Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
Braz J Infect Dis ; 26(6): 102717, 2022.
Article in English | MEDLINE | ID: mdl-36410397

ABSTRACT

The chronic hepatitis C (CHC) treatment is currently based on the use of direct-acting antivirals (DAAs), and patients infected with hepatitis C virus genotype 3 (GT3) have emerged as a more difficult-to-cure population. The NS5A inhibitor daclatasvir (DCV) and sofosbuvir (SOF), an NS5B viral polymerase inhibitor, are among the drugs that compose more effective and safer treatment regimens. The virus genetic variability is related to resistance-associated substitutions (RASs) that adversely impact DAAs effectiveness. The aims of this study were to analyze the association of NS5A and NS5B RASs and other clinical factors with DAAs regimens effectiveness in patients with GT3 CHC infection. This was a prospective cohort study performed in a Brazilian university hospital. Individuals older than 18 years with GT3 CHC treated with SOF + DCV ± ribavirin (RBV) or SOF + peginterferon (PEG) + RBV were included. Blood samples were collected at baseline and post-treatment. A total of 121 patients were included. Sustained virological response rates were 87.6% for the SOF + DCV ± RBV group and 80.0% for the SOF + PEG + RBV arm. Cirrhosis, prior treatment with interferon/PEG + RBV, and baseline NS5A RAS were associated with higher risk of treatment failure. The NS5A analysis suggested that A30K, Y93H, and RAS at site 62 were related to failure. Interestingly, a likely compensatory effect was shown between A30K and A62T. Emergence of Y93H was always associated with RAS at position 62. The RASs dynamics comprehension is an important tool to indicate more effective treatment for GT3 patients.


Subject(s)
Antiviral Agents , Hepatitis C, Chronic , Humans , Antiviral Agents/therapeutic use , Antiviral Agents/pharmacology , Hepatitis C, Chronic/complications , Hepacivirus/genetics , Prospective Studies , Sofosbuvir/therapeutic use , Sofosbuvir/pharmacology , Ribavirin/therapeutic use , Ribavirin/pharmacology , Treatment Outcome , Drug Therapy, Combination , Genotype , Drug Resistance, Viral/genetics
2.
J Antimicrob Chemother ; 76(7): 1874-1885, 2021 06 18.
Article in English | MEDLINE | ID: mdl-33880524

ABSTRACT

BACKGROUND: Current approaches of drug repurposing against COVID-19 have not proven overwhelmingly successful and the SARS-CoV-2 pandemic continues to cause major global mortality. SARS-CoV-2 nsp12, its RNA polymerase, shares homology in the nucleotide uptake channel with the HCV orthologue enzyme NS5B. Besides, HCV enzyme NS5A has pleiotropic activities, such as RNA binding, that are shared with various SARS-CoV-2 proteins. Thus, anti-HCV NS5B and NS5A inhibitors, like sofosbuvir and daclatasvir, respectively, could be endowed with anti-SARS-CoV-2 activity. METHODS: SARS-CoV-2-infected Vero cells, HuH-7 cells, Calu-3 cells, neural stem cells and monocytes were used to investigate the effects of daclatasvir and sofosbuvir. In silico and cell-free based assays were performed with SARS-CoV-2 RNA and nsp12 to better comprehend the mechanism of inhibition of the investigated compounds. A physiologically based pharmacokinetic model was generated to estimate daclatasvir's dose and schedule to maximize the probability of success for COVID-19. RESULTS: Daclatasvir inhibited SARS-CoV-2 replication in Vero, HuH-7 and Calu-3 cells, with potencies of 0.8, 0.6 and 1.1 µM, respectively. Although less potent than daclatasvir, sofosbuvir alone and combined with daclatasvir inhibited replication in Calu-3 cells. Sofosbuvir and daclatasvir prevented virus-induced neuronal apoptosis and release of cytokine storm-related inflammatory mediators, respectively. Sofosbuvir inhibited RNA synthesis by chain termination and daclatasvir targeted the folding of secondary RNA structures in the SARS-CoV-2 genome. Concentrations required for partial daclatasvir in vitro activity are achieved in plasma at Cmax after administration of the approved dose to humans. CONCLUSIONS: Daclatasvir, alone or in combination with sofosbuvir, at higher doses than used against HCV, may be further fostered as an anti-COVID-19 therapy.


Subject(s)
COVID-19 , Pharmaceutical Preparations , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Carbamates , Chlorocebus aethiops , Humans , Imidazoles , Pyrrolidines , RNA, Viral , SARS-CoV-2 , Sofosbuvir/pharmacology , Valine/analogs & derivatives , Vero Cells
3.
J Gen Virol ; 102(1)2021 01.
Article in English | MEDLINE | ID: mdl-33141008

ABSTRACT

Hepatitis C virus (HCV) genotype 3 presents a high level of both baseline and acquired resistance to direct-acting antivirals (DAAs), particularly those targeting the NS5A protein. To understand this resistance we studied a cohort of Brazilian patients treated with the NS5A DAA, daclatasvir and the nucleoside analogue, sofosbuvir. We observed a novel substitution at NS5A amino acid residue 98 [serine to glycine (S98G)] in patients who relapsed post-treatment. The effect of this substitution on both replication fitness and resistance to DAAs was evaluated using two genotype 3 subgenomic replicons. S98G had a modest effect on replication, but in combination with the previously characterized resistance-associated substitution (RAS), Y93H, resulted in a significant increase in daclatasvir resistance. This result suggests that combinations of substitutions may drive a high level of DAA resistance and provide some clues to the mechanism of action of the NS5A-targeting DAAs.


Subject(s)
Antiviral Agents/pharmacology , Carbamates/pharmacology , Drug Resistance, Viral/genetics , Hepacivirus/drug effects , Imidazoles/pharmacology , Pyrrolidines/pharmacology , Valine/analogs & derivatives , Viral Nonstructural Proteins/genetics , Antiviral Agents/therapeutic use , Brazil , Carbamates/therapeutic use , Cell Line, Tumor , Cohort Studies , Drug Resistance, Viral/drug effects , Drug Therapy, Combination , Genotype , Hepacivirus/genetics , Hepacivirus/physiology , Hepatitis C/drug therapy , Hepatitis C/virology , Humans , Imidazoles/therapeutic use , Mutation , Pyrrolidines/therapeutic use , Recurrence , Sofosbuvir/pharmacology , Sofosbuvir/therapeutic use , Valine/pharmacology , Valine/therapeutic use , Viral Nonstructural Proteins/antagonists & inhibitors , Virus Replication/genetics
4.
Viruses ; 11(11)2019 10 31.
Article in English | MEDLINE | ID: mdl-31683616

ABSTRACT

In Brazil, hepatitis C treatment has been evolving significantly with the licensing of direct-acting antivirals (DAAs). However, viral determinants (amino acid substitutions in hepatitis C virus (HCV) genome and infective genotype) associated with host factors (hepatic condition and prior HCV therapy) might limit the achievement of sustained virologic response (SVR). Here, we described two case reports in which the occurrence of HCV NS5A mutations A30K (subtype 3a) and Y93N (subtype 1a) might have influenced daclatasvir (DCV)/sofosbuvir (SOF) combined therapy non-response. Despite high response rates for DAA combined therapies in Brazil, these case reports stated the importance of an investigation about how to manage a DAA treatment failure since a combination of factors, especially the occurrence of resistance substitutions, could impact a rescue therapy with new available antivirals in clinical routine.


Subject(s)
Drug Resistance, Viral/genetics , Hepacivirus/genetics , Hepatitis C, Chronic/drug therapy , Viral Nonstructural Proteins/genetics , Aged , Antiviral Agents/pharmacology , Carbamates , Female , Humans , Imidazoles/pharmacology , Male , Mutation , Pyrrolidines , Sofosbuvir/pharmacology , Treatment Failure , Valine/analogs & derivatives
5.
Ann Hepatol ; 18(6): 816-824, 2019.
Article in English | MEDLINE | ID: mdl-31594756

ABSTRACT

INTRODUCTION AND OBJECTIVES: Direct antiviral agents (DAAs) are very efficient in inhibiting hepatitis C virus and might be used to treat infections caused by other flaviviruses whose worldwide detection has recently increased. The aim of this study was to verify the efficacy of DAAs in inhibiting yellow fever virus (YFV) by using drug repositioning (a methodology applied in the pharmaceutical industry to identify new uses for approved drugs). MATERIALS AND METHODS: Three DAAs were evaluated: daclatasvir, sofosbuvir and ledipasvir or their combinations. For in vitro assays, the drugs were diluted in 100% dimethyl sulfoxide. Vaccine strain 17D and a 17D strain expressing the reporter fluorescent protein were used in the assays. A fast and reliable cell-based screening assay using Vero cells or Huh-7 cells (a hepatocyte-derived carcinoma ell line) was carried out. Two patients who acquired yellow fever virus with acute liver failure were treated with sofosbuvir for one week as a compassionate use. RESULTS: Using a high-content screening assay, we verified that sofosbuvir presented the best antiviral activity against YFV. Moreover, after an off-label treatment with sofosbuvir, the two female patients diagnosed with yellow fever infection displayed a reduction in blood viremia and an improvement in the course of the disease, which was observed in the laboratory medical parameters related to disease evolution. CONCLUSIONS: Sofosbuvir may be used as an option for treatment against YFV until other drugs are identified and approved for human use. These results offer insights into the role of nonstructural protein 5 (NS5) in YFV inhibition and suggest that nonstructural proteins may be explored as drug targets for YFV treatment.


Subject(s)
Antiviral Agents/pharmacology , Benzimidazoles/pharmacology , Fluorenes/pharmacology , Imidazoles/pharmacology , Sofosbuvir/pharmacology , Yellow Fever/drug therapy , Yellow fever virus/drug effects , Animals , Antiviral Agents/therapeutic use , Carbamates , Cell Line, Tumor , Chlorocebus aethiops , Compassionate Use Trials , Drug Repositioning , Female , Humans , In Vitro Techniques , Liver Failure, Acute/etiology , Pyrrolidines , Sofosbuvir/therapeutic use , Valine/analogs & derivatives , Vero Cells , Viral Load/drug effects , Viral Nonstructural Proteins/antagonists & inhibitors , Yellow Fever/complications
6.
PLoS Negl Trop Dis ; 13(1): e0007072, 2019 01.
Article in English | MEDLINE | ID: mdl-30699122

ABSTRACT

Yellow fever virus (YFV) is a member of the Flaviviridae family. In Brazil, yellow fever (YF) cases have increased dramatically in sylvatic areas neighboring urban zones in the last few years. Because of the high lethality rates associated with infection and absence of any antiviral treatments, it is essential to identify therapeutic options to respond to YFV outbreaks. Repurposing of clinically approved drugs represents the fastest alternative to discover antivirals for public health emergencies. Other Flaviviruses, such as Zika (ZIKV) and dengue (DENV) viruses, are susceptible to sofosbuvir, a clinically approved drug against hepatitis C virus (HCV). Our data showed that sofosbuvir docks onto YFV RNA polymerase using conserved amino acid residues for nucleotide binding. This drug inhibited the replication of both vaccine and wild-type strains of YFV on human hepatoma cells, with EC50 values around 5 µM. Sofosbuvir protected YFV-infected neonatal Swiss mice and adult type I interferon receptor knockout mice (A129-/-) from mortality and weight loss. Because of its safety profile in humans and significant antiviral effects in vitro and in mice, Sofosbuvir may represent a novel therapeutic option for the treatment of YF. Key-words: Yellow fever virus; Yellow fever, antiviral; sofosbuvir.


Subject(s)
Antiviral Agents/pharmacology , Drug Resistance, Viral , RNA, Viral/drug effects , Sofosbuvir/pharmacology , Yellow Fever/drug therapy , Yellow fever virus/drug effects , Animals , Chlorocebus aethiops , Disease Models, Animal , Hep G2 Cells , Humans , Mice , Mice, Knockout , RNA, Viral/blood , RNA, Viral/genetics , Vero Cells , Yellow Fever/blood , Yellow Fever/pathology , Yellow Fever/virology , Yellow fever virus/genetics
7.
Braz. j. infect. dis ; Braz. j. infect. dis;22(3): 186-192, May-June 2018. tab, graf
Article in English | LILACS | ID: biblio-974205

ABSTRACT

ABSTRACT Background This study aimed to evaluate the clinical effectiveness in terms of sustained virological response and tolerability of available second generation direct-acting antivirals in Brazilian patients. Methods This was a retrospective observational study conducted in six centers in Southern Brazil. The sample comprised adult patients who were chronically infected with hepatitis C virus, regardless of virus genotype, fibrosis stage, or prior treatment. Statistical analysis was performed to compare the effectiveness among the treatments, and also to uncover the factors influencing the achievement of sustained virological response. Results A total of 296 patients were included in the study, with the majority receiving sofosbuvir with daclatasvir (59%) or sofosbuvir with simeprevir (26%). Overall sustained virological response rates were approximately 91.6%. For genotype 1, sofosbuvir with daclatasvir had an sustained virological response rate of approximately 95%, while the sustained virological response rate of sofosbuvir with simeprevir was 92%; this difference was statistically significant only for subtype 1b. The only treatment used for genotype 3 patients was sofosbuvir with daclatasvir, and lower rates of sustained virological response were observed for this group, compared to genotype 1 (84% versus 95%, p < 0.05). Apart from this difference between genotypes, and a difference between patients who achieved rapid virologic response compared with those who did not, there were no other statistically significant factors associated with sustained virological response. Conclusions The results point to the effectiveness of second-generation direct-acting antivirals in hepatitis C virus Brazilian patients, especially those with genotype 1. Furthermore, that patients with genotype 3 need more attention and adjustments in available treatment options.


Subject(s)
Humans , Male , Female , Middle Aged , Aged , Antiviral Agents/pharmacology , Hepatitis C, Chronic/drug therapy , Reference Values , Ribavirin/pharmacology , Time Factors , Brazil , Logistic Models , Polymerase Chain Reaction , Retrospective Studies , Viral Load , Hepatitis C, Chronic/complications , Dose-Response Relationship, Drug , Simeprevir/pharmacology , Sofosbuvir/pharmacology , Sustained Virologic Response , Imidazoles/pharmacology , Liver Cirrhosis/virology
8.
Braz J Infect Dis ; 22(3): 186-192, 2018.
Article in English | MEDLINE | ID: mdl-29752891

ABSTRACT

BACKGROUND: This study aimed to evaluate the clinical effectiveness in terms of sustained virological response and tolerability of available second generation direct-acting antivirals in Brazilian patients. METHODS: This was a retrospective observational study conducted in six centers in Southern Brazil. The sample comprised adult patients who were chronically infected with hepatitis C virus, regardless of virus genotype, fibrosis stage, or prior treatment. Statistical analysis was performed to compare the effectiveness among the treatments, and also to uncover the factors influencing the achievement of sustained virological response. RESULTS: A total of 296 patients were included in the study, with the majority receiving sofosbuvir with daclatasvir (59%) or sofosbuvir with simeprevir (26%). Overall sustained virological response rates were approximately 91.6%. For genotype 1, sofosbuvir with daclatasvir had an sustained virological response rate of approximately 95%, while the sustained virological response rate of sofosbuvir with simeprevir was 92%; this difference was statistically significant only for subtype 1b. The only treatment used for genotype 3 patients was sofosbuvir with daclatasvir, and lower rates of sustained virological response were observed for this group, compared to genotype 1 (84% versus 95%, p<0.05). Apart from this difference between genotypes, and a difference between patients who achieved rapid virologic response compared with those who did not, there were no other statistically significant factors associated with sustained virological response. CONCLUSIONS: The results point to the effectiveness of second-generation direct-acting antivirals in hepatitis C virus Brazilian patients, especially those with genotype 1. Furthermore, that patients with genotype 3 need more attention and adjustments in available treatment options.


Subject(s)
Antiviral Agents/pharmacology , Hepatitis C, Chronic/drug therapy , Aged , Brazil , Carbamates , Dose-Response Relationship, Drug , Female , Hepatitis C, Chronic/complications , Humans , Imidazoles/pharmacology , Liver Cirrhosis/virology , Logistic Models , Male , Middle Aged , Polymerase Chain Reaction , Pyrrolidines , Reference Values , Retrospective Studies , Ribavirin/pharmacology , Simeprevir/pharmacology , Sofosbuvir/pharmacology , Sustained Virologic Response , Time Factors , Valine/analogs & derivatives , Viral Load
9.
Virus Res ; 244: 64-70, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29113824

ABSTRACT

A strong correlation between Zika virus (ZIKV) infection and severe neurological disease in newborns and occasionally adults has emerged in the Brazilian outbreak. Efficient human cell-based assays are required to test candidate inhibitors of ZIKV replication. The aim of this work was to investigate ZIKV propagation and quantification in different cell lines. The human (U87, A549, Huh7), mosquito (C6/36) and monkey (VERO E6) cell lines tested were all permissive to ZIKV infection. When assessed by plaque forming units (PFU) in three different target cell lines, the maximal production of ZIKV was achieved in Huh7 at day 3 post-infection (6.38±0.44 log10PFU/ml). The C6/36 cell line showed a low and slow production of virus when compared with other cell lines. A549 readout cells generated a larger number of plaques compared to Huh7 but not to VERO E6 cells. ZIKV PFU and RNA titers showed the highest correlation when Huh7 and A549 were used as the producer and readout cells, respectively. Also, U87 cells produced ZIKV RNA titers which were highly correlated with PFU independently from the readout cell line. Using the best virus-cell system, sofosbuvir and ribavirin EC50 were 1.2µM and 1.1µM when measured through plaque assay, and 4.2µM and 5.2µM when measured by quantitative real time PCR (qRT-PCR), respectively. In summary, ZIKV can efficiently infect different human cell lines and rapidly reach peak viral titers. Overall, A549 cells appear to be as efficient as the VERO E6 gold standard for plaque assay allowing the use of human, rather than simian, cells for evaluating candidate anti-ZIKV compounds by the reference assay. The possibility to replace the labor-intensive plaque assay with the more rapid and easy-to-perform qRT-PCR is appealing and warrants further investigation.


Subject(s)
Antiviral Agents/pharmacology , Host Specificity , RNA, Viral/antagonists & inhibitors , Ribavirin/pharmacology , Sofosbuvir/pharmacology , Virus Replication/drug effects , Zika Virus/drug effects , A549 Cells , Animals , Brazil , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , Culicidae , Hepatocytes/drug effects , Hepatocytes/virology , Humans , Microbial Sensitivity Tests , Neuroglia/drug effects , Neuroglia/virology , RNA, Viral/biosynthesis , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction , Vero Cells , Viral Load/drug effects , Viral Plaque Assay , Zika Virus/genetics , Zika Virus/growth & development , Zika Virus/metabolism
10.
Sci Rep ; 7(1): 9409, 2017 08 25.
Article in English | MEDLINE | ID: mdl-28842610

ABSTRACT

Zika virus (ZIKV) causes significant public health concerns because of its association with congenital malformations, neurological disorders in adults, and, more recently, death. Considering the necessity to mitigate ZIKV-associated diseases, antiviral interventions are an urgent necessity. Sofosbuvir, a drug in clinical use against hepatitis C virus (HCV), is among the FDA-approved substances endowed with anti-ZIKV activity. In this work, we further investigated the in vivo activity of sofosbuvir against ZIKV. Neonatal Swiss mice were infected with ZIKV (2 × 107 PFU) and treated with sofosbuvir at 20 mg/kg/day, a concentration compatible with pre-clinical development of this drug. We found that sofosbuvir reduced acute levels of ZIKV from 60 to 90% in different anatomical compartments, such as the blood plasma, spleen, kidney, and brain. Early treatment with sofosbuvir doubled the percentage and time of survival of ZIKV-infected animals. Sofosbuvir also prevented the acute neuromotor impairment triggered by ZIKV. In the long-term behavioural analysis of ZIKV-associated sequelae, sofosbuvir prevented loss of hippocampal- and amygdala-dependent memory. Our results indicate that sofosbuvir inhibits ZIKV replication in vivo, which is consistent with the prospective necessity of antiviral drugs to treat ZIKV-infected individuals.


Subject(s)
Antiviral Agents/pharmacology , Sofosbuvir/pharmacology , Zika Virus Infection/drug therapy , Zika Virus/physiology , Animals , Animals, Newborn , Antiviral Agents/administration & dosage , Chlorocebus aethiops , Memory , Mice , RNA, Viral , Reflex, Righting , Sofosbuvir/administration & dosage , Vero Cells , Virus Replication/drug effects , Zika Virus Infection/mortality
11.
Sci Rep ; 7: 40920, 2017 01 18.
Article in English | MEDLINE | ID: mdl-28098253

ABSTRACT

Zika virus (ZIKV) is a member of the Flaviviridae family, along with other agents of clinical significance such as dengue (DENV) and hepatitis C (HCV) viruses. Since ZIKV causes neurological disorders during fetal development and in adulthood, antiviral drugs are necessary. Sofosbuvir is clinically approved for use against HCV and targets the protein that is most conserved among the members of the Flaviviridae family, the viral RNA polymerase. Indeed, we found that sofosbuvir inhibits ZIKV RNA polymerase, targeting conserved amino acid residues. Sofosbuvir inhibited ZIKV replication in different cellular systems, such as hepatoma (Huh-7) cells, neuroblastoma (SH-Sy5y) cells, neural stem cells (NSC) and brain organoids. In addition to the direct inhibition of the viral RNA polymerase, we observed that sofosbuvir also induced an increase in A-to-G mutations in the viral genome. Together, our data highlight a potential secondary use of sofosbuvir, an anti-HCV drug, against ZIKV.


Subject(s)
Antiviral Agents/pharmacology , Sofosbuvir/pharmacology , Virus Replication/drug effects , Zika Virus/physiology , Antiviral Agents/therapeutic use , Cell Line , Cell Survival/drug effects , DNA-Directed RNA Polymerases/antagonists & inhibitors , DNA-Directed RNA Polymerases/metabolism , Genome, Viral , Humans , Mutation , Sofosbuvir/therapeutic use , Viral Proteins/antagonists & inhibitors , Viral Proteins/metabolism , Zika Virus/genetics , Zika Virus/isolation & purification , Zika Virus Infection/drug therapy , Zika Virus Infection/pathology , Zika Virus Infection/virology
12.
Antivir Ther ; 22(5): 447-451, 2017.
Article in English | MEDLINE | ID: mdl-28085003

ABSTRACT

BACKGROUND: The prevalence of natural polymorphisms associated with resistance to NS5B nucleoside/nucleotide (NI) sofosbuvir is distinct in different geographical regions. In Brazil, direct-acting anti-HCV therapy has recently changed with the introduction of interferon (IFN)-free regimens with sofosbuvir; however, the presence of resistant variants on clinical outcomes remains unknown. The aim of this study was to assess the natural polymorphisms associated with resistance to the NS5B NI sofosbuvir in Brazilian HCV-1 isolates and to compare it with that from other geographical regions. METHODS: Nucleotide sequencing of the HCV NS5B gene was performed in serum samples of 95 therapy-naive Brazilian patients infected with subtype 1a (n=43) and 1b (n=52). The sequences were analysed along with 1,525 NS5B sequences from North America, Europe and Asia retrieved from public HCV databases. RESULTS: In Brazilian HCV-1b patients who have never been exposed to a direct-acting anti-HCV drug, the C316N was detected in 15/52 (28.85%) patients, of these, 2 (3.85%) had single C316N variant, while 13 (25%) presented the double L159F-C316N mutant. A lower rate of L159F-C316N variants was detected in North American (n=9/238; 3.78%, P<0.001), European (n=17/281; 6.05%, P<0.001) and Asian (n=2/173; 1.16%, P<0.001) isolates. No sofosbuvir resistance-associated variants (RAVs) were identified in HCV-1a sequences. CONCLUSIONS: Resistant variants to sofosbuvir were found at different frequencies in worldwide HCV-1b sequences but not in HCV-1a sequences. The high frequency of double mutation L159F-C316N observed in Brazilian HCV-1b patients contrast with the lower rate observed in the three continents studied. The association of these findings and the clinical implications awaits further analysis.


Subject(s)
Antiviral Agents/pharmacology , Drug Resistance, Viral , Genotype , Hepacivirus/genetics , Hepatitis C/epidemiology , Hepatitis C/virology , Viral Nonstructural Proteins/genetics , Amino Acid Substitution , Antiviral Agents/therapeutic use , Brazil/epidemiology , Female , Hepatitis C/drug therapy , Humans , Male , Mutation , Prevalence , Public Health Surveillance , Sofosbuvir/pharmacology , Sofosbuvir/therapeutic use
13.
Hepatology ; 64(5): 1430-1441, 2016 11.
Article in English | MEDLINE | ID: mdl-27474787

ABSTRACT

In 2015, European and U.S. health agencies issued warning letters in response to 9 reported clinical cases of severe bradycardia/bradyarrhythmia in hepatitis C virus (HCV)-infected patients treated with sofosbuvir (SOF) in combination with other direct acting antivirals (DAAs) and the antiarrhythmic drug, amiodarone (AMIO). We utilized preclinical in vivo models to better understand this cardiac effect, the potential pharmacological mechanism(s), and to identify a clinically translatable model to assess the drug-drug interaction (DDI) cardiac risk of current and future HCV inhibitors. An anesthetized guinea pig model was used to elicit a SOF+AMIO-dependent bradycardia. Detailed cardiac electrophysiological studies in this species revealed SOF+AMIO-dependent selective nodal dysfunction, with initial, larger effects on the sinoatrial node. Further studies in conscious, rhesus monkeys revealed an emergent bradycardia and bradyarrhythmia in 3 of 4 monkeys administered SOF+AMIO, effects not observed with either agent alone. Morever, bradycardia and bradyarrhythmia were not observed in rhesus monkeys when intravenous infusion of MK-3682 was completed after AMIO pretreatment. CONCLUSIONS: These are the first preclinical in vivo experiments reported to replicate the severe clinical SOF+AMIO cardiac DDI and provide potential in vivo mechanism of action. As such, these data provide a preclinical risk assessment paradigm, including a clinically relevant nonhuman primate model, with which to better understand cardiovascular DDI risk for this therapeutic class. Furthermore, these studies suggest that not all HCV DAAs and, in particular, not all HCV nonstructural protein 5B inhibitors may exhibit this cardiac DDI with amiodarone. Given the selective in vivo cardiac electrophysiological effect, these data enable targeted cellular/molecular mechanistic studies to more precisely identify cell types, receptors, and/or ion channels responsible for the clinical DDI. (Hepatology 2016;64:1430-1441).


Subject(s)
Amiodarone/pharmacology , Anti-Arrhythmia Agents/pharmacology , Antiviral Agents/pharmacology , Heart/drug effects , Hepacivirus/drug effects , Nucleotides/antagonists & inhibitors , Sofosbuvir/pharmacology , Amiodarone/adverse effects , Animals , Anti-Arrhythmia Agents/adverse effects , Antiviral Agents/adverse effects , Drug Interactions , Guinea Pigs , Heart/physiology , Macaca mulatta , Male , Sofosbuvir/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL