Sunday, January 6, 2013

New therapeutic strategies in HCV: polymerase inhibitors

Review Article
Liver International
New therapeutic strategies in HCV: polymerase inhibitors
 
Ludmila Gerber, Tania M. Welzel, Stefan Zeuzem*
 
Article first published online: 3 JAN 2013
 
 
 
DOI: 10.1111/liv.12068
Gerber, L., Welzel, T. M. and Zeuzem, S. (2013), New therapeutic strategies in HCV: polymerase inhibitors. Liver International, 33: 85–92. doi: 10.1111/liv.12068

Author Information
  1. Klinikum der J.W. Goethe Universität, Frankfurt am Main, Germany
  2.  Correspondence Stefan Zeuzem, MD, Professor of Medicine, Chief \x96 Department of Medicine I, J.W. Goethe University Hospital, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
    Tel: +49 (0)69 6301 6899 or 4544
    Fax: +49 (0)69 6301 6448
antiviral therapy;
hepatitis C;
non-nucleoside polymerase inhibitors;
nucleoside polymerase inhibitors

Abstract
The characterization of the viral life cycle facilitated the development of directly acting antiviral drugs. Among those, several inhibitors of the viral RNA-dependent RNA polymerase have proven effectiveness in clinical trials. The characteristics of different nucleos(t)ide and non-nucleoside polymerase inhibitors, as well as their clinical applications and combinations with other classes of directly acting antiviral drugs are reviewed herein.
 
Abbreviations
DAA directly acting antiviral
eRVR extended rapid virological response
HCV hepatitis C virus
HCC hepatocellular carcinoma
PEG-IFN pegylated interferon
PI protease inhibitors
RVR rapid viral response
RBV ribavirin
SOC standard of care
SVR sustained virological response
 
According to the World Health Organization (WHO), about 3% of the world's population has been infected with the hepatitis C virus (HCV). Of those, approximately 170 million are chronic HCV carriers at risk of developing cirrhosis and hepatocellular carcinoma (HCC) contributing to a large percentage of liver transplantations in Europe and the United States [1].
 
For almost a decade, the combination of pegylated interferon (PEG-IFN) and ribavirin (RBV) has been the standard of care (SOC) producing sustained virological response (SVR) rates of ~50% in treatment-naive patients infected with HCV genotype 1 and 70–90% in patients infected with HCV genotypes 2 and 3 [2]. Recently, this therapeutic backbone has been supplemented by the addition of first- generation protease inhibitors (Telaprevir, Boceprevir) directly targeting the viral NS3/4A protease. This ‘triple therapy’ improved SVR rates to 70–80% in treatment-naive patients infected with HCV genotype 1 [3, 4].
 
The development of directly acting antiviral (DAA) HCV therapeutics was facilitated by the adaptation of HCV to a cell culture system allowing the description of the HCV lifecycle and identification of potential novel drug targets. In addition to protease inhibitors (PI), these targets include inhibitors of the non-structural NS5A enzyme, a protein possibly involved in HCV replication, and nucleoside/nucleotide analogue and non-nucleoside inhibitors of the HCV RNA-dependent RNA polymerase (RdRp/NS5B) [5-7].
 
The following sections provide an overview of novel therapeutic strategies involving nucleoside/nucleotide analogue and non-nucleoside inhibitors of the HCV RNA-dependent RNA polymerase (RdRp/NS5B).
 
Function and biological role of the Hepatitis C Virus (HCV) polymerase inhibitor
A highly structured association of RNA and viral proteins, of cellular proteins and cofactors, and of rearranged intracellular lipid membranes derived from the endoplasmic reticulum are essential for replication of the viral RNA genome [8]. The key enzyme in this process is NS5B, a RNA-dependent RNA polymerase, catalysing the synthesis of a complementary negative-strand RNA by using the positive-strand RNA genome as a template. Numerous RNA strands of positive polarity are produced by NS5B activity from this negative-strand RNA and serve as templates for further replication and polyprotein translation [9].
 
Schematically, the NS5B protein has the shape of a right hand with an active site located within the palm domain and encircled of a thumb and a finger domain (Fig. 1). The thumb domains act as regulator of nucleic acid binding and the catalytic efficiency of the enzyme's active site, while the palm domains coordinate the function of the active site and carry out the nucleotidyl transfer reaction [10].
 
Figure 1. NS5B polymerase structure and molecular target sites. Ribbon model of the NS5B polymerase from PDB structure 2IJN. Palm, thumb and fingers are coloured in red, green and blue respectively, with finger loops coloured in yellow. Active site with bound inhibitor (blue space-filling model) and non-nucleoside inhibitor (NNI) sites 1–4 are indicated.


Figure 1. NS5B polymerase structure and molecular target sites. Ribbon model of the NS5B polymerase from PDB structure 2IJN. Palm, thumb and fingers are coloured in red, green and blue respectively, with finger loops coloured in yellow. Active site with bound inhibitor (blue space-filling model) and non-nucleoside inhibitor (NNI) sites 1–4 are indicated.

Two classes of NS5B inhibitors have been developed: nucleoside/nucleotide and non-nucleoside polymerase inhibitors.

Nucleoside/nucleotide analogue polymerase inhibitors
Nucleoside/nucleotide analogues act as natural polymerase substrates leading to termination of RNA chain elongation by inhibition of the active site of the HCV RdRp.
Mostly synthetic prodrugs of nucleotides are administered to facilitate resorption, and additional steps of intracellular phosphorylation are required to gain full functional activity as a nucleosid triphosphate.
 
Because of high conservation of the active site of the HCV RdRp across all HCV genotypes, these drugs have pan-genotype equivalent antiviral activity in vitro. In vivo data have not yet been completed. In vitro, nucleosidic inhibitors display a low ‘genetic barrier to resistance’ as drug resistance can be observed with single amino acid substitutions. However, because of the poor fitness of resistant variants in the presence of nucleosidic inhibitors, these agents are considered to have a high overall ‘barrier’ to resistance.

Non-nucleoside polymerase inhibitors
Non-nucleoside inhibitors bind outside the active site and target allosteric sites on the surface of the enzyme, downregulating the RdRp activity through induction of conformational changes (Fig.1).
Non-nucleoside inhibitors are so far specific for HCV genotype 1. The efficacy against genotype 1 subtypes, however, may differ. The barrier to resistance of non-nucleoside inhibitors is considered to be low.
 
Four groups of non-nucleoside inhibitors have entered clinical development
(summarized in Table 1).

Table 1. Polymerase inhibitors in clinical development
Efficiency / log decline (log10 IU/ml)
Generic namePhase of developmentMonotherapyCombinationComment
Nucleosidic inhibitors
NM-283 (Idenix)ValopicitabinPhase Idiscontinued0.87 (15 d)3.90-4.56 (IFN)Gastrointestinal side effects
R-1626 (Roche) Phase IIdiscontinued2.6-3.7 (14 d) 5.2 (IFN/R)Neutropenia, lymphopenia, neurotoxicity
INX-189/BMS-986094 (Inhibitex/BMS) Phase IIIdiscontinued0.6-4.25 (7 d)0.75-3.79 (R/7 d)Liver and severe cardiac & renal toxicity
PSI-938/GS-938 (Pharmasset/Gilead) Phase IIIdiscontinued4.8-5.8 (14 d) Hepatotoxicity
Alios-2200 (Alios) Phase Iongoing4.54 (7 d)n.a.
Alios-2158 (Alios) Phase Iongoingn.a.n.a.
R-7128/RG-7128 (Roche)MericitabinePhase IIongoing2.7 (14 d)5.00
PSI-7977/GS-7977 (Pharmasset/Gilead)SofosbuvirPhase IIIongoing4.7 (7 d)6.4 (IFN/R)
Non-nucleosidic inhibitors
Thumb I inhibitors
BILB-1941 (Boehringer Ingelheim) Phase Idiscontinued> 1 (5 d)/gastro-intestinal side effects
MK-3281 (Merk) Phase Idiscontinued3.75 (7 d)/gastro-intestinal side effects
BI-207127 (Boehringer Ingelheim) Phase IIongoing0.6-3.1 (5 d)5.6 (IFN/R)
Thumb II inhibitors
VX-759 (Vertex) Phase Idiscontinued2.5 (10 d)3.7 (VX222)low anti-viral efficacy
VX-916 (Vertex) Phase Idiscontinued1.5 (3 d)/low anti-viral efficacy
GS-9669 (Gilead) Phase Iongoing3.5 (3 d)/
PF-00868554 (Pfizer)FilibuvirPhase IIongoing0.68-2.13 (8 d)3.44-4.43 (IFN/R)
VX-222 (Vertex) Phase IIongoing3.1-3.4 (3 d)ongoing
Palm I inhibitors
ABT-333 (Abbott) Phase Iongoing0.7-1.5 (2 d)3.5-4.0 (IFN/R)
ABT-072 (Abbott) Phase Iongoing1.19-2-3 (2 d)(ABT-450/-333)
ANA-598 (Roche)SetrobuvirPhase IIongoing2.4-2.9 (3 d)outstanding
Palm II inhibitors
HCV-796 (ViroPharma/Wyeth)NesbuvirPhase Idiscontinued1.4 (14 d)/hepatotoxicity
IDX-375 (Idenix) Phase Idiscontinued0.5-1.1 (1 d)/hepatotoxicity
GS-9190 (Gilead)TegobuvirPhase IIongoing1.4-1.7 (8 d)5.7 (9256/IFN/RBV)


Thumb I inhibitors (benzimidazole site): MK-3281 (Merck, Rahway, New Jersey, USA), B ILB1941 and BI 207127 (Boehringer-Ingelheim, Ingelheim am Rhein, Germany) are thumb I site inhibitors. Pre-existing NS5B substitutions known to reduce sensitivity were identified e.g. for BILB1941 at positions V494I/A, I424V and P496A [11].
 
Thumb II inhibitors (thiophene site): filibuvir (PF-00868554/Pfizer, New York, USA), VX-759, VX-916 and VX-222 (Vertex, Cambridge, Massachusetts, USA) bind to the thumb II site. Variants at position M423 known to confer resistance to filibuvir from in vitro studies were also selected in most patients in vivo. For VCH-759 as well as VCH-916, viral breakthroughs with selection of resistant variants conferring high (M423T/V/I) and medium (L419V/M, I482L/V/T, V494A/I) levels of resistance were described [11].
 
Palm I inhibitors (benzothiadiazine site): ABT-333, ABT-072 (both Abbott, Abbott Park, Illinois, USA) and Setrobuvir (ANA598; Anadys/Roche, Basel, Switzerland) are inhibitors of the palm I site. Several variants (M414T, G554D, D559G) were described from in vitro replicon studies to confer resistance to ANA598. Mutations associated with resistance in patients treated with ABT-333 were observed at positions C316Y and Y448C [11].
 
Palm II inhibitors (benzofuran site): nesbuvir (HCV-796/ViroPharma/Wyeth, Exton, Pennsylvania, USA) and IDX-375 (Idenix, Cambridge, Massachusetts, USA) act at the palm II site. The C316Y mutation in NS5B is associated with resistance to HCV-796 in vitro and in vivo [11].

Polymerase inhibitors in clinical trials
Nucleoside and non-nucleoside inhibitors have been investigated in combination with PEG-IFN and RBV and in PEG-IFN-free therapeutic regimens.

Nucleos(t)ide inhibitors
Several drugs in clinical development were discontinued because of clinically significant side effects including valopicitabine (NM-283), R-1626, PSI-938 and BMS-986094 (formerly known as INX-189) (Table 1).
 
Promising clinical data have been presented for the following HCV nucleos(t)ide analogues that have entered phase 2/3 clinical testing: mericitabine (RG-7128; prodrug of the pyrimidine (cytosine) nucleoside analogue PSI-6130) and sofosbuvir (PSI-7977/GS-7977; chirally pure isomer form of PSI-7851; prodrug of the nucleotide pyrimidine (uridine) analogue GS-7411). Early results were recently reported for ALS-2200, a novel nucleotide pyrimidine analogue polymerase inhibitor with a median reduction in HCV-RNA of 4.54 log10 after 7 days of dosing [12].

Nucleos(t)ide inhibitors in clinical trials with interferon

Mericitabine (RG 7128)
The PROPEL study investigated safety, tolerability and efficacy of 8 and 12 weeks of mericitabine (1000 mg BID) in combination with PEG-IFN/RBV in 408 treatment-naive patients with chronic HCV genotype 1 or 4 infection. The rapid viral response (RVR) rates were up to 62% in the treatment arms with mericitabine compared with 18% in the control arm with PEG-IFN/RBV alone. SVR rates, however, did not differ between patients receiving mericitabine in combination with PEG-IFN/RBV and patients who received placebo plus PEG-IFN/RBV for 48 weeks. In the response-guided arms, fewer patients achieved SVR (33–49%) because of high virological relapse rates [13].
 
The JUMP-C trial (phase 2) investigated the safety and efficacy of 24 weeks of response-guided therapy with mericitabine (1000 mg BID), PEG-IFN/RBV in 168 treatment-naive patients with HCV genotype 1 or 4 infection. Patients were randomized (1:1) to response-guided therapy with mericitabine plus PEG-IFN and RBV for 24 or 48 weeks or to placebo, PEG-IFN/RBV for 48 weeks. In the mericitabine treatment arm, therapy was stopped at week 24 in patients with an extended rapid virological response [eRVR], defined as undetectable HCV RNA (<15 IU/ml) from week 4 to 22. Patients without eRVR received PEG-IFN/RBV for another 24 weeks. Overall SVR rates were higher in patients treated with mericitabine plus PEG-IFN/RBV than in patients treated with PEG-IFN/RBV alone (58% vs. 36%). Among 49 patients (60%) who achieved an eRVR with mericitabine plus PEG-IFN/RBV and discontinued therapy at week 24, the SVR rate was 78%. Combination therapy with mericitabine was safe, well-tolerated, demonstrated a high resistance barrier and a low potential for pharmacokinetic drug–drug interactions [14].
 
The MATTERHORN study, another phase 2 trial, is an ongoing study evaluating the efficacy and safety of various combinations of mericitabine and ritonavir-boosted danoprevir and PEG-IFN/RBV in patients with HCV genotype 1 infection.
 
The ongoing phase 2 DYNAMO I and II studies investigate quadruple therapy with mericitabine in combination with the protease inhibitors boceprevir or telaprevir and PEG-IFN/RBV in HCV genotype 1 non-responders to PEG-IFN/RBV.

Sofosbuvir (GS-7977, formerly PSI-7977)
The combination of sofosbuvir (400 mg QD), PEG-IFN and RBV was assessed for 12 weeks in 25 treatment-naive patients infected with HCV genotypes 2/3 in an open-label, uncontrolled pilot study (PROTON). Of the patients, who completed therapy (n = 24), 100% achieved an SVR. Based on these favourable results, the PROTON study was expanded to treatment-naive patients infected with HCV genotype 1. Patients were randomized 2:2:1 to different dose groups of sofosbuvir (200 or 400 mg QD) for 12 weeks plus PEG-IFN/RBV for 24 weeks. Patients without RVR were continued on PEG-IFN/RBV through week 48. Reported SVR rates were 88, 91% and <50% for sofosbuvir 200, 400 mg and the control group respectively [15, 16].
 
The ELECTRON study investigated sofosbuvir (400 mg QD) and RBV for 12 weeks in combination with 0, 4, 8 or 12 weeks PEG-IFN. All patients (n = 10 per group) achieved an SVR after 12 weeks of therapy [17].
 
The ATOMIC study, a Phase 2 randomized open-label trial investigated sofosbuvir in combination with PEG-IFN/RBV in 316 non-cirrhotic HCV genotype 1, 4 and 6 patients. Patients infected with HCV genotype 1 were randomized to 12 or 24 weeks sofosbuvir, PEG-IFN/RBV, or 12 weeks of the triple combination followed by re-randomization (1:1) to receive additional 12 weeks of either sofosbuvir alone or sofosbuvir plus RBV. Also, 16 patients infected with HCV genotypes 4 and 6 were randomized to the 24-week regimen of sofosbuvir plus PEG-IFN/RBV. Results of an interim analysis showed SVR rates of 90% in patients who received 12 weeks of the triple combination [18].
The phase III trial NEUTRINO, a single-arm study, evaluating a 12-week course of sofosbuvir plus PEG-IFN/RBV in 300 patients infected with HCV genotypes 1, 4, 5 and 6 is currently ongoing.

Nucleos(t)ide inhibitors in clinical trials without interferon
Nucleos(t)ide analogues were also investigated in combination with RBV, NS3/4A protease inhibitors, NS5A inhibitors and/or other nucleotide inhibitors in IFN-free treatment regimens.

Mericitabine (RG 7128)
The INFORM-1 study provided first proof of principle that suppression of HCV RNA with an interferon-free combination of mericitabine and danoprevir (NS3/4 protease inhibitor) is effective. Different doses of mericitabine in combination with danoprevir were administered for 14 days to patients infected with HCV genotype 1. At the highest combination doses tested (1000 mg mericitabine and 900 mg danoprevir BID), the median change in HCV RNA concentration from baseline to day 14 was −5.1 log(10) IU/ml in treatment-naive patients and −4.9 log(10) IU/ml in previous null-responders to PEG-IFN/RBV therapy[19].
 
INFORM-SVR, a phase 2b trial, subsequently investigated a 12 or 24 week interferon-free regimen of ritonavir-boosted danoprevir (DNV/r, 100/100 mg), and mericitabine (1000 mg BID) with or without RBV in treatment-naive HCV genotype 1 infected patients. Patients with undetectable HCV RNA at week 2 and week 10 were re-randomized at week 12 to stop therapy or to continue until week 24. Rapid viral response was similar and comparable in both arms (91 and 93% respectively). Owing to high relapse rates, however, the 12-week treatment arm was prematurely stopped. Furthermore, patients randomized to the RBV-free group were offered to continue on PEG-IFN/RBV therapy because of insufficient SVR rates. The data show SVR in 71% of HCV genotype 1b patients, but only in 26% of the genotype 1a-infected patients who received 24 weeks of DNV/r, mericitabine and RBV treatment. Higher SVR rates were reported among patients who were rapid virological responders. Among patients with undetected HCV-RNA at week 2, 80% with genotype 1b and 31% with genotype 1a achieved SVR. IL28B genotype appeared to have less impact on SVR rates.

Breakthrough rates were higher among patients not receiving RBV and showed in all patients, the selection of danoprevir-resistant variants, while only one patient showed the NS5B S282T polymerase mutation associated with resistance to mericitabine. Neutropenia or treatment emergent liver toxicity was not observed [20].
 
The currently ongoing phase II study ANNAPURNA evaluates the safety, tolerability, and efficacy of combination treatment with mericitabine (1000 mg BID), ritonavir-boosted danoprevir (DNV/r 100/100 mg), setrobuvir (ANA-598) and RBV administered for 12–14 or 24–26 weeks to treatment-naive patients or for 24–26 weeks to non-responders to PEG-IFN/RBV therapy with chronic hepatitis C genotype 1 infection.

Sofosbuvir (GS-7977/PSI-7977)
In the ELECTRON trial, the interferon-free combination of sofosbuvir and RBV achieved 100% SVR rates in treatment-naive patients infected with HCV genotype 2/3. In subsequent cohorts, patients with HCV genotype 1 infection and prior non-response to PEG-IFN/RBV received sofosbuvir and RBV for 12 weeks, but showed high relapse rates after the end of 12 weeks of treatment (89%). Reported SVR4 rates in treatment-naive patients infected with genotype 1 or treatment-experienced patients infected with genotype 2/3 receiving sofosbuvir and RBV for 12 weeks were 88 and 80% respectively [17].
 
The QUANTUM study, a phase 2b study, was planned as the first interferon-free, pan-genotypic, all-nucleotide study, combining a pyrimidine and a purine analogue, sofosbuvir and PSI-938 with or without RBV for 12 weeks. However, PSI-938 had to be discontinued because of liver toxicity. An SVR rate of 59% was reported for 17 HCV genotype 1 patients [21].
 
Three ongoing US studies for patients with HCV genotype 2/3 infection are FISSION, POSITRON, and FUSION. FISSION will compare 12 weeks of treatment with sofosbuvir and RBV to 24 weeks of PEG-IFN/RBV therapy. POSITRON evaluates the same combination for 12 weeks in interferon ineligible or intolerant patients. FUSION will examine treatment-experienced patients exploring 12 or 16 weeks of treatment with sofosbuvir and RBV.
 
Furthermore, a phase 3 study (VALENCE) was recently started to investigate the combination of sofosbuvir and RBV for 12 weeks in treatment-naive and treatment-experienced patients with HCV genotype 2 and 3 infection also in Europe.
 
Different oral combinations of sofosbuvir (400 mg QD) plus daclatasvir (BMS-790052, 60 mg QD) with/without RBV were tested in an open-label Phase 2a pharmaceutical cross-collaboration in treatment-naive non-cirrhotic HCV genotype 1-, 2- and 3-infected patients. SVR rates were 88, 100, and 79% respectively [22]. Furthermore, a combination study of simeprevir (TMC435) plus sofosbuvir is ongoing and a phase III study, investigating the combination of sofosbuvir and GS-5885 (NS5A inhibitor), is currently in preparation.

Non-nucleoside inhibitors
Several non-nucleoside inhibitors were discontinued from clinical development for different reasons. BILB1941 and MK-3281 were discontinued because of gastrointestinal side events [23-25], VX-759 and VX-916 showed only low-to-medium antiviral activity [26, 27], nesbuvir (HCV-796) and IDX-375 showed elevation of liver enzymes [27].
 
Currently, BI 207127, VX-222, ABT-072, ABT-333, setrobuvir (ANA-598), filibuvir and tegobuvir (GS-9190) are still investigated in clinical trials.

Non-nucleoside inhibitors in clinical trials with interferon

BI 207127
The SOUND C1 trial evaluated the combination of the non-nucleoside polymerase inhibitor BI 207127 with the protease inhibitor BI 201335 and RBV for 4 weeks followed by BI 201335 and PEG-IFN/RBV in treatment-naive patients with HCV genotype 1 infection. The RVR rates ranged from 73% (genotype 1a) to 100% (genotype 1b). An SVR rate of 94% was achieved in the group receiving a higher dose of BI 207127 (600 mg; TID) [28].

VX-222
The Phase 2 ZENITH study was designed to evaluate the antiviral activity of the protease inhibitor telaprevir and two dose levels of VX-222 administered with or without PEG-IFN/RBV for 12 or 24 weeks in treatment-naive patients with genotype 1 infection. This quad regimen led to SVR rates of 82–93% [29].

Setrobuvir (ANA-598)
Setrobuvir was studied at different doses in combination with PEG-IFN and RBV in treatment-naive and treatment-experienced patients with HCV genotype 1. Both doses of setrobuvir demonstrated a favourable safety and tolerability profile through 12 weeks. SVR was achieved in 8 of 11 patients (73%) who received 24 weeks of treatment [30].

Tegobuvir (GS-9190)
The antiviral activity of tegobuvir and the NS3/4A protease inhibitor GS-9256 alone or in combination with RBV or PEG-IFN/RBV in treatment-naive patients with HCV genotype 1 was assessed in a phase II trial. Patients were randomized to 28 days of tegobuvir/GS-9256, tegobuvir/GS-9256/RBV or tegobuvir/GS-9256/Peg-IFN/RBV, followed by PEG-IFN/RBV for further 44 weeks. Rapid viral response was observed in 100% (14 of 14) of patients receiving tegobuvir/GS-9256/PEG-IFN/RBV [31]. Response-guided quadruple therapy with tegobuvir and GS-9256 plus PEG-IFN/RBV based on a virological response at week 2 enabled to shorten antiviral therapy to 16 weeks and yielded an SVR rate of 95% [32].
 
Because of pancytopenia in quadruple therapy combinations including tegobuvir, the polymerase inhibitors GS-9256 or GS-9451, Peg-IFN and RBV, dosing of tegobuvir was discontinued [33].

Non-nucleoside inhibitors in clinical trials without interferon

BI 207127
SOUND-C2 (n = 362), an open-label, randomized, phase IIb trial investigated safety and efficacy of the combination faldaprevir (BI 201335; protease inhibitor), BI 207127 with or without RBV for 16, 28, 40 weeks in genotype 1-infected treatment-naive patients, including 10% of patients with compensated cirrhosis. Patients were randomized to one of five treatment arms: (A) 120mg QD BI 201335 (1335QD) plus 600mg TID BI 207127 (7127TID) and RBV for 16 weeks, (B) 28 weeks, or (C) 40 weeks; (D) 1335QD plus 600mg BID BI 207127 (7127BID) and RBV for 28 weeks; (E) 1335 plus 7127TID (no RBV) for 28 weeks. Randomization to Arm E was stopped prematurely according to FDA feedback on study design prior to data analysis. The RBV-sparing arm showed substantial but lower response rates than other arms of the trial.
 
This combination therapy achieved a SVR rate of 60% in treatment-naive patients with genotype 1 after 16 weeks of treatment, confirming the potent antiviral activity of this treatment regimen. In patients with liver cirrhosis, an SVR rate of 60% in HCV genotype 1a and up to 83% in HCV genotype 1b-infected patients was reported. The BID (for BI 207127) regimen demonstrated the most favourable safety and tolerability profile with a low rate of adverse event-related study discontinuations. Post-hoc subanalyses by IL28B genotype showed clinically important differences in SVR rates: HCV 1a (IL28B: non-CC) 32% vs. HCV 1a (IL28B: CC) 75% and HCV 1b (IL28B: CC and non-CC) 82–84% [34].

ABT-072 and ABT-333
In the PILOT trial, 11 treatment-naive, non-cirrhotic HCV genotype 1 infected patients expressing the IL28B CC genotype were treated with ABT-450/r (150/100 mg QD), ABT-072 (400 mg QD) and RBV for 12 weeks. The SVR rate was 91%.
 
Another study (Co-Pilot) investigated ABT-450/r in combination with ABT-333 and RBV in 33 treatment-naive HCV genotype-1 infected patients and 17 non-responders to previous PEG-IFN/RBV therapy. Therapy-naive patients were treated with two different doses of ABT-450/r (250/100 mg and 150/100 mg, Arm 1 and 2 respectively) while prior non-responders received open-label ABT-450/r (150/100 mg, Arm 3). ABT-333 was administered in a fixed dose of 400 mg QD across groups. SVR rates in treatment-naïve patients were independent of ABT-450/r dose and IL28B genotype (95 and 93% for arm 1 and 2 respectively. In previous non-responders (Arm 3), the SVR rate was 47% [35].

VX-222
The phase 2 ZENITH study was designed to evaluate the antiviral activity of the protease inhibitor telaprevir and VX-222 in a dual regimen, in addition with RBV as triple or with PEG-IFN/RBV as quadruple combination therapy in treatment-naive patients with genotype 1 infection. A proportion of 38% of patients receiving 100 mg VX-222 plus telaprevir and 50% of patients receiving 400 mg VX-222 plus telaprevir were able to undergo only 12 weeks of therapy because of RVR (undetectable HCV-RNA at week 2 and 8) and achieved SVR rates of 83 and 90% respectively [29].

Tegobuvir (GS-9190)
Another interferon-free study, investigating the combination of tegobuvir, GS-5885 (NS5A inhibitor), GS-9451 (NS3/4A protease inhibitor) and RBV for 12 and 24 weeks in treatment-naive and experienced HCV genotype 1 patients demonstrated SVR rates up to 89% in patients treated for 12 weeks and up to 100% in patients treated for 24 weeks. Viral breakthrough occurred only in GT1a-infected patients [36].

Conclusion
The optimal treatment regimens for patients with chronic hepatitis C must still be defined. In many trials, nucleos(t)ide and non-nucleoside polymerase inhibitors are key components. The high resistance barrier of the nucleoside analogue inhibitors provides a potential backbone in various interferon-free treatment combinations. The non-nucleoside inhibitors have complementary resistance profiles to protease inhibitors and NS5A inhibitors and are also positioned to become relevant components in combination trials. In the near future, polymerase inhibitors as key components could be clinically available if ongoing clinical trials continue to show promising results on efficacy, safety and tolerability.

Acknowledgements
We thank Dr C. Welsch for providing Figure 1.

Disclosure
Financial disclosures: Stefan Zeuzem – Consultancies for Abbott, Achillion, AstraZeneca, BMS, Boehringer-Ingelheim, Gilead, Idenix, Janssen, Merck, Novartis, Presidio, Roche, Santaris, Vertex. Tania M. Welzel – Consultancies for Novartis. Ludmila Gerber has no disclosure.

References
  • 1
    WHO. Hepatitis C: Available at http://www.who.int/csr/disease/hepatitis/whocdscsrlyo2003/en/index3.html. Accessed: August 2012.
  • 2
    Zeuzem S, Berg T, Moeller B, et al. Expert opinion on the treatment of patients with chronic hepatitis C. J Viral Hepat 2009; 16: 7590.
  • 3
    Jacobson IM, McHutchison JG, Dusheiko G, et al. Telaprevir for previously untreated chronic hepatitis C virus infection. N Engl J Med 2011; 364: 240516.
  • 4
    Poordad F, McCone J Jr, Bacon BR, et al. Boceprevir for untreated chronic HCV genotype 1 infection. N Engl J Med 2011; 364: 1195206.
  • 5
    Choo QL, Kuo G, Weiner AJ, et al. Isolation of a cDNA clone derived from a blood-borne non-A, non-B viral hepatitis genome. Science 1989; 244: 35962.
  • 6
    Kim JL, Morgenstern KA, Dwyer MD, et al. Crystal structure of the hepatitis C virus (NS3 protease domain complexed with a synthetic NS4A cofactor peptide. Cell 1996; 87: 34355.
  • 7
    Bressanelli S, Tomei L, Incitti I, et al. Crystal structure of the RNA-dependent RNA polymerase of hepatitis C virus. Proc Nat Acad Sci 1996; 96: 130349.
  • 8
    Moradpour D, Penin F, Rice CM. Replication of hepatitis C virus. Nat Rev Microbiol 2007; 5: 45363.
  • 9
    Lesburg CA, Cable MB, Ferrari E, et al. Crystal structure of the RNA-dependent RNA polymerase from hepatitis C virus reveals a fully encircled active site. Nat Struct Biol 1999; 6: 93743.
  • 10
    Biswal BK, Cherney MM, Wang M, et al. Crystal structures of the RNA-dependent RNA polymerase genotype 2a of hepatitis C virus reveal two conformations and suggest mechanisms of inhibition by non-nucleoside inhibitors. J Biol Chem 2005; 280: 1820210.
  • 11
    Sarrazin C, Zeuzem S. Resistance to direct antiviral agents in patients with hepatitis C virus infection. Gastroenterology 2010; 138: 44762.
  • 12
    de Bruijne J, de Rooij JV, van Vliet A, et al. Phase I study in healthy volunteers and patients with IDX-375, a novel non-nucleoside HCV polymerase inhibitor. Hepatology 2010; 52(Suppl): 1219A.
  • 13
    Jensen DM, Wedemeyer H, Herring RW, et al. High rates of early viral response, promising safety profile and lack resistance-related breakthrough in HCV GT1/4 patients treated with RG7128 plus pegIFN alfa-2a (40KD)/RBV: planned week 12 interim analysis from the PROPEL study. Hepatology 2010; 52(Suppl): 3601A.
  • 14
    Pockros P, Jensen D, Tsai N, et al. SVR-12 among G1/4treatment-naïve patients receiving mericitabine in combination with peg-IFNa-2A7RBV: interim analysis from the JUMP-C study. J Hepatol 2012; 56(Suppl): 4778.
  • 15
    Lawitz E, et al. PSI-7977 PROTON AND ELECTRON: 100% concordance of SVR4 with in HCV GT1, GT2, & GT3. J Hepatol 2012; 56(Suppl): S4.
  • 16
    Lalezari J, Lawitz E, Rodriguez-Torres M, et al. Once daily PSI-7977 plus peginterferon/ribavirin in a Phase 2b trial: rapid virologic suppression in treatment-naive patients with HCV genotype 2/3. J Hepatol 2011; 54(Suppl): S28.
  • 17
    Gane EJ, et al. Once daily GS-7977 plus ribavirin in HCV genotypes 1-3: the ELECTRON. Hepatology 2012; 56(Suppl): 3067A.
  • 18
    Hassanein T, Lawitz E, et al. Once daily sofosbuvir (GS-7977) plus PEG/RBV: high early response rates are maintained during post-treatment follow-up in treatment-naïve patients with HCV genotype 1, 4, and 6 infection in the ATOMIC study. Hepatology 2012; 56(Suppl): 307A.
  • 19
    Gane EJ, Roberts SK, Stedman CA, et al. Oral combination therapy with a nucleoside polymerase inhibitor (RG7128) and danoprevir for chronic hepatitis C genotype 1 infection (INFORM-1): a randomised, double-blind, placebo-controlled, dose-escalation trial. Lancet 2010; 376: 146775.
  • 20
    Gane EJ. Interferon-free treatment with a combination of mericitabine and danoprevir/r with or without ribavirin in treatment naive HCV genotype 1 infected patients. J Hepatol 2012; 56(Suppl): 5556.
  • 21
    Gilead Announces Early Sustained Virologic Response Rates for GS-7977 Plus Ribavirin in Genotype 1 Treatment-Naive Hepatitis C Patients: Available at http://investors.gilead.com/phoenix.zhtml?c&#x00A0;=&#x00A0;69964&p=irol-newsArticle&ID=1684792&highlight. Accessed: October 2012.
  • 22
    Sulkowski M, et al. Potent viral suppression with all-oral combination of daclatasvir (NS5A inhibitor) and GS-7977 (NS5B inhibitor), +/-ribavirin, in treatment-naive patients with chronic HCV GT1, 2, or 3. J Hepatol 2012; 56(Suppl): 560.
  • 23
    Erhardt A, Deterding K, Benhamou Y, et al. Safety, pharmacokinetics and antiviral effect of BILB 1941, a novel hepatitis C virus RNA polymerase inhibitor, after 5 days oral treatment. Antivir Ther 2009; 14: 2332.
  • 24
    Larrey D, Benhamou Y, Lohse AW, et al. Safety, pharmacokinetic and antiviral effect of BI207127, a novel HCV RNA polymerase inhibitor, after 5 days oral treatment in patients with chronic hepatitis C. J Hepatol 2009; 50(Suppl): S3834.
  • 25
    Brainard DM, Anderson MS, Petry A, et al. Safety and antiviral activity of NS5B polymerase inhibitor MK-3281, in treatment-naive genotype 1a, 1b and 3 HCV-infected patients. Hepatology 2009; 50(Suppl): 1026-7A.
  • 26
    Cooper C, Lawitz EJ, Ghali P, et al. Evaluation of VCH-759 monotherapy in hepatitis C infection. J Hepatol 2009; 51: 3946.
  • 27
    Lawitz E, Cooper C, Rodriguez-Torres M, et al. Safety, tolerability and antiviral activity of VCH-916, a novel non-nucleoside HCV polymerase inhibitor in patients with chronic HCV genotype 1 infection. J Hepatol 2009; 50(Suppl): S37.
  • 28
    Zeuzem S, Asselah T, Angus PW, et al. High SVR following IFN-free treatment of chronic HCV GT1 infection for 4 weeks with HCV protease inhibitor BI 201335, polymerase inhibitor BI 207127 and ribavirin, followed by BI 201335 and PegIFN/ribavirin – the SOUND-C1 study. Hepatology 2011; 54(Suppl): 486-7A.
  • 29
    Nelson DR, Gane EJ, Jacobson IM, et al. VX-222/telaprevir in combination with peginterferon-alfa-2a and ribavirin in treatment-naive genotype 1 HCV patients treated for 12 weeks: ZENITH study, SVR12 interim analysis. Hepatology 2011; 54: 1435A.
  • 30
    Lawitz E, Rodriguez-Torres M, DeMicco M, et al. Antiviral activity of ANA598, a potent non-nucleoside polymerase inhibitor, in chronic hepatitis C patients. J Hepatol 2009; 50(Suppl): S384.
  • 31
    Zeuzem S, Buggisch P, Agarwal K, et al. The protease inhibitor, GS-9256, and non-nucleoside polymerase inhibitor tegobuvir alone, with ribavirin, or pegylated interferon plus ribavirin in hepatitis C. Hepatology 2012; 55: 74958.
  • 32
    Nelson DR, Lawitz E, Bain V, et al. High SVR12 with 16 weeks of tegobuvir and GS-9256 with peginterferon-alfa 2a and ribavirin in treatment-naive genotype 1 HCV patients. J Hepatol 2012; 56(Suppl): S67.
  • 33
    Vertex Announces Positive Results from Viral Kinetic Study of the Nucleotide Analogue ALS-2200 in People with Hepatitis C: Available at http://investors.vrtx.com/releasedetail.cfm?releaseid=696581. Accessed: October 2012.
  • 34
    Zeuzem S, Soriano V, Asselah T, et al. Virologic response to an interferon-free regimen of BI 201335 and BI 207127, with and without ribavirin, in treatment-naive patients with chronic genotype-1 HCV infection: week 12 interim results of the SOUND-C2 study. Hepatology 2011; 54(Suppl): 1436A.
  • 35
    Poordad F et al., A 12-week interferon-free regimen of ABT-450/r + ABT-333 + ribavirin achieved SVR12 in more than 90% of treatment-naïve HCV genotype-1-infected subjects and 47% of previous non-responders. J Hepatol 2012; 56(Suppl): S54950.
  • 36
    Sulkowski M, Rodriguez-Torres M, Lawitz E, et al. Complete SVR4 rates in treatment-naïve HCV genotype 1a and 1b patients who achieved vRVR with an interferon-free all-oral regimen. Hepatology 2012; 56(Suppl): 298A.
Copyright © 1999–2012 John Wiley & Sons, Inc. All Rights Reserved.

No comments:

Post a Comment