Hepatitis Delta Infection - Current and New Treatment Options

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

Best Practice & Research Clinical Gastroenterology 31 (2017) 321e327

Contents lists available at ScienceDirect

Best Practice & Research Clinical Gastroenterology


journal homepage: https://ees.elsevier.com/ybega/default.asp

10

Hepatitis delta infection e Current and new treatment options


Menashe Elazar, PhD, Senior Research Scientist a, 1,
Christopher Koh, MD, MHSc, Associate Professor d, 2,
Jeffrey S. Glenn, MD, PhD, Associate Professor of Medicine and Microbiology &
Immunology a, b, c, *
a
Department of Medicine, Division of Gastroenterology and Hepatology, Stanford University School of Medicine, 269 Campus Drive, Stanford, CA 94305,
USA
b
Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
c
Veterans Administration Medical Center, Palo Alto, CA, USA
d
Liver Diseases Branch, National Institute of Diabetes & Digestive & Kidney Diseases, NIH, 10 Center Drive, CRC, 5-2740 Bethesda, MD 20892 USA

a b s t r a c t
Keywords: In humans, hepatitis D virus (HDV) infection only occurs in the presence of a concomitant hepatitis B
Hepatitis delta virus virus (HBV) infection, and induces the most severe form of human viral hepatitis. Even though HDV is
Treatment
spread worldwide and is endemic in some regions, screening and treatment has been often neglected in
Novel therapies
part due to the lack of an effective therapy. Moreover, HDV prevalence rates are increasing in many
countries driven by immigration from areas of high endemicity. Currently, no FDA-approved anti-HDV
therapy is available, although interferon (IFN) alpha therapy has demonstrated benefit in a minority of
patients. In this review, we present a current view of our understanding of the epidemiology, molecular
virology and management of HDV infection. We additionally discuss new treatment approaches in
development and describe the most promising results of recent and ongoing clinical trials of these new
potential agents.
© 2017 Elsevier Ltd. All rights reserved.

Introduction infectiondeither as a de novo co-infection of an HBV naïve patient,


or as a superinfection of a patient already chronically infected with
Hepatitis Delta virus (HDV) was identified in 1977 in a cohort of HBV e a fact explained by HDV's molecular virology (see below).
hepatitis B virus (HBV) patients presenting with severe hepatitis. The presence of HDV is generally associated with worse clinical
Liver biopsies from these patients revealed a novel immunostaining outcomes compared to HBV monoinfection, both in terms of the
pattern [1]. This apparent new antigenic moiety was termed d an- severity of acute disease as well as accelerating the rate of devel-
tigen and was present in liver cell nuclei from chronic HBV patients. opment of the sequelae of chronic disease, including cirrhosis, liver
Although at first considered a new antigenic moiety of HBV, it was failure, HCC, or death [3]. Here, we will briefly review the molecular
quickly recognized as a separate antigen that was associated with virology, epidemiology and pathogenesis of HDV infection and will
HBsAg [1]. Further characterization identified the d antigen as an focus on current and new therapeutic approaches that are under
antigen of an infectious agent distinct from HBV that was associated clinical investigation.
with a low molecular weight RNA genome [2]. HDV infection was
found to always occurs in the presence of an accompanying HBV
Molecular virology

* Corresponding author. Departments of Medicine, Division of Gastroenterology HDV is a single-stranded (-)RNA virus with a 1.7 kb circular
and Hepatology and Microbiology & Immunology, Stanford University School of genome that forms a collapsed rod structure by the self-annealing
Medicine, Stanford, CA, USA. Fax: þ1 650 723 3032. of 74% of its nucleotides [4]. The viral genome codes for one protein
E-mail addresses: menashe@stanford.edu (M. Elazar), Christopher.Koh@nih.gov
(C. Koh), jeffrey.glenn@stanford.edu (J.S. Glenn).
that exists in two forms, known as small and large delta antigen
1
Fax: þ1 650 723 3032. (SHDAg and LHDAg), respectively. These proteins are identical
2
Fax: þ1 301 480 6303. except that the large delta antigen contains an extra 19 amino acids

http://dx.doi.org/10.1016/j.bpg.2017.05.001
1521-6918/© 2017 Elsevier Ltd. All rights reserved.
322 M. Elazar et al. / Best Practice & Research Clinical Gastroenterology 31 (2017) 321e327

at its carboxyl terminus. This extension is the result of a specific the influx of immigrants into western European countries from
RNA editing event that occurs during replication of the genome [5]. areas of high endemicity, such as Turkey, Eastern Europe and Af-
The addition of these 19 amino acids change the carboxy terminus rica [14e18]. Higher prevalence of HDV infection is observed in the
of the protein to possess a CXXX-box motif (where C ¼ cysteine, and eastern Mediterranean basin, Middle East, central and northern
X ¼ one of the last 3 amino acids at the carboxy terminus) that Asia and central western Africa [14]. For example, in Brazil, while
renders the protein a substrate for farnesyltransferase, an enzyme the prevalence in the general population is around 8%, the prev-
that adds a farnesyl group to the cysteine of the CXXX-box [6]. This alence in the Amazonian Basin is 65% in HBsAg positive out-
farnesylation reaction is an essential modification for virion as- patients, with similar observations in other South American
sembly [7]. The complete HDV virus particle consists of a complex countries [19]. In Africa, HDV affects mostly Western and Central
of the viral genome and both delta antigen isoforms, all encapsu- Africa with prevalence ranging from 33% in Mauritania [20] to up
lated by a lipid envelope decorated with HBV surface antigens (S, M to 66% in Gabon [21,22]. In Asia, a mixed picture emerges with
and L HBsAg) proteins, which are provided by a co-infecting HBV some areas demonstrating low HDV prevalence such as Malaysia,
[8]. The HBV proteins present in the lipid envelope provide the Thailand, Philippines and Japan, while a higher prevalence is
means for HDV particle exit and entry into the cell [8]. This observed in India and Pakistan [23]. In Taiwan while prevalence in
dependence on HBV for a source of envelope proteins provides the the general population is around 6%, in IVDU patients, the prev-
molecular explanation for why HDV infection is always accompa- alence has been reported to be 67% [24]. In Mongolia, some studies
nied by HBV co-infection. This supply of HBV surface antigens is the have reported populations with extremely high rates of HDV co-
only helper function provided by HBV. infection (>88%). A recent large nationwide survey using vali-
HDV genome replication proceeds entirely through RNA- dated assays documented ~60% HDV co-infection among HBsAg
dependent RNA replication. There are no DNA intermediates and subjectsdan extraordinarily high prevalence that may help
there is no archiving of HDV into DNA, yet HDV does not code for explain the world's highest rate of hepatocellular carcinoma being
its own RNA-dependent RNA-polymerase. Rather HDV depends on observed in that country [25,26]. It is noteworthy that diagnostics
host cell polymerases for its replication. Interestingly, the virus for anti-HDV infection are not standardized, and although there is
appears to recruit Pol-II, a host DNA-dependent RNA polymerase an international standard for HDV RNA detection to enable con-
to replicate its RNA genome in a RNA dependent manner [9]. It has version to International Units (IU), commercial pangenotypic as-
been hypothesized that the double stranded-like structure of the says for HDV RNA are not readily available in many countries. In
HDV genomic RNA provides a template akin to double stranded addition, many HBsAg-positive patients are simply not screened
DNA that enables the recruitment of Pol-II [10]. Following HDV for HDV. Together these factors combine to potentially underes-
entry and uncoating, the incoming HDV RNA genome is trans- timate HDV prevalence in many countries.
ferred into the nucleus. The RNA is replicated through a so-called
double rolling circle mechanism by Pol-II associated with the Pathogenesis
SHDAg to generate linear multimeric copies of antigenomic RNA,
which undergo cleavage by the antigenomic ribozyme to form From its discovery, it was noted that HDV infection worsens the
linear monomers that are then ligated, forming closed circular liver disease of HBV infected patients when compared to in-
antigenomes that serve as templates for production of genomic dividuals with HBV infection alone [1], however clinically there is
RNA in a similar mechanism [9]. As replication proceeds, an RNA no difference between HBV and HDV induced hepatitis [27]. Indeed,
editing event, mediated by ADAR1, occurs on the antigenomic RNA HDV induces the worst form of hepatitis in humans and is associ-
that modifies the amber stop codon of the reading frame encoding ated with higher risk of liver decompensation and death compared
SHDAg. This results in translation proceeding to the next down- to HBV infection alone [3].
stream stop codon, adding an extra 19 amino acids and yielding HDV infection can occur in one of two ways: coinfection, by
expression of the LHDAg [11]. These extra 19 amino acids which HBV and HDV infection happens simultaneously, or super-
dramatically change the function of the delta antigen. For infection, where HDV infects an individual already chronically
example, while the SHDAg is required for HDV RNA genome infected with HBV [28]. As for acute HBV infection, coinfection in
replication, the LHDAg acts as a potent transdominant inhibitor of adults is less likely to induce chronic hepatitis (>5%) [29], however
HDV replication [12]. Moreover, only the LHDAg isoform is able to development of severe acute hepatitis with a potential to result in
mediate assembly with HBsAg envelope proteins. Thus the above acute liver failure (2e20%) is the major risk [27]. Superinfection on
RNA editing event represents an important molecular switch in the other hand, results in 70e90% of infected individuals devel-
the virus life cycle, serving to turn off RNA replication and turn on oping chronic hepatitis [30]. Chronically infected HDV patients are
packaging of newly synthesized HDV genomes, virus assembly and at high risk of developing cirrhosis [31], with an estimated 80% of
release [13]. infected individuals developing cirrhosis within 30 years after
infection [30]. Additionally, 10e15% of chronically infected patients
Epidemiology may develop cirrhosis within two years of infection [30]. These
observations on the pathogenesis of HDV-infected patients clearly
It is estimated that HDV's worldwide prevalence is 15e20 demonstrate the high health burden of this infection and the need
million people, with an overall average of ~5% of the HBV popu- for the development of better diagnostic tools as well as more
lation harboring HDV, but the percentage of HBV patients co- effective antivirals.
infected with HDV is variable in different geographic regions HDV infection is associated with HCC, although the precise risk
[14]. Some areas have been identified as endemic for HDV infec- estimate is somewhat controversial. In one study, Fattovich et al.
tion, including regions in Africa, South America, Turkey, Southern [31] reported a 3-fold increased risk of developing HCC with a 2-fold
Italy and countries from the former Soviet Union [14]. In the increased risk for death. A similar result was obtained in a US study
United States and Northern Europe HDV prevalence has settled at conducted among veteran patients demonstrating that HDV was an
around 8% in HBsAg positive patients with higher prevalence in independent predictor with 3-fold increased risk for HCC develop-
populations of intravenous drug users (IVDU) and hemophiliacs ment [32]. In a Swedish study, the Standard Incidence Ratio (SIR) to
[15]. This represents a balance between declines from the origi- develop HCC in chronic HDV patients was higher compared to HBV
nally reported prevalences when HDV was first discovered, and infection alone [33]. In contrast, Romeo et al. [34] showed that 15% of
M. Elazar et al. / Best Practice & Research Clinical Gastroenterology 31 (2017) 321e327 323

patients with chronic HDV develop cancer with a relatively low However, this study did not demonstrate any improvement in
annual rate of 2.8%, and that there was no clear correlation with HDV response rates [47], thereby underscoring the need for more effi-
RNA levels [35]. Niro et al. [36] reported 9% of patients with chronic cient therapies. Despite the poor response rates of IFNa therapy in
HDV advanced to HCC, and Buti et al. [37] found that only 3% of HDV patients, the potential for long-term clinical benefits in re-
patients developed HCC. These authors attributed the low rate to the sponders supports a recommendation for treatment in chronically
young age of patients in their study, but all of these studies suggest HDV infected patients for 48 weeks with once weekly dosing of
that liver decompensation and death are a major risk factors in 180 mg PEG-IFNa. However, careful selection of patients for treat-
chronic HDV infected patients [34,36,37]. ment taking into consideration ALT levels, the degree of histologic
fibrosis, and side effects, is recommended. Fig. 1 offer a suggested
Treatment algorithm for HDV management.

Currently, no efficient anti-HDV therapy is available nor is there Potential antiviral targets
an FDA-approved therapy for HDV infection. The wide spectrum of
antiviral activity of interferon (IFN) alpha (a) initially prompted a The unique HDV life cycle and dependence on HBV for its entry
plethora of studies to examine its effect on HDV infection [38e40]. and egress presents several steps that have the potential for anti-
One early study reported a beneficial effect of IFNa with 48 weeks of viral intervention, including entry, replication, viral egress and RNA
treatment, where 71% of patients treated with a high dose (9 million interference (RNAi).
units) had undetectable virus in serum at the end of treatment
(EOT). However, at the end of 6 months post-therapy follow-up, only Replication
43% of subjects maintained RNA negativity and a longer follow-up
extending to 39 months post-therapy, demonstrated virologic Targeting the RNA dependent RNA replication, the genomic or
relapse in all responders [38]. Although the interpretation of these the antigenomic ribozymes, or the ligation step that produces the
results may reflect in part an early HDV RNA assay with suboptimal final circular genomic/antigenomic molecules are all valid sites for
sensitivity, long-term follow-up of the patients in this study (out to antiviral intervention in the HDV life cycle. Inhibition of the host
12 years) demonstrated an increased survival rate in the patients polymerase has not been reported, presumably as it may be too
treated with high dose IFN and overall a better clinical outcome in toxic to interfere with an essential host functions. However, SHDAg
terms of liver disease progression. Additionally, significantly better is an essential cofactor of Pol-II in the HDV replication cycle thus,
clinical outcomes were associated with achieving a 2 log drop in inhibition of the interaction between these two proteins may
HDV RNA [41]. Gunser et al. (2004) conducted a similar study but inhibit viral replication.
extended treatment duration out to two years. In this study, similar Aminoglycosides have been shown to inhibit the ribozyme ac-
trends were observed with 50% of patients achieving undetectable tivity, however this was not translated to inhibition of HDV in cell
HDV RNA levels at EOT, however only 20% of patients remained culture [48,49]. One possible explanation is that all the replication
negative at end of the follow-up period of 6 months [39]. Similar steps occur in the nucleus, thus every candidate molecule will have
outcomes with pegylated interferon a (PEG-IFNa) treatment were to reach the nucleus in order to exert an inhibitory effect on HDV
observed when tested in HDV infected subjects. One year PEG-IFNa replication, which might prove challenging.
treatment resulted in 57 % of patients achieving HDV RNA negativity,
which dropped to 43% at end of follow-up (up to 42 months, median HBV inhibition
16 months) [42], while up to 5 years of treatment resulted in only
42% of patients becoming HDV RNA negative with only 25% The dependence of HDV on HBV for completing its life cycle
remaining negative by the end of 5 years [43]. The subsequent suggests that HBV inhibition will result in inhibition of HDV viral
addition of ribavirin (RBV) to PEG-IFNa did not improve response load. However, several attempts to utilize various anti-HBV nucle-
rates and does not appear to have a role in the therapy of HDV os(t)ide analog therapies to treat HDV have demonstrated little to
infected patients [44]. Thus far, the most comprehensive evaluation no response in chronically infected HDV subjects [50e53]. This is
of PEG-IFNa treatment with long-term outcome reporting has been not surprising as inhibition of the HBV polymerase does not result
conducted by the Hep-Net-International Delta Hepatitis Interven- in HBsAg inhibition, which is the sole required HBV element for
tion Trial (HIDIT) study group. This consortium's first study, the HDV. Attempts to use HBV nucleos(t)ide analog therapy alone or in
HIDIT-1 study, enrolled 90 subjects into one of three arms combination with IFN and PEG-IFN has been attempted with no
comparing PEG-IFNa with adefovir (nucleotide analog) versus success (see Treatment above).
PEG-IFNa versus adefovir alone, and had a primary end point of ALT
normalization and clearance of HDV RNA at the end of 48 weeks of RNA interference
therapy. Unfortunately, only 7% of subjects achieved the primary
end-point in both PEG-IFNa treated study arms while none of those RNA interference (RNAi) is a collective name for small RNA
treated with adefovir alone did [45]. It is noteworthy that in both molecules used to inhibit gene expression in a sequence specific
PEG-IFNa treated groups, 28% of patients cleared the virus within 24 manner.
weeks after completing the treatment while none of the adefovir Since its discovery, RNAi approaches have been evaluated as
alone treated patients achieved viral clearance [45]. A long-term potential therapeutic modalities for various conditions including
follow-up of patients from this study (range of 0.5e5.5 years of infectious diseases and more specifically viral infections. A similar
follow-up) showed that 70% of patients that were treated with approach for HDV treatment may be feasible [54].
either of the PEG-IFNa arms, and who were HDV negative at end of
treatment, were positive at least once during follow-up, with 44% Promising clinical trials
being HDV RNA positive at end of follow-up. This highlights the need
for a close follow-up of patients even when HDV RNA negativity is Several agents have advanced beyond the preclinical stage and
achieved [46]. The consortium's second study, the HIDIT-2 study, are being actively investigated in a variety of clinical trials. The
aimed at determining the benefit of longer treatment (96 week) most advanced of these will be briefly reviewed next. Like other
with PEG-IFNa and tenofovir in chronically infected HDV subjects. viral diseases, combination therapy may ultimately result in the
324 M. Elazar et al. / Best Practice & Research Clinical Gastroenterology 31 (2017) 321e327

Fig. 1. Suggested algorithm for management of HDV infection.

highest response rates as these experimental therapies progress N-terminal of the L-HBsAg are the essential determinant for re-
through the pipeline. ceptor binding [56], and it has recently been demonstrated that a
myristoylated peptide corresponding to this sequence inhibits HBV
HBV entry inhibitor e Myrcludex-B and HDV entry in vitro and in vivo [57,58], through competitive
inhibition of the particle binding to hNTPC [59]. This peptide, called
The HBV entry receptor, human sodium-taurocholate cotrans- Myrcludex-B (Myr), is undergoing evaluation in clinical trials in
porting polypeptide (hNTCP) has been identified as a potential subjects with chronic HBV monoinfection and chronic HDV/HBV co-
target against HBV and HDV [55]. The 48 amino-acids of the preS1 infection. In an early phase clinical study, 24 patients with serum
M. Elazar et al. / Best Practice & Research Clinical Gastroenterology 31 (2017) 321e327 325

HDV RNA levels ranging from 1e3 to 1e5 copies/ml were treated mechanism of action of NAPs in HBV, with inhibition of HBsAg
with Myr (2 mg SQ QD) alone or in combination with PEG-IFNa and secretion in animal models, this raises the question of whether long
compared to PEG-IFNa alone for 24 weeks. A reported interim term administration may increase the risk of HCC [69,74].
analysis at the end of 24 weeks of treatment revealed no significant
changes in HBsAg, the study's primary end-point. However, HDV Viral assembly and release inhibitors e lonafarnib
RNA declined 1.67 (Myr arm), 2.6 (Myr þ PEG-IFNa arm), and 2.2 log
(PEG-IFNa arm), with 2, 5, and 2 subjects in the respective groups HDV requires HBsAg for completing its life cycle successfully,
achieving serum HDV RNA levels below the level of quantitation but this is not sufficient. Prenylation, in particular the covalent
[60]. HBV DNA was inhibited to near or below detectable levels in all addition of a farnesyl prenyl lipid group to the C-terminus of HDV
the patients in the Myr-PEG-IFNa arm while no changes were large antigen (HDLAg) is essential for successful interaction of
observed in the patients from the Myr or PEG-IFNa arms of the study HDLAg with HBsAg and formation of secreted particles, with HBsAg
[60]. Subjects were then continued on PEG-IFNa monotherapy for a [75]. Inhibition of farnesyltransferase was shown to inhibit HDV
total of 48 weeks, which did not appear to provide a significant virion secretion and inhibit infection in vitro [7] and in vivo [76].
benefit, with end of treatment responses comparable to what has This requirement prompted the investigation of farnesyltransferase
been seen historically for PEG-IFNa, and patients generally inhibitors (FTIs) for HDV treatment. The FTI lonafarnib (Sarasar®)
rebounding following cessation of therapy (Hepatitis Delta Inter- was extensively studied for use as a cancer therapy. Its develop-
national Network (HDIN) at AASLD 2016 and [61]. This pilot study ment as an oncology product was terminated due to insufficient
demonstrated that Myr-mediated inhibition of viral entry can have anti-tumor efficacy, but these studies provided an extensive safety
inhibitory activity against HDV, however higher doses, longer data base in over 2000 patients, where the side effects were pre-
treatment or combination with another drug will be required to dominantly gastrointestinal. This presented an ideal repurposing
eradicate the virus. Additionally, all the participants in this study opportunity as a potential anti-HDV agent. Lonafarnib (LNF) has
demonstrated relatively low viremia at baseline and further evalu- now been tested in multiple trials comprising over 100 HDV pa-
ation is needed to determine if similar results can be achieved in tients. In a placebo-controlled proof-of-concept study, 14 patients
patients with higher levels of serum HDV RNA. were treated with either 100 mg BID or 200 mg BID lonafarnib
administered orally for 4 weeks, and followed up for 6 months post
HBsAg secretion inhibitors e REP2055 and REP2139-Ca therapy. Both dosing groups had a drop in serum HDV RNA (0.73 log
and 1.54 log in the low and high dose treatment arms, respectively),
Polymers demonstrating inhibitory activity against a variety of and the changes in HDV RNA were highly correlated with the drug
viruses have been described for many years where their activity concentration measured in patients' sera [77], demonstrating the
appears to be dependent on polymer length and hydrophobicity (see potential utility of this drug for HDV management. Importantly,
review [62]). One such polymer class is the phosphorothioate nucleic there was no evidence of virus resistance, as had been predicted for
acid polymers (PS-ON or NAP) which possesses a broad spectrum such a host targeting antiviral therapy. Following this study, a series
antiviral activity due to its length and amphipathic nature [62]. of studies aiming to optimize drug dose and treatment duration
In vitro, NAPs have been shown to inhibit HIV-1 entry via interaction were conducted. The LOWR HDVd1 (LOnafarnib With and without
with an exposed amphipathic helix in the viral fusion protein gp41 Ritonavir in HDV e 1) study explored higher doses and longer
[63]. Additionally, NAPs have demonstrated inhibitory activity duration of treatment, as well as combination with ritonavir (an
against herpes simplex virus (HSV) [64,65], Arenaviruses [66] and inhibitor of LNF's metabolism). 300 mg LNF PO BID achieved a 2-log
hepatitis C virus (HCV) [67] through interaction of the NAP's hydro- reduction at 4 weeks, but had increased GI side effects. Adminis-
phobic side with amphipathic stretches within proteins from these tering a lower 100 mg dose PO BID, but with ritonavir (RTV) to
pathogens. Preclinical testing of the NAP molecule REP2055 against maximize post-absorbed levels of LNF, resulted in less GI side ef-
duck HBV (DHBV) demonstrated inhibition of HBV infection in fects with better antiviral response e 2.4 log drop at 4 weeks and
addition to post-entry inhibition [68]. This post-entry mechanism 3.2 log at 8 weeks [78]. Similar antiviral responses were seen with
involves the inhibition of DHBsAg secretion and its accumulation in 100 mg LNF PO BID combined with standard PEG-IFNa. In both
liver cells [69], a result that may have a clinical utility in HDV infected cases, the onset of viral inhibition in these combination treatments
patients. REP2139 was developed to address tolerability issues with was more rapid and profound than previously observed with
REP2055 in initial early clinical studies in HBV patients. 3 of 12 sub- PEG-IFNa alone in the context of the HIDIT-2 study. The LOWR-2
jects had HBsAg levels decline below the limit of quantitation after study aimed to identify the optimal combinations of LNF and
being treated with weekly IV infusions of REP2139 for 20e35 weeks, RTV, with or without PEG-IFNa, to maximize efficacy and tolera-
which was accompanied by 4e6 log reductions in HBV DNA [70]. bility. Although still ongoing, interim results have identified regi-
These promising results in HBV moninfected patients have resulted mens (e.g. 50 mg PO BID þ 100 mg RTV PO BID, and 25 mg PO
in a small cohort study testing REP2139 in HBV/HDV coinfected pa- BID þ 100 mg RTV PO BID þ PEG-IFNa) that appear to show good
tients. REP2139 was administered once weekly at a dose of 500 mg antiviral activity in a significant number of patients, and are suffi-
IV for 15 weeks followed by an additional 15 weeks of weekly ciently well tolerated to enable treatment for as long as needed to
REP2139 250 mg IV combined with PEG-IFNa, and then transitioning achieve HDV RNA negativity. Regimens bracketing those explored
to PEG-IFNa monotherapy for another 33 weeks (REP301- in LOWR HDV-2 were explored in LOWR HDV-3 and LOWR HDV-4.
ClinicalTrials.gov # NCT02233075). 4/12 patients exhibited large LOWR HDV-3 explored single daily doses of LNF (50 mg vs. 75 mg
drops in HBsAg (up to 5 logs) at week 15, which were accompanied by vs. 100 mg) þ RTV (100 mg) for 12 vs. 24 weeks, and LOWR HDV-4
HDV RNA going below the limit of detection [71], and several addi- sought to determine the efficacy of rapid step-wise dose escalation
tional patients were reported below the limit of quantitation after to 100 mg LNF þ 100 mg RTV, each PO BID [79]. The final results of
addition of PEG-IFNa [72]. Upon withdrawal of therapy, however, 5/ both will soon be available. In addition to a progressive decline of
12 patients became HDV RNA positive again (A. Valiant et al. the 12th HDV RNA on LNF therapy, another mechanism has emerged
HDIN at AASLD Boston, USA 2016 and [73]). A long-term follow-up whereby LNF treated patients can achieve HDV RNA negativity. In
study of these patients is underway to assess for maintenance of particular, when LOWR-1 and LOWR-2 patients were monitored
response. While these early results are promising, evidence is still post treatment, approximately 20% of patients who did not clear
lacking for the safety of NAPs for long term administration. Given the HDV RNA on 3e6 months of LNF treatment experienced
326 M. Elazar et al. / Best Practice & Research Clinical Gastroenterology 31 (2017) 321e327

THerapeutic Alt Normalizing ChangeS (THANCS) accompanied by References


serum HDV RNA negativity. After as little as 6 months of achieving
such normalized ALT and HDV RNA negativity, regression in fibrosis [1] Rizzetto M, Canese MG, Arico S, Crivelli O, Trepo C, Bonino F, et al. Immu-
nofluorescence detection of new antigen-antibody system (delta/anti-delta)
was observed on liver biopsy [80]. associated to hepatitis B virus in liver and in serum of HBsAg carriers. Gut
1977;18:997e1003.
Type III interferon [2] 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;77:6124e8.
Although shown to have anti-HDV efficacy, PEG-IFNa has sig- [3] Rizzetto M. Hepatitis D virus: introduction and epidemiology. Cold Spring
nificant side effects, attributed in part to the fact that its receptors Harb Perspect Med 2015;5, a021576.
[4] Griffin BL, Chasovskikh S, Dritschilo A, Casey JL. Hepatitis delta antigen re-
are widely expressed throughout the body. In contrast, the re- quires a flexible quasi-double-stranded RNA structure to bind and condense
ceptors for Type-III IFNs (such as IFNl) have a more restricted hepatitis delta virus RNA in a ribonucleoprotein complex. J Virol 2014;88:
localization on specific cell types that include the liver [81]. It was 7402e11.
[5] Casey JL. Control of ADAR1 editing of hepatitis delta virus RNAs. Curr Top
therefore (successfully) hypothesized that PEG-IFNl could be active Microbiol Immunol 2012;353:123e43.
against a human hepatitis virus, such as HCV, yet with less side [6] Glenn JS, Watson JA, Havel CM, White JM. Identification of a prenylation site in
effects than PEG-IFNa. Similarly, PEG-IFNl may also be able to delta virus large antigen. Science 1992;256:1331e3.
[7] Bordier BB, Marion PL, Ohashi K, Kay MA, Greenberg HB, Casey JL, et al. A
inhibit HDV with less side effects than PEG-IFNa, and a clinical trial
prenylation inhibitor prevents production of infectious hepatitis delta virus
is under way to examine this possibility (ClinicalTrials.gov particles. J Virol 2002;76:10465e72.
NCT02765802). If successful as a monotherapy, IFNl can also be [8] Bonino F, Heermann KH, Rizzetto M, Gerlich WH. Hepatitis delta virus: protein
considered as an attractive combination therapeutic with the above composition of delta antigen and its hepatitis B virus-derived envelope. J Virol
1986;58:945e50.
mentioned investigational drugs. [9] Sureau C, Negro F. The hepatitis delta virus: replication and pathogenesis. J
Hepatol 2016;64:S102e16.
Concluding remarks [10] Chang J, Nie X, Chang HE, Han Z, Taylor J. Transcription of hepatitis delta virus
RNA by RNA polymerase II. J Virol 2008;82:1118e27.
[11] Luo GX, Chao M, Hsieh SY, Sureau C, Nishikura K, Taylor J. A specific base
HDV infections pose a major health problem world-wide. The transition occurs on replicating hepatitis delta virus RNA. J Virol 1990;64:
fast disease progression and lack of appropriate treatment results 1021e7.
[12] Glenn JS, White JM. trans-dominant inhibition of human hepatitis delta virus
in an increased risk of end stage liver complications. It is clear that genome replication. J Virol 1991;65:2357e61.
PEG-IFNa treatment is not sufficient to alleviate this problem, and [13] Chao M, Hsieh SY, Taylor J. Role of two forms of hepatitis delta virus antigen:
that new approaches for HDV management are in need. The recent evidence for a mechanism of self-limiting genome replication. J Virol 1990;64:
5066e9.
developments of new candidate therapeutics to treat HDV are very [14] Noureddin M, Gish R. Hepatitis delta: epidemiology, diagnosis and manage-
promising, suggesting that we may have new therapeutic ap- ment 36 years after discovery. Curr Gastroenterol Rep 2014;16:365.
proaches in coming years. [15] Ciancio A, Rizzetto M. Chronic hepatitis D at a standstill: where do we go from
here? Nat Rev Gastroenterol Hepatol 2014;11:68e71.
[16] Reinheimer C, Doerr HW, Berger A. Hepatitis delta: on soft paws across
Germany. Infection 2012;40:621e5.
Practice points [17] Servant-Delmas A, Le Gal F, Gallian P, Gordien E, Laperche S. Increasing
prevalence of HDV/HBV infection over 15 years in France. J Clin Virol 2014;59:
126e8.
 Hepatitis delta is the worst form of human viral hepatitis.
[18] William Tong CY, Asher R, Toby M, Ngui SL, Tettmar K, Ijaz S, et al.
 Patients with hepatitis B should be screened for hepatitis A re-assessment of the epidemiology and patient characteristics of
delta virus (HDV). hepatitis D virus infection in inner city London. J Infect 2013;66:521e7.
 HDV-infected patients should be considered for treatment [19] Crispim MA, Fraiji NA, Campello SC, Schriefer NA, Stefani MM, Kiesslich D.
Molecular epidemiology of hepatitis B and hepatitis delta viruses circulating
with interferon alpha or experimental therapies. in the Western Amazon region, North Brazil. BMC Infect Dis 2014;14:94.
[20] Lunel-Fabiani F, Mansour W, Amar AO, Aye M, Le Gal F, Malick FZ, et al. Impact
of hepatitis B and delta virus co-infection on liver disease in Mauritania: a
cross sectional study. J Infect 2013;67:448e57.
[21] Andernach IE, Leiss LV, Tarnagda ZS, Tahita MC, Otegbayo JA, Forbi JC, et al.
Characterization of hepatitis delta virus in sub-Saharan Africa. J Clin Microbiol.
2014;52:1629e36.
Research agenda [22] Makuwa M, Mintsa-Ndong A, Souquiere S, Nkoghe D, Leroy EM, Kazanji M.
Prevalence and molecular diversity of hepatitis B virus and hepatitis delta
 Updated prevalences of HDV co-infection should be virus in urban and rural populations in northern Gabon in central Africa. J Clin
Microbiol. 2009;47:2265e8.
determined in most countries. [23] Rizzetto M, Ciancio A. Epidemiology of hepatitis D. Semin Liver Dis 2012;32:
 Promising new therapies in development need to be 211e9.
proven safe and effective and are likely to change man- [24] Chang SY, Yang CL, Ko WS, Liu WC, Lin CY, Wu CH, et al. Molecular epide-
miology of hepatitis D virus infection among injecting drug users with and
agement for this serious unmet medical need. without human immunodeficiency virus infection in Taiwan. J Clin Microbiol
 Detailed studies focusing on the natural long-term history 2011;49:1083e9.
outcomes for HDV-infected patients can help highlight the [25] Takahashi M, Nishizawa T, Gotanda Y, Tsuda F, Komatsu F, Kawabata T, et al.
High prevalence of antibodies to hepatitis A and E viruses and viremia of
danger of not treating patients. hepatitis B, C, and D viruses among apparently healthy populations in
Mongolia. Clin Diagn Lab Immunol 2004;11:392e8.
[26] Oyunsuren T, Kurbanov F, Tanaka Y, Elkady A, Sanduijav R, Khajidsuren O,
et al. High frequency of hepatocellular carcinoma in Mongolia; association
with mono-, or co-infection with hepatitis C, B, and delta viruses. J Med Virol
Conflict of interest
2006;78:1688e95.
[27] Caredda F, Rossi E, d'Arminio Monforte A, Zampini L, Re T, Meroni B, et al.
Elazar: Equity interest in Eiger BioPharmaceuticals, Inc.; Koh: Hepatitis B virus-associated coinfection and superinfection with delta agent:
none; Glenn: Equity interest in Eiger BioPharmaceuticals, Inc. indistinguishable disease with different outcome. J Infect Dis 1985;151:
925e8.
[28] Negro F. Hepatitis D virus coinfection and superinfection. Cold Spring Harb
Acknowledgement Perspect Med 2014;4, a021550.
[29] Rizzetto M. Hepatitis D: virology, clinical and epidemiological aspects. Acta
Gastroenterol Belg 2000;63:221e4.
None.
M. Elazar et al. / Best Practice & Research Clinical Gastroenterology 31 (2017) 321e327 327

[30] Yurdaydin C, Idilman R, Bozkaya H, Bozdayi AM. Natural history and treat- [58] Gripon P, Cannie I, Urban S. Efficient inhibition of hepatitis B virus infection by
ment of chronic delta hepatitis. J Viral Hepat 2010;17:749e56. acylated peptides derived from the large viral surface protein. J Virol 2005;79:
[31] Fattovich G, Giustina G, Christensen E, Pantalena M, Zagni I, Realdi G, et al. 1613e22.
Influence of hepatitis delta virus infection on morbidity and mortality in [59] Barrera A, Guerra B, Notvall L, Lanford RE. Mapping of the hepatitis B virus
compensated cirrhosis type B. The European Concerted Action on Viral Hep- pre-S1 domain involved in receptor recognition. J Virol 2005;79:9786e98.
atitis (Eurohep). Gut 2000;46:420e6. [60] Bogomolov P, Alexandrov A, Voronkova N, Macievich M, Kokina K,
[32] Kushner T, Serper M, Kaplan DE. Delta hepatitis within the Veterans Affairs Petrachenkova M, et al. Treatment of chronic hepatitis D with the entry in-
medical system in the United States: prevalence, risk factors, and outcomes. J hibitor myrcludex B: first results of a phase Ib/IIa study. J Hepatol 2016;65:
Hepatol 2015;63:586e92. 490e8.
[33] Ji J, Sundquist K, Sundquist J. A population-based study of hepatitis D virus as [61] Bogomolov P, Voronkova N, Schoeneweis K, Schwab M, Lempp FA, Haag M,
potential risk factor for hepatocellular carcinoma. J Natl Cancer Inst 2012;104: et al. A proof-of-concept Phase IIa clinical trial to treat chronic HBV/HDV with
790e2. the entry inhibitor myrcludex B. The Liver Meeting. Boston, USA: Hepatology;
[34] Romeo R, Del Ninno E, Rumi M, Russo A, Sangiovanni A, de Franchis R, et al. A 2016. p. 121A.
28-year study of the course of hepatitis Delta infection: a risk factor for [62] Vaillant A. Nucleic acid polymers: broad spectrum antiviral activity, antiviral
cirrhosis and hepatocellular carcinoma. Gastroenterology 2009;136:1629e38. mechanisms and optimization for the treatment of hepatitis B and hepatitis D
[35] Romeo R, Foglieni B, Casazza G, Spreafico M, Colombo M, Prati D. High serum infection. Antiviral Res 2016;133:32e40.
levels of HDV RNA are predictors of cirrhosis and liver cancer in patients with [63] Vaillant A, Juteau JM, Lu H, Liu S, Lackman-Smith C, Ptak R, et al. Phosphor-
chronic hepatitis delta. PLoS One 2014;9, e92062. othioate oligonucleotides inhibit human immunodeficiency virus type 1
[36] Niro GA, Smedile A, Ippolito AM, Ciancio A, Fontana R, Olivero A, et al. fusion by blocking gp41 core formation. Antimicrob Agents Chemother
Outcome of chronic delta hepatitis in Italy: a long-term cohort study. J Hep- 2006;50:1393e401.
atol 2010;53:834e40. [64] Bernstein DI, Goyette N, Cardin R, Kern ER, Boivin G, Ireland J, et al. Amphi-
[37] Buti M, Homs M, Rodriguez-Frias F, Funalleras G, Jardi R, Sauleda S, et al. pathic DNA polymers exhibit antiherpetic activity in vitro and in vivo. Anti-
Clinical outcome of acute and chronic hepatitis delta over time: a long-term microb Agents Chemother 2008;52:2727e33.
follow-up study. J Viral Hepat 2011;18:434e42. [65] Guzman EM, Cheshenko N, Shende V, Keller MJ, Goyette N, Juteau JM, et al.
[38] Farci P, Mandas A, Coiana A, Lai ME, Desmet V, Vaneyken P, et al. Treatment of Amphipathic DNA polymers are candidate vaginal microbicides and block
chronic hepatitis-D with interferon-alfa-2a. New Engl J Med 1994;330:88e94. herpes simplex virus binding, entry and viral gene expression. Antivir Ther
[39] Gunsar F, Akarca US, Ersoz G, Kobak AC, Karasu Z, Yuce G, et al. Two-year 2007;12:1147e56.
interferon therapy with or without ribavirin in chronic delta hepatitis. Antivir [66] Lee AM, Rojek JM, Gundersen A, Stroher U, Juteau JM, Vaillant A, et al. Inhi-
Ther 2005;10:721e6. bition of cellular entry of lymphocytic choriomeningitis virus by amphipathic
[40] Yurdaydin C, Bozkaya H, Karaaslan H, Onder FO, Erkan OE, Yalcin K, et al. A DNA polymers. Virology 2008;372:107e17.
pilot study of 2 years of interferon treatment in patients with chronic delta [67] Matsumura T, Hu Z, Kato T, Dreux M, Zhang YY, Imamura M, et al. Amphi-
hepatitis. J Viral Hepat 2007;14:812e6. pathic DNA polymers inhibit hepatitis C virus infection by blocking viral entry.
[41] Farci P, Roskams T, Chessa L, Peddis G, Mazzoleni AP, Scioscia R, et al. Long- Gastroenterology 2009;137:673e81.
term benefit of interferon alpha therapy of chronic hepatitis D: regression of [68] Noordeen F, Vaillant A, Jilbert AR. Nucleic acid polymers inhibit duck hepatitis
advanced hepatic fibrosis. Gastroenterology 2004;126:1740e9. B virus infection in vitro. Antimicrob Agents Chemother 2013;57:5291e8.
[42] Castelnau C, Le Gal F, Ripault MP, Gordien E, Martinot-Peignoux M, Boyer N, [69] Noordeen F, Scougall CA, Grosse A, Qiao Q, Ajilian BB, Reaiche-Miller G, et al.
et al. Efficacy of peginterferon alpha-2b in chronic hepatitis delta: relevance of Therapeutic antiviral effect of the nucleic acid polymer REP 2055 against
quantitative RT-PCR for follow-up. Hepatology 2006;44:728e35. persistent duck hepatitis B virus infection. PLoS One 2015;10, e0140909.
[43] Heller T, Rotman Y, Koh C, Clark S, Haynes-Williams V, Chang R, et al. [70] Al-Mahtab M, Bazinet M, Vaillant A. Safety and efficacy of nucleic acid poly-
Long-term therapy of chronic delta hepatitis with peginterferon alfa. Aliment mers in monotherapy and combined with immunotherapy in treatment-
Pharmacol Ther 2014;40:93e104. Naive Bangladeshi patients with HBeAg+ chronic hepatitis B infection. PLoS
[44] Niro GA, Ciancio A, Gaeta GB, Smedile A, Marrone A, Olivero A, et al. Pegylated One 2016;11, e0156667.
interferon alpha-2b as monotherapy or in combination with ribavirin in [71] Bazinet M, Pantea V, Cebotarescu V, Cojuhari L, Jimbei P, Vaillant A. Significant
chronic hepatitis delta. Hepatology 2006;44:713e20. reduction of HBsAg and HDV RNA by the nucleic acid polymer REP 2139 in
[45] Wedemeyer H, Yurdaydin C, Dalekos GN, Erhardt A, Cakaloglu Y, caucasian patients with chronic HBV/HDV co-infection. EASL. Vienna, Austria:
Degertekin H, et al. Peginterferon plus adefovir versus either drug alone for J Hepatol 2015:S257e8.
hepatitis delta. N Engl J Med 2011;364:322e31. [72] Vaillant A, Pantea V, Cebotarescu V, Cojuhari L, Jimbei P, Albrecht J, et al.
[46] Heidrich B, Yurdaydin C, Kabacam G, Ratsch BA, Zachou K, Bremer B, et al. Late HBsAg and HDV RNA reduction with REP 2139-Ca and peg-INF alpha 2a in
HDV RNA relapse after peginterferon alpha-based therapy of chronic hepatitis chronic HBV/HDV infection. APASAL. Tokyo, Japan: Hepatol Int 2016. p. S54.
delta. Hepatology 2014;60:87e97. [73] Bazinet M, Pantea V, Cebotarescu V, Cojuhari L, Jimbei P, Albrecht J, et al.
[47] Wedemeyer H, Yurdaydin C, Ernst S, Caruntu FA, Curescu MG, Yalcin K, et al. Initial follow up results from the REP 301 trial: safety and efficacy of REP
Prolonged therapy of Hepatitis Delta for 96 weeks with pegylated-interferon- 2139-Ca and pegylated interferon alpha-2a in caucasian patients with chronic
a-2a plus tenofovir of placebo does not prevent HDV RNA relapse after HBV/HDV co-infection. AASLD. Boston, USA: Hepatology 2016. p. 912A.
treatment: the Hidit-2 study group. J Hepatol 2014;60:S2e3. [74] Wang Y, Cui F, Lv Y, Li C, Xu X, Deng C, et al. HBsAg and HBx knocked into the
[48] Chia JS, Wu HL, Wang HW, Chen DS, Chen PJ. Inhibition of hepatitis delta virus p21 locus causes hepatocellular carcinoma in mice. Hepatology 2004;39:
genomic ribozyme self-cleavage by aminoglycosides. J Biomed Sci 1997;4: 318e24.
208e16. [75] Einav S, Glenn JS. Prenylation inhibitors: a novel class of antiviral agents. J
[49] Rogers J, Chang AH, von Ahsen U, Schroeder R, Davies J. Inhibition of the self- Antimicrob Chemother 2003;52:883e6.
cleavage reaction of the human hepatitis delta virus ribozyme by antibiotics. J [76] Bordier BB, Ohkanda J, Liu P, Lee SY, Salazar FH, Marion PL, et al. In vivo
Mol Biol 1996;259:916e25. antiviral efficacy of prenylation inhibitors against hepatitis delta virus. J Clin
[50] Kabacam G, Onder FO, Yakut M, Seven G, Karatayli SC, Karatayli E, et al. Invest 2003;112:407e14.
Entecavir treatment of chronic hepatitis D. Clin Infect Dis 2012;55:645e50. [77] Koh C, Canini L, Dahari H, Zhao X, Uprichard SL, Haynes-Williams V, et al. Oral
[51] Yurdaydin C, Bozkaya H, Onder FO, Senturk H, Karaaslan H, Akdogan M, et al. prenylation inhibition with lonafarnib in chronic hepatitis D infection: a
Treatment of chronic delta hepatitis with lamivudine vs lamivudine + proof-of-concept randomised, double-blind, placebo-controlled phase 2A
interferon vs interferon. J Viral Hepat 2008;15:314e21. trial. Lancet Infect Dis 2015;15:1167e74.
[52] Lau DT, Doo E, Park Y, Kleiner DE, Schmid P, Kuhns MC, et al. Lamivudine for [78] Yurdaydin C, Idilman R, Choong I, Kalkan C, Keskin O, Karakaya MF, et al.
chronic delta hepatitis. Hepatology 1999;30:546e9. Optimizing the prenylation inhibitor lonafarnib using ritonavir boosting in
[53] Niro GA, Ciancio A, Tillman HL, Lagget M, Olivero A, Perri F, et al. Lamivudine patients with chronic delta hepatitis. The International Liver Congress. Vienna,
therapy in chronic delta hepatitis: a multicentre randomized-controlled pilot Austria: J Hepatol 2015. p. S252.
study. Aliment Pharmacol Ther 2005;22:227e32. [79] Wedemeyer H, Port K, Deterding K, Wranke A, Kirschner J, Martins EB, et al. A
[54] Chang J, Taylor JM. Susceptibility of human hepatitis delta virus RNAs to small phase 2 study of titrating-dose lonafarnib plus ritonavir in patients with
interfering RNA action. J Virol 2003;77:9728e31. chronic hepatitis D: interim results from the lonafarnib with ritonavir in
[55] Yan H, Zhong G, Xu G, He W, Jing Z, Gao Z, et al. Sodium taurocholate HDV-4 (LOWR HDV-4) study. The Liver Meeting. Boston, MA: Hepatology;
cotransporting polypeptide is a functional receptor for human hepatitis B and 2016. p. 121A.
D virus. Elife 2012;1, e00049. [80] Yurdaydin C, Idilman R, Kalkan R, Karakaya F, Kartal AC, Keskin O, et al. The
[56] Engelke M, Mills K, Seitz S, Simon P, Gripon P, Schnolzer M, et al. Charac- prenylation inhibitor lonafarnib can induce post-treatment Alt Flares with
terization of a hepatitis B and hepatitis delta virus receptor binding site. viral clearance in patients with chronic delta hepatitis. The Liver Meeting.
Hepatology 2006;43:750e60. Boston, MA: Hepatology; 2016. p. 927A.
[57] Lutgehetmann M, Mancke LV, Volz T, Helbig M, Allweiss L, Bornscheuer T, [81] Lasfar A, Abushahba W, Balan M, Cohen-Solal KA. Interferon lambda: a new
et al. Humanized chimeric uPA mouse model for the study of hepatitis B and D sword in cancer immunotherapy. Clin Dev Immunol 2011;2011:349575.
virus interactions and preclinical drug evaluation. Hepatology 2012;55:
685e94.

You might also like