480
Views
7
CrossRef citations to date
0
Altmetric
Editorial

Ribavirin in chronic hepatitis C: past and future

, , , &
Pages 249-253 | Published online: 10 Jan 2014

Pegylated IFN-α and ribavirin is, at present, the standard of care for chronic hepatitis C, leading to a sustained virological response (SVR) in 45–80% of the patients depending on the genotype Citation[1–3]. Ribavirin is a key factor of this therapeutic strategy, doubling the SVR rate obtained with pegylated interferon alone Citation[2,4]. Ribavirin mainly acts by preventing relapses, especially in genotype 1 patients Citation[5]. The near future for the treatment of chronic hepatitis C is specifically targeted antiviral therapy (STAT-C). However, STAT-C and, notably, protease inhibitors are still used in combination with pegylated interferon and ribavirin. The absence of ribavirin in the drug combination resulted in a relapse rate of 48% Citation[6]. Finally, pharmacological and pharmacodynamic data showed that the current use of ribavirin according to bodyweight (BW) can lead to inadequate individual exposure to ribavirin, which in turn impacts the SVR, suggesting that optimizing individual ribavirin dose by monitoring the drug blood levels might be a key strategy to improve the SVR rate, particularly in difficult-to-treat patients Citation[7–10].

Clinical past of ribavirin

Ribavirin is a guanosine analog synthesized in the 1970s that has a broad-spectrum activity against DNA and RNA viruses. It was initially approved in clinics to treat respiratory syncytial virus infection in children and then Lassa fever Citation[11–13]. In 1991, Reichard et al. were the first to show that ribavirin alone can reduce serum alanine aminotransferase activity in chronic hepatitis C patients Citation[14]. In 1994, Brillanti et al. demonstrated that ribavirin associated with IFN-α gave promising results Citation[15]; these results were confirmed in 1995 by the first randomized study Citation[16], showing that ribavirin could double the SVR rate obtained with IFN-α alone. The benefit was particularly significant for genotype 1 patients. The antiviral effect of ribavirin and pegylated IFN-α was also found to be synergistic Citation[1,2], with ribavirin preventing relapses and breakthroughs Citation[5].

Ribavirin maintenance dose seems to be crucial for SVR. Data from two pivotal clinical trials highlighted that the total daily dose of ribavirin (>10.6 mg/kg of BW), especially during the first 3 months, was predictive of SVR Citation[1,17], and that ribavirin had to be administered for the full treatment duration Citation[5]. A higher starting dose of ribavirin reduced relapse in a controlled trial where patients were randomized to receive either 800 mg or 1000–1200 mg of ribavirin Citation[3]. In a pilot study, Lindhal et al. suggested that the early use of very high doses of ribavirin (mean daily dose: 2.5 g [1.6–3.6 g]) led to a higher SVR rate in genotype 1 patients Citation[18].

Ribavirin mechanisms of action

So far, the mechanisms of action of ribavirin are not well understood. The antiviral mechanism is probably the best documented. Once ribavirin enters the cell, it is phosphorylated to ribavirin mono-, di- and tri-phosphate (RMP, RDP and RTP). The erroneous incorporation of RTP into replicating RNA strands may inhibit chain elongation and cause chain termination Citation[19].

The inhibition of inosine monophosphate dehydrogenase (IMPDH), which is an enzyme essential for the de novo synthesis of GTP, is a potential mechanism of action: it may inhibit RNA synthesis and promote the incorporation of RTP in place of GTP. However, ribavirin’s impact on IMPDH is weaker than that of mycophenolate mofetil (MMF). In the replicon system, the addition of guanosine in excess completely restores the HCV replication rate with MMF, but only partially with ribavirin Citation[20].

The mutagenesis hypothesis has been recently prioritized. Ribavirin would lower the viral infectivity by virus mutagenesis. This mutagenesis might be higher in the presence of interferon. In the HCV replicon system, ribavirin was shown to inhibit the ability of progeny subgenomic replicons to transfect new cells, although the replication rate remained unaffected Citation[21,22]. However, a recent in vivo study undermined this hypothesis Citation[23].

The most attractive hypothesis was recently proposed by Feld et al.Citation[24]. The authors suggested that ribavirin enhances the response of interferon-stimulated genes to pegylated interferon and that pretreatment with ribavirin may heighten this induction, making cells more responsive to interferon and increasing the production of endogenous interferon.

Following the onset of double therapy with interferon and ribavirin, one can observe a two-phase profile of plasma RNA decline: a rapid first phase decline observed for 1–2 days Citation[25] attributed to a reduction in production and or release of new virions due to interferon action, and a slower second phase attributed to the elimination of infected cells. The influence of ribavirin on the first-phase decline of HCV RNA is weak and transient (0.5 log) and only observed in patients treated with ribavirin alone Citation[26]. The impact on the second slope is more probable and would involve mutagenesis and more likely enhancement of the interferon-stimulated genes’ response.

Pharmacokinetics of ribavirin

Individual factors probably influence ribavirin’s pharmacokinetics. There is no significant correlation between ribavirin exposure and total or weight-standardized dose. This reflects the large interindividual variability of the dose–concentration relationship. In a study by Bruchfeld et al., the effect of renal function on ribavirin clearance was only apparent when creatinine clearence was less than 34 ml/min Citation[27]. Other studies showed that ribavirin clearance was affected by BW, gender, age and serum creatinine, but that these four covariates only explained 27–40% of its interindividual variability Citation[27,28]. It is possible that the liver plays a role, although Glue et al. showed that hepatic dysfunction had no substantial influence on the apparent clearance of ribavirin Citation[29]. Finally, high-fat meals can increase ribavirin bioavailability by 46% compared with the fasting state Citation[30].

Data from the literature showed that, following a single oral dose, the plasma concentration of ribavirin exhibits a three-phase profile: a rapid absorption phase with a mean time to the maximum concentration (tmax) of approximately 1.5 h; a rapid distribution phase (half-life of ∼3.7 h); and a long terminal elimination phase, the last measurable concentration time point being at approximately 100 h post-dose Citation[19,31].

Ribavirin & anemia

Hemolytic anemia is the major adverse event that impairs the ability of patients to remain on full doses of the drug.

In nucleated cells, ribavirin phosphorylation is reversible, as opposed to anucleated blood cells where its concentrations can exceed that found in plasma by 60-fold. RTP enters in competition with ATP and the oxydative stress results in the alteration of the erythrocyte membrane fluidity and extravascular hemolysis. Ribavirin half-life in the body is 40 days, equal to the half-life of erythocytes before their destruction in the spleen Citation[32]. This hemolytic mechanism is reversible and dose dependent. Moreover, interferon contributes to anemia by its bone marrow-suppressive effect. The prevalence of anemia during standard bitherapy is high, with hemoglobin (Hb) less than 11 g/dl in 30% and less than 10 g/dl in 9–13% of patients Citation[1,2].

In total, 10–15% of patients present with severe anemia, with a decline of more than 5 g/dl in Hb levels. In real life, anemia is responsible for 36% of the HCV treatment interruptions Citation[33]. Erythropoietin improves the quality of life and can help to maintain the full dose of ribavirin by significantly increasing the Hb level, with acceptable tolerance Citation[34,35]. However, its impact on SVR has not been demonstrated Citation[36].

Monitoring ribavirin blood levels

The aforementioned clinical trials all emphasized the crucial role of the cumulative dose of ribavirin on SVR, especially during the first 3 months Citation[1,2,17]. Anemia is linked to ribavirin concentration, even if this relationship is highly variable Citation[7,9]. Moreover, ribavirin has a typical profile for drug monitoring: its half-life is long and there is a large interindividual variability of its dose–concentration relationship, that is, of its pharmacokinetics, despite dose adjustment on BW Citation[7,9,37].

It has recently been demonstrated that patients with genotype non-2 non-3 HCV who are partial or even complete early responders (HCV RNA level <2 logs IU or undetectable at week 12) have a probability of only 27 and 60% of SVR, respectively, and may greatly benefit from early monitoring of ribavirin concentrations. On the contrary, rapid virological responders (RVRs; HCV RNA undetectable at week 4), who represent only 16% of genotype 1 patients, have a high probability (80–90%) of SVR. These patients, who are very sensitive to the effect of interferon, may not so much require the maintenance of an optimal ribavirin exposure Citation[38,39].

A recent study in 35 HCV–HIV-coinfected patients suggested that the influence of ribavirin concentration on the SVR could be linked to the HCV genotype (i.e., clear influence on the virus response only for genotypes 1 or 4) Citation[40]. However, a Swedish study, the NORDynamic trial, conducted in a large population of 382 genotype 2- and 3-monoinfected patients found a statistical relationship between residual ribavirin concentrations at week 4 of treatment and SVR. The positive predictive value for SVR at week 4 was 91% for a ribavirin concentration more than 2 mg/l (p = 0.02) Citation[41]. Therefore, ribavirin residual concentrations seem to have an impact on SVR in all genotypes but this impact is probably greater in genotype 1 patients.

In the literature, the monitoring of ribavirin blood levels was commonly based on trough concentration measurements at weeks 4, 12 or 24 after the initiation of treatment Citation[7,10]. However, multiparametric statistical analyses showed that the relationship between trough concentration and SVR was less influential than the viral load and of course the genotype Citation[7]. A trough concentration above 2 mg/l at week 4 was predictive of SVR Citation[10]. Arase et al. demonstrated that high ribavirin trough concentrations at week 8 (>3.5 mg/l) were linked with a higher rate of ribavirin discontinuation because of anemia and suggested not to exceed this limit, considered as a security test Citation[42]. Another attempt of a single-point monitoring strategy, measuring at week 12 ribavirin concentration, 2–4 h after the morning dose (close to the peak concentration), showed a significant relationship with SVR in genotype 1 patients Citation[8]. However, all these approaches did not allow early ribavirin dose adjustment.

A patient’s global exposure to ribavirin, as evaluated by the AUC, seems more pertinent in terms of exposure–effect relationship than any single time point. We recently showed, in patients infected by genotype 1 HCV, that ribavirin plasma exposure after the first dose (i.e., interdose AUC0–12h or abbreviated AUC0–4h) was significantly and strongly linked with SVR, whereas AUCs determined at week 12 and 24 and trough concentrations at day 0 and week 12 were not Citation[9]. After the first dose, AUC0–12h was only slightly affected by the elimination phase (due to the very long half-life of the drug), while abbreviated AUC0–4h was clearly limited to ribavirin absorption and distribution.

Finally, based on receiver operating characteristic (ROC) curve analysis, we proposed a minimum AUC0–4h threshold of 1755 µg.h/l at day 0 as a target for ribavirin dose adjustment Citation[9]. On a practical purpose, AUC0–4h can be estimated using three blood samples (0.5, 1 and 2 h after the first dose) and Bayesian estimation.

The relationship between ribavirin concentration and early virological response (EVR) is unclear. In a retrospective study, Donnerer et al. found no correlation between ribavirin levels at week 12 and EVR Citation[43]. By contrast, recent findings in HCV–HIV-coinfected patients suggested that a high ribavirin concentration could improve the EVR rate Citation[44]. Ribavirin plasma AUC0–12h or AUC0–4h after the first dose were significantly linked to EVR, but to a lesser extent than to SVR and RVR Citation[9]. Explanations for these discrepancies could be the low positive predictive value of EVR for SVR, the retrospective design of most of the studies and, in some of them, the lack of information on the time of sample collection.

Several analytical methods have been developed for the determination of ribavirin in biological fluids Citation[45], including radioimmunoassays, capillary electrophoresis or liquid chromatography coupled with mass-spectrometric or UV detection.

The choice of plasma or serum for ribavirin concentration measurements might be important: ribavirin concentrations would be more reproducible in plasma than in serum in intensive sampling pharmacokinetic studies Citation[7]. This apparent discrepancy might be due to a poorer stability of serum levels during the phase of blood clotting at ambient temperature. Indeed, as ribavirin can be exchanged between blood cells and serum or plasma in the sampling tube, the samples must be immediately put on ice, taken to the laboratory within 2 h and then immediately centrifuged at +4°C. After that, the plasma or serum samples can be stored at -20°C. Another assumption recently made by Dahari et al. is that measuring intra-erythrocyte ribavirin concentrations could be a more relevant approach than measuring plasma levels, based on the assumption that intra-erythrocyte ribavirin accumulation early on during treatment would be higher in patients more likely to achieve SVR Citation[46]. However, the irreversible intra-erythrocyte accumulation of RTP is probably not representative of the drug turnover in other target cells, such as hepatocytes, where ribavirin activation into triphosphate metabolites is reversible. The intra-erythrocyte determination of ribavirin should certainly be further investigated, as there is no evidence so far that it is a better approach than that of plasma concentrations.

Conclusion

Ribavirin remains a key factor of the standard of care of chronic hepatitis C. In the era of specifically targeted antiviral therapy for HCV (STAT-C) therapy, convincing data suggest that there is no getting away from ribavirin in the near-future in combination therapies for chronic hepatitis C. The purpose should be thus to optimize ribavirin utilization in double and then triple therapies. The best way is probably to individually adjust the dose so that each patient could reach the highest probability of SVR with limited side effects. Global exposure after the first dose of ribavirin is an early and robust pharmacokinetic predictor of SVR Citation[9]. Therapeutic drug monitoring based on this approach will be tested soon in a French prospective, randomized fixed-dose versus concentration-controlled trial. The monitoring of ribavirin might also be useful to prevent relapses and drug mutations in future tri-therapies Citation[6].

The ability of ribavirin trough concentration at weeks 4 or 12 to predict SVR seems to be weaker Citation[37], but measurement of ribavirin trough concentration at week 4 and 8 could be useful to prevent the potential toxicity of ribavirin when administered at high doses in certain individuals in order to reach the target AUC and to check patient compliance.

Ribavirin concentrations can be measured either in plasma or in serum, by either high-performance liquid chromatography coupled to UV or mass-spectrometry detection, but the key points are to keep blood samples on ice and reduce the time between blood sampling and centrifugation to less than 2 h.

Financial & competing interests disclosure

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

No writing assistance was utilized in the production of this manuscript.

References

  • Manns MP, McHutchison JG, Gordon SC et al. Peginterferon α-2b plus ribavirin compared with interferon α-2b plus ribavirin for initial treatment of chronic hepatitis C: a randomised trial. Lancet358, 958–965 (2001).
  • Fried MW, Shiffman ML, Reddy KR et al. Peginterferon α-2a plus ribavirin for chronic hepatitis C virus infection. N. Engl. J. Med.347, 975–982 (2002).
  • Hadziyannis SJ, Sette H Jr, Morgan TR et al. Peginterferon-α and ribavirin combination therapy in chronic hepatitis C: a randomized study of treatment duration and ribavirin dose. Ann. Intern. Med.140, 346–355 (2004).
  • Heathcote J, Zeuzem S. Pegylated interferon monotherapy for chronic hepatitis C. Semin. Liver Dis.24(Suppl. 2), 39–45 (2004).
  • Bronowicki JP, Ouzan D, Asselah T et al. Effect of ribavirin in genotype 1 patients with hepatitis C responding to pegylated interferon α-2a plus ribavirin Gastroenterology131, 1040–1048 (2006).
  • Dusheiko GM, Hezode C, Pol S. Treatment of chronic hepatitis C with telaprevir in combination with peginterferon α 2A with or without ribavirin: further interim analysis. Results of the Prove 2 study. J. Hepatol.48(Suppl. 2), S26 (2008).
  • Jen J, Laughlin M, Chung C et al. Ribavirin dosing in chronic hepatitis C: application of population pharmacokinetic–pharmacodynamic models. Clin. Pharmacol. Ther.72, 349–361 (2002).
  • Larrat S, Stanke-Labesque F, Plages A, Zarski JP, Bessard G, Souvignet C. Ribavirin quantification in combination treatment of chronic hepatitis C. Antimicrob. Agents Chemother.47, 124–129 (2003).
  • Loustaud-Ratti V, Alain S, Rousseau A et al. Ribavirin exposure after the first dose is predictive of sustained virological response in chronic hepatitis C. Hepatology47, 1453–1461 (2008).
  • Maynard M, Pradat P, Gagnieu MC, Souvignet C, Trepo C. Prediction of sustained virological response by ribavirin plasma concentration at week 4 of therapy in hepatitis C virus genotype 1 patients. Antivir. Ther.13(4), 607–611 (2008).
  • Fernandez H, Banks G, Smith R. Ribavirin: a clinical overview. Eur. J. Epidemiol.2, 1–14 (1986).
  • Murata Y, Falsey AR. Respiratory syncytial virus infection in adults. Antivir. Ther.12, 659–670 (2007).
  • Snell NJ. Ribavirin current status of a broad spectrum antiviral agent. Expert Opin. Pharmacother.2, 1317–1324 (2001).
  • Reichard O, Andersson J, Schvarcz R, Weiland O. Ribavirin treatment for chronic hepatitis C. Lancet337, 1058–1061 (1991).
  • Brillanti S, Garson J, Foli M et al. A pilot study of combination therapy with ribavirin plus interferon α for interferon α-resistant chronic hepatitis C. Gastroenterology107, 812–817 (1994).
  • Di Bisceglie AM, Conjeevaram HS, Fried MW et al. Ribavirin as therapy for chronic hepatitis C. A randomized, double-blind, placebo-controlled trial. Ann. Intern. Med.123, 897–903 (1995).
  • McHutchison JG, Manns M, Patel K et al. International Hepatitis Interventional Therapy Group. Adherence to combination therapy enhances sustained response in genotype-1-infected patients with chronic hepatitis C. Gastroenterology123, 1061–1069 (2002).
  • Lindahl K, Stahle L, Bruchfeld A, Schvarcz R. High-dose ribavirin in combination with standard dose peginterferon for treatment of patients with chronic hepatitis C. Hepatology41(2), 275–279 (2005).
  • Dixit NM, Perelson AS. The metabolism, pharmacokinetics and mechanisms of antiviral activity of ribavirin against hepatitis C virus. Cell Mol. Life Sci.63, 832–842 (2006).
  • Markland W, McQuaid TJ, Jain J, Kwong AD. Broad-spectrum antiviral activity of the IMP dehydrogenase inhibitor VX-497: a comparison with ribavirin and demonstration of antiviral additivity with a interferon. Antimicrob. Agents Chemother.44(4), 859–866 (2000).
  • Zhou S, Liu R, Baroudy BM, Malcolm BA, Reyes GR The effect of ribavirin and IMPDH inhibitors on hepatitis C virus subgenomic replicon RNA. Virology310, 333–342 (2003).
  • Asahina Y, Izumi N, Enomoto N et al. Mutagenic effects of ribavirin and response to interferon/ribavirin combination therapy in chronic hepatitis C. J. Hepatol.43, 623–629 (2005).
  • Chevaliez S, Brillet R, Lazaro E, Hezode C, Pawlotsky JM. Analysis of ribavirin mutagenicity in human hepatitis C virus infection. J. Virol.81, 7732–7741 (2007).
  • Feld JJ, Nanda S, Huang Y et al. Hepatic gene expression during treatment with peginterferon and ribavirin: identifying molecular pathways for treatment response. Hepatology46, 1548–1563 (2007).
  • Neumann AU, Lam NP, Dahari H et al. Hepatitis C viral dynamics in vivo and the antiviral efficacy of interferon-α therapy. Science282, 103–107 (1998).
  • Pawlotsky JM, Dahari H, Neumann AU et al. Antiviral action of ribavirin in chronic hepatitis C. Gastroenterology126, 703–714 (2004).
  • Bruchfeld A, Lindahl K, Schvarcz R, Ståhle L. Dosage of ribavirin in patients with hepatitis C should be based on renal function: a population pharmacokinetic analysis. Ther. Drug Monit.24, 701–708 (2002).
  • Jen JF, Glue P, Gupta S, Zambas D, Hajian G. Population pharmacokinetic and pharmacodynamic analysis of ribavirin in patients with chronic hepatitis C. Ther. Drug Monit.22, 555–565 (2000).
  • Glue P, Schenker S, Gupta S, Clement RP, Zambas D, Salfi M. The single dose pharmacokinetics of ribavirin in subjects with chronic liver disease. Br. J. Clin. Pharmacol.49, 417–421 (2000).
  • Wade JR, Snoeck E, Duff F, Lamb M, Jorga K. Pharmacokinetics of ribavirin in patients with hepatitis C virus. Br. J. Clin. Pharmacol.62, 710–714 (2006).
  • Glue P. The clinical pharmacology of ribavirin. Semin. Liver Dis.19, 17–24 (1999).
  • De Franceschi L, Fattovich G, Turrini F. Hemolytic anemia induced by ribavirin therapy in patients with chronic hepatitis C virus infection: role of membrane oxidative damage. Hepatology31(4), 997–1004 (2000).
  • Gaeta GB, Precone DF, Felaco FM et al. Premature discontinuation of interferon plus ribavirin for adverse effects: a multicentre survey in ‘real world’ patients with chronic hepatitis C. Aliment. Pharmacol. Ther.16, 1633–1639 (2002).
  • Dieterich DT, Wasserman R, Bräu N et al. Once-weekly epoetin α improves anemia and facilitates maintenance of ribavirin dosing in hepatitis C virus-infected patients receiving ribavirin plus interferon α. Am. J. Gastroenterol.98, 2491–2499 (2003).
  • Afdhal NH, Dieterich DT, Pockros PJ et al.; Proactive Study Group. Epoetin α maintains ribavirin dose in HCV-infected patients: a prospective, double-blind, randomized controlled study. Gastroenterology126, 1302–1311 (2004).
  • Shiffman ML, Salvatore J, Hubbard S et al. Treatment of chronic hepatitis C virus genotype 1 with peginterferon, ribavirin, and epoetin α. Hepatology46, 371–379 (2007).
  • Crespo M, Pou L, Esteban JI et al. Early monitoring of ribavirin serum concentration is not useful to optimize hepatitis C virus treatment in HIV-coinfected patients. Antivir. Ther.12, 1217–1223 (2007).
  • Marcellin P, Jensen DM, Hadzyiannis SJ, Ferenci P. Differnciation of early virologic response (EVR) into RVR, complete EVR (cEVR) and partial EVR(pEVR), allows a more precise prediction of SVR in HCV genotype 1 patients treated with peginterferon α 2a and ribavirin. Hepatology46, 818A (2007) (Abstract).
  • Ferenci P, Fried MW, Shiffman ML et al. Predicting sustained virological responses in chronic hepatitis C patients treated with peginterferon α-2a (40 KD)/ribavirin. J. Hepatol.43, 425–433 (2005).
  • Nicot F, Legrand-Abravanel F, Lafont T et al. Serum concentrations of ribavirin and pegylated interferon and viral responses in patients infected with HIV and HCV. J. Med. Virol.80, 1523–1529 (2008).
  • Christensen PB, Alsio AA, Buhl MR et al. Ribavirin concentration at W4 is an independent predictor for sustained virological response after treatment of hepatitis C genotype 2/3 (Nordynamic trial). J. Hepatol.48(Suppl. 2), S6–S7 (2008).
  • Arase Y, Ikeda K, Tsubota A et al. Significance of serum ribavirin concentration in combination therapy of interferon and ribavirin for chronic hepatitis C. Intervirology48, 138–144 (2005).
  • Donnerer J, Grahovac M, Stelzl E et al. Ribavirin levels and haemoglobin decline in early virological responders and non-responders to hepatitis C virus combination therapy. Pharmacology76, 136–140 (2006).
  • Rendón AL, Núñez M, Romero M et al. Early monitoring of ribavirin plasma concentrations may predict anemia and early virologic response in HIV/hepatitis C virus-coinfected patients. J. Acquir. Immune Defic. Syndr.39, 401–405 (2005).
  • Bosch ME, Sánchez AJ, Rojas FS, Ojeda CB. Ribavirin: analytical determinations since the origin until today. J. Pharm. Biomed. Anal.45, 185–193 (2007).
  • Dahari H, Markatou M, Zeremski M et al. Early ribavirin pharmacokinetics, HCV RNA and alanine aminotransferase kinetics in HIV/HCV co-infected patients during treatment with pegylated interferon and ribavirin. J. Hepatol.47, 23–30 (2007).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.