Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Feb 1.
Published in final edited form as: HIV Med. 2013 Sep 11;15(2):108–115. doi: 10.1111/hiv.12086

Virologic Response Rates of Telaprevir-based Hepatitis C Triple Therapy in Patients with and without HIV Co-infection

V Martel-Laferrière 1, S Brinkley 2, K Bichoupan 1, S Posner 1, A Stivala 1, P Perumalswami 1, TD Schiano 1, M Sulkowski 2, DT Dieterich 1, AD Branch 1
PMCID: PMC3947307  NIHMSID: NIHMS519948  PMID: 24025147

Abstract

Objectives

Pegylated-interferon/ribavirin dual therapy for hepatitis C virus (HCV) has a lower sustained virologic response (SVR) rate in HIV/HCV-co-infected patients than in HCV mono-infected patients, but little is known about the relative effectiveness of teleprevir-based triple therapy in the two groups.

Methods

Data on 33 co-infected and 116 mono-infected patients were analyzed on an intention-to-treat basis. SVR12 was defined as undetectable HCV RNA at week-12 post-end-of-treatment, severe anemia as hemoglobin ≤89 g/L or a drop ≥45 g/L, and advanced fibrosis/cirrhosis as Fib-4 ≥3.25. All co-infected patients had well-controlled HIV.

Results

The groups were similar in age, gender, percentage with Fib-4 ≥3.25, and HCV viral load, but differed in previous treatment response, with more co-infected patients non responders/treatment intolerant (75.8% vs. 50.0%, <0.01). During treatment, the percentages of patients with undetectable HCV RNA were similar, but, surprisingly, tended to be higher in co-infected patients. SVR12 rates were 60.6% (co-infected) vs. 42.2% (mono-infected), p=0.06. In multivariable analysis, SVR12 was associated with HIV infection (OR: 3.55, p<0.01), African American race (OR: 0.37, p=0.03) and previous treatment response (OR: 0.46, p=0.03). Rates of severe anemia (45.5% vs. 58.6%, p=0.18) were similar in the two groups, but rash (15.2% vs. 34.5%, p=0.03) and rectal symptoms (12.1% vs. 43.1%, p<0.01) were less common in co-infected patients.

Conclusions

Virologic responses of co- and mono-infected patients did not differ significantly, but tended to be higher in co-infected patients, who had a 60.6% SVR12 rate. Telaprevir-based triple therapy is a promising option for co-infected patients with well-controlled HIV.

Keywords: hepatitis C virus, HIV, co-infection, telaprevir, side effects

Introduction

Due to shared routes of transmission, 30% of European and American HIV-infected patients are co-infected with the hepatitis C virus (HCV) and 8% of HCV-positive patients are co-infected with HIV (1, 2). Compared to HCV mono-infection, HIV/HCV co-infection results in accelerated liver fibrosis progression in patients with a detectable HIV viral load, although it is not accelerated in patients with undetectable HIV viral load (3). Mortality is higher in co-infected patients, underscoring the urgent need for more effective treatments (4). Numerous studies have demonstrated lower sustained virologic response (SVR) rates in co-infected patients receiving dual therapy with pegylated-interferon (pegIFN) and ribavirin (RBV). Among co-infected patients receiving dual therapy, SVR rates vary from 14% to 38% in patients with genotype 1 HCV and from 44% to 72% in patients with genotype 2–3 HCV (59). These SVR rates are about 10–20% lower than among genotype-matched mono-infected patients (10).

In May 2011, the approval of the first two direct-acting antiviral drugs, telaprevir (TVR) and boceprevir, significantly changed the treatment of patients infected with HCV genotype 1. For HCV mono-infected patients previously naïve to treatment, the SVR rate in clinical trials of TVR-based triple therapy increased to 72–75% (11, 12). For patients who had previously relapsed after achieving an end of treatment (EOT) response during dual therapy, the SVR rate increased to 83% (13). Difficult to treat populations also had improved outcomes, with SVR rates reaching 59% for previous partial responders and 29% for previous null responders (13).

TVR-based triple therapy has not yet received Food and Drug Administration approval for HIV/HCV co-infected patients, but a phase III trial is underway. A phase II study of 60 co-infected patients who were naive to HCV treatment at the start of the trial has been completed. The SVR rate was 74% among the 38 people in the TVR arm, which is similar to the rate in treatment naïve mono-infected patients (14). Among the patients in the TVR arm of this phase II trial, discontinuations for adverse events were rare. Two patients discontinued all medications, and one discontinued TVR. Eighteen percent developed an hemoglobin level < 100 g/L. One patient developed severe anemia, defined as an hemoglobin between 70 and 89 g/L or a decrease in hemoglobin from baseline by > 45 g/L. Thirty-four percent developed mild to moderate rash, compared to 23% of patients on dual therapy (without TVR).

Although results of the Phase II trial are promising, it is a small study carried out in the setting of a prospective clinical trial. Effectiveness in clinical practice often falls short of the efficacy reported in clinical trials, and side effects may be worse. The primary objective of the current study is to compare virologic responses to off-label TVR-based therapy in HIV/HCV co-infected vs. HCV mono-infected patients in a real-life setting. The secondary objectives are to assess the safety and tolerability of TVR-based triple therapy in co-infected patients by analyzing rates of severe anemia, hospitalization, rash, and rectal symptoms.

Methods

Study design and patients

This study is a medical record review of patients who began TVR in combination with pegIFN and RBV before December 2011 at the Icahn School of Medicine at Mount Sinai (MSSM), New York, NY and John Hopkins University (JHU), Baltimore, MD. Seventeen co-infected patients and 116 mono-infected patients were treated at MSSM and 16 co-infected patients were treated at JHU. None of the patients had previously received a liver transplant and none was treated for more than 48 weeks. All patients were treated with weight-based ribavirin dosing. TVR was administered at a dose of 750 mg three times a day, except when co-administered with efavirenz where a dose of 1,125 mg three times a day was used. Mono-infected patients who were naïve to HCV treatment or who previously relapsed after treatment with pegIFN/RBV, who were not cirrhotic and who had undetectable levels of HCV RNA at week-4 and -12 were eligible for a shortened treatment course of 24 weeks [response guided therapy (RGT)]. For this subset of patients, week-24 data were carried forward for the week-48 analysis (EOT). Co-infected patients were not considered for RGT.

Data were collected at baseline, week-4, -12, -24, -48 and 12 weeks post-EOT. Data recorded included demographics, baseline medical conditions and medications, previous HCV treatment history, laboratory values, adverse events including rash, rectal symptoms, emergency room visits and hospitalizations. To evaluate the workload associated with treatment planning, pre-treatment medical visits were counted. The education visit for injection learning was not counted, as it is standard of care for all patients. HCV and HIV viral loads were measured by RT-PCR (Roche Cobas Ampliprep Cobas Taqman version 2.0). HCV viral load below the lower limit of detection (18 IU/mL) was recorded as “undetectable”. HCV virologic failure was defined as a viral load > 1000 IU/mL at week-4 or -12 or a detectable HCV viral load after week-24. Patients were considered to have advanced fibrosis or cirrhosis if they had a Fib-4 score ≥3.25 (15). Anemia was defined as an hemoglobin < 135 g/L in men and < 120 g/L in women. Based on theDAIDS criteria, severe anemia was defined as an hemoglobin ≤ 89 g/L and/or a decrease ≥ 45 g/L(16). IRB approval was received from each institution.

Statistical analysis

Chi-square or Fisher exact tests were used for discrete variables and students T tests or Mann-Whitney U tests were used for continuous variables. Baseline characteristics of the HIV/HCV co-infected patients from MSSM and JHU were compared to each other and were combined because no statistically significant differences were found. Treatment outcomes were evaluated on an intention-to-treat basis. Patients lost to follow up were considered to have a virologic failure.

Data of co- and mono-infected patients were included in a multivariable logistic regression analysis of factors independently associated with SVR12. Factors known to influence SVR during dual therapy were included: age, sex, race (African American or not), previous treatment response [favourable (naïve or relapser) vs. not favourable (non-responder or treatment intolerant)], body mass index (BMI), logarithm of the HCV viral load, fibrosis stage (Fib-4 score below vs. above 3.25), and HIV infection status. To confirm the comparability of the patients, the treatment site was also evaluated. The multivariable model was built using forward and backward likelihood ratios. Entry and exit p-values were both fixed at 0.10 and factors with p-values below 0.05 in the final model were retained. Data were analyzed in SPSS (Chicago, IL, version 20). A p-value below 0.05 was considered statistically significant.

Results

The baseline characteristics of the study group are presented in Table 1. Mono- and co-infected patients were similar in age, sex, BMI, HCV viral load, and the percentage with advanced fibrosis/cirrhosis. The groups were different in their racial composition, with a higher percentage of African Americans in the co-infected group (42.4 vs. 16.4%, p<0.01). The previous treatment responses were also different, with more non-responders and patients who were intolerant to dual therapy in the co-infected group (75.8% vs. 50.0%, p<0.01). Co-infected patients had significantly more pre-treatment visits than mono-infected patients (median: 2 (range: 1–4) vs. 1 (15); p<0.01).

Table 1.

Baseline characteristics

Co-infected (N = 33) Mono- infected (N = 116) p-value
Age, year, median (IQR) 57 (52–59) 56 (51–61) 0.78*

Male (% of total) 26 (78.8%) 78 (67.2%) 0.20§

African American (% of total) 14 (42.4%) 19 (16.4%) <0.01§

Previous response to dual therapy (% of total) <0.01§,θ
Favorable group 8 (24.3%) 58 (50.0%)
 Naïve 3 (9.1%) 35 (30.2%)
 Relapser 5 (15.2%) 23 (19.8%)
Non favorable group 25 (75.8%) 58 (50.0%)
 Non-responder 21 (63.6%) 51 (44.0%)
 Intolerant 4 (12.1%) 7 (6.0%)

Advanced fibrosis/cirrhosis (% of total) 16 (48.5%) 40/112 (35.7%) 0.18§

Baseline HCV viral load, log10 IU/mL, median (IQR) 6.46 (5.92–7.00) 6.47 (5.91–6.74) 0.44

Number of pre-treatment visits, median (range) 2 (1–4) 1 (1–5) <0.01*
CD4+ T cell count, cells/μL, median (IQR) 478 (362–763) - -

HIV VL (% of total)
Undetectable/< 20 copies/mL 24/30 (80%) - -
< 1000 copies/mL 30/30(100%)

ART regimen switch required before triple therapy (% of total) 17 (51.5%) - -
*

Mann-Whitney U Test,

§

Chi-Square,

θ

Chi-square calculated by comparing favorable and non favorable groups,

Student T-test

Fisher’s Exact Test

IQR: interquartile range, VL: viral load, ART: antiretroviral therapy

At baseline, the median CD4+ T cell count of the co-infected patients was 478 cells/μL [inter quartile range (IQR), 362–763; min, 194; max, 1346]. Eighty percent had an undetectable HIV viral load and the remainder a viral load < 1000 copies/mL. All were on HIV antiretroviral therapy. Tenofovir and emtricitabine were the most common reverse transcriptase inhibitors. They were generally combined with ritonavir-boosted atazanavir, efavirenz, and/or raltegravir. Before initiating TVR-based therapy, 51.5% of the patients had to switch their HIV regimen out of concern about potential drug-drug interactions with TVR.

Virologic responses are presented in Fig. 1. A similar percentage of co- and mono-infected patients had undetectable HCV RNA at week-4 (42.4% vs. 43.1%, p=0.94), week-12 (69.7% vs. 65.5%, p=0.65), week-24 (75.8% vs. 57.8 %, p=0.06) and at the EOT (69.7% vs. 54.3%, p=0.11). There was a trend toward a better SVR12 rate in co-infected patients (60.6% vs. 42.2%, p=0.06). Among co-infected patients, CD4+ T cell counts were similar in patients with and without SVR12: median, 466 cells/μL (with) vs. 614 cells/μL (without), p=0.83.

Figure 1.

Figure 1

Virologic treatment responses

Co-infected patients had similar rates of on-treatment response and a significantly better SVR12 rate than in mono-infected patients.

RVR: rapid virologic response, EVR: early virologic response, eRVR: extended rapid virologic response, EOT: end of treatment, SVR12: sustained virologic response 12 weeks post-treatment

In univariable logistic regression analysis, there was a trend for an association between the SVR rate and race, previous HCV antiviral treatment response, and HIV status. In the multivariable model, HIV infection was positively associated with SVR [odds ratio (OR)= 3.55; 95% CI: 1.44–8.75] and African American race and an unfavourable response to previous dual therapy (non-response or treatment intolerance) were inversely related to SVR (OR= 0.37,95% CI: 0.15–0.92 and OR=0.46, 95% CI: 0.23–0.93, respectively) (Table 2).

Table 2.

Univariable and multivariable analysis with SVR12 as the outcome

OR univariable p-value univariable OR multivariable p-value multivariable
Age, year 1.00 (0.97–1.04) 0.89
Sex, female 1.02 (0.51–2.06) 0.95
HIV infected 2.10 (0.96–4.63) 0.07 3.55 (1.44–8.75) < 0.01
Race, African American 0.50 (0.22–1.12) 0.09 0.37 (0.15–0.92) 0.03
Response, non- responder/intolerant 0.55 (0.29–1.06) 0.07 0.46 (0.23–0.93) 0.03
Log HCV VL 0.91 (0.60–1.37) 0.65
BMI, kg/m2, 0.97 (0.90–1.04) 0.34
Advanced fibrosis/cirrhosis 0.63 (0.32–1.25) 0.19
Diabetes 0.62 (0.24–1.58) 0.31
Treatment site, JHU 2.09 (0.72–6.10) 0.18

OR: Odds ratio; Log HCV VL: logarithm of hepatitis C viral load; body mass index; JHU: John Hopkins University

Side effects and major adverse events were common in both co-infected and mono-infected patients (Table 3). There was no statistically significant difference in the proportions with a hospitalization (27.2% vs. 18.1%, p=0.25), emergency department visit (18.2% vs. 13.8%, p=0.53), anemia (87.8% vs. 91.4%, p=0.54), or severe anemia (45.5% vs. 58.6%, p=0.18). Rash and rectal symptoms were less common among co-infected patients (p= 0.03 and p<0.01, respectively). Treatment had to be discontinued because of side effects in a similar percentage of patients in the two groups. Early discontinuation of TVR (with continuation of pegIFN/RBV) occurred in 6.0% of co-infected patients vs. 0.9% of mono-infected patients, p=0.12; early discontinuation of all three drugs occurred in 21.1% vs. 12.9%, p=0.27; and early discontinuation of any drug occurred in 24.2% vs. 13.8%, p=0.18.

Table 3.

Discontinuations and side effects

Co-infected (N = 33) Mono-infected (N = 116) p-value
Discontinuation due to side effects (% of total)
-Telaprevir only 2 (6.0%) 1(0.9%) 0.12
-All three drugs 7 (21.2%) 15 (12.9%) 0.27
-Any drug discontinuation 8 (24.2%)* 16 (13.8%) 0.18

Hospitalization (% of total)
-Number of patients 8 (24.2%) 18 (15.5%) 0.24§
-Number of events 9 (27.2%) 21 (18.1%) 0.25§

Emergency room visits (% of total)
-Number of patients 6 (18.2%) 12 (10.3%) 0.22§
-Number of events 6 (18.2%) 16 (13.8%) 0.53§

Anemia (% of total) 29 (87.8%) 106 (91.4%) 0.54§

Severe anemia (% of total) 15 (45.5%) 68 (58.6%) 0.18§

Rash (% of total) 5 (15.2%) 40 (34.5%) 0.03§

Rectal symptoms (% of total) 4 (12.1%) 50 (43.1%) <0.01§
*

One co-infected patient initially discontinued telaprevir only. Two weeks later, he discontinued pegylated-interferon and ribavirin.

§

Chi-Square,

Fisher’s Exact Test

Based on the SVR12 rates in clinical trials, we expected an SVR12 rate of 60.7% in our population of mono-infected patients (1113), which is significantly higher than the 42.2% we observed, p<0.01 (Table 4). The lower overall rate in our study was largely due to the unexpectedly poor outcomes in two subgroups: patients who were previously naïve to treatment and patients who relapsed after an EOT response (p= 0.02 and p=0.01, respectively, compared to comparable groups in published trials). Among treatment naive patients, 10/35 (28.5%) discontinued triple-therapy early due to side effects. These patients accounted for 62.5% of all mono-infected patients who discontinued for side effects. Among former relapsers, 4/23 (17.4%) relapsed again after EOT. The previous non-responders had a response rate similar to comparable patients in clinical trials (p=0.30). Nineteen mono-infected patients were eligible for RGT. Thirteen (68.4%) achieved SVR12, whereas three relapsed and three were lost to follow up.

Table 4.

Comparison between SVR12 rates in our study compared to clinical trials

Phase III clinical trials Current study p-value
Naïves (n=35) 73.5%(11, 12) 51.4% 0.02§
Relapsers (n=23) 83%(13) 47.8% 0.01§
Non-responders/intolerants (n= 58) 44%(13) 34.5% 0.30§
Total (n=116)* 60.7% 42.2% <0.01§
§

Chi-Square

*

Projection of a matched population for previous treatment response

Discussion

To our knowledge, our study provides the first information about the effectiveness of TVR-based triple therapy in HIV/HCV co-infected patients receiving care outside of a clinical trial. Consistent with the outcome achieved in a phase II trial in 38 treatment-naïve patients (SVR24 rate of 74%)(14), the SVR12 rate of our co-infected patients was 60.6%, a promising result. Our findings are particularly encouraging because 42.4 % of the co-infected patients were African American, and 48.5% had advanced fibrosis/cirrhosis, factors associated with lower SVR rates among co-infected patients receiving dual therapy (17, 18). Even though the patients in our study received triple therapy without the structure and resources of a clinical trial, the SVR12 rate was higher than reported in major clinical trials of dual therapy for co-infected patients with genotype 1 HCV, which had SVR rates of 14–38% (59). Our data are consistent with those of an ongoing study of treatment-experienced co-infected patients (19). This study has an unusual design that includes a 4 week lead-in phase of pegIFN/RBV followed by 12 weeks of TVR-based triple therapy and a final 32 or 56 weeks of dual therapy. At week 12 of triple therapy, 88% of the patients were HCV viral load undetectable (19). In the co-infected phase II trial, 79% of patients were undetectable after 12 weeks of treatment and this translated in an SVR rate of 74% (14). These findings and our results suggest that triple-therapy is likely to be more effective in patients than dual-therapy in co-infected patients with well-controlled HIV. In contrast to the promising (60.6%) SVR12 rate observed in our co-infected patients, the lower (42.2%) rate we observed in our mono-infected patients was disappointing, although the difference between the two groups was not statistically significant (p=0.06).

The higher SVR12 rate among the co-infected patients may reflect a selection bias. Adherence was not measured in this study, and may have been lower in the mono-infected patients. All of the co-infected patients were receiving combination anti-retroviral therapy and maintained HIV viral loads below 1000 copies/mL. This prior experience with managing treatment side effects and ability to maintain adherence may have contributed to the high SVR12 rate of the co-infected patients. As formal possibilities to explain the higher SVR rate in the co-infected patients, one can also speculate that the HIV medications boosted levels of the HCV medications, giving an advantage to the co-infected patients, and/or that the altered immunological status of the co-infected patients somehow promoted HCV suppression in the unusual setting of TVR-based therapy. Further studies are needed to explore these possibilities. The mechanism of action of TVR almost certainly enhanced the SVR rate compared to dual therapy in the co-infected patients. TVR is a direct-acting antiviral agent that targets the HCV NS3/NS4A protease. It does not rely on host antiviral defences for efficacy. The direct antiviral effect of TVR may be especially important for HIV/HCV co-infected patients because these individuals have a residual immunological impairment, even when HIV is well controlled. In our study, CD4+ T counts were not associated with SVR.

Compared to results in Phase III trials, our results in HCV mono-infected patients were significantly lower than expected among previous relapsers and naïve patients. Several factors likely contributed to these differences. Our study analyzed outcome of clinical care provided in a real-life setting. Clinical practice is less controlled than clinical trials, and may include a broader range of patients. The discontinuation rates of all three drugs for side effects were higher in our patients than in the phase III trials (12.9–21.2% vs. 6–10%) (1113). The high rate of treatment discontinuation was especially pronounced for the previously treatment-naive mono-infected patients. One explanation for the higher discontinuation rates in treatment-naïve patients compared to patients who previously failed therapy is the possibility that patients who experienced severe side effects on dual therapy declined triple therapy. In addition, those who experienced mild side effects during dual therapy may have used this experience to cope with the side effects of triple therapy. Among patients receiving RGT, the SVR rate was 68.4%, significantly better than in the other mono-infected patients (37.1%, p=0.01), but lower than in Phase III trials (92%; p=0.08)(12).

In terms of safety, major events such as hospitalizations, emergency room visits, severe anemia and transfusion occurred at similar rates in co- and mono-infected patients. One death occurred in the mono-infected group. The patient had liver cirrhosis and died of cerebral hemorrhage after falling while riding in a bus that was involved in an accident eight weeks after the end of his treatment. Co-infected patients had lower rates of rectal symptoms and rash. The rate of rash observed in the mono-infected group is similar to the ones seen in the registration trials (34.5 vs. 34–37%) (11, 13, 14). The immunological impairment of the co-infected patients may have contributed to their lower incidence of rash. In a separate study, a reduced incidence of rash was observed in patients with advanced fibrosis/cirrhosis, a condition associated with reduced immune function (20). Both immunological impairment and experience with drug side effects may have contributed to the lower incidence of rectal complaints expressed by the co-infected patients.

When considering TVR-based therapy for HIV/HCV co-infected patients, possible drug-drug interactions need to be included in the treatment plan. TVR is a potent inhibitor of the cytochrome P450 3A4, which leads to many problematic drug-drug interactions (21). A limited number of HIV medications have been studied and proven to be safe with TVR. Tenofovir, emtricitabine, raltegravir, rilpivirine, etravirine and ritonavir-boosted atazanavir do not significantly alter TVR pharmacokinetics (2224), while efavirenz decreases the TVR Cmin and the area under the curve. An increased dose of TVR (1,125 mg three times daily) is recommended for combination with efavirenz (25). The HIV regimen of 51.5% of our co-infected patients was switched prior to starting TVR to avoid drug-drug interactions. The need for a medication adjustment may explain the higher number of pre-treatment visits among the co-infected patients compared to the mono-infected patients. Changing HIV regimens can be emotionally difficult for patients, especially when the established regimen is working well. For patients with multiple HIV resistance mutations, it may be difficult to find an alternative combination that is compatible with TVR. Treatment guidelines can be used to identify possible options, but input from a co-infection specialist may be required (26, 27). Boceprevir has more drug-drug interactions than TVR and is only considered suitable for use in combination with raltegravir and NRTIs, thereby significantly limiting its use in co-infected patients (28).

The main limitations of this study are the small number of co-infected patients and the dependence on medical records. Data about HIV parameters were not always available, IL28b polymorphisms were unknown for most patients, and because HCV genotyping was not repeated to confirm historical data, information about HCV subtype (1a vs. 1b) was lacking for some patients. The strengths of the study include the information it provides about the effectiveness of TVR-based triple therapy outside clinical trials, the likely generalizability of the results to other real-world settings, the analysis of contemporaneously-treated co- and mono-infected patients, and the inclusion of co-infected patients who had failed prior HCV treatment.

Conclusion

In conclusion, co-infected patients had similar rates of on-treatment response and SV12 than mono-infected patients when treated with TVR-based triple therapy. TVR was associated with numerous adverse events, but there were not significantly more events in the co- than in the mono-infected population. A phase III study of TVR in co-infected is currently on-going and will hopefully lead to the official approval of TVR in co-infected patients.

Acknowledgments

Source of Funding: This work was supported by the National Institutes of Health DA031095 and DK090317 to ADB and National Institute on Drug Abuse K24 DA034621/DA/NIDA and R01 DA016065-10/DA/NIDA to MS. The contents of this publication are solely the responsibility of the authors and do not necessarily represent the official views of the National Institutes of Health. Valérie Martel-Laferrière was funded by the 2011 AMMI Canada/Pfizer Post-Residency Fellowship and the 2012 Grant of the CHUM Foundation.

Footnotes

Disclosures:

Sherilyn Brinkley received consulting fees from Merck, Vertex Pharamaceuticals, Jansen and Kadmon and payment for lectures from Merck, Vertex Pharmaceuticals, Kadmon and Roche/Genentech. Thomas D. Schiano is a paid lecturer, consultant, paid lecturer, and a participant in the DSMB of companies that include Bristol-Myers Squibb/Sanofi-Aventis Partnership, Novartis, Pfizer, and Salix Pharmaceuticals, Inc. Mark Sulkowski received consulting fees and research grants from AbbVie, Bristol-Myers Squibb, Boehringer Ingelheim Pharmaceuticals, Gilead, Janssen, Merck, Idenix, and Vertex Pharmaceuticals. He received fees for participation in review activities from Pfizer. Douglas T. Dieterich serves as a paid lecturer, consultant, and is a member on scientific advisory boards of companies which either develop or assess medicines used for the treatment of viral hepatitis. These companies include Gilead Sciences, Boehringer Ingelheim, Novartis, Vertex Pharmaceuticals, Achillion, Tibotec, Idenix, Merck, Kadmon, Bayer Healthcare, Roche/Genentech and Bristol-Myers Squibb.

Valerie Martel-Laferriere, Kian Bichoupan, Shai Posner, Alicia Stivala, Ponni Perumalswami and Andrea D. Branch have no relevant disclosures.

References

  • 1.Rockstroh JK, Mocroft A, Soriano V, et al. Influence of hepatitis C virus infection on HIV-1 disease progression and response to highly active antiretroviral therapy. J Infect Dis. 2005;192:992–1002. doi: 10.1086/432762. [DOI] [PubMed] [Google Scholar]
  • 2.Ghany MG, Strader DB, Thomas DL, Seeff LB. Diagnosis, management, and treatment of hepatitis C: an update. Hepatology. 2009;49:1335–74. doi: 10.1002/hep.22759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Brau N, Salvatore M, Rios-Bedoya CF, et al. Slower fibrosis progression in HIV/HCV-coinfected patients with successful HIV suppression using antiretroviral therapy. J Hepatol. 2006;44:47–55. doi: 10.1016/j.jhep.2005.07.006. [DOI] [PubMed] [Google Scholar]
  • 4.Ly KN, Xing J, Klevens RM, Jiles RB, Ward JW, Holmberg SD. The increasing burden of mortality from viral hepatitis in the United States between 1999 and 2007. Ann Intern Med. 2012;156:271–8. doi: 10.7326/0003-4819-156-4-201202210-00004. [DOI] [PubMed] [Google Scholar]
  • 5.Laguno M, Murillas J, Blanco JL, et al. Peginterferon alfa-2b plus ribavirin compared with interferon alfa-2b plus ribavirin for treatment of HIV/HCV co-infected patients. AIDS. 2004;18:F27–36. doi: 10.1097/00002030-200409030-00003. [DOI] [PubMed] [Google Scholar]
  • 6.Carrat F, Bani-Sadr F, Pol S, et al. Pegylated interferon alfa-2b vs standard interferon alfa-2b, plus ribavirin, for chronic hepatitis C in HIV-infected patients: a randomized controlled trial. JAMA. 2004;292:2839–48. doi: 10.1001/jama.292.23.2839. [DOI] [PubMed] [Google Scholar]
  • 7.Chung RT, Andersen J, Volberding P, et al. Peginterferon Alfa-2a plus ribavirin versus interferon alfa-2a plus ribavirin for chronic hepatitis C in HIV-coinfected persons. N Engl J Med. 2004;351:451–9. doi: 10.1056/NEJMoa032653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Torriani FJ, Rodriguez-Torres M, Rockstroh JK, et al. Peginterferon Alfa-2a plus ribavirin for chronic hepatitis C virus infection in HIV-infected patients. N Engl J Med. 2004;351:438–50. doi: 10.1056/NEJMoa040842. [DOI] [PubMed] [Google Scholar]
  • 9.Nunez M, Miralles C, Berdun MA, et al. Role of weight-based ribavirin dosing and extended duration of therapy in chronic hepatitis C in HIV-infected patients: the PRESCO trial. AIDS Res Hum Retroviruses. 2007;23:972–82. doi: 10.1089/aid.2007.0011. [DOI] [PubMed] [Google Scholar]
  • 10.Ghany M, Nelson D, Strader D, Thomas D, Seeff L. An Update on Treatment of Genotype 1 Chronic Hepatitis C Virus Infection: Practice Guideline by the American Association for the Study of Liver Diseases. Hepatology. 2011;54:1433–44. doi: 10.1002/hep.24641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Jacobson IM, McHutchison JG, Dusheiko G, et al. Telaprevir for previously untreated chronic hepatitis C virus infection. N Engl J Med. 2011;364:2405–16. doi: 10.1056/NEJMoa1012912. [DOI] [PubMed] [Google Scholar]
  • 12.Sherman KE, Flamm SL, Afdhal NH, et al. Response-guided telaprevir combination treatment for hepatitis C virus infection. N Engl J Med. 2011;365:1014–24. doi: 10.1056/NEJMoa1014463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Zeuzem S, Andreone P, Pol S, et al. Telaprevir for retreatment of HCV infection. N Engl J Med. 2011;364:2417–28. doi: 10.1056/NEJMoa1013086. [DOI] [PubMed] [Google Scholar]
  • 14.Sulkowski MS, Sherman KE, Dieterich DT, et al. Combination Therapy With Telaprevir for Chronic Hepatitis C Virus Genotype 1 Infection in Patients With HIV: A Randomized Trial. Ann Intern Med. 2013 doi: 10.7326/0003-4819-159-2-201307160-00654. [DOI] [PubMed] [Google Scholar]
  • 15.Vallet-Pichard A, Mallet V, Nalpas B, et al. FIB-4: an inexpensive and accurate marker of fibrosis in HCV infection. comparison with liver biopsy and fibrotest. Hepatology. 2007;46:32–6. doi: 10.1002/hep.21669. [DOI] [PubMed] [Google Scholar]
  • 16. [accessed on February 7, 2013];Table for Grading the Severity of Adult and Pediatric Adverse Events. 2004 Dec; Version 1.0. (Clarification dated August 2009). http://rsctech-rescom/safetyandpharmacovigilance/gradingtablesaspx.
  • 17.Mira JA, Garcia-Rey S, Rivero A, et al. Response to pegylated interferon plus ribavirin among HIV/hepatitis C virus-coinfected patients with compensated liver cirrhosis. Clin Infect Dis. 2012;55:1719–26. doi: 10.1093/cid/cis779. [DOI] [PubMed] [Google Scholar]
  • 18.Zeremski M, Dimova RB, Makeyeva J, et al. IL28B polymorphism, pretreatment CXCL10 and HCV RNA levels predict treatment response in racially diverse HIV/HCV co-infected and HCV mono-infected patients. J Acquir Immune Defic Syndr. 2012 doi: 10.1097/QAI.0b013e31828323c1. [DOI] [PubMed] [Google Scholar]
  • 19.Cotte L, Braun J, Lascoux-Combe C, et al. High Early Virological Response with Telaprevir-Pegylated-Interferon-Ribavirin in Treatment-experienced Hepatitis C Virus Genotype 1/HIV Co-infected Patients: ANRS HC26 TelapreVIH Study. 20th Conference on Retroviruses and Opportunistic Infections; Atlanta, USA. March 3–6, 2013. [Google Scholar]
  • 20.Bichoupan K, Giannattasio E, Martel-Laferriere V, et al. Side effects and adverse outcomes of telaprevir-based triple therapy in HCV-positive patients with and without advanced fibrosis: real life experience. Hepatology. 2012:56. [Google Scholar]
  • 21.INCIVEK (telaprevir) Film Coated Tablets, for oral use. Revised: 04/13.
  • 22.Heeswijk Rv, Garg V, Boogaerts G, et al. The Pharmacokinetic Interaction Between Telaprevir & Raltegravir in Healthy Volunteers 51st ICAAC. 2011. p. Abstract A1–1738a. [Google Scholar]
  • 23.Heeswijk Rv, Vandevoorde A, Boogaerts G, et al. Pharmacokinetic Interactions between ARV Agents and the Investigational HCV Protease Inhibitor TVR in Healthy Volunteers. 18th Conference on Retroviruses and Opportunistic Infections (CROI); 2011. p. Abstract 119. [Google Scholar]
  • 24.Kakuda T, Leopold L, Nijs S, et al. Telaprevir and Etravirine or Rilpivirine. 13th HIV PK Workshop; 2012. p. Abstract O-18. [Google Scholar]
  • 25.Garg V, Chandorkar G, Yang Y, et al. The effect of CYP3A inhibitors and inducers on the pharmacokinetics of telaprevir in healthy volunteers. Br J Clin Pharmacol. 2013;75:431–9. doi: 10.1111/j.1365-2125.2012.04345.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the use of antiretroviral agents in HIV-1-infected adults and adolescents. Department of Health and Human Services; Available at http://aidsinfo.nih.gov/ContentFiles/AdultandAdolescentGL.pdf. [Google Scholar]
  • 27.Reiss P, Battegay M, Clumeck N, et al. European AIDS Cilnical Society Guidelines. 2011. pp. 1–60. Version 6. [Google Scholar]
  • 28. [Access on February 22, 2013];Victrelis (boceprevir) Capsules for oral use: Full prescribing information. :26. www.merck.com/product/usa/pi_circulars/v/vitrelis/vitrelis_pi.pdf. Revised: 12/2012.

RESOURCES