New HCV therapies on the horizon

New HCV therapies on the horizon

REVIEW 10.1111/j.1469-0691.2010.03430.x New HCV therapies on the horizon J. Vermehren and C. Sarrazin Medizinische Klinik 1, Klinikum der J. W. Goet...

187KB Sizes 3 Downloads 84 Views

REVIEW

10.1111/j.1469-0691.2010.03430.x

New HCV therapies on the horizon J. Vermehren and C. Sarrazin Medizinische Klinik 1, Klinikum der J. W. Goethe-Universita¨t, Frankfurt am Main, Germany

Abstract Improved understanding of the hepatitis C virus (HCV) life cycle has led to the discovery of numerous potential targets for antiviral therapy. HCV polyprotein processing and replication have been identified as the most promising viral targets. However, viral entry and fusion, RNA translation, virus assembly and release and several host cell factors may provide alternative attractive targets for future anti-HCV therapies. Inhibitors of the HCV NS3/4A protease are currently the most advanced in clinical development. Monotherapy with protease inhibitors has shown high antiviral activity, but is associated with frequent selection of resistant HCV variants, often resulting in viral breakthrough. However, there is encouraging evidence from phase 2/3 trials indicating that the addition of a protease inhibitor (e.g. telaprevir and boceprevir) to pegylated interferon-a/ribavirin substantially improves sustained virological response rates in both treatment-naı¨ve and treatment-experienced patients with HCV genotype 1. Nucleos(t)ide inhibitors of the HCV NS5B polymerase have shown variable antiviral activity against different HCV genotypes, but seem to have a higher genetic barrier to resistance than protease inhibitors. In addition, several allosteric binding sites have been identified for non-nucleoside inhibitors of the NS5B polymerase. However, the development of a substance with high antiviral activity and a high genetic barrier to resistance seems to be difficult. Among the different host cell-targeting compounds in early clinical development, cyclophilin inhibitors have shown the most promising results. Although advances have also been made in improving interferons, combinations of antiviral agents with different mechanisms of action may lead to the eventual possibility of interferon-free regimens. Keywords: Chronic hepatitis C, direct-acting antiviral, protease inhibitor, polymerase inhibitor, NS5A inhibitor, cyclophilin inhibitor, review Article published online: 18 November 2010 Clin Microbiol Infect 2011; 17: 122–134 Corresponding author: C. Sarrazin, Medizinische Klinik 1, Klinikum der J.W. Goethe-Universita¨t, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany E-mail: [email protected]

Introduction Chronic hepatitis C represents a major health burden, affecting approximately 170 million people worldwide [1]. With the current standard of care (SOC), a combination of pegylated interferon (PEG-IFN)-a plus weight-based ribavirin (RBV), viral eradication can be achieved in fewer than half of all patients with hepatitis C virus (HCV) genotype 1 and in approximately 80% of patients with HCV genotype 2 or 3 [2–5]. Treatment may take up to 72 weeks [6], but therapyassociated side effects such as pancytopenia, flu-like symptoms or depression are commonly observed, and may lead to early treatment discontinuation. Improved understanding of the HCV life cycle in recent years has supported the development of direct-acting

antiviral (DAA) agents that specifically target post-translational processing and HCV replication [7–10]. Inhibitors of HCV entry, HCV RNA translation and virus assembly and release are mostly still in preclinical development, whereas several host cell-targeting compounds rate among the more promising therapeutic approaches in recent anti-HCV drug discoveries. Finally, improvements have also been made in the development of new, long-acting interferons with the potential for more favourable safety and efficacy profiles. This review will give an overview of recent developments in the HCV drug pipeline. DAA agents, host cell-targeting compounds and new interferons that have already been evaluated in clinical trials will be discussed. Other treatment approaches, such as toll-like receptor agonists and therapeutic vaccines, are beyond the scope of this article.

ª2011 The Authors Clinical Microbiology and Infection ª2011 European Society of Clinical Microbiology and Infectious Diseases

Vermehren and Sarrazin

CMI

NS3/4A protease inhibitors

The NS3/4A protease has been recognized as an important target for anti-HCV therapy because of its key role within the HCV life cycle (e.g. cleavage of the genome-encoded polyprotein and inactivation of cellular proteins required for innate immunity). NS3/4A protease inhibitors have shown strong antiviral efficacy but a low genetic barrier to resistance in early clinical studies. They can be divided into two chemical classes: macrocyclic inhibitors, and linear, tetrapeptide a-ketoamide derivates (Fig. 1).

Telaprevir

The orally bioavailable, linear, ketoamide protease inhibitor telaprevir was initially investigated in a 14-day randomized, placebo-controlled monotherapy study in patients with HCV genotype 1 [12]. In this study, the greatest reduction in HCV RNA was achieved in patients who received 750 mg of telaprevir every 8 h, with a median viral load decline of

(b)

NS3 protease inhibitors

5

4.6

4.4

4.3

4.2

3.8 3.9

4

3.9

3.6

3.3

3.3

2.9

3 2.1

1.8

2 1

Nucleos(t)ide polymerase inhibitors

4

5.4 Mean or median HCV RNA decline

3

2.7 1.9

2

1

0

0.7

3.7

4 3.1

2.9

3

2.5 2.1

1.9

2

1

0.6

ily da ce on m 0 10 84 X1

Cyclophilin inhibitors 3

3

2.3

2.1

1.7

2

1.5

1

IV

*

BW kg

ic

g/

tw

m

g in in

15

m lib Si

50 0 ia

Al in

riv Al

is

po

da ily e

ce on g m

00 0 ir 1

m 0 35

SC

Y6

da ily

ily es tim

re th g

g m 3 23

72 D AZ

e

e th re

in ily da e

95

on c g m

da

da i es tim

T1 G

g m 10 0 00 52 50

BM

S-

82 43

93

ly

s pa

on ce

tie

da i

nt

ly

ily da e ic tw

g m

S79

3

ob uv Te g

T33 AB

ir 1

60 0

20

m

g

g

tw

tw

ic

ic e

e

da

da ily

ily

ily da es

m 80 0

AN

A5 98

BM

VX

-7

59

VX

-2

80 0

22

m

g

75 0

th

re

m

e

g

tw

tim

ic

e

es tim re e th g

m 00 ir 3

da ily

ily da

ily da es tim e re th g m 80 0

3.9

3.6

4

0

BI

Fi lib uv

27 71

ID

5

0

20

g

on g m 0 40 1) 85 -7 SI (P 7 97 I-7 PS

NS5A inhibitors and others

Mean or median HCV RNA decline

Mean or median HCV RNA decline

(d)

Non-nucleoside polymerase inhibitors

5

ily ce

e ic tw g m 00 15 28 71 G R

Bo

(c)

da

da

ce p Te rev la ir D pre 400 an vi op r 7 mg re 50 th vi r 2 mg ree 00 th tim TM m ree es g d Va C43 thr time ail y e ni 5 pr 20 e ti s da m e BI vi 0 m e ily BM 20 r 7 g sd S- 13 00 on aily 65 35 mg ce tw da PH 003 24 2 0 m ice ily X 17 40 g da 0 o ily G 66 m n S80 g ce 9 tw d 0 G 256 m ice aily Sg 9 20 tw da M 451 0 m ice ily Kg 4 AC 51 00 tw dail y m ic AB H 72 -1 40 g o e d T62 0 45 nc aily 5 m I e 0/ rit DX 60 g o da on 32 0 i n av 0 mg ce ly 4 ir d 20 00 twi ail c m 0/ e y 10 g o da n 0 i m ce ly g on dai ce ly da ily

ily

0

30

Mean or median HCV RNA decline

6

123

The clinical proof-of-concept for NS3/4A protease inhibitors was obtained with ciluprevir, a macrocyclic NS3/4A protease inhibitor that showed substantial antiviral activity in patients with HCV genotype 1 [11]. However, the development of ciluprevir was stopped because of serious cardiotoxicity in a monkey model.

Antivirals Targeting HCV Polyprotein Processing

(a)

New HCV therapies

FIG. 1. Antiviral activity of (a) NS3/4A protease inhibitors, (b) nucleos(t)ide analogue NS5B polymerase inhibitors, (c) non-nucleoside analogue NS5B polymerase inhibitors and (d) NS5A and other inhibitors during monotherapy for 3–14 days in patients with chronic hepatitis C, modified from [9]. No head-to-head studies were conducted. *Alinia was administered at a dose of of 500 mg twice daily for 24 weeks in genotype 4 patients only. BW, body weight; HCV, hepatitis C virus; IV, intravenous. ª2011 The Authors Clinical Microbiology and Infection ª2011 European Society of Clinical Microbiology and Infectious Diseases, CMI, 17, 122–134

124

Clinical Microbiology and Infection, Volume 17 Number 2, February 2011

4.4 log10 IU/mL HCV RNA from baseline. Viral rebound caused by selected drug-resistant mutants occurred in the majority of patients during treatment [13]. Mutations associated with clinical resistance to telaprevir were identified at four positions of the NS3 catalytic domain — V36A/M/L, T54A, R155K/M/S/T and A156S (all three conferring lowlevel to medium-level resistance) and A156T/V (conferring high-level resistance) [13–16]. In two short-term studies of telaprevir, given in combination with PEG-IFN-a-2a ± RBV for 2–4 weeks, an even more pronounced viral decline, with a reduced frequency of resistance mutations and no viral breakthrough, was observed [17,18]. Subsequently, two large phase 2b placebo-controlled trials of telaprevir in combination with PEG-IFN-a ± RBV were conducted in treatment-naı¨ve patients with HCV genotype 1 in the USA (PROVE 1; n = 250) [19] and in Europe (PROVE 2; n = 323) [20]. Overall, sustained virologic response (SVR) rates of up to 67–69% were achieved in the telaprevir-containing study arms vs. 41–46% in patients who received SOC. In PROVE 1, only patients who achieved an extended rapid virological response (eRVR) (HCV RNA negative at week 4 and thereafter) were eligible for a shortened treatment duration. The exploratory study arms with 12 weeks of telaprevir plus PEG-IFN-a ± RBV were associated with high relapse rates, especially in patients without RBV (up to 48%), indicating that 12 weeks of overall therapy was insufficient to achieve favourable treatment outcomes, and highlighting the importance of adding RBV, because of improved antiviral activity, reduced relapse rates, and a smaller number of patients with selection of resistant variants and viral breakthrough. Rates of treatment discontinuation attributable to adverse events were higher in the telaprevircontaining study arms than in patients treated with SOC alone (12–21% vs. 7–11%). Moreover, discontinuation rates increased to nearly 30% in patients who received telaprevir for 24 weeks. Thus, in subsequent studies, telaprevir dosing was restricted to a maximum of 12 weeks. For more convenient application and to increase treatment adherence, telaprevir every 12 h at a dose of 1125 mg was investigated in a small open-label phase 2 study, conducted in treatment-naı¨ve patients with HCV genotype 1 (study C208). In this study, telaprevir was also given in combination with PEG-IFN-a-2b and RBV. Equally high SVR rates were observed with telaprevir every 12 h vs. every 8 h, and with PEG-IFN-a-2b vs. PEG-IFN-2a [21]. However, differences between the two PEG-IFN formulations may only become visible in a more difficult-to-treat patient population (i.e. patients with high viral load, advanced fibrosis or prior non-response to PEG-IFN-a therapy). Telaprevir was also

CMI

investigated in patients infected with HCV genotypes 2, 3 and 4, with varying success [22,23]. The two pivotal phase 3 studies, known as ADVANCE and ILLUMINATE, were conducted in more than 1500 treatment-naı¨ve patients infected with HCV genotype 1, and showed SVR rates of 75% and 72% following triple therapy for 12 weeks followed by PEG-IFN-a-2a/RBV for an additional 12 or 36 weeks, respectively. Thus, SVR could be increased by approximately 30% as compared with control patients treated with SOC for 48 weeks (44%). Patients who received triple combination therapy for 8 weeks, followed by SOC for 16 or 40 weeks, still achieved an SVR rate of 69% (ADVANCE). Precise plans for the management of adverse events, especially the frequently observed rashes, were developed for the phase 3 studies, and rates of discontinuation attributable to adverse events were somewhat lower than in the PROVE trials (7–8% vs. 4% in patients receiving SOC) [24,25]. In the ADVANCE study, a response-guided therapy approach was applied, with a shortened treatment period of 24 weeks, in patients who achieved an eRVR, whereas all other patients were treated for 48 weeks. In the ILLUMINATE study, no benefit of a longer treatment duration (48 weeks vs. 24 weeks) in eRVR patients could be demonstrated (Fig. 2a). Together, these findings show that an eRVR was achieved in 57–65% of treatment-naı¨ve patients with HCV genotype 1. Thus, the majority of patients were eligible for shortened treatment duration, with equally high SVR rates as those obtained with the standard therapy duration of 48 weeks. Telaprevir in combination with PEG-IFN-a-2a ± RBV was also investigated in treatment-experienced HCV genotype 1 patients (PROVE 3, n = 453; and study 107, n = 117) [26]. Overall, significantly higher SVR rates were achieved in prior relapsers who received triple combination therapy than in those receiving SOC (up to 69–97% vs. 20%). Patients with prior breakthrough or non-response achieved SVR rates in the ranges of 57–86% and 38–39%, respectively, as compared with 20% and 9% when given SOC. Prior non-responders, including partial non-responders (‡2 log10 decline but detectable HCV RNA at treatment week 12) and null-responders (<2 log10 decline in HCV RNA at week 12), experienced the highest rates of viral breakthrough during triple combination therapy. A phase 3 study (REALIZE) of telaprevir evaluated the efficacy of a 12-week triple combination treatment regimen followed by 36 weeks of SOC, or a 4-week lead-in with SOC, followed by 12 weeks of triple combination therapy, followed by 32 weeks of SOC in treatment-experienced genotype 1 patients (partial non-responders, null-responders and relapsers) vs. SOC for 48 weeks. SVR rates for the three

ª2011 The Authors Clinical Microbiology and Infection ª2011 European Society of Clinical Microbiology and Infectious Diseases, CMI, 17, 122–134

Vermehren and Sarrazin

(b)

66

59

38

48 PR

PR ±

B3 2

PR

4/

PR

PR 4/

12

B4 4 PR 4/

PR 4/ PR

48

21

PR

*SVR data are shown for patients with eRVR only

RESPOND-2

63

20

48 PR

PR T1 48 2P R 44 4/

PR

T1 2

48

24 PR

PR T1 2

T1 2

PR

48

17

66

PR

44

SPRINT-2

B4 4

SVR (%)

64

66

PR

REALIZE

ILLUMINATE*

69

2 T8 4/4 PR 8 24 /4 8

T1 2

87.5

ADVANCE 75

125

Boceprevir

100 90 80 70 60 50 40 30 20 10 0

±

92

B2 4

Telaprevir

100 90 80 70 60 50 40 30 20 10 0

PR

SVR (%)

(a)

New HCV therapies

PR

CMI

FIG. 2. Sustained viral response (SVR) results of recent telaprevir (ADVANCE, ILLUMINATE and REALIZE) and boceprevir (SPRINT-2 and RESPOND-2) trials. No head-to-head studies were conducted. (a) ADVANCE and ILLUMINATE were conducted in treatment-naı¨ve patients with hepatitis C virus (HCV) genotype 1. The ADVANCE treatment arms were as follows: (i) 12 weeks of telaprevir (T) plus PEG-IFN-a-2a and ribavirin (PR) followed by PR for 12 or 36 weeks, based on treatment response at weeks 4 and 12; (ii) 8 weeks of T plus PR followed by PR for 16 or 40 weeks, based on treatment response at weeks 4 and 12; and (iii) PR alone for 48 weeks. In the ILLUMINATE trial, patients who achieved an extended rapid virological response (eRVR) were randomized to receive 12 weeks of T plus PR followed by PR for (i) 12 weeks or (ii) 24 weeks. Treatment-experienced patients with HCV genotype 1 (REALIZE) received: (i) 12 weeks of T plus PR followed by PR for 36 weeks; (ii) a 4-week PR lead-in followed by T plus PR for 12 weeks, followed by PR alone for 32 weeks; or (iii) PR alone for 48 weeks. In the REALIZE study, relapsers and non-responder (null-responder and partial non-responder) patients were enrolled. (b) The SPRINT-2 treatment arms in treatment-naı¨ve patients with HCV genotype 1 were as follows: (i) 4-week lead-in with PEG-IFN-a-2b plus ribavirin (PR) followed by 44 weeks of PR plus boceprevir (B); (ii) 4-week lead-in with PR followed by PR plus B for 24 weeks or followed by PR plus B for 24 weeks and PR for an additional 20 weeks, based on treatment response at weeks 8 and 12; and (iii) PR alone for 48 weeks. A similar treatment schedule was used in treatment-experienced genotype 1 patients (RESPOND-2): (i) 4-week lead-in with PR followed by PR plus B for 44 weeks; (ii) 4-week lead in with PR followed by PR plus B for 32 weeks or followed by PR plus B for 32 weeks and PR for an additional 12 weeks, based on treatment response at weeks 8 and 12; and (iii) PR alone for 48 weeks.

treatment arms were 64%, 66% and 17%, respectively. Prior relapsers achieved SVR rates of 83–88%, whereas prior nullresponders achieved SVR rates in the range of 29–33%. Discontinuation because of adverse events occurred in 4% of the combined telaprevir study arms and in 3% of the control group. [27]. During triple therapy with telaprevir plus PEG-IFN-a/RBV, viral breakthrough associated with the selection of resistant variants was observed in 1–5% of treatment-naı¨ve patients and in up to 25% of previous non-responders. Moreover, variants conferring resistance to NS3 protease inhibitors were also detected in patients with viral relapse after the end of treatment. The long-term significance of these resistant variants is unknown but, generally, a decline in frequency over time was observed. However, in single patients, low to medium levels of the V36 and A156 variants were observed for up to 4 years after telaprevir therapy [28]. Boceprevir

In a phase 1 study, administration of boceprevir (200 or 400 mg every 8 h), a peptidomimetic orally bioavailable

a-ketoamide HCV protease inhibitor, with or without PEGIFN-a-2b, resulted in mean maximum reductions of HCV RNA of up to 1.61 log10 and 2.88 log10 IU/mL, respectively, in treatment-experienced patients infected with HCV genotype 1 [29]. Subsequently, a number of mutations conferring boceprevir resistance at low to medium levels were discovered in vivo: V36M/A, T54A/S, V55A, R155K/T, A156S and V170A [30–33]. As for telaprevir, reduced antiviral activities of boceprevir in HCV genotype 2/3-infected patients were reported. In a phase 2 clinical trial (SPRINT-1), boceprevir (800 mg every 8 h) in combination with PEG-IFN-a-2b and RBV was investigated in treatment-naı¨ve patients with HCV genotype 1 (n = 595) [34]. Patients received either all three drugs in combination for 28 or 48 weeks or for 24 or 44 weeks after a previous 4-week lead-in phase of PEG-IFN-a-2b/RBV, or SOC for 48 weeks. SVR rates ranged from 54% for 28 weeks of triple therapy to 75% after a 4-week lead-in with PEG-IFN-a-2b/RBV followed by 44 weeks of triple therapy, as compared with 38% in the control group. Treatment discontinuations attributable to adverse events occurred in

ª2011 The Authors Clinical Microbiology and Infection ª2011 European Society of Clinical Microbiology and Infectious Diseases, CMI, 17, 122–134

126

Clinical Microbiology and Infection, Volume 17 Number 2, February 2011

9–19% of patients receiving boceprevir as compared with 8% in the control group. In patients with prior non-response to SOC therapy, the addition of boceprevir to PEG-IFN-a and RBV gave only slightly increased SVR rates as compared with standard therapy in a phase 2b study with suboptimal boceprevir/RBV dosing (14% vs. 2%) [35]. The 4-week lead-in phase with PEG-IFN-a/RBV was introduced into boceprevir phase 2 trials to decrease the probability of resistance development. During the lead-in, a viral decline of >2 log10 IU/mL was induced in the majority of treatment-naive patients, resulting in undetectable HCV RNA levels in 60–64% of patients after an additional 4 weeks of triple therapy (week 8 of overall treatment), whereas only 37–39% of patients without lead-in therapy achieved undetectable HCV RNA levels after 4 weeks of triple therapy. Numerically, lower breakthrough rates and higher SVR rates were observed in patients with a lead-in period than in those without induction therapy. The lead-in period may help in the selection of the optimal triple-therapy duration for both treatment-naı¨ve and treatment-experienced patients. However, in virological non-sustained responders treated with telaprevir-based triple therapy, no benefit of a lead-in-phase was observed [27]. In subsequent phase 3 studies, a larger number of treatment-naı¨ve (SPRINT-2; n = 1097) and treatment-experienced (RESPOND-2; n = 403; partial non-responders and relapsers only) patients with HCV genotype 1 infection were enrolled to receive a 4-week lead-in treatment with PEG-IFN-a-2b/ RBV alone, followed by either 44 weeks of triple combination therapy or a response-guided schedule with the possibility of stopping therapy at week 28 or week 36 of overall treatment duration, respectively. SVR rates were 63–66% in treatment-naı¨ve patients and 59–66% in treatment-experienced patients, as compared with 38% and 21% in the respective control groups (Fig. 2b). Treatment discontinuations attributable to adverse events occurred in 12–16% of treatment-naı¨ve patients and in 8–12% of treatment-experienced patients, as compared with 16% and 3% in the respective control groups [36,37]. Other protease inhibitors

A large number of NS3/4A protease active site inhibitors are currently in early clinical development (Table 1a): danoprevir (RG 7227/ITMN191), TMC435, vaniprevir (MK-7009), BI 201335, narlaprevir (SCH900518), BMS-650032, PHX 1766, ACH-1625, IDX320, ABT-450, MK-5172, GS9256 and GS-9451 have all been tested in phase 1 and/or phase 2 clinical trials [38–52]. Overall, these compounds exhibit high antiviral activity in HCV genotype 1 patients, and

CMI

recently reported phase 2 results showed SVR rates comparable to those achieved with telaprevir and boceprevir [48– 50]. Potential advantages of the newer antiviral compounds include improved pharmacokinetics (once-daily to twice-daily dosing), broader genotypic activity and better tolerability. In addition, ritonavir boosting is being investigated for a number of protease inhibitors, including narlaprevir and danoprevir, to reduce side effects and to enhance patient exposure to the latter agents, thereby potentially overcoming resistance and allowing less frequent dosing. The resistance profiles of all NS3 protease inhibitors currently in clinical development overlap (Table 2) [9].

Antivirals Targeting HCV Replication NS5B polymerase inhibitors

Two classes of NS5B polymerase inhibitors can be distinguished: nucleoside or nucleotide analogue inhibitors, which mimic the natural substrates of the RNA-dependent RNA polymerase and are incorporated into the elongated RNA, where they act as chain terminators; and non-nucleoside analogue inhibitors, representing a heterogeneous group of compounds that bind to different allosteric enzyme sites, resulting in a conformational protein change before the elongation complex is formed. Nucleos(t)ide inhibitors

Nucleos(t)ide inhibitors potentially show similar efficacies against all HCV genotypes, because the active centre of NS5B is a highly conserved region. The first nucleoside analogue in development was valopicitabine (NM283, 2¢-C-methylcytidine/NM107). However, valopicitabine showed only weak antiviral activity, and further drug development was stopped because of gastrointestinal side effects [53]. The nucleoside analogue R1626 (4¢-azidocytidine/PSI-6130) showed marked antiviral activity when given at high doses alone or in combination with PEG-IFN-a-2a/RBV. No viral breakthrough was reported for either monotherapy or combination therapy. However, further development was stopped because of severe adverse events, including severe lymphocytopenia with lethal outcomes [54,55]. Among the nucleos(t)ide analogues still in development, RG 7128 is the most advanced. Week 12 interim results of a phase 2 trial of RG 7128 in combination with PEG-IFN-a-2a/ RBV in treatment-naı¨ve genotype 1 or 4 patients have shown early virological response rates of >80% and no resistancerelated viral breakthrough [56]. SVR rates have to be awaited. Other nucleotide analogue inhibitors that are

ª2011 The Authors Clinical Microbiology and Infection ª2011 European Society of Clinical Microbiology and Infectious Diseases, CMI, 17, 122–134

Vermehren and Sarrazin

CMI

New HCV therapies

127

TABLE 1. New hepatitis C virus therapies in the pipeline (a) Drug name NS3/4A protease inhibitors Ciluprevir (BILN 2061) Boceprevir (SCH503034) Telaprevir (VX-950) Danoprevir (RG 7227) TMC435 Vaniprevir (MK-7009) BI 201335 BMS-650032 GS-9256 ABT-450 Narlaprevir (SCH900518) PHX 1766 ACH-1625 IDX320 MK-5172 VX-985 GS-9451 Nucleos(t)ide NS5B polymerase inhibitors Valopicitabine (NM283) RG 7128 IDX184 R1626 PSI-7977 Non-nucleoside NS5B polymerase inhibitors BILB 1941 BI 207127 MK-3281 Filibuvir (PF-00868554) VX-916 VX-222 VX-759 ANA598 ABT-333 ABT-072 Nesbuvir (HCV-796) Tegobuvir (GS-9190) IDX375 NS5A inhibitors BMS-790052 BMS-824393 PPI-461 AZD7295

Company

Target/active drug

Study phase

Boehringer Ingelheim Merck Vertex InterMune/Roche Tibotec/Medivir Merck Boehringer Ingelheim Bristol-Myers Squibb Gilead Abbott/Enanta Merck Phenomix Achillion Idenix Merck Vertex Gilead

Active Active Active Active Active Active Active Active Active Active Active Active Active Active Active Active Active

site/macrocyclic site/linear site/linear site/macrocyclic site/macrocyclic site/macrocyclic site/linear site site site site/linear site site site/macrocyclic site/macrocyclic site site

Stopped Phase 3 Phase 3 Phase 2 Phase 2 Phase 2 Phase 2 Phase 2 Phase 2 Phase 2 On hold Phase 1 Phase 1 On hold Phase 1 Phase 1 Phase 1

Idenix/Novartis Roche/Pharmasset Idenix Roche Pharmasset

Active Active Active Active Active

site/NM107 site/PSI-6130 site site/R1479 site

Stopped Phase 2 On hold Stopped Phase 2

Boehringer Ingelheim Boehringer Ingelheim Merck Pfizer Vertex Vertex Vertex Anadys Abbott Abbott ViroPharma/Wyeth Gilead Idenix

NNI NNI NNI NNI NNI NNI NNI NNI NNI NNI NNI NNI NNI

Bristol-Myers Squibb Bristol-Myers Squibb Presidio AstraZeneca

NS5A NS5A NS5A NS5A

Novartis Scynexis Debiopharm/Novartis Romark Migenix

Cyclophilin inhibitor Cyclophilin inhibitor Cyclophilin inhibitor PKR induction? a-Glucosidase inhibitor

Stopped Phase 1 Phase 2 Phase 2 Phase 2

Biolex ZymoGenetics/BMS HGS/Novartis

Interferon receptor type 1 Interferon receptor type 3 Interferon receptor type 1

Phase 2 Phase 2 Stopped

site site site site site site site site site site site site site

1/thumb 1 1/thumb 1 1/thumb 1 2/thumb 2 2/thumb 2 2/thumb 2 2/thumb 2 3/palm 1 3/palm 1 3/palm 1 4/palm 2 4/palm 2 4/palm 2

Stopped Phase 2 Stopped Phase 2 On hold Phase 2 Phase 1 Phase 2 Phase 2 Phase 2 Stopped Phase 2 Phase 1

domain 1 inhibitor inhibitor inhibitor inhibitor

Phase Phase Phase Phase

2 1 1 1

(b) Indirect inhibitors/unknown mechanism of action NIM811 SCY-635 Alisporivir (Debio 025) Nitazoxanide Celgosivir New interferons Locteron (BLX-883) PEG-rIL-29 (pegylated interferon-k) Joulferon (albinterferon-a-2b) Double-stranded-RNA-activated protein kinase. PKR.

TABLE 2. Mutations conferring resistance to hepatitis C virus NS3 protease inhibitors; modified from [9] V36A/M Telaprevir (linear)

T54S/A

V55A

Q80R/K

R155K/T/Q

A156S

A156T/V

D168A/V/T/H

V170A/T a

a

Boceprevir (linear)

a

Narlaprevir (linear) Ciluprevir (macrocyclic) Danoprevir (macrocyclic)

a

a

Vaniprevir (macrocyclic) TMC435 (macrocyclic) BI-201335 (linear) aMutations

associated with resistance in vitro but not described in patients

ª2011 The Authors Clinical Microbiology and Infection ª2011 European Society of Clinical Microbiology and Infectious Diseases, CMI, 17, 122–134

128

Clinical Microbiology and Infection, Volume 17 Number 2, February 2011

CMI

currently in clinical development include PSI-7977 (a chirally pure isomer form of PSI-7851) and IDX184 [57,58].

inhibitors include ABT-072 and ABT-333, both of which have entered phase 2 clinical trials [70].

Allosteric, non-nucleoside inhibitors

Non-nucleoside site 4 inhibitors (palm 2/benzofuran site)

The structure of the NS5B polymerase resembles a characteristic ‘right-hand motif’, consisting of finger, palm and thumb domains. At least four different allosteric binding sites have been identified for the inhibition of the NS5B polymerase by non-nucleoside inhibitors: a benzimidazole (thumb 1)-binding site, a thiophene (thumb 2)-binding site, a benzothiadiazine (palm 1)-binding site, and a benzofuran (palm 2)-binding site [59,60]. As non-nucleoside inhibitors bind more distantly to the active site of NS5B, resistant mutations occur more frequently than for nucleos(t)ide analogues.

Monotherapy with HCV-796 (nesbuvir) showed low antiviral activity in HCV genotype 1-infected patients, and resulted in selection of resistant variants and viral breakthrough in several patients. Further drug development was suspended because of abnormal liver enzyme elevations in a subsequent phase 2 study [71]. GS-9190 (tegobuvir) displayed low antiviral activity in a phase 1 clinical study, and drug-resistant variants were observed [72]. However, GS-9190 has entered phase 2 clinical trials, both in combination with SOC and with the protease inhibitor GS-9256 ± RBV. Finally, IDX375 also binds to the palm pocket, and is currently in phase 1 clinical trials [73].

Non-nucleoside site 1 inhibitors (thumb 1/benzimidazole site)

BI 207127, BILB 1941 and MK-3281 are non-nucleoside site 1 inhibitors that have been shown to exhibit low to medium antiviral activity in phase 1 clinical trials [61–63]. Four weeks of BI 207127 (600 mg three times daily) in combination with PEG-IFN-a-2a/RBV resulted in a median reduction of 5.6 log10 HCV RNA in treatment-naı¨ve genotype 1 patients, and no viral breakthrough was observed [64]. Results of further studies have to be awaited. The further development of BILB 1941 and MK-3281 was halted because of gastrointestinal adverse events. Non-nucleoside site 2 inhibitors (thumb 2/thiophene site)

The non-nucleoside site 2 inhibitor filibuvir (PF-00868554) has shown medium antiviral activity in phase 1 clinical trials. Interim results from a phase 1 trial investigating different doses of filibuvir in combination with PEG-IFN-a-2a/RBV for 4 weeks followed by SOC for 44 weeks have shown similar SVR (at 12 weeks of follow-up) results for the different filibuvir groups and for the SOC group, indicating that longer dosing with filibuvir is needed [65]. Other site 2 inhibitors include VX-759, VX-916 and VX-222 [66–68]. However, only VX-222 has progressed to phase 2 development. Non-nucleoside site 3 inhibitors (palm 1/benzothiadiazine site)

A phase 2 trial of ANA598 in combination with PEG-IFN-a2a/RBV in treatment-naı¨ve patients with HCV genotype 1 is currently ongoing. It was recently reported that 75% of patients who received 400 mg of ANA598 twice daily achieved undetectable HCV RNA levels at treatment week 12 (complete early virological response (cEVR)) [69]. However, SVR rates have to be awaited. Other site 3 palm 1

NS5A inhibitors

Inhibitors of NS5A are potentially active against all HCV genotypes. BMS-790052 binds to domain I of the NS5A protein, which is crucial for the regulation of HCV replication, assembly and release. Following promising results from a phase 1 study, BMS-790052 is currently under investigation in a number of phase 2 clinical trials. Recently, interim data were released from a phase 2 study investigating different doses of BMS-790052 in combination with SOC. Rapid virological response rates were 83% and 92% in patients who received 10 and 60 mg BMS-790052 once daily, respectively. In addition, undetectable HCV RNA levels at week 12 (cEVR) were observed in 83% of patients [74]. Other NS5A inhibitors that are in early clinical development include BMS-824393, AZD7295 and PPI-461 [75–77].

DAA Combination Therapies Interferon-associated side effects may limit treatment success in a large number of patients. Therefore, one of the primary future goals of DAA therapy is to control viral replication or even to achieve SVR in interferon-free regimens. In the INFORM-1 study, combinations of different doses of a polymerase inhibitor (RG 7128) and the NS3/4A protease inhibitor danoprevir (RG 7227/ITMN191) were tested in both treatment-naı¨ve and treatment-experienced patients with HCV genotype 1 for up to 2 weeks. Up to 63% of treatment-naı¨ve patients achieved undetectable HCV RNA levels after 2 weeks of combination therapy, and only one patient experienced viral rebound without exhibiting resistance mutations [78]. HCV RNA negativity rates were somewhat lower in treatment-experienced patients (25%).

ª2011 The Authors Clinical Microbiology and Infection ª2011 European Society of Clinical Microbiology and Infectious Diseases, CMI, 17, 122–134

Vermehren and Sarrazin

CMI

A number of other drug combinations are currently being investigated. Dual, triple and quadruple therapy with GS9256 and GS-9190 alone or in combination with RBV or PEG-IFN-a/RBV for up to 28 days revealed the importance of RBV for the enhancement of antiviral activity and reduction of viral breakthrough [79]. Interferon-sparing triple combination therapy with BI 201335 and BI 207127 (400 or 600 mg three times daily) plus RBV resulted in undetectable HCV RNA after 4 weeks in all patients treated with the higher dose of BI 207127 [80]. Finally, in an ongoing trial of HCV genotype 1 null-responders treated with BMS-790052 plus BMS-650032 alone or in combination with PEG-IFN-a/ RBV for 24 weeks, 14/21 patients achieved cEVR, whereas all patients with viral breakthrough (n = 6) were genotype 1a patients, indicating that not all DAAs may be equally effective in both HCV genotype 1a and 1b patients [81]. Overcoming drug resistance will be the primary challenge in DAA combination therapy, and drug combinations that have a genetic barrier of four or more mutations may be required [82]. Future trials need to address the tolerability and safety of long-term DAA administration, and whether SVR can be achieved without the addition of PEG-IFN-a/ RBV.

Emerging Mechanisms Emerging potential drug candidates in future anti-HCV therapies include inhibitors of HCV entry, internal ribosome entry site inhibitors, inhibitors of HCV assembly and release, and a-glucosidase inhibitors. However, potential compounds are currently still in preclinical or very early clinical development. Host cells may provide additional targets for antiviral therapies, and several host cell-targeting inhibitors have entered clinical development, with promising results. Cyclophilin inhibitors

Cyclophilins are ubiquitous proteins in human cells, and are involved in protein folding. Moreover, cyclophilins participate in HCV replication as functional regulators of the HCV NS5B polymerase. The cyclophilin inhibitor Debio 025 (alisporivir) showed antiviral activity in patients infected with different HCV genotypes (1–4) during monotherapy and in combination studies with PEG-IFN-a. Maximum log10 changes in HCV RNA of up to 4.75 were observed in HCV genotype 1 or 4 patients who received 1000 mg of Debio 025 in combination with PEG-IFN-a-2a/RBV for 4 weeks [83]. There is one case report of a genotype 3a patient who achieved SVR following

New HCV therapies

129

4 weeks of Debio 025 1000 mg/day as monotherapy [84]. No viral breakthrough has been observed in clinical studies so far. However, selection of HCV variants resistant to Debio 025 with mutations clustering in the NS5A gene was observed in the HCV replicon system [85,86]. SCY-635 is another non-immunosuppressive analogue of cyclosporin A that exhibits potent suppression of HCV. Different doses of SCY-635 were investigated in patients infected with HCV genotype 1, and a mean maximum decline of 2.3 log10 IU/mL in viral load was observed after 15 days of 900-mg SCY-635 monotherapy [87]. No viral rebound was observed during SCY-635 therapy. However, minimal evidence of resistance selection was observed within NS5B [88]. Silibinin

Silymarin, an extract of milk thistle (Silybum marianum) with antioxidant activity, has been used as self-medication for liver diseases for centuries [89]. Silibinin is one of the six major flavonolignans in silymarin. Although its mechanism of action and treatment efficacy are not yet fully understood, it was recently reported that silibinin inhibited HCV RNA-dependent RNA polymerase activity [90]. However, its antiviral action includes blocking of virus entry and transmission, possibly by targeting the host cell [91]. Intravenous silibinin was investigated in non-responders to prior IFN-based antiviral therapy, and led to a significant decline in HCV RNA of between 0.55 and 3.02 log10 IU/mL after 7 days, and a further decrease after an additional 7 days in combination with PEG-IFN-a-2a/RBV, in the range of 1.63–4.85 log10 IU/mL [92]. Studies in larger patient cohorts, including resistance analyses, are underway. Nitazoxanide

Nitazoxanide (alinia) is a thiazolide that has been shown in vitro to have activity against both hepatitis B virus and HCV. The antiviral method of action of nitazoxanide is still unknown, but it is believed that it inhibits viral glycoproteins at the post-translational level (i.e. PKR induction). A phase 2 study of 500 mg of nitazoxanide twice daily for 12 weeks, followed by nitazoxanide, PEG-IFN-a-2a ± RBV for 36 weeks, yielded an SVR rate of 79% in treatment-naı¨ve genotype 4 patients [93]. However, a similar regimen in treatment-naı¨ve genotype 1 patients yielded a week 12 SVR rate of only 44% [94]. Further studies are warranted.

New Interferons Albinterferon-a-2b (Joulferon) is a recombinant protein consisting of interferon-a-2b fused to human albumin that can

ª2011 The Authors Clinical Microbiology and Infection ª2011 European Society of Clinical Microbiology and Infectious Diseases, CMI, 17, 122–134

130

Clinical Microbiology and Infection, Volume 17 Number 2, February 2011

be administered every 2 weeks, owing to a longer half-life than that of the currently marketed pegylated interferons. The efficacy and safety of albinterferon-a-2b administered every 2 weeks in combination with RBV for 48 weeks in patients infected with HCV genotype 1, and for 24 weeks in patients infected with HCV genotype 2/3, were recently studied in two large phase 3 clinical trials (ACHIEVE 1 and ACHIEVE 2/3) that were designed to demonstrate the noninferiority of these regimens as compared with SOC [95,96]. Both studies achieved the primary objective. In patients with HCV genotype 1, intention-to-treat SVR rates of 51%, 48.2% and 47.3% were achieved for PEG-IFN-a-2a, albinterferon-a2b (900 lg) and albinterferon-a-2b (1200 lg), respectively. For genotype 2/3-infected patients, intention-to-treat SVR rates were 84.8%, 79.8% and 80%, respectively. The safety profile was similar between the different treatment regimens, indicating that albinterferon-a-2b was not better tolerated than PEG-IFN-a-2a. Owing to concerns expressed by the regulatory authorities regarding the benefit/risk ratio, further development of albinterferon was recently suspended. Locteron is a controlled-release formulation of interferona-2b that can be administered every 2 weeks. Week 12 interim results from a phase 2b study have shown a similar reduction in HCV RNA and a reduction of flu-like symptoms by 57% for locteron at a dose of 480 lg as compared with PEG-IFN-a-2b in treatment-naı¨ve patient with HCV genotype 1 [97]. SVR results have to be awaited. PEG-IFN-k (PEG-rIL-29) is a type III interferon that binds to a unique receptor with a more limited distribution than the type I interferon receptor used by interferon-a. In a phase 1b trial, 56 treatment-naı¨ve and SOC relapser patients were enrolled to receive different doses of PEG-IFN-k (ranging from 0.5 to 3.0 lg/kg) administered every 2 weeks or weekly with or without daily RBV for 4 weeks. Antiviral activity was seen at all dose levels, with a mean maximum change of HCV RNA from baseline of 3.27 log10 IU/mL in treatment-naı¨ve patients treated with weekly PEG-IFN-k (1.5 lg/kg) plus RBV [98]. Phase 2 trials are currently ongoing.

Conclusions A number of advances have been made in the development of new therapeutic options for the treatment of chronic hepatitis C. In particular, the challenges of treating patients with HCV genotype 1 who achieve SVR rates in the range of 40– 50% with the current SOC have been met by the development of DAA agents. Although monotherapy with DAAs may not be suitable, owing to the selection of drug-resistant

CMI

HCV variants followed by viral breakthrough, convincing results have been achieved for the combination of new antiviral agents with SOC. Results from phase 2 clinical studies of the currently most advanced compounds, telaprevir and boceprevir, have indicated that the addition of an HCV protease inhibitor to PEG-IFN-a/RBV significantly improves SVR rates in both treatment-naı¨ve and treatment-experienced patients. In addition, response-guided treatment schedules have been successfully applied [19,20,26]. Thus, two of the major goals of anti-HCV therapy, i.e. higher SVR rates and shorter treatment duration based on RVR results, can be achieved. The pivotal phase 3 trials of telaprevir and boceprevir in treatment-naı¨ve patients infected with HCV genotype 1 recently confirmed these promising results, showing SVR rates ranging from 72% to 75% in telaprevir-treated patients and from 63% to 66% in boceprevir-treated patients [24,25,37]. Most HCV polymerase inhibitors have shown low to medium antiviral activities during monotherapy studies. Therefore, the future role of HCV polymerase inhibitors has yet to be determined, and SVR data from larger studies with triple combination therapies have to be awaited. Unlike protease inhibitors, which are mostly active in patients with HCV genotype 1 only, polymerase inhibitors bind to the highly conserved centre of the HCV polymerase, with the possible advantage of being equally effective for different HCV genotypes. With the addition of new antiviral compounds to standard therapy, a number of additional side effects unrelated to SOC have been observed, and increased treatment discontinuation rates have been reported. In addition, frequent dosing intervals (i.e. three times daily) may lead to increased non-adherence outside of clinical trials, and more patientfriendly formulations need to be developed. One of the major challenges of future anti-HCV treatment regimens will be the emerging field of DAA drug resistance. Relatively low genetic barriers to resistance have been reported for HCV protease inhibitors and non-nucleoside polymerase inhibitors as compared with nucleos(t)ide HCV polymerase inhibitors, which appear to have a high genetic barrier to resistance [9]. However, HCV eradication may not be achieved by monotherapy with either substance group, and drug combinations with non-cross-resistance patterns have to be further evaluated. In addition, pre-existing resistant variants and their potential long-term persistence have to be taken into account for the selection of optimal treatment and retreatment strategies. Although the addition of PEG-IFN-a and RBV substantially reduces the frequency of viral breakthrough caused by resistance development,

ª2011 The Authors Clinical Microbiology and Infection ª2011 European Society of Clinical Microbiology and Infectious Diseases, CMI, 17, 122–134

Vermehren and Sarrazin

CMI

combinations of different classes of DAA agents may eventually replace interferon-based regimens. Several trials addressing this issue are currently underway. In addition, the future of hepatitis C therapy will probably involve individually tailored treatment durations, based on viral kinetics, IL28B polymorphisms and other parameters. Besides compounds that target HCV polyprotein processing or HCV replication, inhibitors of HCV entry and host cell-targeting inhibitors (e.g. cyclophilin inhibitors) represent further interesting strategies for future anti-HCV therapies. Indeed, the cyclophilin inhibitor alisporivir (Debio 025) has exhibited substantial antiviral efficacy with and without SOC in early clinical trials. As interferon and RBV are likely to remain the backbone of anti-HCV therapy in the near future, the development of new interferon formulations is also underway, and preliminary study results have shown that these may facilitate antiviral therapy without providing additional antiviral efficacy.

Transparency Declaration C. Sarrazin has served as a clinical investigator, consultant and/or member of speakers’ bureaus for Abbott, BMS, Boehringer Ingelheim, Gilead, Merck, Novartis, Roche, Tibotec, and Vertex. J. Vermehren has nothing to disclose.

References 1. Barnes E. The need for an HCV vaccine. WHO Factsheet 2010. Available at: http://www.who.int/vaccine_research/diseases/viral_cancers/en/index2.html (last accessed 1 August 2010).. 2. Fried MW, Shiffman ML, Reddy KR et al. Peginterferon alfa-2a plus ribavirin for chronic hepatitis C virus infection. N Engl J Med 2002; 347: 975–982. 3. Hadziyannis SJ, Sette H Jr, Morgan TR et al. Peginterferon-alpha2a and ribavirin combination therapy in chronic hepatitis C: a randomized study of treatment duration and ribavirin dose. Ann Intern Med 2004; 140: 346–355. 4. Manns MP, McHutchison JG, Gordon SC et al. Peginterferon alfa-2b plus ribavirin compared with interferon alfa-2b plus ribavirin for initial treatment of chronic hepatitis C: a randomised trial. Lancet 2001; 358: 958–965. 5. Zeuzem S, Hultcrantz R, Bourliere M et al. Peginterferon alfa-2b plus ribavirin for treatment of chronic hepatitis C in previously untreated patients infected with HCV genotypes 2 or 3. J Hepatol 2004; 40: 993–999. 6. Farnik H, Lange CM, Sarrazin C et al. Meta-analysis shows extended therapy improves response of patients with chronic hepatitis C virus genotype 1 infection. Clin Gastroenterol Hepatol 2010; 8: 884– 890. 7. Kim JL, Morgenstern KA, Lin C et al. Crystal structure of the hepatitis C virus NS3 protease domain complexed with a synthetic NS4A cofactor peptide. Cell 1996; 87: 343–355.

New HCV therapies

131

8. Moradpour D, Penin F, Rice CM. Replication of hepatitis C virus. Nat Rev Microbiol 2007; 5: 453–463. 9. Sarrazin C, Zeuzem S. Resistance to direct antiviral agents in patients with hepatitis C virus infection. Gastroenterology 2010; 138: 447–462. 10. Wakita T, Pietschmann T, Kato T et al. Production of infectious hepatitis C virus in tissue culture from a cloned viral genome. Nat Med 2005; 11: 791–796. 11. Lamarre D, Anderson PC, Bailey M et al. An NS3 protease inhibitor with antiviral effects in humans infected with hepatitis C virus. Nature 2003; 426: 186–189. 12. Reesink HW, Zeuzem S, Weegink CJ et al. Rapid decline of viral RNA in hepatitis C patients treated with VX-950: a phase Ib, placebo-controlled, randomized study. Gastroenterology 2006; 131: 997– 1002. 13. Sarrazin C, Kieffer TL, Bartels D et al. Dynamic hepatitis C virus genotypic and phenotypic changes in patients treated with the protease inhibitor telaprevir. Gastroenterology 2007; 132: 1767–1777. 14. Lin C, Lin K, Luong YP et al. In vitro resistance studies of hepatitis C virus serine protease inhibitors, VX-950 and BILN 2061: structural analysis indicates different resistance mechanisms. J Biol Chem 2004; 279: 17508–17514. 15. Lin C, Kwong AD, Perni RB. Discovery and development of VX-950, a novel, covalent, and reversible inhibitor of hepatitis C virus NS3.4A serine protease. Infect Disord Drug Targets 2006; 6: 3–16. 16. Lin K, Perni RB, Kwong AD et al. VX-950, a novel hepatitis C virus (HCV) NS3-4A protease inhibitor, exhibits potent antiviral activities in HCV replicon cells. Antimicrob Agents Chemother 2006; 50: 1813– 1822. 17. Forestier N, Reesink HW, Weegink CJ et al. Antiviral activity of telaprevir (VX-950) and peginterferon alfa-2a in patients with hepatitis C. Hepatology 2007; 46: 640–648. 18. Lawitz E, Rodriguez-Torres M, Muir AJ et al. Antiviral effects and safety of telaprevir, peginterferon alfa-2a, and ribavirin for 28 days in hepatitis C patients. J Hepatol 2008; 49: 163–169. 19. McHutchison JG, Everson GT, Gordon SC et al. Telaprevir with peginterferon and ribavirin for chronic HCV genotype 1 infection. N Engl J Med 2009; 360: 1827–1838. 20. Hezode C, Forestier N, Dusheiko G et al. Telaprevir and peginterferon with or without ribavirin for chronic HCV infection. N Engl J Med 2009; 360: 1839–1850. 21. Forns X, Marcellin P, Ferenci P et al. On-treatment response-guided therapy with telaprevir q8h or q12h combined with peginterferon alfa-2a or peginterferon alfa-2b and ribavirin in treatment-naive genotype 1 hepatitis C (study C208). J Hepatol 2010; 52: S26. 22. Benhamou Y, Moussalli J, Ratziu V et al. Activity of telaprevir monotherapy or in combination with peginterferon-alfa-2a and ribavirin in treatment-naive genotype 4 hepatitis-C-patients: final results of study C210. Hepatology 2010; 52: 719A. 23. Foster GR, Hezode C, Bronowicki JP et al. Activity of telaprevir alone or in combination with peginterferon alfa-2a and ribavirin in treatment-naive genotype 2 and 3 hepatitis-C patients: final results of study C209. J Hepatol 2010; 52: S27. 24. Jacobson IM, McHutchison JG, Dusheiko G et al. Telaprevir in combination with peginterferon and ribavirin in genotype 1 HCV treatment-naive patients: final results of phase 3 ADVANCE study. Hepatology 2010; 52: 427A. 25. Sherman KE, Flamm SL, Afdhal NH et al. Telaprevir in combination with peginterferon alfa-2a and ribavirin for 24 or 48 weeks in treatment-naive genotype 1 HCV patients who achieved an extended rapid viral response: final results of phase 3 ILLUMINATE study. Hepatology 2010; 52: 401A. 26. McHutchison JG, Manns MP, Muir AJ et al. Telaprevir for previously treated chronic HCV infection. N Engl J Med 2010; 362: 1292–1303.

ª2011 The Authors Clinical Microbiology and Infection ª2011 European Society of Clinical Microbiology and Infectious Diseases, CMI, 17, 122–134

132

Clinical Microbiology and Infection, Volume 17 Number 2, February 2011

27. Vertex Pharmaceuticals Inc. 65% of people whose prior treatment for hepatitis C was unsuccessful achieved SVR (viral cure) with telaprevir-based therapy in phase 3 REALIZE study. Press Release 2010. Available at: http://investors.vrtx.com/releasedetail.cfm?ReleaseID=505239 (last accessed 18 October 2010).. 28. Susser S, Forestier N, Vermehren J et al. Decline of detectable resistance mutations within the HCV NS3 protease quasispecies during long-term follow-up after treatment with telaprevir. J Hepatol 2010; 52: S300. 29. Sarrazin C, Rouzier R, Wagner F et al. SCH 503034, a novel hepatitis C virus protease inhibitor, plus pegylated interferon alpha-2b for genotype 1 nonresponders. Gastroenterology 2007; 132: 1270–1278. 30. Qiu P, Sanfiorenzo V, Curry S et al. Identification of HCV protease inhibitor resistance mutations by selection pressure-based method. Nucleic Acids Res 2009; 37: e74. 31. Susser S, Welsch C, Wang Y et al. Characterization of resistance to the protease inhibitor boceprevir in hepatitis C virus-infected patients. Hepatology 2009; 50: 1709–1718. 32. Tong X, Bogen S, Chase R et al. Characterization of resistance mutations against HCV ketoamide protease inhibitors. Antiviral Res 2008; 77: 177–185. 33. Vermehren J, Susser S, Karey U et al. Clonal analysis of mutations selected in the HCV NS3 protease domain of genotype 1 nonresponders sequentially treated with boceprevir (SCH503034) and/or pegylated interferon alfa-2b (PEG-IFN alfa-2b). Hepatology 2009; 50: 1040A. 34. Kwo PY, Lawitz EJ, McCone J et al. Efficacy of boceprevir, an NS3 protease inhibitor, in combination with peginterferon alfa-2b and ribavirin in treatment-naive patients with genotype 1 hepatitis C infection (SPRINT-1): an open-label, randomised, multicentre phase 2 trial. Lancet 2010; 376: 705–716. 35. Schiff E, Poordad F, Jacobson I et al. Boceprevir combination therapy in null responders: response dependent on interferon responsiveness. J Hepatol 2008; 48: S46. 36. Bacon BR, Gordon SC, Lawitz E et al. HCV RESPOND-2 final results: high sustained virologic response among genotype 1 previous nonresponders and relapsers to peginterferon/ribavirin when re-treated with boceprevir plus pegintron (peginterferon alfa-2b)/ribavirin. Hepatology 2010; 52: 430A. 37. Poordad F, McCone J, Bacon BR et al. Boceprevir (BOC) combined with peginterferon alfa-2b/ribavirin (P/R) for treatment-naive patients with hepatitis C virus (HCV) genotype (G) 1: SPRINT-2 final results. Hepatology 2010; 52: 402A. 38. Detishin V, Haazen W, Robison H et al. Virological response, safety, and pharmacokinetic profile following single- and multipledose administration of ACH-0141625 protease inhibitor to healthy volunteers and HCV genotype-1 patients. J Hepatol 2010; 52: S468. 39. Forestier N, Larrey D, Marcellin P et al. Antiviral activity and safety of ITMN-191 in combination with peginterferon alfa-2a and ribavirin in patients with chronic hepatitis C virus (HCV). J Hepatol 2009; 50: S35. 40. Hotho D, de Bruijne J, O’Farrell AM et al. Accelerated clinical trial design to assess the safety, tolerability and anti-viral activity of PHX1766, a novel HCV NS3/4A protease inhibitor, in healthy volunteers and chronic hepatitis C patients. Hepatology 2009; 50: 1031A. 41. Pasquinelli C, Eley T, Villegas C et al. Safety, tolerability, pharmacokinetics and antiviral activity following single- and multiple-dose administration of BMS-650032, a novel HCV NS3 inhibitor, in subjects with chronic genotype 1 hcv infection. Hepatology 2009; 50: 411A. 42. Sulkowski M, Ferenci P, Emanoil C et al. SILEN-C1: early antiviral activity and safety of BI 201335 combined with peginterferon alfa-2a and ribavirin in treatment-naive patients with chronic genotype 1 HCV infection. Hepatology 2009; 50: 2A.

CMI

43. Brainard DM, Petry A, van Dyck K et al. Safety and antiviral activity of MK-5172, a novel HCV NS3/4A protease inhibitor with potent activity against known resistance mutants, in genotype 1 and 3 HCVinfected patients. Hepatology 2010; 52: 706A. 44. Goldwater R, DeMicco MP, Zong J et al. Safety, pharmacokinetics, and antiviral activity of single oral doses of the HCV NS3 protease inhibitor GS 9256. Hepatology 2010; 52: 717A. 45. Lawitz E, Hill JM, Marbury TC et al. Three-day, dose-ranging study of the HCV NS3 protease inhibitor GS-9451. Hepatology 2010; 52: 714A. 46. de Bruijne J, Bergmann JF, Reesink HW et al. Antiviral activity of narlaprevir combined with ritonavir and pegylated interferon in chronic hepatitis C patients. Hepatology 2010; 52: 1590–1599. 47. Terrault NA, Cooper C, Balart LA et al. Phase II randomised, partially-blind, parallel-group study of oral danoprevir (RG7227) with PegIFNalfa-2a (PEGASYS) plus ribavirin in treatment-naive genotype 1 patients with CHC: results of planned week 12 interim analysis of the ATLAS study. Hepatology 2010; 52: 335A. 48. Manns MP, Gane EJ, Rodriguez-Torres M et al. Sustained viral response (SVR) rates in genotype 1 treatment-naive patients with chronic hepatitis C (CHC) infection treated with vaniprevir (MK7009), a NS3/4A protease inhibitor, in combination with pegylated interferon alfa-2a and ribavirin for 28 days. Hepatology 2010; 52: 361A. 49. Berg T, Dieterich DT, Lalezari JP et al. Virological response and safety of 4 weeks treatment with the protease inhibitor BI 201335 combined with 48 weeks of peginterferon alfa 2a and ribavirin for treatment of HCV GT-1 patients who failed peginterferon/ribavirin. Hepatology 2010; 52: 704A. 50. Fried MW, Buti M, Dore GJ et al. Efficacy and safety of TMC435 in combination with peginterferon alfa-2a and ribavirin in treatmentnaive genotype-1 HCV patients: 24-week interim results from the PILLAR study. Hepatology 2010; 52: 403A. 51. Reesink HW, van Vliet A, Mu¨hr F et al. Antiviral activity, safety and pharmacokinetics of IDX320, a novel macrocyclic HCV protease inhibitor, in a 3-day proof-of-concept study in patients with chronic hepatitis C. Hepatology 2010; 52: 882A. 52. Lawitz E, Gaultier F, Poordad F et al. 4-week virologic response and safety of ABT-450 given with low-dose ritonavir (ABT-450/r) in combination with pegylated interferon alpha-2a and ribavirin (SOC) after 3-day monotherapy in genotype 1 (GT1) HCV-infected treatmentnaive subjects. Hepatology 2010; 52: 878A. 53. Lawitz E, Nguyen T, Younes Z et al. Clearance of HCV RNA with valopicitabine (NM283) plus peg-interferon in treatment-naive patients with HCV-1 infection: results at 24 and 48 weeks. J Hepatol 2007; 46: S9. 54. Pockros PJ, Nelson D, Godofsky E et al. R1626 plus peginterferon Alfa-2a provides potent suppression of hepatitis C virus RNA and significant antiviral synergy in combination with ribavirin. Hepatology 2008; 48: 385–397. 55. Roberts SK, Cooksley G, Dore GJ et al. Robust antiviral activity of R1626, a novel nucleoside analog: a randomized, placebo-controlled study in patients with chronic hepatitis C. Hepatology 2008; 48: 398– 406. 56. Jensen DM, Wedemeyer H, Herring RW et al. High rates of early viral response, promising safety profile and lack of resistance-related breakthrough in HCV GT 1/4 patients treated with RG7128 plus PEGIFN alfa-2a (40 KD)/RBV: planned week 12 interim analysis from the propel study. Hepatology 2010; 52: 360A. 57. Lalezari JP, O’Riordan W, Poordad F et al. A phase IIa study of IDX184 in combination with pegylated interferon (pegIFN) and ribavirin (RBV) in treatment-naive HCV genotype 1-infected subjects. Hepatology 2010; 52: 337A.

ª2011 The Authors Clinical Microbiology and Infection ª2011 European Society of Clinical Microbiology and Infectious Diseases, CMI, 17, 122–134

Vermehren and Sarrazin

CMI

58. Lawitz E, Lalezari JP, Rodriguez-Torres M et al. High rapid virologic response (RVR) with PSI-7977 qd plus PEG-IFN/RBV in a 28-day phase 2a trial. Hepatology 2010; 52: 706A. 59. Beaulieu PL. Non-nucleoside inhibitors of the HCV NS5B polymerase: progress in the discovery and development of novel agents for the treatment of HCV infections. Curr Opin Investig Drugs 2007; 8: 614–634. 60. Lesburg CA, Cable MB, Ferrari E et al. Crystal structure of the RNAdependent RNA polymerase from hepatitis C virus reveals a fully encircled active site. Nat Struct Biol 1999; 6: 937–943. 61. 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: 1026A. 62. 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: 23–32. 63. Larrey DG, Benhamou Y, Lohse AW et al. BI 207127 is a potent HCV RNA polymerase inhibitor during 5 days monotherapy in patients with chronic hepatitis C. Hepatology 2009; 50: 1044A. 64. Larrey DG, Lohse AW, de Ledinghen V et al. 4 week therapy with the non-nucleoside polymerase inhibitor BI207127 in combination with peginterferon-alfa2a and ribavirin in treatment naive and treatment experienced chronic HCV GT1 patients. J Hepatol 2010; 52: S466. 65. Jacobson IM, Pockros PJ, Lalezari J et al. Virologic response rates following 4 weeks of filibuvir in combination with pegylated interferon alfa-2a and ribavirin in chronically-infected HCV genotype-1 patients. J Hepatol 2010; 52: S465. 66. Cooper C, Lawitz EJ, Ghali P et al. Evaluation of VCH-759 monotherapy in hepatitis C infection. J Hepatol 2009; 51: 39–46. 67. 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: S37. 68. Rodriguez-Torres M, Lawitz E, Conway B et al. Safety and antiviral activity of the HCV non-nucleoside polymerase inhibitor VX-222 in treatment-naive genotype 1 HCV-infected patients. J Hepatol 2010; 52: S14. 69. Lawitz E, Rodriguez-Torres M, Rustgi VK et al. Safety and antiviral activity of ANA598 in combination with pegylated interferon alfa-2a plus ribavirin in treatment-naive genotype-1 chronic HCV patients. Hepatology 2010; 52: 334A. 70. Rodriguez-Torres M, Lawitz E, Cohen D et al. Treatment-naive, HCV genotype 1-infected subjects show significantly greater HCV RNA decreases when treated with 28 days of ABT-333 plus peginterferon and ribavirin compared to peginterferon and ribavirin alone. Hepatology 2009; 50: 5A. 71. Kneteman NM, Howe AY, Gao T et al. HCV796: a selective nonstructural protein 5B polymerase inhibitor with potent anti-hepatitis C virus activity in vitro, in mice with chimeric human livers, and in humans infected with hepatitis C virus. Hepatology 2009; 49: 745– 752. 72. Harris J, Bae A, Sun SC et al. Antiviral response and resistance analysis of treatment-naive HCV infected subjects receiving single and multiple doses of GS-9190. Hepatology 2010; 52: 722A. 73. de Bruijne J, van de Wetering de Rooij J, van Vliet AA et al. Phase I study in healthy volunteers and patients with IDX375, a novel nonnucleoside HCV polymerase inhibitor. Hepatology 2010; 52: 1219A. 74. Pol S, Everson GT, Ghalib R et al. Once-daily NS5A inhibitor (BMS790052) plus peginterferon-alpha-2a and ribavirin produces high rates of extended rapid virologic response in treatment-naive HCV-genotype 1 subjects: phase 2a trial. J Hepatol 2010; 52: S462.

New HCV therapies

133

75. Gane EJ, Foster GR, Cianciara J et al. Antiviral activity, pharmakokinetics, and tolerability of AZD7295, a novel NS5A inhibitor, in a placebo-controlled multiple ascending dose study in HCV genotype 1 and 3 patients. J Hepatol 2010; 52: S464. 76. Colonno R, Peng E, Bencsik M et al. Identification and characterization of PPI-461, a potent and selective HCV NS5A inhibitor with activity against all HCV genotypes. J Hepatol 2010; 52: S14. 77. Nettles R, Wang X, Quadri S et al. BMS-824393 is a potent hepatitis C virus NS5A inhibitor with substantial antiviral activity when given as monotherapy in subjects with chronic G1 HCV infection. Hepatology 2010; 52: 1203A. 78. 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: 1467–1475. 79. Zeuzem S, Buggisch P, Agarwal A et al. Dual, triple, and quadruple combination treatment with a protease inhibitor (GS-9256) and a polymerase inhibitor (GS-9190) alone and in combination with ribavirin (RBV) or PegIFN/RBV for up to 28 days in treatment naive, genotype 1 HCV subjects. Hepatology 2010; 52: 400A. 80. Zeuzem S, Asselah T, Angus PW et al. Strong antiviral activity and safety of IFN-sparing treatment with the protease inhibitor BI 201335, the HCV polymerase inhibitor BI 207127 and ribavirin in patients with chronic hepatitis C. Hepatology 2010; 52: 876A. 81. Lok AS, Gardiner DF, Lawitz E et al. Combination therapy with BMS790052 and BMS-650032 alone or with PEGIFN/RBV results in undetectable HCV RNA through 12 weeks of therapy in HCV genotype 1 null responders. Hepatology 2010; 52: 877A. 82. Rong L, Dahari H, Ribeiro RM et al. Rapid emergence of protease inhibitor resistance in hepatitis C virus. Sci Transl Med 2010; 2: 30ra32. 83. Flisiak R, Feinman SV, Jablkowski M et al. The cyclophilin inhibitor Debio 025 combined with PEG IFNalpha2a significantly reduces viral load in treatment-naive hepatitis C patients. Hepatology 2009; 49: 1460–1468. 84. Horban A, Berak H, Kolakowska-Rzadzka A et al. Sustained virological response after 4-week treatment with debio 025 monotherapy or coadministered with PEG-IFN alpha 2a in HCV genotype 1 and 3 patients. J Hepatol 2010; 52: S294. 85. Coelmont L, Gallay P, Bobardt M et al. Particular in vitro anti-HCV activities and resistance profile of the cyclophilin inhibitor debio 025. J Hepatol 2009; 50: S36. 86. Coelmont L, Kaptein S, Paeshuyse J et al. Debio 025, a cyclophilin binding molecule, is highly efficient in clearing hepatitis C virus (HCV) replicon-containing cells when used alone or in combination with specifically targeted antiviral therapy for HCV (STAT-C) inhibitors. Antimicrob Agents Chemother 2009; 53: 967. 87. Hopkins S, Heuman D, Gavis E et al. Safety, plasma pharmakokinetics, and anti-viral activity of SCY-635 in adult patients with chronic hepatitis C virus infection. J Hepatol 2009; 50: S36. 88. Hopkins S, Mosier S, Harris R et al. Resistance selection following 15 days of monotherapy with SCY-635, a non-immunosuppressive cyclophilin inhibitor with potent anti-HCV activity. J Hepatol 2010; 52: S15. 89. Gazak R, Walterova D, Kren V. Silybin and silymarin—new and emerging applications in medicine. Curr Med Chem 2007; 14: 315–338. 90. Ahmed-Belkacem A, Ahnou N, Barbotte L et al. Silibinin and related compounds are direct inhibitors of hepatitis C virus RNA-dependent RNA polymerase. Gastroenterology 2010; 138: 1112–1122. 91. Wagoner J, Negash A, Kane OJ et al. Multiple effects of silymarin on the hepatitis C virus lifecycle. Hepatology 2010; 51: 1912–1921. 92. Ferenci P, Scherzer TM, Kerschner H et al. Silibinin is a potent antiviral agent in patients with chronic hepatitis C not responding to pegy-

ª2011 The Authors Clinical Microbiology and Infection ª2011 European Society of Clinical Microbiology and Infectious Diseases, CMI, 17, 122–134

134

Clinical Microbiology and Infection, Volume 17 Number 2, February 2011

lated interferon/ribavirin therapy. Gastroenterology 2008; 135: 1561– 1567. 93. Rossignol JF, Elfert A, El Gohary Y et al. Improved virologic response in chronic hepatitis C genotype 4 treated with nitazoxanide, peginterferon, and ribavirin. Gastroenterology 2009; 136: 856–862. 94. Bacon BR, Shiffman ML, Lim JK et al. Phase 2 randomized, doubleblind, placebo-controlled study of nitazoxanide plus peginterferon and ribavirin in HCV genotype 1 naive patients: week 12 sustained virologic response rate. J Hepatol 2010; 52: S463. 95. Nelson DR, Benhamou Y, Chuang WL et al. Randomized trial of albinterferon alfa-2b for the treatment of patients with chronic hepatitis C virus genotype 2 or 3. Gastroenterology 2010; 139: 1267– 1276.

CMI

96. Zeuzem S, Sulkowski MS, Lawitz EJ et al. Randomized trial of albinterferon alfa-2b for the treatment of patients with chronic hepatitis C virus genotype 1. Gastroenterology 2010; 139: 1257–1266. 97. Long WA, Takov D, Tchernev K et al. Q2week controlled-releaseinterferon-alpha2b+ribavirin reduces flu-like symptoms >50% and provides equivalent efficacy in comparison to weekly pegylated-interferon-alpha2b+ribavirin in treatment-naive-genotype-1-chronic-hepatitis C: results from EMPOWER, a randomized-open-label-12-weekcomparison in 133 patients. J Hepatol 2010; 52: S467. 98. Muir AJ, Shiffman ML, Zaman A et al. Phase 1b study of pegylated interferon lambda 1 with or without ribavirin in patients with chronic genotype 1 hepatitis C virus infection. Hepatology 2010; 52: 822–832.

ª2011 The Authors Clinical Microbiology and Infection ª2011 European Society of Clinical Microbiology and Infectious Diseases, CMI, 17, 122–134