Next Article in Journal
Reliable Polymerase Chain Reaction Methods for Screening for Porcine Endogenous Retroviruses-C (PERV-C) in Pigs
Previous Article in Journal
Ad6-Based GM-CSF Expressing Vector Displays Oncolytic and Immunostimulatory Effects in an Immunocompetent Syrian Hamster Model of Cholangiocarcinoma
Previous Article in Special Issue
Elimination of HCV Infection: Recent Epidemiological Findings, Barriers, and Strategies for the Coming Years
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Comprehensive Review of Antiviral Therapy for Hepatitis C: The Long Journey from Interferon to Pan-Genotypic Direct-Acting Antivirals (DAAs)

1
SIcilian Network for Therapy, Epidemiology and Screening In Hepatology (SINTESI), 90127 Palermo, Italy
2
Department of Oncology and Hematology, Azienda Ospedaliero-University Hospital of Mod, 41124 Modena, Italy
3
Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, 41100 Modena, Italy
4
Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
5
Department of Medicine, Buccheri-La Ferla Hospital, 90123 Palermo, Italy
*
Author to whom correspondence should be addressed.
Viruses 2025, 17(2), 163; https://doi.org/10.3390/v17020163
Submission received: 30 December 2024 / Revised: 19 January 2025 / Accepted: 20 January 2025 / Published: 24 January 2025
(This article belongs to the Special Issue Hepatitis C Virus: From Epidemiology to Treatment)

Abstract

:
The treatment landscape for hepatitis C virus (HCV) infection has transformed over the past few decades, evolving from the limited efficacy of interferon (IFN) monotherapy to the highly successful pan-genotypic direct-acting antivirals (DAAs) used today. Initially, alpha-interferon monotherapy, introduced in the 1990s, was the standard treatment, yet it provided low sustained virological response (SVR) rates and caused significant adverse effects, limiting its utility. The development of pegylated interferon (peg-IFN) improved the pharmacokinetic profile of IFN, allowing for less frequent dosing and modestly improved response rates. When combined with ribavirin, peg-IFN achieved higher SVR rates, especially in non-genotype 1 HCV infections, but the combination also brought additional side effects, such as anemia and depression. The advent of the first-generation DAAs, such as telaprevir and boceprevir, marked a significant milestone. Combined with peg-IFN and ribavirin, these protease inhibitors boosted response rates in patients with genotype 1 HCV. However, high rates of adverse effects and drug resistance remained challenges. Second-generation DAAs, like sofosbuvir and ledipasvir, introduced IFN-free regimens with improved safety profiles and efficacy. The most recent advances are pan-genotypic DAAs, including glecaprevir-pibrentasvir and sofosbuvir-velpatasvir, which offer high SVR rates across all genotypes, shorter treatment durations, and fewer side effects. Current pan-genotypic regimens represent a cornerstone in HCV therapy, providing an accessible and effective solution globally.

1. Introduction

The World Health Organization (WHO) estimates that 50 million people with chronic hepatitis C virus (HCV) infection live in the world, and 1 million new infections occur annually, with a global prevalence of approximately 1%. HCV is a leading cause of chronic liver disease, including cirrhosis and hepatocellular carcinoma (HCC) [1,2].
Chronic HCV infection is often asymptomatic, progressing over decades without detection. Without intervention, approximately 20–30% of chronically infected individuals develop liver cirrhosis, and up to 5% may develop HCC [3,4,5].
This progression highlights the need for effective antiviral therapy to prevent long-term liver damage and associated mortality. Antiviral therapy has been shown to reduce the incidence of cirrhosis, liver failure, and HCC by achieving sustained virological response (SVR), essentially curing the infection [6,7].
The development of antiviral therapy for HCV has been transformative. Treatment has evolved from interferon-based regimens, which were only modestly effective and associated with severe side effects, to highly effective direct-acting antivirals (DAAs) that provide high SVR rates with minimal adverse effects [8]. The latest pan-genotypic DAAs, effective across all HCV genotypes, have simplified treatment protocols, making them accessible and effective worldwide [9,10].
This review aims to provide a comprehensive overview of the evolution of antiviral therapy for HCV, tracing the journey from early interferon-based regimens to the development of pan-genotypic DAAs. By examining each treatment stage’s efficacy, safety, and limitations, it seeks to highlight the pivotal advances in HCV therapy and discuss the implications for global health and HCV elimination efforts.
The main purpose of the review is to describe the progressive and rapid effectiveness of drugs for all patients with hepatitis C, a disease for which, until 35 years ago, the etiological agent was unknown and had no cure. In light of these powerful innovations, DAAs should be available not only for developed countries, but also and at less onerous costs, for developing countries where the prevalence of HCV infection and liver disease is very high.

2. The Era of Alpha-Interferon Monotherapy

In the early 1990s, alpha-interferon (IFN) monotherapy emerged as the first-line treatment for HCV infection, marking a milestone in the management of a disease that was often asymptomatic but could progress to severe liver damage over time [11].

3. Mechanism of Action of Alpha-Interferon

IFN is a cytokine produced by immune cells in response to viral infections. It functions by inducing an “antiviral state” within host cells. The antiviral effects are mediated through several pathways: alpha-interferon binds to receptors on the surface of target cells, activating intracellular signaling pathways, such as the JAK-STAT pathway, which leads to the transcription of interferon-stimulated genes (ISGs). These genes produce antiviral proteins that inhibit viral replication at various stages, including entry, RNA transcription, protein synthesis, and release. Interferon also enhances the immune response by promoting the activity of natural killer (NK) cells and cytotoxic T lymphocytes, which play critical roles in clearing infected cells [12].

4. Clinical Efficacy and Challenges

While alpha-interferon represented a significant advancement, the therapy was limited by low response rates and high rates of relapse post-treatment. Early studies showed that ALT levels were normalized in 35–40% of patients, and biopsy also demonstrated histological improvement in lobular and periportal inflammation. Unfortunately, relapse was a common problem in these studies, occurring in 49–57% of patients [13,14,15].
The poor response rates were compounded by the significant side effects associated with interferon therapy. Common adverse effects included flu-like symptoms (fever, fatigue, myalgia), neuropsychiatric symptoms (depression, irritability), and hematologic changes, such as leukopenia and thrombocytopenia, which often required dose adjustments or even discontinuation of therapy [16].

5. Understanding the Limitations

Research during this era highlighted the inherent limitations of alpha-interferon in treating HCV. Unlike other viruses that rely on intracellular replication machinery (such as the hepatitis B virus), HCV has a high mutation rate, resulting in quasi-species or closely related viral variants that can evade immune responses. This genetic variability made HCV particularly resistant to interferon monotherapy, which could not completely suppress viral replication [17,18,19].
Additionally, the need for frequent subcutaneous administration, often three times weekly, the limited efficacy, and the high side effect rates of alpha-interferon monotherapy underscored the need for other treatments that could enhance effectiveness and safety.

6. Pegylated Interferon Monotherapy

The introduction of pegylated interferon (peg-IFN) in the early 2000s represented a pivotal advancement in HCV treatment. Chemically modifying IFN with polyethylene glycol (PEG) offered a more stable pharmacokinetic profile, allowing for once-weekly dosing while enhancing efficacy in suppressing HCV replication [20].

7. Mechanism and Pharmacokinetics of Pegylation

Pegylation is a process that involves attaching PEG, a large, inert molecule, to the interferon protein. This modification shields the interferon from rapid degradation and clearance, particularly by the kidneys, and reduces its immunogenicity, thereby prolonging its half-life. The PEG molecule increases the interferon’s size, slowing its absorption and creating a controlled release mechanism that maintains stable blood levels over the week. The two primary peg-IFN formulations differ slightly in their PEG structure: peg-IFN alfa-2a uses a larger, branched PEG molecule, which provides more consistent plasma concentrations, while peg-IFN alfa-2b uses a smaller, linear PEG molecule, which is absorbed more rapidly but still offers a significant half-life extension compared to standard IFN [21]. This prolonged and stable pharmacokinetic profile was beneficial for HCV treatment, as it maintained adequate drug levels without the peaks and troughs associated with standard IFN. This not only provided continuous antiviral action but also minimized the frequency of side effects that tended to arise during the peaks in standard IFN dosing [22].

8. Challenges and Limitations of Peg-IFN Monotherapy

While peg-IFN significantly improved over standard IFN, its efficacy as a monotherapy was limited. SVR rates with peg-IFN monotherapy remained suboptimal, particularly in genotype 1 patients, with approximately 60–70% of genotype 1 patients experiencing viral relapse after treatment [23,24].

9. Clinical Implications of Peg-IFN Monotherapy

Despite its limitations, peg-IFN monotherapy marked a significant step forward in HCV treatment and set the stage for combination therapy with ribavirin. The enhanced pharmacokinetics and improved SVR rates of peg-IFN compared to standard IFN provided the foundation for subsequent treatment advancements. Peg-IFN also highlighted the need for genotype-specific approaches, as genotypes 1 and 4 proved more challenging to treat with peg-IFN monotherapy [25,26], while genotypes 2 and 3 were more responsive [27]. These findings underscored the genetic diversity of HCV and informed subsequent treatment guidelines and strategies [28,29].

10. Combination Therapy with Pegylated Interferon and Ribavirin

The combination of peg-IFN with ribavirin marked a significant milestone in HCV treatment, substantially improving SVR rates compared to peg-IFN alone. Introduced in the early 2000s, this combination became the standard of care and remained the cornerstone of HCV therapy for over a decade until the advent of DAAs. Ribavirin, a nucleoside analog with broad-spectrum antiviral activity, enhanced the effectiveness of peg-IFN, particularly in achieving SVR across various HCV genotypes, though challenges persisted, especially for genotypes 1 and 4 [30].

11. Mechanism of Action of Ribavirin and Its Role in Improving SVR

Ribavirin’s mechanism of action is complex. It is thought to exert its antiviral effects through multiple pathways, including (1) the direct inhibition of HCV replication, (2) inhibition of host inosine monophosphate dehydrogenase (IMPDH) enzyme, (3) mutagenesis induction to drive a rapidly replicating virus beyond the threshold to error catastrophe, and (4) immunomodulation by inducing a Th1 immune response. Specifically, ribavirin monophosphate (RMP) is converted to ribavirin triphosphate (RTP) forms, and binding of RTP to the nucleotide binding site of RNA polymerase prevents binding of the correct nucleotides, leading to reduced viral replication or the production of defective virions. Additionally, ribavirin monophosphate is a competitive inhibitor of IMPDH, an enzyme involved in the de novo synthesis of guanine nucleotides. This could lead to a decline in viral protein synthesis and limit the replication of viral genomes. Finally, ribavirin enhances the immune response against HCV by modulating the balance of T-helper (Th1/Th2) cells, favoring a Th1-dominant, cell-mediated immune response, which is more effective in clearing the virus [31].
In combination with peg-IFN, ribavirin enhances the SVR by increasing the likelihood of viral eradication, particularly when administered over an extended period [32].

12. SVR Rates Across Different HCV Genotypes

HCV genotype plays a crucial role in determining treatment response. Peg-IFN alfa-2b plus ribavirin achieved significantly higher SVR rates compared to interferon alfa-2b plus ribavirin across all HCV genotypes (Table 1). However, response rates varied by genotype. For patients with genotype 1, the most prevalent and hardest-to-treat genotype, the SVR rate was around 42% with combination therapy, compared to lower rates with interferon alone. This represented a meaningful improvement but left a substantial proportion of genotype 1 patients without an effective cure.
In contrast, patients with genotypes 2 and 3 responded much more favorably to the combination therapy, with SVR rates of around 76–82%. This high efficacy in genotypes 2 and 3 led to shorter treatment durations (24 weeks instead of 48 weeks), given that these genotypes were more susceptible to the antiviral effects of the combined therapy. Genotype 4, more prevalent in regions such as the Middle East and North Africa, exhibited intermediate SVR rates of approximately 55–60%, presenting additional challenges in achieving complete viral eradication compared to genotypes 2 and 3 [33,34,35,36,37,38,39,40,41].

13. Adverse Effects of Ribavirin, Especially Hematological Toxicity

While ribavirin enhanced treatment efficacy, it also introduced significant adverse effects, particularly hematological toxicity. The most notable side effect is hemolytic anemia, which occurs because ribavirin accumulates in red blood cells, causing oxidative damage that leads to hemolysis [42]. Ribavirin can also exacerbate the neuropsychiatric side effects associated with interferon, such as depression and irritability, impacting patient adherence [43,44].

14. The Introduction of Direct-Acting Antivirals (DAAs): First Generation

The introduction of DAAs marked a turning point in HCV treatment, revolutionizing approaches with enhanced efficacy compared to prior therapies. The first-generation DAAs, notably protease inhibitors such as boceprevir and telaprevir, were designed to target viral replication, significantly advancing the treatment landscape for chronic HCV patients.

15. Mechanism of Action: NS3/4A Protease Inhibition

Boceprevir and telaprevir became FDA-approved in 2011 as the first DAAs targeting the HCV NS3/4A protease, a crucial enzyme in viral replication [45,46]. By binding to the active site of this protease, boceprevir and telaprevir effectively turn off the protease’s function, halting the replication process. Unlike previous interferon-based therapies, these protease inhibitors offered a mechanism of action specific to the virus, opening the door to targeted antiviral therapy and transforming the treatment paradigm by focusing on viral pathways rather than general immune system modulation [47,48,49].

16. Improvements in Sustained Virological Response (SVR) Rates

One of the key benefits of first-generation DAAs was their impact on SVR rates, a crucial indicator of HCV treatment efficacy. Clinical trials demonstrated that adding boceprevir or telaprevir to the existing standard of care—typically PEG-IFN and ribavirin—dramatically improved SVR rates, especially in patients with genotype 1 HCV, the most challenging to treat with IFN-based therapies alone (Table 2). For example, telaprevir-based regimens achieved SVR rates between 67% and 79% in treatment-naïve genotype 1 patients, a substantial improvement over the approximate 40% SVR rates seen with PEG-IFN and ribavirin alone [50,51,52,53,54,55,56,57].

17. Challenges and Limitations: Resistance, Complex Dosing, and Drug–Drug Interactions

Despite their efficacy, first-generation DAAs presented significant challenges, including the risk of viral resistance, adverse effects, and complex treatment regimens. Resistance was a notable issue; since these DAAs targeted a single viral enzyme, mutations in the NS3/4A protease could render the drugs less effective. Resistance-associated variants (RAVs) often emerged during treatment, particularly in patients who experienced incomplete viral suppression. This challenge emphasized the need for combination therapies targeting multiple viral components to reduce the risk of resistance [58].
Another major limitation of first-generation DAAs was their reliance on interferon. Boceprevir and telaprevir were not administered as standalone therapies; rather, they were combined with PEG-IFN and ribavirin, which brought additional adverse effects and lengthy treatment durations. IFN-based regimens were associated with side effects such as fatigue, flu-like symptoms, and hematologic abnormalities, often making adherence challenging for patients. Moreover, the DAAs themselves introduced side effects, including anemia, rash, and gastrointestinal disturbances, further complicating treatment [59,60,61].
Finally, first-generation DAAs also required intricate dosing schedules, demanding strict patient adherence. The potential for drug–drug interactions exacerbated this complexity, as boceprevir and telaprevir were metabolized by the liver enzyme CYP3A. This led to interactions with many other medications, particularly those affecting hepatic function. The narrow therapeutic window of these drugs made managing comorbid conditions challenging and sometimes necessitated additional monitoring or dose adjustments for concomitant medications [62,63,64,65].
  • Biological insights that led to development of DAAs
The development of DAAs was driven by several pivotal biological insights into the virus’s life cycle and molecular biology. These insights enabled the identification of highly specific therapeutic targets within the virus (Table 3).
  • Understanding the HCV Genome and Polyprotein Processing
The HCV genome is a positive-sense single-stranded RNA (~9.6 kb) that encodes a single polyprotein. This polyprotein is cleaved into structural proteins (core, E1, E2) and non-structural (NS) proteins (NS3, NS4A, NS4B, NS5A, and NS5B) by host and viral proteases. The viral NS proteins were identified as critical components for replication and virion assembly, making them ideal targets for DAAs.
2.
Role of NS3/4A Protease in Viral Maturation.
NS3/4A is a serine protease that cleaves the HCV polyprotein at specific junctions, producing functional proteins required for viral replication. Inhibiting NS3/4A blocks the processing of the polyprotein, halting replication at an early stage. Protease inhibitors (e.g., Glecaprevir, Voxilaprevir) were designed to bind the NS3/4A active site, preventing substrate processing.
3.
RNA-Dependent RNA Polymerase as the Engine of Replication
NS5B is an RNA-dependent RNA polymerase responsible for synthesizing the viral RNA genome. It is highly conserved across HCV genotypes. NS5B became a key target for therapeutic intervention, as inhibiting it would directly halt genome replication. Nucleoside inhibitors (e.g., Sofosbuvir) mimic natural nucleotides and are incorporated into the viral RNA, causing chain termination. Non-nucleoside inhibitors (e.g., Dasabuvir) bind allosterically to NS5B, reducing its enzymatic activity.
4.
Multifunctional Role of NS5A in Viral Replication and Assembly
NS5A is a phosphoprotein involved in multiple steps of the HCV life cycle, including replication, assembly, and interactions with host factors. NS5A inhibitors disrupt the replication complex and virion assembly, resulting in a reduction in infectious virus production. Drugs like Ledipasvir, Velpatasvir, and Pibrentasvir were designed to bind NS5A, leveraging its role as a central hub in viral replication.
5.
Role of Host–Virus Interactions
HCV relies on host factors, such as lipid metabolism pathways and cellular chaperones, to facilitate its replication and assembly. Early therapies targeted host factors (e.g., interferons), but the high specificity of DAAs for viral proteins minimized off-target effects and adverse reactions. The focus on direct viral targets reduced dependency on host-directed strategies, improving efficacy and tolerability.
6.
Genotype-Specific Variations.
HCV exhibits genetic diversity with seven major genotypes and numerous subtypes, which affect drug sensitivity and resistance patterns. Understanding genotype-specific differences informed the design of pan-genotypic DAAs, such as Sofosbuvir and Velpatasvir, which target conserved regions of the virus.
7.
Viral Resistance Mechanisms.
HCV’s high replication rate and lack of proofreading by NS5B polymerase lead to the rapid generation of quasispecies, promoting resistance. Combination therapy targeting multiple viral proteins simultaneously was developed to prevent resistance and enhance treatment efficacy.
8.
Advances in Structural Biology and Drug Design
High-resolution crystal structures of HCV proteins (e.g., NS3/4A, NS5A, and NS5B) provided a detailed understanding of their active sites and interaction dynamics. Structure-based drug design enabled the development of DAAs with high specificity and potency [66,67,68,69,70,71,72,73,74,75,76,77].
These insights into the molecular and structural biology of HCV, combined with advances in medicinal chemistry, allowed the development of DAAs that transformed HCV therapy. The ability to target multiple stages of the virus life cycle, combined with the advent of pan-genotypic agents, has resulted in cure rates exceeding 95% for most patients.

18. Efficacy and Improved Tolerability in IFN-Free Regimens

Clinical trials revealed that sofosbuvir, combined with ribavirin or other DAAs, achieved high SVR rates across multiple HCV genotypes. For instance, in genotype 1 patients—a group previously challenging to treat—sofosbuvir-based regimens demonstrated SVR rates exceeding 90% when combined with other DAAs such as ledipasvir [78]. Genotype 2 and 3 patients also showed excellent responses, with SVR rates around 93–97% when sofosbuvir was combined with ribavirin alone, eliminating the need for IFN [79]. This shift allowed for shorter, simpler treatment courses with fewer side effects, increasing patient adherence and satisfaction.

19. Impact on Treatment Adherence and Sustained Virological Response (SVR)

The shift from IFN-based to IFN-free regimens profoundly affected treatment adherence and SVR rates. IFN has been associated with a range of side effects, including flu-like symptoms, depression, and hematologic abnormalities, often leading to early treatment discontinuation. Sofosbuvir-based IFN-free regimens addressed this issue by providing a more tolerable therapy with fewer adverse events, enabling patients to complete their treatment courses. The high efficacy and low side effect profile of sofosbuvir made it feasible to offer treatment to a wider range of patients, including those previously ineligible for IFN-based regimens, such as those with advanced liver disease or liver transplantation [80,81,82,83,84].

20. Efficacy, Tolerability, and Reduced Treatment Duration of Combination Therapies

The combination therapies in second- and third-generation DAAs offer superior efficacy, with SVR rates often surpassing 95% and, in some cases, approaching 100%, particularly in treatment-naïve patients (Table 4). The ledipasvir–sofosbuvir combination, for example, showed exceptional efficacy for HCV genotype 1 patients, achieving SVR rates of 94–100% depending on prior treatment history and the presence of cirrhosis. This combination also demonstrated remarkable tolerability, with mild side effects such as headache and fatigue being the most reported, significantly improving the severe adverse effects of interferon-based therapies [85,86,87,88].
Another potent combination, daclatasvir–asunaprevir, has also proven effective across genotypes. Studies show that these combinations maintain high efficacy and tolerability, particularly for genotypes 1 and 3, which were historically challenging to treat. These regimens improve patient comfort and quality of life and enhance adherence, as patients are less likely to discontinue due to adverse effects [89,90].
One of the most transformative aspects of modern DAA therapies is the reduced treatment duration. Earlier regimens required 24–48 weeks of treatment, whereas modern DAA combinations can achieve high SVR rates in as little as 8–12 weeks for most patients. This shorter duration is especially beneficial for patients with complex medical profiles or limited ability to adhere to long treatment courses.
Finally, DAA therapies are also highly effective in populations previously considered difficult to treat, including patients with cirrhosis or liver transplant, those co-infected with HIV, and individuals who have relapsed after prior treatment attempts [91,92].

21. Pan-Genotypic DAAs: The Latest Generation

The development of pan-genotypic DAAs marks a significant advance in HCV therapy by introducing regimens effective across all major HCV genotypes. Unlike earlier treatments that required genotype-specific regimens, pan-genotypic DAAs, such as glecaprevir-pibrentasvir [93,94] and sofosbuvir-velpatasvir [95,96,97], are designed to target multiple genotypes, simplifying the treatment landscape. This shift reduces the need for genotype testing before initiating therapy, enabling faster, more accessible treatment on a global scale. These drugs not only achieve high SVR rates across genotypes but also offer improved tolerability and shorter treatment durations (8 or 12 weeks), even for hard-to-treat patient populations [98].

22. High SVR Rates, Improved Tolerability, and Simplified Treatment Regimens

Pan-genotypic DAAs deliver high SVR rates across HCV genotypes, even among patient populations traditionally considered challenging to treat (Table 5). Clinical trials have shown that the sofosbuvir–velpatasvir combination [99,100,101] and glecaprevir–pibrentasvir combination [102,103] achieve SVR rates exceeding 95% in patients across all major genotypes, including those with cirrhosis and previous treatment failure. The combination therapy of Sofosbuvir, Velpatasvir, and Voxilaprevir represents a powerful option, particularly for patients who have previously failed treatment with other antiviral regimens [104].
These pan-genotypic regimens are associated with minimal side effects, typically limited to mild symptoms such as headache, fatigue, and nausea. This improved tolerability represents a major shift from the adverse effect profiles of older treatments, particularly interferon-based regimens, which often caused significant side effects leading to treatment discontinuation. The reduced adverse event profile of pan-genotypic DAAs contributes to better patient adherence and overall treatment success, as patients are less likely to experience side effects that could otherwise hinder their ability to complete the regimen [105,106,107,108,109,110].

23. Current Recommendations and Future Perspectives

Current guidelines for hepatitis C virus (HCV) treatment from major liver societies, including the American Association for the Study of Liver Diseases (AASLD) and the European Association for the Study of the Liver (EASL) emphasize the use of pan-genotypic DAAs for simplified, highly effective treatment across all HCV genotypes [9,10]. These guidelines recommend DAAs like glecaprevir-pibrentasvir and sofosbuvir-velpatasvir as first-line treatments due to their high SVR rates, minimal side effects, and shorter treatment durations, which often range from 8 to 12 weeks for most patient populations. The guidelines also suggest routine HCV screening for all adults and prioritize treatment for high-risk groups, including those with advanced liver disease and individuals co-infected with HIV.

24. Remaining Challenges and Future Directions

Despite advances in HCV therapy, challenges persist. Reinfection, especially among populations with higher-risk behaviors (such as injection drug users), poses a significant obstacle to long-term viral elimination. Addressing this requires effective antiviral treatments and public health measures, such as harm reduction programs and access to preventive care [111,112,113,114]. Additionally, limited accessibility to DAAs in low-resource settings hampers global efforts to eradicate HCV. High treatment costs and inadequate healthcare infrastructure in some regions continue to restrict access, making it challenging to achieve universal HCV elimination [115].
Looking ahead, the focus is on making HCV treatment even more accessible and effective. Pan-genotypic regimens and simplified dosing protocols reduce the need for extensive pre-treatment testing and make therapy more accessible in diverse clinical settings.
As part of a global elimination effort, there is growing emphasis on community-based screening, decentralizing treatment, and developing point-of-care diagnostics to identify and treat HCV infections more rapidly [116].
Finally, a major unsolved problem remains the high cost of drugs. Reducing drug costs requires a comprehensive approach that addresses economic, regulatory and market factors [117].
Policy options could include the following:
  • The production of generic drugs by encouraging early patent expiration or negotiating compulsory licenses to allow the production of low-cost generic versions.
  • Centralized purchasing by governments or health organizations that can adopt large-scale supply models, negotiating directly with manufacturers to secure lower prices.
  • Market competition to promote the entry of more manufacturers by offering tax incentives or reducing regulatory barriers, increasing competition and lowering prices.
  • Public–private partnerships to collaborate with pharmaceutical companies to develop and produce drugs, ensuring affordable prices in exchange for research support.
  • Public funding to invest in the research and development of treatments that are not subject to the profit-driven models of private companies.
  • Price controls to set maximum price caps for essential medicines or impose regulations on pharmaceutical companies’ profit margins.
  • Tax exemptions and incentives to reduce taxes or duties on raw materials and finished medicines to lower production and distribution costs.
These options must be adapted to local regulatory, economic and health contexts, while balancing affordability, innovation and sustainability for health systems.
Through these strategies, and with continued support from international health organizations, there is optimism that HCV can be eliminated as a public health threat within the coming decades.

Author Contributions

Conceptualization, L.D.M., V.C. and V.D.M.; methodology, L.D.M. and F.C.; resources L.D.M. and C.L.M.; data curation, L.D.M., S.C., E.F., G.L., R.N. and C.L.M.; writing—original draft preparation, L.D.M.; writing—review and editing, F.C., V.C. and V.D.M.; supervision, V.D.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

Vincenza Calvaruso served on the advisory board of Abbvie, Gilead, Ipsen, and Advanz. Vito Di Marco received research support from Abbvie and Gilead, and served on the advisory boards of Abbvie, Gilead, and Ipsen. Fabio Cartabellotta received research support from Abbvie and Gilead. The other authors have no disclosures to declare.

References

  1. World Health Organization. Global Hepatitis Report 2017; World Health Organization: Geneva, Switzerland, 2017. [Google Scholar]
  2. Polaris Observatory HCV Collaborators. Global change in hepatitis C virus prevalence and cascade of care between 2015 and 2020: A modelling study. Lancet Gastroenterol. Hepatol. 2022, 7, 396–415. [Google Scholar] [CrossRef] [PubMed]
  3. Alter, M.J.; Margolis, H.S.; Krawczynski, K.; Judson, F.N.; Mares, A.; Alexander, W.; Hu, P.Y.; Miller, J.K.; Gerber, M.A.; Sampliner, R.E.; et al. The natural history of community-acquired hepatitis C in the United States. The Sentinel Counties Chronic non-A, non-B Hepatitis Study Team. N. Engl. J. Med. 1992, 327, 1899–1905. [Google Scholar] [CrossRef]
  4. Davis, G.L.; Alter, M.J.; El-Serag, H.; Poynard, T.; Jennings, L.W. Aging of hepatitis C virus (HCV)-infected persons in the United States: A multiple cohort model of HCV prevalence and disease progression. Gastroenterology 2010, 138, 513–521. [Google Scholar] [CrossRef] [PubMed]
  5. Alter, M.J. Epidemiology of hepatitis C virus infection. World J. Gastroenterol. 2007, 13, 2436–2441. [Google Scholar] [CrossRef]
  6. Ioannou, G.N.; Feld, J.J. What Are the Benefits of a Sustained Virologic Response to Direct-Acting Antiviral Therapy for Hepatitis C Virus Infection? Gastroenterology 2019, 156, 446–460. [Google Scholar] [CrossRef] [PubMed]
  7. Shiratori, Y.; Ito, Y.; Yokosuka, O.; Imazeki, F.; Nakata, R.; Tanaka, N.; Arakawa, Y.; Hashimoto, E.; Hirota, K.; Yoshida, H.; et al. Antiviral therapy for cirrhotic hepatitis C: Association with reduced hepatocellular carcinoma development and improved survival. Ann. Intern. Med. 2005, 142, 105–114. [Google Scholar] [CrossRef]
  8. Dore, G.J.; Feld, J.J. Hepatitis C virus therapeutic development: In pursuit of “perfectovir”. Clin. Infect. Dis. 2015, 60, 1829–1836. [Google Scholar] [CrossRef] [PubMed]
  9. Bhattacharya, D.; Aronsohn, A.; Price, J.; Lo Re, V.; AASLD-IDSA HCV Guidance Panel. Hepatitis C Guidance 2023 Update: AASLD-IDSA Recommendations for Testing, Managing, and Treating Hepatitis C Virus Infection. Clin. Infect. Dis. 2023, 2023, ciad319. [Google Scholar] [CrossRef]
  10. European Association for the Study of the Liver. EASL recommendations on treatment of hepatitis C: Final update of the series. J. Hepatol. 2020, 73, 1170–1218. [Google Scholar] [CrossRef]
  11. Hoofnagle, J.H.; Mullen, K.D.; Jones, D.B.; Rustgi, V.; Di Bisceglie, A.; Peters, M.; Waggoner, J.G.; Park, Y.; Jones, E.A. Treatment of chronic non-A, non-B hepatitis with recombinant human alpha interferon. A preliminary report. N. Engl. J. Med. 1986, 315, 1575–1578. [Google Scholar] [CrossRef]
  12. Samuel, C.E. Antiviral actions of interferons. Clin. Microbiol. Rev. 2001, 14, 778–809. [Google Scholar] [CrossRef] [PubMed]
  13. Di Bisceglie, A.M.; Martin, P.; Kassianides, C.; Lisker-Melman, M.; Murray, L.; Waggoner, J.; Goodman, Z.; Banks, S.M.; Hoofnagle, J.H. Recombinant interferon alfa therapy for chronic hepatitis C. A randomized, double-blind, placebo-controlled trial. N. Engl. J. Med. 1989, 321, 1506. [Google Scholar] [CrossRef]
  14. Davis, G.L.; Balart, L.A.; Schiff, E.R.; Lindsay, K.; Bodenheimer, H.C.; Perrillo, R.P.; Carey, W.; Jacobson, I.M.; Payne, J.; Dienstag, J.L.; et al. Treatment of chronic hepatitis C with recombinant interferon alfa. A multicenter randomized, controlled trial. N. Engl. J. Med. 1989, 321, 1501–1506. [Google Scholar] [CrossRef]
  15. Davis, G.L. Recombinant alpha-interferon treatment of non-A, and non-B (type C) hepatitis: Review of studies and recommendations for treatment. J. Hepatol. 1990, 11 (Suppl. S1), S72–S77. [Google Scholar] [CrossRef]
  16. Dusheiko, G. Side effects of alpha interferon in chronic hepatitis C. Hepatology 1997, 26 (Suppl. S1), 112S–121S. [Google Scholar] [CrossRef]
  17. Pawlotsky, J.M.; Germanidis, G.; Neumann, A.U.; Pellerin, M.; Frainais, P.-O.; Dhumeaux, D. Interferon resistance of hepatitis C virus genotype 1b: Relationship to nonstructural 5A gene quasispecies mutations. J. Virol. 1998, 72, 2795–2805. [Google Scholar] [CrossRef]
  18. von Wagner, M.; Lee, J.H.; Rüster, B.; Kronenberger, B.; Sarrazin, C.; Roth, W.K.; Zeuzem, S. Dynamics of hepatitis C virus quasispecies turnover during interferon-alpha treatment. J. Viral Hepat. 2003, 10, 413–422. [Google Scholar] [CrossRef] [PubMed]
  19. Paterson, M.; Laxton, C.; Goldin, R.D.; Ackrill, A.M.; Foster, G.R. Selection of HCV NS5Aquasispecies during IFN therapy in patients with chronic HCV. Dig. Dis. Sci. 2001, 46, 1399–1408. [Google Scholar] [CrossRef] [PubMed]
  20. Zeuzem, S.; Heathcote, J.E.; Martin, N.; Nieforth, K.; Modi, M. Peginterferon alfa-2a (40 kDa) monotherapy: A novel agent for chronic hepatitis C therapy. Expert Opin. Investig. Drugs 2000, 10, 2201–2213. [Google Scholar]
  21. Zeuzem, S.; Welsch, C.; Herrmann, E. Pharmacokinetics of peginterferons. Semin. Liver Dis. 2003, 23 (Suppl. S1), 23–28. [Google Scholar] [CrossRef] [PubMed]
  22. Zaman, A.; Fennerty, M.B.; Keeffe, E.B. Systematic review: Peginterferon vs. standard interferon in the treatment of chronic hepatitis C. Aliment. Pharmacol. Ther. 2003, 18, 661–670. [Google Scholar] [CrossRef] [PubMed]
  23. Pockros, P.J.; Carithers, R.; Desmond, P.; Dhumeaux, D.; Fried, M.W.; Marcellin, P.; Shiffman, M.L.; Minuk, G.; Reddy, K.R.; Reindollar, R.W.; et al. Efficacy and safety of two-dose regimens of peginterferon alpha-2a compared with interferon alpha-2a in chronic hepatitis C: A multicenter, randomized controlled trial. Am. J. Gastroenterol. 2004, 99, 1298–1305. [Google Scholar] [CrossRef] [PubMed]
  24. Lurie, Y.; Rouzier-Panis, R.; Webster, G.J.; Dusheiko, G.M.; Laughlin, M.; Jackson, M.L.; Oren, R. Optimal dosing frequency of pegylated interferon alfa-2b monotherapy for chronic hepatitis C virus infection. Clin. Gastroenterol. Hepatol. 2005, 3, 610–615. [Google Scholar] [CrossRef] [PubMed]
  25. Kamal, S.M. Improving outcome in patients with hepatitis C virus genotype 4. Am. J. Gastroenterol. 2007, 102, 2582–2588. [Google Scholar] [CrossRef] [PubMed]
  26. Kamal, S.M.; Kassim, S.K.; Ahmed, A.I.; Mahmoud, S.; A Bahnasy, K.; A Hafez, T.; Aziz, I.A.; Fathelbab, I.F.; Mansour, H.M. Host and viral determinants of the outcome of exposure to HCV infection genotype 4: A large longitudinal study. Am. J. Gastroenterol. 2014, 109, 199–211. [Google Scholar] [CrossRef]
  27. Yada, M.; Masumoto, A.; Yamashita, N.; Motomura, K.; Koyanagi, T.; Sakamoto, S. Immediate virological response predicts the success of short-term peg-interferon monotherapy for chronic hepatitis C. World J. Gastroenterol. 2010, 16, 1506–1511. [Google Scholar] [CrossRef]
  28. Kawaoka, T.; Hayes, C.N.; Ohishi, W.; Ochi, H.; Maekawa, T.; Abe, H.; Tsuge, M.; Mitsui, F.; Hiraga, N.; Imamura, M.; et al. Predictive value of the IL28B polymorphism on the effect of interferon therapy in chronic hepatitis C patients with genotypes 2a and 2b. J. Hepatol. 2011, 54, 408–414. [Google Scholar] [CrossRef]
  29. Ochi, H.; Maekawa, T.; Abe, H.; Hayashida, Y.; Nakano, R.; Imamura, M.; Hiraga, N.; Kawakami, Y.; Aimitsu, S.; Kao, J.-H.; et al. IL-28B predicts response to chronic hepatitis C therapy--fine-mapping and replication study in Asian populations. J. Gen. Virol. 2011, 92, 1071–1081. [Google Scholar] [CrossRef]
  30. McHutchison, J.G.; Fried, M.W. Current therapy for hepatitis C: Pegylated interferon and ribavirin. Clin. Liver Dis. 2003, 7, 149–161. [Google Scholar] [CrossRef]
  31. Te, H.S.; Randall, G.; Jensen, D.M. Mechanism of action of ribavirin in the treatment of chronic hepatitis C. Gastroenterol. Hepatol. 2007, 3, 218–225. [Google Scholar]
  32. Feld, J.J.; Hoofnagle, J.H. Mechanism of action of interferon and ribavirin in treatment of hepatitis C. Nature 2005, 436, 967–972. [Google Scholar] [CrossRef] [PubMed]
  33. McHutchison, J.G.; Gordon, S.C.; Schiff, E.R.; Shiffman, M.L.; Lee, W.M.; Rustgi, V.K.; Goodman, Z.D.; Ling, M.-H.; Cort, S.; Albrecht, J.K. Interferon alfa-2b alone or in combination with ribavirin as initial treatment for chronic hepatitis C. Hepatitis Interventional Therapy Group. N. Engl. J. Med. 1998, 339, 1485–1489. [Google Scholar] [CrossRef] [PubMed]
  34. Poynard, T.; Marcellin, P.; Lee, S.S.; Niederau, C.; Minuk, G.S.; Ideo, G.; Bain, V.; Heathcote, J.; Zeuzem, S.; Trepo, C.; et al. Randomised trial of interferon alpha2b plus ribavirin for 48 weeks or for 24 weeks versus interferon alpha2b plus placebo for 48 weeks for treatment of chronic infection with hepatitis C virus. International Hepatitis Interventional Therapy Group (IHIT). Lancet 1998, 352, 1426–1432. [Google Scholar] [CrossRef]
  35. Lindsay, K.L.; Trepo, C.; Heintges, T.; Shiffman, M.L.; Gordon, S.C.; Hoefs, J.C.; Schiff, E.R.; Goodman, Z.D.; Laughlin, M.; Yao, R.; et al. A randomized double- blind trial comparing pegylated interferon alfa-2b to inter-feron alfa-2b as initial treatment for chronic hepatitis C. Hepatology 2001, 34, 395–403. [Google Scholar] [CrossRef] [PubMed]
  36. Zeuzem, S.; Feinman, S.V.; Rasenack, J.; Heathcote, E.J.; Lai, M.-Y.; Gane, E.; O’Grady, J.; Reichen, J.; Diago, M.; Lin, A.; et al. Peginterferon alfa- 2a in patients with chronic hepatitis C. N. Engl. J. Med. 2000, 343, 1666–1672. [Google Scholar] [CrossRef]
  37. Heathcote, E.J.; Shiffman, M.L.; Cooksley, W.G.E.; Dusheiko, G.M.; Lee, S.S.; Balart, L.; Reindollar, R.; Reddy, R.K.; Wright, T.L.; Lin, A.; et al. Peginterferon alfa-2a in patients with chronic hepatitis C and cirrhosis. N. Engl. J. Med. 2000, 343, 1673–1680. [Google Scholar] [CrossRef]
  38. Manns, M.P.; McHutchison, J.G.; Gordon, S.C.; Rustgi, V.K.; Shiffman, M.; Reindollar, R.; Goodman, Z.D.; Koury, K.; Ling, M.-H.; Albrecht, J.K. 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. [Google Scholar] [CrossRef]
  39. Fried, M.W.; Shiffman, M.L.; Reddy, K.R.; Smith, C.; Marinos, G.; Gonçales, F.L.J.; Häussinger, D.; Diago, M.; Carosi, G.; Dhumeaux, D.; et al. Peginterferon alfa-2a plus ribavirin for chronic hepatitis C virus infection. N. Engl. J. Med. 2002, 347, 975–982. [Google Scholar] [CrossRef]
  40. Kamal, S.M.; El Kamary, S.S.; Shardell, M.D.; Hashem, M.; Ahmed, I.N.; Muhammadi, M.; Sayed, K.; Moustafa, A.; Hakem, S.A.; Ibrahiem, A.; et al. Pegylated interferon alpha-2b plus ribavirin in patients with genotype 4 chronic hepatitis C: The role of rapid and early virologic response. Hepatology 2007, 46, 1732–1740. [Google Scholar] [CrossRef]
  41. McHutchison, J.G.; Lawitz, E.J.; Shiffman, M.L.; Muir, A.J.; Galler, G.W.; McCone, J.; Nyberg, L.M.; Lee, W.M.; Ghalib, R.H.; Schiff, E.R.; et al. Peginterferon alfa-2b or alfa-2a with ribavirin for treatment of hepatitis C infection. N. Engl. J. Med. 2009, 361, 580–593. [Google Scholar] [CrossRef]
  42. Kowdley, K.V. Hematologic side effects of interferon and ribavirin therapy. J. Clin. Gastroenterol. 2005, 39 (Suppl. S1), S3–S8. [Google Scholar] [CrossRef] [PubMed]
  43. Minami, T.; Kishikawa, T.; Sato, M.; Tateishi, R.; Yoshida, H.; Koike, K. Meta-analysis:mortality and serious adverse events of peginterferon plus ribavirin therapy for chronic hepatitis C. J. Gastroenterol. 2013, 48, 254–268. [Google Scholar] [CrossRef]
  44. Udina, M.; Castellví, P.; Moreno-España, J.; Navinés, R.; Valdés, M.; Forns, X.; Langohr, K.; Solà, R.; Vieta, E.; Martín-Santos, R. Interferon-induced depression in chronic hepatitis C: A systematic review and meta-analysis. J. Clin. Psychiatry 2012, 73, 1128–1138. [Google Scholar] [CrossRef]
  45. Lam, J.T.; Jacob, S. Boceprevir: A recently approved protease inhibitor for hepatitis C virus infection. Am. J. Health Syst. Pharm. 2012, 69, 2135–2139. [Google Scholar] [CrossRef] [PubMed]
  46. Muir, A.J. Telaprevir for the treatment of chronic hepatitis C infection. Expert Rev. Anti-Infect. Ther. 2011, 9, 1105–1114. [Google Scholar] [CrossRef]
  47. Klibanov, O.M.; Vickery, S.B.; Olin, J.L.; Smith, L.S.; Williams, S.H. Boceprevir: A novel NS3/4 protease inhibitor for the treatment of hepatitis C. Pharmacotherapy 2012, 32, 173–190. [Google Scholar] [CrossRef] [PubMed]
  48. Smith, L.S.; Nelson, M.; Naik, S.; Woten, J. Telaprevir: An NS3/4A protease inhibitor for the treatment of chronic hepatitis C. Ann. Pharmacother. 2011, 45, 639–648. [Google Scholar] [CrossRef]
  49. Jacobson, I.M.; Pawlotsky, J.M.; Afdhal, N.H.; Dusheiko, G.M.; Forns, X.; Jensen, D.M.; Poordad, F.; Schulz, J. A practical guide for the use of boceprevir telaprevir for the treatment of hepatitis C. J. Viral Hepat. 2012, 19 (Suppl. S2), 1–26. [Google Scholar] [CrossRef]
  50. McHutchison, J.G.; Everson, G.T.; Gordon, S.C.; Jacobson, I.M.; Sulkowski, M.; Kauffman, R.; McNair, L.; Alam, J.; Muir, A.J. Telaprevir with peginterferon and ribavirin for chronic HCV genotype 1 infection. N. Engl. J. Med. 2009, 360, 1827–1838. [Google Scholar] [CrossRef] [PubMed]
  51. Hézode, C.; Forestier, N.; Dusheiko, G.; Ferenci, P.; Pol, S.; Goeser, T.; Bronowicki, J.-P.; Bourlière, M.; Gharakhanian, S.; Bengtsson, L.; et al. Telaprevir and peginterferon with or without ribavirin for chronic HCV infection. N. Engl. J. Med. 2009, 360, 1839–1850. [Google Scholar] [CrossRef]
  52. McHutchison, J.G.; Manns, M.P.; Muir, A.J.; Terrault, N.A.; Jacobson, I.M.; Afdhal, N.H.; Heathcote, E.J.; Zeuzem, S.; Reesink, H.W.; Garg, J.; et al. Telaprevir for previously treated chronic HCV infection. N. Engl. J. Med. 2010, 362, 1292–1303. [Google Scholar] [CrossRef] [PubMed]
  53. Jacobson, I.M.; McHutchison, J.G.; Dusheiko, G.; Di Bisceglie, A.M.; Reddy, K.R.; Bzowej, N.H.; Marcellin, P.; Muir, A.J.; Ferenci, P.; Flisiak, R.; et al. Telaprevir for previously untreated chronic hepatitis C virus infection. N. Engl. J. Med. 2011, 364, 2405–2416. [Google Scholar] [CrossRef] [PubMed]
  54. Zeuzem, S.; Andreone, P.; Pol, S.; Lawitz, E.; Diago, M.; Roberts, S.; Focaccia, R.; Younossi, Z.; Foster, G.R.; Horban, A.; et al. Telaprevir for retreatment of HCV infection. N. Engl. J. Med. 2011, 364, 2417–2428. [Google Scholar] [CrossRef]
  55. Kwo, P.Y.; Lawitz, E.J.; McCone, J.; Schiff, E.R.; Vierling, J.M.; Pound, D.; Davis, M.N.; Galati, J.S.; Gordon, S.C.; Ravendhran, N.; 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. [Google Scholar] [CrossRef] [PubMed]
  56. Poordad, F.; McCone, J., Jr.; Bacon, B.R.; Bruno, S.; Manns, M.P.; Sulkowski, M.S.; Jacobson, I.M.; Reddy, K.R.; Goodman, Z.D.; Boparai, N.; et al. Boceprevir for untreated chronic HCV genotype 1 infection. N. Engl. J. Med. 2011, 364, 1195–1206. [Google Scholar] [CrossRef]
  57. Bacon, B.R.; Gordon, S.C.; Lawitz, E.; Marcellin, P.; Vierling, J.M.; Zeuzem, S.; Poordad, F.; Goodman, Z.D.; Sings, H.L.; Boparai, N.; et al. Boceprevir for previously treated chronic HCV genotype 1 infection. N. Engl. J. Med. 2011, 364, 1207–1217. [Google Scholar] [CrossRef]
  58. Sarrazin, C. The importance of resistance to direct antiviral drugs in HCV infection in clinical practice. J. Hepatol. 2016, 64, 486–504. [Google Scholar] [CrossRef]
  59. Hézode, C. Boceprevir and telaprevir for the treatment of chronic hepatitis C: Safety management in clinical practice. Liver Int. 2012, 32 (Suppl. S1), 32–38. [Google Scholar] [CrossRef] [PubMed]
  60. Patel, P.; Malik, K.; Krishnamurthy, K. Cutaneous Adverse Events in Chronic Hepatitis C Patients Treated with New Direct-Acting Antivirals: A Systematic Review and Meta-Analysis. J. Cutan. Med. Surg. 2016, 20, 58–66. [Google Scholar] [CrossRef]
  61. Mauss, S.; Hueppe, D.; Alshuth, U. Renal impairment is frequent in chronic hepatitis C patients under triple therapy with telaprevir or boceprevir. Hepatology 2014, 59, 46–48. [Google Scholar] [CrossRef]
  62. Back, D.; Else, L. The importance of drug-drug interactions in the DAA era. Dig. Liver Dis. 2013, 45 (Suppl. S5), S343–S348. [Google Scholar] [CrossRef]
  63. Talavera Pons, S.; Lamblin, G.; Boyer, A.; Sautou, V.; Abergel, A. Drug interactions and protease inhibitors used in the treatment of hepatitis C: How to manage? Eur. J. Clin. Pharmacol. 2014, 70, 775–789. [Google Scholar] [CrossRef] [PubMed]
  64. Burger, D.; Back, D.; Buggisch, P.; Buti, M.; Craxí, A.; Foster, G.; Klinker, H.; Larrey, D.; Nikitin, I.; Pol, S.; et al. Clinical management of drug-drug interactions in HCV therapy: Challenges and solutions. J. Hepatol. 2013, 58, 792–800. [Google Scholar] [CrossRef] [PubMed]
  65. Kiser, J.J.; Burton, J.R., Jr.; Everson, G.T. Drug-drug interactions during antiviral therapy for chronic hepatitis C. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 596–606. [Google Scholar] [CrossRef] [PubMed]
  66. Pawlotsky, J.M. New hepatitis C therapies: The toolbox, strategies, and challenges. Gastroenterology 2014, 146, 1176–1192. [Google Scholar] [CrossRef]
  67. Feeney, E.R.; Chung, R.T. Antiviral treatment of hepatitis C. BMJ 2014, 348, g3308. [Google Scholar] [CrossRef]
  68. Abraham, G.M.; Spooner, L.M. Sofosbuvir in the treatment of chronic hepatitis C: New dog, new tricks. Clin. Infect. Dis. 2014, 59, 411–415. [Google Scholar] [CrossRef]
  69. Feld, J.J. Interferon-free strategies with a nucleoside/nucleotide analogue. Semin. Liver Dis. 2014, 34, 37–46. [Google Scholar] [CrossRef]
  70. Noell, B.C.; Besur, S.V.; deLemos, A.S. Changing the face of hepatitis C management—The design and development of sofosbuvir. Drug Des. Dev. Ther. 2015, 9, 2367–2374. [Google Scholar]
  71. Gutierrez, J.A.; Lawitz, E.J.; Poordad, F. Interferon-free, direct-acting antiviral therapy for chronic hepatitis C. J. Viral Hepat. 2015, 22, 861–870. [Google Scholar] [CrossRef]
  72. Keating, G.M. Ledipasvir/Sofosbuvir: A review of its use in chronic hepatitis C. Drugs 2015, 75, 675–685. [Google Scholar] [CrossRef] [PubMed]
  73. Fourati, S.; Guedj, J.; Chevaliez, S.; Nguyen, T.H.T.; Roudot-Thoraval, F.; Ruiz, I.; Soulier, A.; Scoazec, G.; Varaut, A.; Poiteau, L.; et al. Viral kinetics analysis and virological characterization of treatment failures in patients with chronic hepatitis C treated with sofosbuvir and an NS5A inhibitor. Aliment. Pharmacol. Ther. 2018, 47, 665–673. [Google Scholar] [CrossRef] [PubMed]
  74. Buti, M.; Riveiro-Barciela, M.; Esteban, R. Management of direct-acting antiviral agent failures. J. Hepatol. 2015, 63, 1511–1522. [Google Scholar] [CrossRef]
  75. Cuenca-Lopez, F.; Rivero, A.; Rivero-Juárez, A. Pharmacokinetics and pharmacodynamics of sofosbuvir and ledipasvir for the treatment of hepatitis C. Expert Opin. Drug Metab. Toxicol. 2017, 13, 105–112. [Google Scholar] [CrossRef] [PubMed]
  76. Uchida, Y.; Kouyama, J.I.; Naiki, K.; Sugawara, K.; Inao, M.; Imai, Y.; Nakayama, N.; Mochida, S. Development of rare resistance-associated variants that are extremely tolerant against NS5A inhibitors during daclatasvir/asunaprevir therapy by a two-hit mechanism. Hepatol. Res. 2016, 46, 1234–1246. [Google Scholar] [CrossRef]
  77. Pearlman, B.L.; Hinds, A.E. Review article: Novel antivirals for hepatitis C-sofosbuvir/velpatasvir/voxilaprevir, glecaprevir/pibrentasvir. Aliment. Pharmacol. Ther. 2018, 48, 914–923. [Google Scholar] [CrossRef] [PubMed]
  78. Lawitz, E.; Mangia, A.; Wyles, D.; Rodriguez-Torres, M.; Hassanein, T.; Gordon, S.C.; Schultz, M.; Davis, M.N.; Kayali, Z.; Reddy, K.R.; et al. Sofosbuvir for previously untreated chronic hepatitis C infection. N. Engl. J. Med. 2013, 368, 1878–1887. [Google Scholar] [CrossRef]
  79. Jacobson, I.M.; Gordon, S.C.; Kowdley, K.V.; Yoshida, E.M.; Rodriguez-Torres, M.; Sulkowski, M.S.; Shiffman, M.L.; Lawitz, E.; Everson, G.; Bennett, M.; et al. Sofosbuvir for hepatitis C genotype 2 or 3 in patients without treatment options. N. Engl. J. Med. 2013, 368, 1867–1877. [Google Scholar] [CrossRef] [PubMed]
  80. Lawitz, E.; Jacobson, I.M.; Nelson, D.R.; Zeuzem, S.; Sulkowski, M.S.; Esteban, R.; Brainard, D.; McNally, J.; Symonds, W.T.; McHutchison, J.G.; et al. Development of sofosbuvir for the treatment of hepatitis C virus infection. Ann. N. Y. Acad. Sci. 2015, 1358, 56–67. [Google Scholar] [CrossRef]
  81. Younossi, Z.M.; Stepanova, M.; Nader, F.; Jacobson, I.M.; Gane, E.; Nelson, D.; Lawitz, E.; Hunt, S.L. Patient-reported outcomes in chronic hepatitis C patients with cirrhosis treated with sofosbuvir-containing regimens. Hepatology 2014, 59, 2161–2169. [Google Scholar] [CrossRef]
  82. Curry, M.P.; Forns, X.; Chung, R.T.; Terrault, N.A.; Brown, R., Jr.; Fenkel, J.M.; Gordon, F.; O’Leary, J.; Kuo, A.; Schiano, T.; et al. Sofosbuvir and ribavirin prevent recurrence of HCV infection after liver transplantation: An open-label study. Gastroenterology 2015, 148, 100–107. [Google Scholar] [CrossRef] [PubMed]
  83. Gane, E.J.; Agarwal, K. Directly acting antivirals (DAAs) for the treatment of chronic hepatitis C virus infection in liver transplant patients: “A flood of opportunity”. Am. J. Transplant. 2014, 14, 994–1002. [Google Scholar] [CrossRef]
  84. Charlton, M.; Gane, E.; Manns, M.P.; Brown, R.S.; Curry, M.P.; Kwo, P.Y.; Fontana, R.J.; Gilroy, R.; Teperman, L.; Muir, A.J.; et al. Sofosbuvir and ribavirin for treatment of compensated recurrent hepatitis C virus infection after liver transplantation. Gastroenterology 2015, 148, 108–117. [Google Scholar] [CrossRef] [PubMed]
  85. Afdhal, N.; Zeuzem, S.; Kwo, P.; Chojkier, M.; Gitlin, N.; Puoti, M.; Romero-Gomez, M.; Zarski, J.-P.; Agarwal, K.; Buggisch, P.; et al. Ledipasvir and sofosbuvir for untreated HCV genotype 1 infection. N. Engl. J. Med. 2014, 370, 1889–1898. [Google Scholar] [CrossRef]
  86. Afdhal, N.; Reddy, K.R.; Nelson, D.R.; Lawitz, E.; Gordon, S.C.; Schiff, E.; Nahass, R.; Ghalib, R.; Gitlin, N.; Herring, R.; et al. Ledipasvir and sofosbuvir for previously treated HCV genotype 1 infection. N. Engl. J. Med. 2014, 370, 1483–1493. [Google Scholar] [CrossRef] [PubMed]
  87. Lawitz, E.; Poordad, F.F.; Pang, P.S.; Hyland, R.H.; Ding, X.; Mo, H.; Symonds, W.T.; McHutchison, J.G.; Membreno, F.E. Sofosbuvir and ledipasvir fixed-dose combination with and without ribavirin in treatment-naive and previously treated patients with genotype 1 hepatitis C virus infection (LONESTAR): An open-label, randomised, phase 2 trial. Lancet 2014, 383, 515–523. [Google Scholar] [CrossRef] [PubMed]
  88. Kowdley, K.V.; Gordon, S.C.; Reddy, K.R.; Rossaro, L.; Bernstein, D.E.; Lawitz, E.; Shiffman, M.L.; Schiff, E.; Ghalib, R.; Ryan, M.; et al. Ledipasvir and sofosbuvir for 8 or 12 weeks for chronic HCV without cirrhosis. N. Engl. J. Med. 2014, 370, 1879–1888. [Google Scholar] [CrossRef]
  89. Poordad, F.; Sievert, W.; Mollison, L.; Bennett, M.; Tse, E.; Bräu, N.; Levin, J.; Sepe, T.; Lee, S.S.; Angus, P.; et al. Fixed-dose combination therapy with daclatasvir, asunaprevir, and beclabuvir for noncirrhotic patientswith HCV genotype 1 infection. JAMA 2015, 313, 1728–1735. [Google Scholar] [CrossRef]
  90. Muir, A.J.; Poordad, F.; Lalezari, J.; Everson, G.; Dore, G.J.; Herring, R.; Sheikh, A.; Kwo, P.; Hézode, C.; Pockros, P.J.; et al. Daclatasvir in combination with asunaprevir and beclabuvirfor hepatitis C virus genotype 1 infection with compensated cirrhosis. JAMA 2015, 313, 1736–1744. [Google Scholar] [CrossRef]
  91. Naggie, S.; Cooper, C.; Saag, M.; Workowski, K.; Ruane, P.; Towner, W.J.; Marks, K.; Luetkemeyer, A.; Baden, R.P.; Sax, P.E.; et al. Ledipasvir and Sofosbuvir for HCV in Patients Coinfected with HIV-1. N. Engl. J. Med. 2015, 373, 705–713. [Google Scholar] [CrossRef]
  92. Liao, H.T.; Tan, P.; Huang, J.W.; Yuan, K.F. Ledipasvir + Sofosbuvir for Liver Transplant Recipients with Recurrent Hepatitis C: A Systematic Review and Meta-Analysis. Transplant. Proc. 2017, 49, 1855–1863. [Google Scholar] [CrossRef]
  93. Cotter, T.G.; Jensen, D.M. Glecaprevir/pibrentasvir for the treatment of chronic hepatitis C: Design, development, and place in therapy. Drug Des. Dev. Ther. 2019, 13, 2565–2577. [Google Scholar] [CrossRef] [PubMed]
  94. Hubbard, H.; Lawitz, E. Glecaprevir + pibrentasvir (ABT493 + ABT-530) for the treatment of Hepatitis C. Expert Rev. Gastroenterol. Hepatol. 2018, 12, 9–17. [Google Scholar] [CrossRef] [PubMed]
  95. Jackson, W.E.; Everson, G.T. Sofosbuvir and velpatasvir for the treatment of hepatitis C. Expert Rev. Gastroenterol. Hepatol. 2017, 11, 501–505. [Google Scholar] [CrossRef]
  96. Chahine, E.B.; Sucher, A.J.; Hemstreet, B.A. Sofosbuvir/Velpatasvir: The First Pangenotypic Direct-Acting Antiviral Combination for Hepatitis C. Ann. Pharmacother. 2017, 51, 44–53. [Google Scholar] [CrossRef] [PubMed]
  97. Lee, R.; Kottilil, S.; Wilson, E. Sofosbuvir/velpatasvir: A pangenotypic drug to simplify HCV therapy. Hepatol. Int. 2017, 11, 161–170. [Google Scholar] [CrossRef] [PubMed]
  98. Toyoda, H. Sofosbuvir/velpatasvir and glecaprevir/pibrentasvir: How should we choose among these two latest and probably the last regimens? Hepatol. Res. 2022, 52, 815–816. [Google Scholar] [CrossRef]
  99. Feld, J.J.; Jacobson, I.M.; Hézode, C.; Asselah, T.; Ruane, P.J.; Gruener, N.; Abergel, A.; Mangia, A.; Lai, C.-L.; Chan, H.L.Y.; et al. Sofosbuvir and Velpatasvir for HCV Genotype 1, 2, 4, 5, and 6 Infection. N. Engl. J. Med. 2015, 373, 2599–2607. [Google Scholar] [CrossRef]
  100. Foster, G.R.; Afdhal, N.; Roberts, S.K.; Bräu, N.; Gane, E.J.; Pianko, S.; Lawitz, E.; Thompson, A.; Shiffman, M.L.; Cooper, C.; et al. Sofosbuvir and Velpatasvir for HCV Genotype 2 and 3 Infection. N. Engl. J. Med. 2015, 373, 2608–2617. [Google Scholar] [CrossRef]
  101. Curry, M.P.; O’Leary, J.G.; Bzowej, N.; Muir, A.J.; Korenblat, K.M.; Fenkel, J.M.; Reddy, K.R.; Lawitz, E.; Flamm, S.L.; Schiano, T.; et al. Sofosbuvir and Velpatasvir for HCV in Patients with Decompensated Cirrhosis. N. Engl. J. Med. 2015, 373, 2618–2628. [Google Scholar] [CrossRef]
  102. Forns, X.; Lee, S.S.; Valdes, J.; Lens, S.; Ghalib, R.; Aguilar, H.; Felizarta, F.; Hassanein, T.; Hinrichsen, H.; Rincon, D.; et al. Glecaprevir plus pibrentasvir for chronic hepatitis C virus genotype 1, 2, 4, 5, or 6 infection in adults with compensated cirrhosis (EXPEDITION-1): A single-arm, open-label, multicentre phase 3 trial. Lancet Infect. Dis. 2017, 17, 1062–1068. [Google Scholar] [CrossRef] [PubMed]
  103. Zeuzem, S.; Foster, G.R.; Wang, S.; Asatryan, A.; Gane, E.; Feld, J.J.; Asselah, T.; Bourlière, M.; Ruane, P.J.; Wedemeyer, H.; et al. Glecaprevir-Pibrentasvir for 8 or 12 Weeks in HCV Genotype 1 or 3 Infection. N. Engl. J. Med. 2018, 378, 354–369. [Google Scholar] [CrossRef]
  104. Bourlière, M.; Gordon, S.C.; Flamm, S.L.; Cooper, C.L.; Ramji, A.; Tong, M.; Ravendhran, N.; Vierling, J.M.; Tran, T.T.; Pianko, S.; et al. Sofosbuvir, Velpatasvir, and Voxilaprevir for Previously Treated HCV Infection. N. Engl. J. Med. 2017, 37, 2134–2146. [Google Scholar] [CrossRef] [PubMed]
  105. Feld, J.J.; Forns, X.; Dylla, D.E.; Kumada, H.; de Ledinghen, V.; Wei, L.; Brown, R.S.; Flisiak, R.; Lampertico, P.; Thabut, D.; et al. Safety analysis of glecaprevir/pibrentasvir in patients with markers of advanced liver disease in clinical and real-world cohorts. J. Viral Hepat. 2022, 29, 1050–1061. [Google Scholar] [CrossRef]
  106. Forns, X.; Feld, J.J.; Dylla, D.E.; Pol, S.; Chayama, K.; Hou, J.; Heo, J.; Lampertico, P.; Brown, A.; Bondin, M.; et al. Safety of Patients with Hepatitis C Virus Treated with Glecaprevir/Pibrentasvir from Clinical Trials and Real-World Cohorts. Adv. Ther. 2021, 38, 3409–3426. [Google Scholar] [CrossRef] [PubMed]
  107. Takehara, T.; Izumi, N.; Mochida, S.; Genda, T.; Fujiyama, S.; Notsumata, K.; Tamori, A.; Suzuki, F.; Suri, V.; Mercier, R.; et al. Sofosbuvir-velpatasvir in adults with hepatitis C virus infection and compensated cirrhosis in Japan. Hepatol. Res. 2022, 52, 833–840. [Google Scholar] [CrossRef]
  108. Flamm, S.; Lawitz, E.; Borg, B.; Charlton, M.; Landis, C.; Reddy, K.R.; Shiffman, M.; Alsina, A.; Chang, C.; Ravendhran, N.; et al. Efficacy and Safety of Sofosbuvir/Velpatasvir Plus Ribavirin in Patients with Hepatitis C Virus-Related Decompensated Cirrhosis. Viruses 2023, 15, 2026. [Google Scholar] [CrossRef]
  109. Llaneras, J.; Riveiro-Barciela, M.; Lens, S.; Diago, M.; Cachero, A.; García-Samaniego, J.; Conde, I.; Arencibia, A.; Arenas, J.; Gea, F.; et al. Effectiveness and safety of sofosbuvir/velpatasvir/voxilaprevir in patients with chronic hepatitis C previously treated with DAAs. J. Hepatol. 2019, 71, 666–672. [Google Scholar] [CrossRef]
  110. Ruiz-Cobo, J.C.; Llaneras, J.; Forns, X.; Moya, A.G.; Amiel, I.C.; Arencibia, A.; Diago, M.; García-Samaniego, J.; Castellote, J.; Llerena, S.; et al. Real-life effectiveness of sofosbuvir/velpatasvir/voxilaprevir in hepatitis C patients previously treated with sofosbuvir/velpatasvir or glecaprevir/pibrentasvir. Aliment. Pharmacol. Ther. 2024, 60, 201–211. [Google Scholar] [CrossRef]
  111. Reiberger, T.; Lens, S.; Cabibbo, G.; Nahon, P.; Zignego, A.L.; Deterding, K.; Elsharkawy, A.M.; Forns, X. EASL position paper on clinical follow-up after HCV cure. J. Hepatol. 2024, 81, 326–344. [Google Scholar] [CrossRef]
  112. Harney, B.L.; Sacks-Davis, R.; van Santen, D.K.; Traeger, M.W.; Wilkinson, A.L.; Asselin, J.; Fairley, C.K.; Roth, N.; Bloch, M.; Matthews, G.V.; et al. Hepatitis C virus reinfection incidence among gay and bisexual men with HIV in Australia from 2016 to 2020. Liver Int. 2024, 44, 1024–1031. [Google Scholar] [CrossRef] [PubMed]
  113. Young, J.; Wang, S.; Lanièce Delaunay, C.; Cooper, C.L.; Cox, J.; Gill, M.J.; Hull, M.; Walmsley, S.; Wong, A.; Klein, M.B. The rate of hepatitis C reinfection in Canadians coinfected with HIV and its implications for national elimination. Int. J. Drug Policy 2023, 114, 103981. [Google Scholar] [CrossRef] [PubMed]
  114. Karasz, A.; Merchant, K.; Singh, R.; Thomas, A.; Borsuk, C.; McKee, D.; Duryea, P.; Kim, A.Y.; Mehta, S.; Norton, B.L.; et al. The experience of re-infection among people who inject drugs successfully treated for hepatitis C. J. Subst. Use Addict. Treat. 2023, 146, 208937. [Google Scholar] [CrossRef] [PubMed]
  115. Cui, F.; Blach, S.; Manzengo Mingiedi, C.; Gonzalez, M.A.; Alaama, A.S.; Mozalevskis, A.; Séguy, N.; Rewari, B.B.; Chan, P.-L.; Le, L.-V.; et al. Global reporting of progress towards elimination of hepatitis B and hepatitis C. Lancet Gastroenterol. Hepatol. 2023, 8, 332–342. [Google Scholar] [CrossRef]
  116. Razavi, H. Polaris Observatory-supporting informed decision-making at the national, regional, and global levels to eliminate viral hepatitis. Antivir. Ther. 2022, 27, 13596535221083179. [Google Scholar] [CrossRef]
  117. Tu, S.S.; Kottilil, S.; Mattingly, T.J. Leveraging Old Hepatitis C Therapies. N. Engl. J. Med. 2025, 392, 2–4. [Google Scholar] [CrossRef]
Table 1. Interferon-based therapy.
Table 1. Interferon-based therapy.
Authors Ref Year GenotypesTreatment RegimensWeeks of TherapyNumber of PatientsSVR (%)
McHutchison et al. [33]19981,2,3IFN α-2b 3 MU tiw242316
IFN α-2b 3 MU tiw4822513
IFN α-2b 3 MU tiw plus RBV 1000–1200 mg/day2422831
IFN α-2b 3 MU tiw plus RBV 1000–1200 mg/day4822838
Poynard[34]19981,2,3,4IFN α-2b 3 MU tiw plus RBV 1000–1200 mg/day 48 277 43
IFN α-2b 3 MU tiw plus RBV 1000–1200 mg/day2427735
IFN α-2b 3 MU tiw plus Placebo 48 278 19
Lindsay et al.[35]20011,2,3,4PEG-IFN α-2b 0.5 μg/kg 4831518
PEG-IFN α-2b 1.0 μg/kg4829725
PEG-IFN α-2b 1.5 μg/kg 4830423
IFN α-2b 3 MU tiw4830312
Zeuzem et al.[36]20001,2,3,4PEG-IFN α-2a 180 μg4826739
IFN α-2a 6 and then 3 MU tiw 4826419
Heathcote et al.
All patients stage 3/4
[37]20001,2,3,4PEG-IFN α-2a 90 μg 489615
PEG-IFN α-2a 180 μg488730
IFN α-2a 3 MU tiw488812
Manns et al.[38]20011,2,3,4PEG-IFN α-2b 1.5 μg/kg plus RBV 800 mg/day4851154
PEG-IFN α-2b 1.5 μg/kg for 4 weeks then 0.5 μg/kg for 44 weeks plus RBV 1000–1200 mg/day4851447
IFN α-2b 3 MU tiw plus RBV 1000–1200 mg/day4850547
Fried et al.[39]20021,2,3,4PEG-IFN α-2a 180 μg plus RBV 1000–1200 mg/day4845356
PEG-IFN α-2a 180 μg4822429
IFN α-2b 3 MU tiw plus RBV 1000–1200 mg/day4844444
Kamal et al. [40]20074PEG-IFN α-2b 1.5 μg/kg plus RBV (10.6 mg/kg/day)485056
PEG-IFN α-2b 1.5 μg/kg plus RBV (10.6 mg/kg/day)246986
PEG-IFN α-2b 1.5 μg/kg plus RBV (10.6 mg/kg/day)367976
PEG-IFN α-2b 1.5 μg/kg plus RBV (10.6 mg/kg/day)4816058
McHutchison et al. [41]20091PEG-IFN α-2b 1.5 μg/kg plus RBV 800–1400 mg/day48101940
PEG-IFN α-2b 1.0 μg/kg plus RBV 800–1400 mg/day48101538
PEG-IFN α-2a 180 μg plus RBV 800–1400 mg/day48103541
Notes: MU = mega units, tiw = three times a week.
Table 2. Therapy with first-generation direct-acting antivirals (DAAs).
Table 2. Therapy with first-generation direct-acting antivirals (DAAs).
Authors Ref Year GenotypesVirological StatusTreatment RegimensWeeks of TherapyNumber of PatientsSVR (%)
McHutchison GJ et al. [50]20091untreated Placebo for 12 weeks plus peginterferon alfa-2a and ribavirin for 48 weeks487541
telaprevir, peginterferon alfa-2a and ribavirin for 12 weeks 121735
telaprevir for 12 weeks plus peginterferon alfa-2a and ribavirin for 24 weeks247961
telaprevir for 12 weeks plus peginterferon alfa-2a and ribavirin for 48 weeks. 487967
Hézode C et al. [51] 1untreated telaprevir, peginterferon alfa-2a and ribavirin for 12 weeks, followed by peginterferon alfa-2a and ribavirin for 12 more weeks. 248169
telaprevir, peginterferon alfa-2a and ribavirin for 12 weeks,128248
telaprevir and peginterferon alfa-2a for 12 weeks,127836
peginterferon alfa-2a and ribavirin for 48 weeks.488146
McHutchison JG et al. [52] 1previously treatedtelaprevir for 12 weeks plus peginterferon alfa-2a and ribavirin for 24 weeks; 2411551
telaprevir for 24 weeks plus peginterferon alfa-2a and ribavirin for 48 weeks4811353
telaprevir for 24 weeks plus peginterferon alfa-2a and ribavirin for 24 weeks2411124
peginterferon alfa-2a and ribavirin for 48 weeks 4811414
Jacobson IM et al. [53] 1untreated telaprevir combined with peginterferon alfa-2a and ribavirin for 12 weeks (T12PR group), followed by peginterferon-ribavirin alone for 12 weeks if HCV RNA was undetectable at weeks 4 and 12 or for 36 weeks if HCV RNA was detectable at either time point;24–3636375
telaprevir with peginterferon-ribavirin for 8 weeks and placebo with peginterferon-ribavirin for 4 weeks (T8PR group), followed by 12 or 36 weeks of peginterferon-ribavirin on the basis of the same HCV RNA criteria; 36–4836469
placebo with peginterferon-ribavirin for 12 weeks, followed by 36 weeks of peginterferon-ribavirin 4836144
Zeuzem S et al. [54] 1previously treatedthe T12PR48 group, received telaprevir for 12 weeks and peginterferon plus ribavirin for a total of 48 weeks;4826683
T12PR48 group, received 4 weeks of peginterferon plus ribavirin followed by 12 weeks of telaprevir and peginterferon plus ribavirin for a total of 48 weeks; 4826488
peginterferon plus ribavirin for 48 weeks. 4813224
Kwo PY et al. [55] 1untreated peginterferon alfa-2b plus ribavirin for 48 weeks 4810438
peginterferon alfa-2b plus ribavirin for 4 weeks, followed by peginterferon alfa-2b, ribavirin, and boceprevir for 24 weeks2810354
peginterferon alfa-2b plus ribavirin for 4 weeks followed by peginterferon alfa-2b, ribavirin, and boceprevir for 44 weeks 4810356
peginterferon alfa-2b plus ribavirin and boceprevir for 28 weeks 2810767
peginterferon alfa-2b plus ribavirin and boceprevir for 48 weeks. 4810375
Poordad F et al. [56] 1untreated peginterferon alfa-2b and ribavirin were administered for 4 weeks (the lead-in period) and subsequently, placebo plus peginterferon-ribavirin for 44 weeks;4836338
peginterferon alfa-2b and ribavirin were administered for 4 weeks (the lead-in period) and subsequently boceprevir plus peginterferon-ribavirin for 24 weeks, and those with a detectable HCV RNA level between weeks 8 and 24 received placebo plus peginterferon-ribavirin for an additional 20 weeks4836863
peginterferon alfa-2b and ribavirin for 4 weeks (the lead-in period) and subsequently boceprevir plus peginterferon-ribavirin for 44 weeks.4836666
Bacon BR et al. [57] 1previously treatedpeginterferon alfa-2b and ribavirin for 4 weeks (the lead-in period) and subsequently placebo plus peginterferon-ribavirin for 44 weeks;488021
peginterferon alfa-2b and ribavirin for 4 weeks (the lead-in period) and subsequently boceprevir plus peginterferon-ribavirin for 32 weeks, and patients with a detectable HCV RNA level at week 8 received placebo plus peginterferon-ribavirin for an additional 12 weeks; 4816259
peginterferon alfa-2b and ribavirin were administered for 4 weeks (the lead-in period) and subsequently received boceprevir plus peginterferon-ribavirin for 44 weeks.4816166
Table 3. DAAs of the second and third generation with the protein inhibited, typical combination and year of approval.
Table 3. DAAs of the second and third generation with the protein inhibited, typical combination and year of approval.
DAAProtein InhibitedTypical CombinationYear of Approval
SofosbuvirNS5B (polymerase)Sofosbuvir + Ledipasvir2013
LedipasvirNS5ASofosbuvir + Ledipasvir2014
OmbitasvirNS5AOmbitasvir + Paritaprevir + Dasabuvir2014
ParitaprevirNS3/4A (protease)Ombitasvir + Paritaprevir + Dasabuvir2014
DasabuvirNS5B (non-nucleoside)Ombitasvir + Paritaprevir + Dasabuvir2014
AsunaprevirNS3/4A (protease)Beclabuvir + Daclatasvir + Asunaprevir2014
DaclatasvirNS5ASofosbuvir + Daclatasvir2015
BeclabuvirNS5B (non-nucleoside)Beclabuvir + Daclatasvir + Asunaprevir2015
VelpatasvirNS5ASofosbuvir + Velpatasvir2016
ElbasvirNS5AGrazoprevir + Elbasvir2016
GrazoprevirNS3/4A (protease)Grazoprevir + Elbasvir2016
GlecaprevirNS3/4A (protease)Glecaprevir + Pibrentasvir2017
PibrentasvirNS5AGlecaprevir + Pibrentasvir2017
VoxilaprevirNS3/4A (protease)Sofosbuvir + Velpatasvir + Voxilaprevir2017
Table 4. Therapy with second-generation direct-acting antivirals (DAAs).
Table 4. Therapy with second-generation direct-acting antivirals (DAAs).
Authors Ref Year GenotypesVirological StatusTreatment RegimensWeeks of TherapyNumber of PatientsSVR
(%)
Afdhal N et al. [84]20141untreated sofosbuvir-ledipasvir 12 21499
sofosbuvir-ledipasvir plus ribavirin 1221797
sofosbuvir-ledipasvir 2421798
sofosbuvir-ledipasvir plus ribavirin 2421799
Afdhal N et al. [85]20141previously treatedsofosbuvir-ledipasvir 12 10994
sofosbuvir-ledipasvir plus ribavirin 1211196
sofosbuvir-ledipasvir 2410999
sofosbuvir-ledipasvir plus ribavirin 2411199
Lawitz E et al.[86]20141untreated (Cohort A)sofosbuvir-ledipasvir 8 2095
sofosbuvir-ledipasvir plus ribavirin 821100
sofosbuvir-ledipasvir 121995
1previously treated with a protease-inhibitor regimen (Cohort B)sofosbuvir-ledipasvir 121995
sofosbuvir-ledipasvir plus ribavirin 1220100
Kowdley KV et al. [87]20141untreatedsofosbuvir-ledipasvir 821594
sofosbuvir-ledipasvir plus ribavirin 821693
sofosbuvir-ledipasvir 1221595
Poordard F et al.[88]20151untreateddaclatasvir—asunaprevir beclabuvir1231292
previously treateddaclatasvir—asunaprevir-beclabuvir1210389
Muir AJ et al. [89]20151untreated cirrhosisdaclatasvir—asunaprevir-beclabuvir125793
daclatasvir—asunaprevir-beclabuvir plus ribavirin 125598
previously treated cirrhosis daclatasvir—asunaprevir-beclabuvir124586
daclatasvir—asunaprevir-beclabuvir plus ribavirin 124593
Table 5. Therapy with pan-genotypic direct-acting antivirals (DAAs).
Table 5. Therapy with pan-genotypic direct-acting antivirals (DAAs).
Authors Ref Year GenotypesVirological StatusTreatment RegimensWeeks of TherapyNumber of PatientsSVR
(%)
Feld JJ et al. [99]20151,2,4,5,6 untreated (68%)
previously treated (32%)
Sofosbuvir–Velpatasvir 1262499
Placebo121160
Foster GR et al. [100]20152untreated
previously treated
Sofosbuvir–Velpatasvir 1213499
Sofosbuvir–Ribavirin 1213294
3untreated
previously treated
Sofosbuvir–Velpatasvir 1227795
Sofosbuvir–Ribavirin 2427580
Curry MP et al. [101]20151,2,3,4,5,6untreated and previously treated decompensated cirrhosis Sofosbuvir–Velpatasvir129083
Sofosbuvir–Velpatasvir plus Ribavirin 129894
Sofosbuvir–Velpatasvir249286
Forns X et al.[102]20171,2,4,5,6untreated and previously treated compensated cirrhosis Glecaprevir-Pibrentasvir 1214699
Zeuzem S et al.[103] 1untreated
previously treated
Glecaprevir-Pibrentasvir 835199
Glecaprevir-Pibrentasvir 1235299
3untreated Glecaprevir-Pibrentasvir 1223395
Sofosbuvir-Daclatasvir1211597
Glecaprevir-Pibrentasvir 815795
Bourlière M et al. [104] 1
(Polaris-1)
previously treated with a regimen containing an NS5A inhibitor Sofosbuvir–Velpatasvir–Voxilaprevir 1226396
Placebo 121520
1,2,3 (Polaris-4)previously treated without a regimen containing an NS5A inhibitor Sofosbuvir–Velpatasvir–Voxilaprevir 1218298
Sofosbuvir–Velpatasvir 1215190
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Di Marco, L.; Cannova, S.; Ferrigno, E.; Landro, G.; Nonni, R.; Mantia, C.L.; Cartabellotta, F.; Calvaruso, V.; Di Marco, V. A Comprehensive Review of Antiviral Therapy for Hepatitis C: The Long Journey from Interferon to Pan-Genotypic Direct-Acting Antivirals (DAAs). Viruses 2025, 17, 163. https://doi.org/10.3390/v17020163

AMA Style

Di Marco L, Cannova S, Ferrigno E, Landro G, Nonni R, Mantia CL, Cartabellotta F, Calvaruso V, Di Marco V. A Comprehensive Review of Antiviral Therapy for Hepatitis C: The Long Journey from Interferon to Pan-Genotypic Direct-Acting Antivirals (DAAs). Viruses. 2025; 17(2):163. https://doi.org/10.3390/v17020163

Chicago/Turabian Style

Di Marco, Lorenza, Simona Cannova, Emanuele Ferrigno, Giuseppe Landro, Rosario Nonni, Claudia La Mantia, Fabio Cartabellotta, Vincenza Calvaruso, and Vito Di Marco. 2025. "A Comprehensive Review of Antiviral Therapy for Hepatitis C: The Long Journey from Interferon to Pan-Genotypic Direct-Acting Antivirals (DAAs)" Viruses 17, no. 2: 163. https://doi.org/10.3390/v17020163

APA Style

Di Marco, L., Cannova, S., Ferrigno, E., Landro, G., Nonni, R., Mantia, C. L., Cartabellotta, F., Calvaruso, V., & Di Marco, V. (2025). A Comprehensive Review of Antiviral Therapy for Hepatitis C: The Long Journey from Interferon to Pan-Genotypic Direct-Acting Antivirals (DAAs). Viruses, 17(2), 163. https://doi.org/10.3390/v17020163

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop