:43037.

    107 Williams V, Brichler S, Radjef N, et al. Hepatitis delta virus proteinsrepress hepatitis B virus enhancers and activate the alpha/betainterferon-inducible MxA gene.J Gen Virol2009; 90:275967.

    108 Gordien E, Rosmorduc O, Peltekian C, Garreau F, Brchot C,Kremsdorf D. Inhibition of hepatitis B virus replication by theinterferon-inducible MxA protein.J Virol2001; 75:268491.

    109 Castelnau C, Le Gal F, Ripault MP, et al. Effi cacy of peginterferonalpha-2b in chronic hepatitis delta: relevance of quantitative RT-PCRfor follow-up. Hepatology2006; 44:72835.

    110 Mathurin P, Thibault V, Kadidja K, et al. Replication status andhistological features of patients with triple (B, C, D) and dual (B, C)hepatic infections.J Viral Hepat2000; 7:1522.

    111 Raimondo G, Brunetto MR, Pontisso P, et al. Longitudinalevaluation reveals a complex spectrum of virological profiles inhepatitis B virus/hepatitis C virus-coinfected patients. Hepatology2006; 43:10007.

    112 Schaper M, Rodriguez-Frias F, Jardi R, et al. Quantitativelongitudinal evaluations of hepatitis delta virus RNA and hepatitis Bvirus DNA shows a dynamic, complex replicative profile in chronichepatitis B and D.J Hepatol2010; 52:65864.

    113 Verme G, Amoroso P, Lettieri G, et al. A histological study ofhepatitis delta virus liver disease. Hepatology1986; 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 intransgenic mice.J Virol1994; 68:105258.

    115 Wu JC, Chen TZ, Huang YS, et al. Natural history of hepatitis Dviral superinfection: significance of viremia detected by polymerasechain reaction. Gastroenterology1995; 108:796802.

    116 Actis GC, Maran E, Rosina F, et al. Natural killer response toexogenous interferon in delta hepatitis: boost or depression defined

    within the first week of therapy. Digestion1987; 37:5158.

  • 8/12/2019 Hepatitis D review from the Lancet

    13/13

    Seminar

    117 Nisini R, Paroli M, Accapezzato D, et al. Human CD4+ T-cellresponse to hepatitis delta virus: identification of multiple epitopes

    and characterization of T-helper cytokine profiles.J Virol1997;71:224151.

    118 Aslan N, Yurdaydin C, Bozkaya H, et al. Analysis and function ofdelta hepatitis virus-specific cellular immune responses.J Hepatol2003; 38:1516.

    119 Wedemeyer H, Ciner A, Yurdaydin C, et al. Differential cytokinepattern of HDV-specific cellular immune responses distinguishestreatment responder and nonresponder to PEG-IFN-2a treatment:results from the HEP-NET/International HIDIT-1 study.J Hepatol2007; 46 (suppl 1):S1314.

    120 Huang YH, Tao MH, Hu CP, Syu WJ, Wu JC. Identification ofnovel HLA-A*0201-restricted CD8+ T-cell epitopes on hepatitis deltavirus.J Gen Virol2004; 85:308998.

    121 Aslan N, Yurdaydin C, Wiegand J, et al. Cytotoxic CD4 T cellsin viral hepatitis.J Viral Hepat2006; 13:50514.

    122 Hsu SC, Syu WJ, Sheen IJ, Liu HT, Jeng KS, Wu JC. Variedassembly and RNA editing effi ciencies between genotypes I and IIhepatitis D virus and their implications. Hepatology2002;35:66572.

    123 Shih HH, Shih C, Wang HW, Su CW, Sheen IJ, Wu JC. Pro-205 oflarge hepatitis delta antigen and Pro-62 of major hepatitis B surfaceantigen influence the assembly of different genotypes of hepatitis Dvirus.J Gen Virol2010; 91:100412.

    124 Shih HH, Jeng KS, Syu WJ, et al. Hepatitis B surface antigen levelsand sequences of natural hepatitis B virus variants influence theassembly and secretion of hepatitis D virus.J Virol2008;82:225064.

    125 Huang C, Chang SC, Yang HC, Chien CL, Chang MF.Clathrin-mediated post-Golgi membrane traffi cking in themorphogenesis of hepatitis delta virus.J Virol2009; 83:1231424.

    126 Wang YC, Huang CR, Chao M, Lo SJ. The C-terminal sequence ofthe large hepatitis delta antigen is variable but retains the ability tobind clathrin. Virol J2009; 6:31.

    127 Mederacke I, Bremer B, Heidrich B, et al. Establishment of a novelquantitative hepatitis D virus (HDV) RNA assay using the Cobas

    TaqMan platform to study HDV RNA kinetics.J Clin Microbiol2010;48:202229.

    128 Manesis EK, Schina M, Le Gal F, et al. Quantitative analysis ofhepatitis D virus RNA and hepatitis B surface antigen serum levelsin chronic delta hepatitis improves treatment monitoring.Antivir Ther2007; 12:38188.

    129 Le Gal F, Gordien E, Affolabi D, et al. Quantification of hepatitisdelta virus RNA in serum by consensus real-time PCR indicatesdifferent patterns of virological response to interferon therapy inchronically infected patients.J Clin Microbiol2005; 43:236369.

    130 Boyd A, Lacombe K, Miailhes P, et al. Longitudinal evaluation ofviral interactions in treated HIV-hepatitis B co-infected patientswith additional hepatitis C and D virus.J Viral Hepat2010;17:6576.

    131 Hadziyannis SJ. Use of alpha-interferon in the treatment of chronicdelta hepatitis.J Hepatol1991; 13(suppl 1):S2126.

    132 Farci P, Roskams T, Chessa L, et al. Long-term benefit of interferonalpha therapy of chronic hepatitis D: regression of advanced hepatic

    fibrosis. Gastroenterology2004; 126:174049.133 Lau DT, Kleiner DE, Park Y, Di Bisceglie AM, Hoofnagle JH.

    Resolution of chronic delta hepatitis after 12 years of interferon alfatherapy. Gastroenterology1999; 117:122933.

    134 Di Marco V, Giacchino R, Timitilli A, et al. Long-terminterferon-alpha treatment of children with chronic hepatitis delta:a multicentre study.J Viral Hepat1996; 3:12328.

    135 Yurdaydin C, Bozkaya H, Karaaslan H, et al. A pilot study of 2 yearsof interferon treatment in patients with chronic delta hepatitis.J Viral Hepat2007; 14:81216.

    136 Niro GA, Ciancio A, Gaeta GB, et al. Pegylated interferon alpha-2bas monotherapy or in combination with ribavirin in chronic

    hepatitis delta. Hepatology2006; 44:71320.137 Erhardt A, Gerlich W, Starke C, et al. Treatment of chronic hepatitis

    delta with pegylated interferon-alpha2b. Liver Int2006; 26:80510.

    138 Wedemeyer H, Yurdaydin C, Dalekos GN, et al. Peginterferon plusadefovir versus either drug alone for hepatitus delta. N Engl J Med2011; 364:32231.

    139 Yurdaydin C, Bozkaya H, Onder FO, et al. Treatment of chronicdelta hepatitis with lamivudinevslamivudine + interferon vsinterferon.J Viral Hepat2008; 15:31421.

    140 Gunsar F, Akarca US, Ersoz G, et al. Two-year interferon therapywith or without ribavirin in chronic delta hepatitis. Antivir Ther2005; 10:72126.

    141 Lau DT, Doo E, Park Y, et al. Lamivudine for chronic delta hepatitis.Hepatology1999; 30:54649.

    142 Niro GA, Ciancio A, Tillman HL, et al. Lamivudine therapy inchronic delta hepatitis: a multicentre randomized-controlled pilotstudy. Aliment Pharmacol Ther2005; 22:22732.

    143 Wolters LM, van Nunen AB, Honkoop P, et al. Lamivudine-highdose interferon combination therapy for chronic hepatitis Bpatients co-infected with the hepatitis D virus.J Viral Hepat2000;7:42834.

    144 Canbakan B, Senturk H, Tabak F, et al. Effi cacy of interferonalpha-2b and lamivudine combination treatment in comparison tointerferon alpha-2b alone in chronic delta hepatitis: a randomizedtrial.J Gastroenterol Hepatol2006; 21:65763.

    145 Yurdaydin C, Bozkaya H, Gurel S, et al. Famciclovir treatmentof chronic delta hepatitis.J Hepatol2002; 37:26671.

    146 Garripoli A, Di Marco V, Cozzolongo R, et al. Ribavirin treatmentfor chronic hepatitis D: a pilot study. Liver1994; 14:15457.

    147 Kaymakoglu S, Karaca C, Demir K, et al. Alpha interferon andribavirin combination therapy of chronic hepatitis D.Antimicrob Agents Chemother2005; 49:113538.

    148 Onder F, Yakut M, Idilman R, et al. Entecavir may be beneficial ina subset of patients with chronic delta hepatitis. Hepatology2009;50(suppl 4):735A (abstr).

    149 Mansour W, Ducancelle A, Le Gal F, et al. Resolution of chronichepatitis Delta after 1 year of combined therapy with pegylatedinterferon, tenofovir and emtricitabine.J Clin Virol2010;47:9799.

    150 Sheldon J, Ramos B, Toro C, et al. Does treatment of hepatitis Bvirus (HBV) infection reduce hepatitis delta virus (HDV) replicationin HIV-HBV-HDV-coinfected patients? Antivir Ther2008;13:97102.

    151 Vietheer PT, Netter HJ, Sozzi T, Bartholomeusz A. Failure of thelamivudine-resistant rtM204I hepatitis B virus mutants to effi cientlysupport hepatitis delta virus secretion.J Virol2005; 79:657073.

    152 Yurdaydin C, Kabacam G, Cakaloglu Y, et al. Effi cacy of pegylatedinterferon-based treatment in patients with cirrhosis due to chronicdelta hepatitis: comparison with non-cirrhotic patients. Hepatology2009; 50(suppl 4):736A (abstr).

    153 OGrady JG, Alexander GJ, Hayllar KM, Williams R. Early indicatorsof prognosis in fulminant hepatic failure. Gastroenterology1989;97:43945.

    154 Bordier BB, Ohkanda J, Liu P, et al. In vivo antiviral effi cacy ofprenylation inhibitors against hepatitis delta virus.J Clin Invest2003; 112:40714.

    155 Glebe D, Urban S, Knoop EV, et al. Mapping of the hepatitis B virusattachment site by use of infection-inhibiting preS1 lipopeptidesand tupaia hepatocytes. Gastroenterology2005; 129:23445.

    156 Gripon P, Cannie I, Urban S. Effi cient inhibition of hepatitis Bvirus infection by acylated peptides derived from the large viralsurface protein.J Virol2005; 79:161322.