--------------------------------CONTRAINDICATIONS-----------------------------­ EPCLUSA and ribavirin combination regimen is contraindicated in patients for whom ribavirin is contraindicated (4) HIGHLIGHTS OF PRESCRIBING INFORMATION These highlights do not include all the information needed to use EPCLUSA safely and effectively. See full prescribing information for EPCLUSA. -------------------------WARNINGS AND PRECAUTIONS---------------------­ Bradycardia with amiodarone coadministration: Serious symptomatic bradycardia may occur in patients taking amiodarone, particularly in patients also receiving beta blockers, or those with underlying cardiac comorbidities and/or advanced liver disease. Coadministration of amiodarone with EPCLUSA is not recommended. In patients without alternative viable treatment options, cardiac monitoring is recommended. (5.1, 7.3) EPCLUSA® (sofosbuvir and velpatasvir) tablets, for oral use Initial U.S. Approval: 2016 -------------------------------INDICATIONS AND USAGE------------------------­ EPCLUSA is a fixed-dose combination of sofosbuvir, a hepatitis C virus (HCV) nucleotide analog NS5B polymerase inhibitor, and velpatasvir, an HCV NS5A inhibitor, and is indicated for the treatment of adult patients with chronic HCV genotype 1, 2, 3, 4, 5 or 6 infection (1): • without cirrhosis or with compensated cirrhosis • with decompensated cirrhosis for use in combination with ribavirin -------------------------------ADVERSE REACTIONS----------------------------­ • The most common adverse reactions (incidence greater than or equal to 10%, all grades) observed with treatment with EPCLUSA for 12 weeks are headache and fatigue. (6.1) • The most common adverse reactions (incidence greater than or equal to 10%, all grades) observed with treatment with EPCLUSA and ribavirin for 12 weeks in patients with decompensated cirrhosis are fatigue, anemia, nausea, headache, insomnia and diarrhea. (6.1) ------------------------DOSAGE AND ADMINISTRATION----------------------­ • Recommended dosage: One tablet (400 mg of sofosbuvir and 100 mg of velpatasvir) taken orally once daily with or without food (2.1) • See recommended treatment regimen and duration in patients with genotypes 1, 2, 3, 4, 5, or 6 HCV in table below: (2.1) Recommended Patient Population Treatment Regimen To report SUSPECTED ADVERSE REACTIONS, contact Gilead Sciences, Inc. at 1-800-GILEAD-5 or FDA at 1-800-FDA-1088 or www.fda.gov/medwatch. Patients without cirrhosis and patients with compensated EPCLUSA for 12 weeks cirrhosis (Child-Pugh A) Patients with decompensated EPCLUSA + ribavirin for cirrhosis (Child-Pugh B and C) 12 weeks • A dosage recommendation cannot be made for patients with severe renal impairment or end stage renal disease (2.2) ---------------------------------DRUG INTERACTIONS--------------------------­ • P-gp inducers and/or moderate to potent CYP inducers (e.g., rifampin, St. John’s wort, carbamazepine): May decrease concentrations of sofosbuvir and/or velpatasvir. Use of EPCLUSA with P-gp inducers and/or moderate to potent CYP inducers is not recommended (5.2, 7) • Consult the full prescribing information prior to use for potential drug interactions (5.1, 5.2, 7) -----------------------DOSAGE FORMS AND STRENGTHS-------------------­ Tablets: 400 mg sofosbuvir and 100 mg velpatasvir (3) See 17 for PATIENT COUNSELING INFORMATION and FDA-approved patient labeling. Revised: 06/2016 FULL PRESCRIBING INFORMATION: CONTENTS* 1 INDICATIONS AND USAGE 2 DOSAGE AND ADMINISTRATION 2.1 Recommended Dosage 2.2 No Dosage Recommendations in Severe Renal Impairment and End Stage Renal Disease 3 DOSAGE FORMS AND STRENGTHS 4 CONTRAINDICATIONS 5 WARNINGS AND PRECAUTIONS 5.1 Serious Symptomatic Bradycardia When Sofosbuvir Is Coadministered with Amiodarone and Another HCV Direct Acting Antiviral 5.2 Risk of Reduced Therapeutic Effect Due to Concomitant Use of EPCLUSA with Inducers of P-gp and/or Moderate to Potent Inducers of CYP 5.3 Risks Associated with Ribavirin and EPCLUSA Combination Treatment 6 ADVERSE REACTIONS 6.1 Clinical Trials Experience 6.2 Postmarketing Experience 7 DRUG INTERACTIONS 7.1 Potential for Other Drugs to Affect EPCLUSA 7.2 Potential for EPCLUSA to Affect Other Drugs 7.3 Established and Potentially Significant Drug Interactions 7.4 Drugs without Clinically Significant Interactions with EPCLUSA 8 USE IN SPECIFIC POPULATIONS 8.1 Pregnancy 8.2 Lactation 8.3 Females and Males of Reproductive Potential 8.4 Pediatric Use 8.5 Geriatric Use 8.6 Renal Impairment 8.7 Hepatic Impairment 10 OVERDOSAGE 11 DESCRIPTION 12 CLINICAL PHARMACOLOGY 12.1 Mechanism of Action 12.2 Pharmacodynamics 12.3 Pharmacokinetics 12.4 Microbiology 13 NONCLINICAL TOXICOLOGY 13.1 Carcinogenesis, Mutagenesis, Impairment of Fertility 14 CLINICAL STUDIES 14.1 Description of Clinical Trials 14.2 Clinical Trials in Subjects without Cirrhosis and Subjects with Compensated Cirrhosis 14.3 Clinical Trials in Subjects with Decompensated Cirrhosis 16 HOW SUPPLIED/STORAGE AND HANDLING 17 PATIENT COUNSELING INFORMATION * Sections or subsections omitted from the full prescribing information are not listed. 1 Reference ID: 3951919 FULL PRESCRIBING INFORMATION 1 INDICATIONS AND USAGE EPCLUSA is indicated for the treatment of adult patients with chronic hepatitis C virus (HCV) genotype 1, 2, 3, 4, 5 or 6 infection [see Dosage and Administration (2.1) and Clinical Studies (14)]: • without cirrhosis or with compensated cirrhosis • with decompensated cirrhosis for use in combination with ribavirin 2 DOSAGE AND ADMINISTRATION 2.1 Recommended Dosage The recommended dosage of EPCLUSA is one tablet taken orally once daily with or without food [see Clinical Pharmacology (12.3)]. One tablet of EPCLUSA contains 400 mg of sofosbuvir and 100 mg of velpatasvir. Table 1 shows the recommended treatment regimen and duration based on patient population. Table 1 Recommended Treatment Regimen in Patients with Genotype 1, 2, 3, 4, 5 or 6 HCV Patient Population Treatment Regimen and Duration Patients without cirrhosis and patients with compensated cirrhosis (Child-Pugh A) Patients with decompensated cirrhosis (Child-Pugh B or C) EPCLUSA 12 weeks EPCLUSA + ribavirina 12 weeks a. When administered with EPCLUSA, the recommended dosage of ribavirin is based on weight (administered with food): 1000 mg per day for patients less than 75 kg and 1200 mg for those weighing at least 75 kg, divided and administered twice daily. The starting dosage and on-treatment dosage of ribavirin can be decreased based on hemoglobin and creatinine clearance. For ribavirin dosage modifications, refer to the ribavirin prescribing information. 2.2 No Dosage Recommendations in Severe Renal Impairment and End Stage Renal Disease No dosage recommendation can be given for patients with severe renal impairment (estimated Glomerular Filtration Rate [eGFR] less than 30 mL/min/1.73 m2) or with end stage renal disease (ESRD), due to higher exposures (up to 20-fold) of the predominant sofosbuvir metabolite [see Use in Specific Populations (8.6) and Clinical Pharmacology (12.3)]. 3 DOSAGE FORMS AND STRENGTHS Each EPCLUSA tablet contains 400 mg of sofosbuvir and 100 mg of velpatasvir. The tablets are pink, diamond-shaped, film-coated, and debossed with “GSI” on one side and “7916” on the other side. Gilead Sciences Reference ID: 3951919 2 4 CONTRAINDICATIONS EPCLUSA and ribavirin combination regimen is contraindicated in patients for whom ribavirin is contraindicated. Refer to the ribavirin prescribing information for a list of contraindications for ribavirin. 5 WARNINGS AND PRECAUTIONS 5.1 Serious Symptomatic Bradycardia When Sofosbuvir Is Coadministered with Amiodarone and Another HCV Direct Acting Antiviral Postmarketing cases of symptomatic bradycardia and cases requiring pacemaker intervention have been reported when amiodarone is coadministered with sofosbuvir in combination with daclatasvir or simeprevir. A fatal cardiac arrest was reported in a patient taking amiodarone who was coadministered a sofosbuvir-containing regimen (HARVONI (ledipasvir/sofosbuvir)). Bradycardia has generally occurred within hours to days, but cases have been observed up to 2 weeks after initiating HCV treatment. Patients also taking beta blockers, or those with underlying cardiac comorbidities and/or advanced liver disease may be at increased risk for symptomatic bradycardia with coadministration of amiodarone. Bradycardia generally resolved after discontinuation of HCV treatment. The mechanism for this effect is unknown. Coadministration of amiodarone with EPCLUSA is not recommended. For patients taking amiodarone who have no other alternative viable treatment options and who will be coadministered EPCLUSA: • Counsel patients about the risk of symptomatic bradycardia. • Cardiac monitoring in an in-patient setting for the first 48 hours of coadministration is recommended, after which outpatient or self-monitoring of the heart rate should occur on a daily basis through at least the first 2 weeks of treatment. Patients who are taking EPCLUSA who need to start amiodarone therapy due to no other alternative viable treatment options should undergo similar cardiac monitoring as outlined above. Due to amiodarone’s long half-life, patients discontinuing amiodarone just prior to starting EPCLUSA should also undergo similar cardiac monitoring as outlined above. Patients who develop signs or symptoms of bradycardia should seek medical evaluation immediately. Symptoms may include near-fainting or fainting, dizziness or lightheadedness, malaise, weakness, excessive tiredness, shortness of breath, chest pains, confusion, or memory problems [see Adverse Reactions (6.2) and Drug Interactions (7.3)]. 5.2 Risk of Reduced Therapeutic Effect Due to Concomitant Use of EPCLUSA with Inducers of P-gp and/or Moderate to Potent Inducers of CYP Drugs that are inducers of P-gp and/or moderate to potent inducers of CYP2B6, CYP2C8, or CYP3A4 (e.g., rifampin, St. John’s wort, carbamazepine) may significantly Gilead Sciences Reference ID: 3951919 3 decrease plasma concentrations of sofosbuvir and/or velpatasvir, leading to potentially reduced therapeutic effect of EPCLUSA. The use of these agents with EPCLUSA is not recommended [see Drug Interactions (7.3)]. 5.3 Risks Associated with Ribavirin and EPCLUSA Combination Treatment If EPCLUSA is administered with ribavirin, the warnings and precautions for ribavirin apply to this combination regimen. Refer to the ribavirin prescribing information for a full list of the warnings and precautions for ribavirin [see Dosage and Administration (2.1)]. 6 ADVERSE REACTIONS The following serious adverse reactions are described below and elsewhere in labeling: • Serious Symptomatic Bradycardia When Sofosbuvir Is Coadministered with Amiodarone and Another HCV Direct Acting Antiviral [see Warnings and Precautions (5.1)]. 6.1 Clinical Trials Experience Because clinical trials are conducted under widely varying conditions, adverse reaction rates observed in the clinical trials of a drug cannot be directly compared to rates in the clinical trials of another drug and may not reflect the rates observed in practice. If EPCLUSA is administered with ribavirin, refer to the prescribing information for ribavirin for a description of ribavirin-associated adverse reactions. Adverse Reactions in Subjects without Cirrhosis or with Compensated Cirrhosis The adverse reactions data for EPCLUSA in patients without cirrhosis or with compensated cirrhosis were derived from three Phase 3 clinical trials (ASTRAL-1, ASTRAL-2, and ASTRAL-3) which evaluated a total of 1035 subjects infected with genotype 1, 2, 3, 4, 5, or 6 HCV, without cirrhosis or with compensated cirrhosis, who received EPCLUSA for 12 weeks. EPCLUSA was studied in placebo- and activecontrolled trials [see Clinical Studies (14.2)]. The proportion of subjects who permanently discontinued treatment due to adverse events was 0.2% for subjects who received EPCLUSA for 12 weeks. The most common adverse reactions (adverse events assessed as causally related by the investigator and at least 10%) were headache and fatigue in subjects treated with EPCLUSA for 12 weeks. Adverse reactions, all grades, observed in greater than or equal to 5% of subjects receiving 12 weeks of treatment with EPCLUSA in ASTRAL-1 include headache (22%), fatigue (15%), nausea (9%), asthenia (5%), and insomnia (5%). Of subjects receiving EPCLUSA who experienced these adverse reactions, 79% had an adverse reaction of mild severity (Grade 1). With the exception of asthenia, each of these adverse reactions occurred at a similar frequency or more frequently in subjects treated with placebo compared to subjects treated with EPCLUSA (asthenia: 3% versus 5% for the placebo and EPCLUSA groups, respectively). Gilead Sciences Reference ID: 3951919 4 The adverse reactions observed in subjects treated with EPCLUSA in ASTRAL-2 and ASTRAL-3 were consistent with those observed in ASTRAL-1. Irritability was also observed in greater than or equal to 5% of subjects treated with EPCLUSA in ASTRAL-3. Adverse Reactions in Subjects with Decompensated Cirrhosis The safety assessment of EPCLUSA in subjects infected with genotype 1, 2, 3, 4 or 6 HCV with decompensated cirrhosis was based on one Phase 3 trial (ASTRAL-4) including 87 subjects who received EPCLUSA with ribavirin for 12 weeks. All 87 subjects had Child-Pugh B cirrhosis at screening. On the first day of treatment with EPCLUSA with ribavirin, 6 subjects and 4 subjects were assessed to have Child-Pugh A and Child-Pugh C cirrhosis, respectively [see Clinical Studies (14.3)]. The most common adverse reactions (adverse events assessed as causally related by the investigator, all grades with frequency of 10% or greater) in the 87 subjects who received EPCLUSA with ribavirin for 12 weeks were fatigue (32%), anemia (26%), nausea (15%), headache (11%), insomnia (11%), and diarrhea (10%). Of subjects who experienced these adverse reactions, 98% had adverse reactions of mild to moderate in severity. A total of 4 (5%) subjects permanently discontinued EPCLUSA with ribavirin due to an adverse event; there was no adverse event leading to discontinuation that occurred in more than 1 subject. Decreases in hemoglobin to less than 10 g/dL and 8.5 g/dL during treatment were observed in 23% and 7% of subjects treated with EPCLUSA with ribavirin for 12 weeks, respectively. Ribavirin was permanently discontinued in 17% of subjects treated with EPCLUSA with ribavirin for 12 weeks, due to adverse reactions. Less Common Adverse Reactions Reported in Clinical Trials The following adverse reactions occurred in less than 5% of subjects without cirrhosis or with compensated cirrhosis treated with EPCLUSA for 12 weeks and are included because of a potential causal relationship. Rash: In the ASTRAL-1 study, rash occurred in 2% of subjects treated with EPCLUSA and in 1% of subjects treated with placebo. No serious adverse reactions of rash occurred and all rashes were mild or moderate in severity. Depression: In the ASTRAL-1 study, depressed mood occurred in 1% of subjects treated with EPCLUSA and was not reported by any subject taking placebo. No serious adverse reactions of depressed mood occurred and all events were mild or moderate in severity. The following adverse reactions occurred in less than 10% of subjects with decompensated cirrhosis (ASTRAL-4) treated with EPCLUSA with ribavirin for 12 weeks and are included because of a potential causal relationship. Gilead Sciences Reference ID: 3951919 5 Rash: Rash occurred in 5% of subjects treated with EPCLUSA with ribavirin. No serious adverse reactions of rash occurred and all rashes were mild or moderate in severity. Laboratory Abnormalities Lipase Elevations: In ASTRAL-1, isolated, asymptomatic lipase elevations of greater than 3xULN were observed in 3% and 1% of subjects treated with EPCLUSA and placebo for 12 weeks, respectively; and in 6% and 3% of subjects treated with EPCLUSA in ASTRAL-2 and ASTRAL-3, respectively. In the Phase 3 trial of subjects with decompensated cirrhosis (ASTRAL-4), lipase was assessed when amylase values were greater than or equal to 1.5xULN. Isolated, asymptomatic lipase elevations of greater than 3xULN were observed in 2% of subjects treated with EPCLUSA with ribavirin for 12 weeks. Creatine Kinase: In ASTRAL-1, isolated, asymptomatic creatine kinase elevations greater than or equal to 10xULN were reported in 1% and 0% of subjects treated with EPCLUSA and placebo for 12 weeks, respectively; and in 2% and 1% of subjects treated with EPCLUSA in ASTRAL-2 and ASTRAL-3, respectively. In the Phase 3 trial with decompensated cirrhosis (ASTRAL-4), isolated, asymptomatic creatine kinase elevations greater than or equal to 10xULN were reported in 1% of subjects treated with EPCLUSA with ribavirin for 12 weeks. Indirect Bilirubin: Increases in indirect bilirubin up to 3 mg/dL above baseline were noted among HIV-1/HCV coinfected subjects treated with EPCLUSA and an atazanavir/ritonavir-based antiretroviral regimen. The elevated indirect bilirubin values were not associated with clinical adverse events and all subjects completed 12 weeks of EPCLUSA without dose adjustment or treatment interruption of either EPCLUSA or HIV antiretroviral agents. 6.2 Postmarketing Experience The following adverse reactions have been identified during post approval use of sofosbuvir. Because postmarketing reactions are reported voluntarily from a population of uncertain size, it is not always possible to reliably estimate their frequency or establish a causal relationship to drug exposure. Cardiac Disorders Serious symptomatic bradycardia has been reported in patients taking amiodarone who initiated treatment with sofosbuvir in combination with another HCV direct acting antiviral [see Warnings and Precautions (5.1) and Drug Interactions (7.3)]. Gilead Sciences Reference ID: 3951919 6 7 DRUG INTERACTIONS 7.1 Potential for Other Drugs to Affect EPCLUSA Sofosbuvir and velpatasvir are substrates of drug transporters P-gp and BCRP while GS-331007 (the predominant circulating metabolite of sofosbuvir) is not. In vitro, slow metabolic turnover of velpatasvir by CYP2B6, CYP2C8, and CYP3A4 was observed. Drugs that are inducers of P-gp and/or moderate to potent inducers of CYP2B6, CYP2C8, or CYP3A4 (e.g., rifampin, St. John’s wort, carbamazepine) may decrease plasma concentrations of sofosbuvir and/or velpatasvir, leading to reduced therapeutic effect of EPCLUSA. The use of these agents with EPCLUSA is not recommended [see Warnings and Precautions (5.2) and Clinical Pharmacology (12.3)]. EPCLUSA may be coadministered with P-gp, BCRP, and CYP inhibitors. 7.2 Potential for EPCLUSA to Affect Other Drugs Velpatasvir is an inhibitor of drug transporters P-gp, breast cancer resistance protein (BCRP), OATP1B1, OATP1B3, and OATP2B1. Coadministration of EPCLUSA with drugs that are substrates of these transporters may increase the exposure of such drugs. 7.3 Established and Potentially Significant Drug Interactions Table 2 provides a listing of established or potentially clinically significant drug interactions. The drug interactions described are based on studies conducted with either EPCLUSA, the components of EPCLUSA (sofosbuvir and velpatasvir) as individual agents, or are predicted drug interactions that may occur with EPCLUSA [see Warnings and Precautions (5.1, 5.2) and Clinical Pharmacology (12.3)]. Table 2 Potentially Significant Drug Interactions: Alteration in Dose or Regimen May Be Recommended Based on Drug Interaction Studies or Predicted Interactiona Concomitant Drug Class: Drug Name Acid Reducing Agents: Antacids (e.g., aluminum and magnesium hydroxide) c H 2 -receptor antagonists (e.g., famotidine) Proton-pump inhibitors (e.g., omeprazole) c Effect on b Concentration ↓ velpatasvir Clinical Effect/Recommendation Velpatasvir solubility decreases as pH increases. Drugs that increase gastric pH are expected to decrease concentration of velpatasvir. Separate antacid and EPCLUSA administration by 4 hours. H 2 -receptor antagonists may be administered simultaneously with or 12 hours apart from EPCLUSA at a dose that does not exceed doses comparable to famotidine 40 mg twice daily. Coadministration of omeprazole or other proton-pump inhibitors is not recommended. If it is considered medically necessary to coadminister, EPCLUSA should be administered with food and taken 4 hours before omeprazole 20 mg. Use with other proton pumpinhibitors has not been studied. Gilead Sciences Reference ID: 3951919 7 Antiarrhythmics: amiodarone Effect on amiodarone, sofosbuvir, and velpatasvir concentrations unknown digoxin ↑ digoxin Anticancers: topotecan Anticonvulsants: carbamazepine phenytoin phenobarbital oxcarbazepine Antimycobacterials: rifabutin c rifampin rifapentine HIV Antiretrovirals: c efavirenz ↑ topotecan c Coadministration of amiodarone with EPCLUSA may result in serious symptomatic bradycardia. The mechanism of this effect is unknown. Coadministration of amiodarone with EPCLUSA is not recommended; if coadministration is required, cardiac monitoring is recommended [see Warnings and Precautions (5.1) and Adverse Reactions (6.2)]. Therapeutic concentration monitoring of digoxin is recommended when coadministered with EPCLUSA. Refer to digoxin prescribing information for monitoring and dose modification recommendations for concentration increases of less than 50%. Coadministration is not recommended. ↓ sofosbuvir ↓ velpatasvir Coadministration is not recommended. ↓ sofosbuvir ↓ velpatasvir Coadministration is not recommended. ↓ velpatasvir Coadministration of EPCLUSA with efavirenz-containing regimens is not recommended. Regimens containing tenofovir DF ↑ tenofovir tipranavir/ritonavir ↓ sofosbuvir ↓ velpatasvir ↓ sofosbuvir ↓ velpatasvir Monitor for tenofovir-associated adverse reactions in patients receiving EPCLUSA concomitantly with a regimen containing tenofovir DF. Refer to the prescribing information of the tenofovir DF-containing product for recommendations on renal monitoring. Coadministration is not recommended. Herbal Supplements: St. John’s wort (Hypericum perforatum) HMG-CoA Reductase Inhibitors: c rosuvastatin atorvastatin ↑ rosuvastatin ↑ atorvastatin Coadministration is not recommended. Coadministration of EPCLUSA with rosuvastatin may significantly increase the concentration of rosuvastatin, which is associated with increased risk of myopathy, including rhabdomyolysis. Rosuvastatin may be administered with EPCLUSA at a dose that does not exceed 10 mg. Coadministration of EPCLUSA with atorvastatin is expected to increase the concentrations of atorvastatin, which is associated with increased risk of myopathy, including rhabdomyolysis. Monitor closely for HMGCoA reductase inhibitor-associated adverse reactions, such as myopathy and rhabdomyolysis. Gilead Sciences Reference ID: 3951919 8 DF = disoproxil fumarate a. This table is not all inclusive. b. ↓ = decrease, ↑ = increase c. These interactions have been studied in healthy adults. 7.4 Drugs without Clinically Significant Interactions with EPCLUSA Based on drug interaction studies conducted with the components of EPCLUSA (sofosbuvir or velpatasvir) or EPCLUSA, no clinically significant drug interactions have been observed with the following drugs [see Clinical Pharmacology (12.3)]: • EPCLUSA: atazanavir/ritonavir, cyclosporine, darunavir/ritonavir, dolutegravir, elvitegravir/cobicistat/emtricitabine/tenofovir alafenamide, emtricitabine, raltegravir or rilpivirine • Sofosbuvir: ethinyl estradiol/norgestimate, methadone, or tacrolimus • Velpatasvir: ethinyl estradiol/norgestimate, ketoconazole, or pravastatin. See Table 2 for use of EPCLUSA with certain HIV antiretroviral regimens [see Drug Interactions (7.3)]. 8 USE IN SPECIFIC POPULATIONS 8.1 Pregnancy Risk Summary If EPCLUSA is administered with ribavirin, the combination regimen is contraindicated in pregnant women and in men whose female partners are pregnant. Refer to the ribavirin prescribing information for more information on ribavirin-associated risks of use during pregnancy. No adequate human data are available to establish whether or not EPCLUSA poses a risk to pregnancy outcomes. In animal reproduction studies, no evidence of adverse developmental outcomes was observed with the components of EPCLUSA (sofosbuvir or velpatasvir) at exposures greater than those in humans at the recommended human dose (RHD) [see Data]. During organogenesis in the mouse, rat and rabbit, systemic exposures (AUC) to velpatasvir were approximately 31 (mice), 6 (rats), and 0.4 (rabbits) times the exposure in humans at the RHD, while exposures to the predominant circulating metabolite of sofosbuvir (GS-331007) were approximately 4 (rats) and 10 (rabbits) times the exposure in humans at the RHD. In rat pre/postnatal development studies, maternal systemic exposures (AUC) to velpatasvir and GS-331007 were approximately 5 times the exposures of each component in humans at the RHD. The background risk of major birth defects and miscarriage for the indicated population is unknown. In the U.S. general population, the estimated background risk of major birth defects and miscarriage in clinically recognized pregnancies is 2–4% and 15–20%, respectively. Data Sofosbuvir: Sofosbuvir was administered orally to pregnant rats (up to 500 mg/kg/day) and rabbits (up to 300 mg/kg/day) on gestation days 6 to 18 and 6 to 19, respectively, Gilead Sciences Reference ID: 3951919 9 and also to rats (oral doses up to 500 mg/kg/day) on gestation day 6 to lactation/post­ partum day 20. No significant effects on embryo-fetal (rats and rabbits) or pre/postnatal (rats) development were observed at the highest doses tested. The systemic exposures (AUC) of the predominant circulating metabolite of sofosbuvir (GS-331007) during gestation were approximately 4 (rats) and 10 (rabbits) times the exposure in humans at the RHD. Velpatasvir: Velpatasvir was administered orally to pregnant mice (up to 1000 mg/kg/day), rats (up to 200 mg/kg/day) and rabbits (up to 300 mg/kg/day) on gestation days 6 to 15, 6 to 17, and 7 to 20, respectively, and also to rats (oral doses up to 200 mg/kg) on gestation day 6 to lactation/post-partum day 20. No significant effects on embryo-fetal (mice, rats, and rabbits) or pre/postnatal (rats) development were observed at the highest doses tested. The systemic exposures (AUC) of velpatasvir during gestation were approximately 31 (mice), 6 (rats), and 0.4 (rabbits) times the exposure in humans at the RHD. 8.2 Lactation Risk Summary It is not known whether the components of EPCLUSA and its metabolites are present in human breast milk, affect human milk production, or have effects on the breastfed infant. The predominant circulating metabolite of sofosbuvir (GS-331007) was the primary component observed in the milk of lactating rats administered sofosbuvir, without effect on nursing pups. When administered to lactating rats, velpatasvir was detected in the milk of lactating rats and in the plasma of nursing pups without effects on the nursing pups [see Data]. The development and health benefits of breastfeeding should be considered along with the mother’s clinical need for EPCLUSA and any potential adverse effects on the breastfed child from EPCLUSA or from the underlying maternal condition. If EPCLUSA is administered with ribavirin, the nursing mother’s information for ribavirin also applies to this combination regimen. Refer to the ribavirin prescribing information for more information on use during lactation. Data Sofosbuvir: No effects of sofosbuvir on growth and postnatal development were observed in nursing pups at the highest dose tested in rats. Maternal systemic exposure (AUC) to the predominant circulating metabolite of sofosbuvir (GS-331007) was approximately 5 times the exposure in humans at the RHD, with exposure of approximately 2% that of maternal exposure observed in nursing pups on lactation day 10. In a lactation study, sofosbuvir metabolites (primarily GS-331007) were excreted into the milk of lactating rats following administration of a single oral dose of sofosbuvir (20 mg/kg) on lactation day 2, with milk concentrations of approximately 10% that of maternal plasma concentrations observed 1 hour post-dose. Gilead Sciences Reference ID: 3951919 10 Velpatasvir: No effects of velpatasvir on growth and postnatal development were observed in nursing pups at the highest dose tested in rats. Maternal systemic exposure (AUC) to velpatasvir was approximately 5 times the exposure in humans at the RHD. Velpatasvir was present in the milk (approximately 173% that of maternal plasma concentrations) of lactating rats following a single oral dose of velpatasvir (30 mg/kg), and systemic exposure (AUC) in nursing pups was approximately 4% that of maternal exposure on lactation day 10. 8.3 Females and Males of Reproductive Potential If EPCLUSA is administered with ribavirin, the information for ribavirin with regard to pregnancy testing, contraception, and infertility also applies to this combination regimen. Refer to ribavirin prescribing information for additional information. 8.4 Pediatric Use Safety and effectiveness of EPCLUSA have not been established in pediatric patients. 8.5 Geriatric Use Clinical trials of EPCLUSA included 156 subjects aged 65 and over (12% of total number of subjects in the Phase 3 clinical trials). No overall differences in safety or effectiveness were observed between these subjects and younger subjects, and other reported clinical experience has not identified differences in responses between the elderly and younger patients, but greater sensitivity of some older individuals cannot be ruled out. No dosage adjustment of EPCLUSA is warranted in geriatric patients [see Clinical Pharmacology (12.3)]. 8.6 Renal Impairment No dosage adjustment of EPCLUSA is required for patients with mild or moderate renal impairment. The safety and efficacy of EPCLUSA have not been established in patients with severe renal impairment (eGFR less than 30 mL/min/1.73 m2) or ESRD requiring hemodialysis. No dosage recommendation can be given for patients with severe renal impairment or ESRD [see Dosage and Administration (2.2) and Clinical Pharmacology (12.3)]. Refer to ribavirin prescribing information regarding use of ribavirin in patients with renal impairment. 8.7 Hepatic Impairment No dosage adjustment of EPCLUSA is required for patients with mild, moderate, or severe hepatic impairment (Child-Pugh Class A, B, or C) [see Adverse Reactions (6.1), Clinical Pharmacology (12.3) and Clinical Studies (14)]. Clinical and hepatic laboratory monitoring (including direct bilirubin), as clinically indicated, is recommended for patients with decompensated cirrhosis receiving treatment with EPCLUSA and ribavirin [see Adverse Reactions (6.1)]. 10 OVERDOSAGE No specific antidote is available for overdose with EPCLUSA. If overdose occurs the patient must be monitored for evidence of toxicity. Treatment of overdose with Gilead Sciences Reference ID: 3951919 11 EPCLUSA consists of general supportive measures including monitoring of vital signs as well as observation of the clinical status of the patient. Hemodialysis can efficiently remove the predominant circulating metabolite of sofosbuvir, GS-331007, with an extraction ratio of 53%. Hemodialysis is unlikely to result in significant removal of velpatasvir since velpatasvir is highly bound to plasma protein. 11 DESCRIPTION EPCLUSA is a fixed-dose combination tablet containing sofosbuvir and velpatasvir for oral administration. Sofosbuvir is a nucleotide analog HCV NS5B polymerase inhibitor and velpatasvir is an NS5A inhibitor. Each tablet contains 400 mg sofosbuvir and 100 mg velpatasvir. The tablets include the following inactive ingredients: copovidone, croscarmellose sodium, magnesium stearate, and microcrystalline cellulose. The tablets are film-coated with a coating material containing the following inactive ingredients: iron oxide red, polyethylene glycol, polyvinyl alcohol, talc, and titanium dioxide. Sofosbuvir: The IUPAC name for sofosbuvir is (S)-Isopropyl 2-((S)-(((2R,3R,4R,5R)-5­ (2,4-dioxo-3,4-dihydropyrimidin-1(2H)-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2­ yl)methoxy)-(phenoxy)phosphorylamino)propanoate. It has a molecular formula of C 22 H 29 FN 3 O 9 P and a molecular weight of 529.45. It has the following structural formula: H N O O O O O HN P O O HO O N F Sofosbuvir is a white to off-white crystalline solid with a solubility of at least 2 mg/mL across the pH range of 2–7.7 at 37 oC and is slightly soluble in water. Velpatasvir: The IUPAC name for velpatasvir is Methyl {(1R)-2-[(2S,4S)-2-(5-{2­ [(2S,5S)-1-{(2S)-2-[(methoxycarbonyl)amino]-3-methylbutanoyl}-5-methylpyrrolidin-2-yl]­ 1,11-dihydro[2]benzopyrano[4',3':6,7]naphtho[1,2-d]imidazol-9-yl}-1H-imidazol-2-yl)-4­ (methoxymethyl)pyrrolidin-1-yl]-2-oxo-1-phenylethyl}carbamate. It has a molecular formula of C 49 H 54 N 8 O 8 and a molecular weight of 883.0. It has the following structural formula: Gilead Sciences Reference ID: 3951919 12 H3CO O NH O O N N H N N N N H O HN H3CO O OCH3 Velpatasvir is practically insoluble (less than 0.1 mg/mL) above pH 5, slightly soluble (3.6 mg/mL) at pH 2, and soluble (greater than 36 mg/mL) at pH 1.2. 12 CLINICAL PHARMACOLOGY 12.1 Mechanism of Action EPCLUSA is a fixed-dose combination of sofosbuvir and velpatasvir which are direct-acting antiviral agents against the hepatitis C virus [see Microbiology (12.4)]. 12.2 Pharmacodynamics Cardiac Electrophysiology The effect of sofosbuvir 400 mg (recommended dosage) and 1200 mg (three times the recommended dosage) on QTc interval was evaluated in an active-controlled (moxifloxacin 400 mg) thorough QT trial. At a dose three times the recommended dose, sofosbuvir does not prolong QTc to any clinically relevant extent. The effect of velpatasvir 500 mg (five times the recommended dosage) was evaluated in an active-controlled (moxifloxacin 400 mg) thorough QT trial. At a dose five times the recommended dose, velpatasvir does not prolong QTc interval to any clinically relevant extent. 12.3 Pharmacokinetics The pharmacokinetic properties of the components of EPCLUSA are provided in Table 3. The multiple dose pharmacokinetic parameters of sofosbuvir and its metabolite, GS-331007, and velpatasvir are provided in Table 4. Table 3 Pharmacokinetic Properties of the Components of EPCLUSA Absorption T max (h) a Effect of moderate meal (relative to fasting) a Effect of high fat meal (relative to fasting) Distribution % Bound to human plasma proteins Blood-to-plasma ratio Metabolism Metabolism Sofosbuvir Velpatasvir 0.5–1 ↑ 60% ↑ 78% 3 ↑ 34% ↑ 21% 61-65 0.7 >99.5 0.52-0.67 Cathepsin A CES1 HINT1 CYP2B6 CYP2C8 CYP3A4 Gilead Sciences Reference ID: 3951919 13 Elimination Major route of elimination t 1/2 (h) SOF: metabolism b GS-331007 : glomerular filtration and active tubular secretion SOF: 0.5 b GS-331007 : 25 e 80 14 c d % Of dose excreted in urine d % Of dose excreted in feces Biliary excretion as parent (77%) 15 0.4 94 CES1 = carboxylesterase 1; HINT1 = histidine triad nucleotide-binding protein 1 a. Values refer to mean systemic exposure. Moderate meal = ~600 kcal, 30% fat; high fat meal = ~800 kcal, 50% fat. EPCLUSA can be taken with or without food. b. GS-331007 is the primary circulating nucleoside metabolite of SOF. c. t 1/2 values refer to median terminal plasma half-life. 14 14 d. Single dose administration of [ C] SOF or [ C] VEL in mass balance studies. e. Predominantly as GS-331007. Table 4 Multiple Dose Pharmacokinetic Parameters of Sofosbuvir and its Metabolite, GS-331007, and Velpatasvir Following Oral Administration of EPCLUSA in HCV-Infected Adults Parameter Mean (%CV) C max (nanogram per mL) AUC tau (nanogram•hr per mL) C trough (nanogram per mL) a b c Sofosbuvir GS-331007 Velpatasvir 567 (30.7) 898 (26.7) 259 (54.3) 1268 (38.5) 14372 (28.0) 2980 (51.3) NA NA 42 (67.3) CV = coefficient of variation; NA = not applicable a. From Population PK analysis, N=666 b. From Population PK analysis, N=1029 c. From Population PK analysis, N=1025 Sofosbuvir and GS-331007 AUC 0-24 and C max were similar in healthy adult subjects and subjects with HCV infection. Relative to healthy subjects (N=331), velpatasvir AUC 0-24 and C max were 37% lower and 42% lower, respectively, in HCV-infected subjects. Velpatasvir AUC increases in a greater than proportional manner from 5 to 50 mg and in a less than proportional manner from 50 to 450 mg in healthy volunteers. However, velpatasvir exhibited more than or near dose-proportional increase in exposures 25 mg to 150 mg in HCV-infected patients when coadministered with sofosbuvir. Sofosbuvir and GS-331007 AUCs are near dose-proportional over the dose range of 200 mg to 1200 mg. Specific Populations Pediatric Patients: The pharmacokinetics of sofosbuvir or velpatasvir in pediatric patients has not been established [see Use in Specific Populations (8.4)]. Geriatric Patients: Population pharmacokinetic analysis in HCV-infected subjects showed that within the age range (18 to 82 years) analyzed, age did not have a Gilead Sciences Reference ID: 3951919 14 clinically relevant effect on the exposure to sofosbuvir, GS-331007 or velpatasvir [see Use in Specific Populations (8.5)]. Patients with Renal Impairment: The pharmacokinetics of sofosbuvir were studied in HCV negative subjects with mild (eGFR between 50 to less than 80 mL/min/1.73 m2), moderate (eGFR between 30 to less than 50 mL/min/1.73 m2), severe renal impairment (eGFR less than 30 mL/min/1.73 m2), and subjects with ESRD requiring hemodialysis following a single 400 mg dose of sofosbuvir. Relative to subjects with normal renal function (eGFR greater than 80 mL/min/1.73 m2), the sofosbuvir AUC 0-inf was 61%, 107%, and 171% higher in subjects with mild, moderate, and severe renal impairment, while the GS-331007 AUC 0-inf was 55%, 88%, and 451% higher, respectively. In subjects with ESRD, relative to subjects with normal renal function, sofosbuvir and GS-331007 AUC 0-inf was 28% and 1280% higher when sofosbuvir was dosed 1 hour before hemodialysis compared with 60% and 2070% higher when sofosbuvir was dosed 1 hour after hemodialysis, respectively. A 4 hour hemodialysis session removed approximately 18% of administered dose [see Dosage and Administration (2.2) and Use in Specific Populations (8.6)]. The pharmacokinetics of velpatasvir were studied with a single dose of 100 mg velpatasvir in HCV negative subjects with severe renal impairment (eGFR less than 30 mL/min by Cockcroft-Gault). No clinically relevant differences in velpatasvir pharmacokinetics were observed between healthy subjects and subjects with severe renal impairment. Patients with Hepatic Impairment: The pharmacokinetics of sofosbuvir were studied following 7-day dosing of 400 mg sofosbuvir in HCV-infected subjects with moderate and severe hepatic impairment (Child-Pugh Class B and C, respectively). Relative to subjects with normal hepatic function, the sofosbuvir AUC 0-24 were 126% and 143% higher in moderate and severe hepatic impairment, while the GS-331007 AUC 0-24 were 18% and 9% higher, respectively. Population pharmacokinetics analysis in HCV-infected subjects indicated that cirrhosis (including decompensated cirrhosis) had no clinically relevant effect on the exposure of sofosbuvir and GS-331007 [see Use in Specific Populations (8.7)]. The pharmacokinetics of velpatasvir were studied with a single dose of 100 mg velpatasvir in HCV negative subjects with moderate and severe hepatic impairment (Child-Pugh Class B and C). Velpatasvir plasma exposure (AUC inf ) was similar in subjects with moderate hepatic impairment, severe hepatic impairment, and control subjects with normal hepatic function. Population pharmacokinetics analysis in HCVinfected subjects indicated that cirrhosis (including decompensated cirrhosis) had no clinically relevant effect on the exposure of velpatasvir [see Use in Specific Populations (8.7)]. Gilead Sciences Reference ID: 3951919 15 Race: Population pharmacokinetics analysis in HCV-infected subjects indicated that race had no clinically relevant effect on the exposure of sofosbuvir, GS-331007 or velpatasvir. Gender: Population pharmacokinetics analysis in HCV-infected subjects indicated that gender had no clinically relevant effect on the exposure of sofosbuvir, GS-331007 or velpatasvir. Drug Interaction Studies After oral administration of EPCLUSA, sofosbuvir is rapidly absorbed and subject to extensive first-pass hepatic extraction (hydrolysis followed by sequential phosphorylation) to form the pharmacologically active triphosphate. In clinical pharmacology studies, both sofosbuvir and the primary circulating metabolite GS-331007 (dephosphorylated nucleotide metabolite) were monitored for purposes of pharmacokinetic analyses. Sofosbuvir and velpatasvir are substrates of drug transporters P-gp and BCRP while GS-331007 is not. Velpatasvir is also transported by OATP1B1 and OATP1B3. In vitro, slow metabolic turnover of velpatasvir by CYP2B6, CYP2C8, and CYP3A4 was observed. Inducers of P-gp and/or moderate to potent inducers of CYP2B6, CYP2C8, or CYP3A4 (e.g., rifampin, St. John’s wort, carbamazepine) may decrease plasma concentrations of sofosbuvir and/or velpatasvir, leading to reduced therapeutic effect of EPCLUSA [see Warnings and Precautions (5.2) and Drug Interactions (7.3)]. Coadministration with drugs that inhibit P-gp and/or BCRP may increase sofosbuvir and/or velpatasvir plasma concentrations without increasing GS-331007 plasma concentration. Drugs that inhibit CYP2B6, CYP2C8, or CYP3A4 may increase plasma concentration of velpatasvir. Velpatasvir is an inhibitor of drug transporter P-gp, BCRP, OATP1B1, OATP1B3, and OATP2B1, and its involvement in drug interactions with these transporters is primarily limited to the process of absorption. At clinically relevant concentration, velpatasvir is not an inhibitor of hepatic transporters OATP1A2 or OCT1, renal transporters OCT2, OAT1, OAT3 or MATE1, or CYP or UGT1A1 enzymes. Sofosbuvir and GS-331007 are not inhibitors of drug transporters P-gp, BCRP, OATP1B1, OATP1B3 and OCT1 and GS-331007 is not an inhibitor of OAT1, OAT3, OCT2, and MATE1. Sofosbuvir and GS-331007 are not inhibitors or inducers of CYP or UGT1A1 enzymes. The effects of coadministered drugs on the exposure of sofosbuvir, GS-331007, and velpatasvir are shown in Table 5. The effects of sofosbuvir, velpatasvir or EPCLUSA on the exposure of coadministered drugs are shown in Table 6. Gilead Sciences Reference ID: 3951919 16 Table 5 Coadmini­ stered Drug Atazanavir/ ritonavir + emtricitabine/ tenofovir DF Cyclosporine Darunavir/ ritonavir + emtricitabine/ tenofovir DF Efavirenz/ emtricitabine/ b tenofovir DF Elvitegravir/ cobicistat/ emtricitabine/ tenofovir c alafenamide Elvitegravir/ cobicistat/ emtricitabine/ d tenofovir DF Famotidine Drug Interactions: Changes in Pharmacokinetic Parameters for Sofosbuvir, GS-331007, and Velpatasvir in the Presence of the Coadministered Druga Dose of Coadmini­ stered Drug (mg) 300/100 + 200/300 once daily 600 single dose 800/100 + 200/300 once daily 600/200/300 once daily 150/150/200/10 once daily 150/150/200/300 once daily 40 single dose simultaneously with EPCLUSA SOF Dose (mg) 400 once daily VEL Dose (mg) 100 once daily 400 single dose ND ND 100 single dose 400 once daily 400 once daily 400 once daily 400 once daily 400 single dose 100 once daily 100 once daily 100 once daily 100 once daily 100 single dose Mean Ratio (90% CI) of Sofosbuvir, GS-331007 and Velpatasvir PK With/Without Coadministered Drug No Effect=1.00 N 24 C max AUC C min sofosbuvir 1.12 (0.97, 1.29) 1.22 (1.12, 1.33) NA GS-331007 1.21 (1.12, 1.29) 1.32 (1.27, 1.36) 1.42 (1.37, 1.49) velpatasvir 1.55 (1.41, 1.71) 2.42 (2.23, 2.64) 4.01 (3.57, 4.50) sofosbuvir 2.54 (1.87, 3.45) 4.53 (3.26, 6.30) NA GS-331007 0.60 (0.53, 0.69) 1.04 (0.90, 1.20) NA velpatasvir 1.56 (1.22, 2.01) 2.03 (1.51, 2.71) NA sofosbuvir 0.62 (0.54, 0.71) 0.72 (0.66, 0.80) NA GS-331007 1.04 (0.99, 1.08) 1.13 (1.08, 1.18) 1.13 (1.06, 1.19) velpatasvir 0.76 (0.65, 0.89) 0.84 (0.72, 0.98) 1.01 (0.87, 1.18) sofosbuvir 1.38 (1.14, 1.67) 0.97 (0.83, 1.14) NA GS-331007 0.86 (0.80, 0.93) 0.90 (0.85, 0.96) 1.01 (0.95, 1.07) velpatasvir 0.53 (0.43, 0.64) 0.47 (0.39, 0.57) 0.43 (0.36, 0.52) sofosbuvir 1.23 (1.07, 1.42) 1.37 (1.24, 1.52) NA GS-331007 1.29 (1.25, 1.33) 1.48 (1.43, 1.53) 1.58 (1.52, 1.65) velpatasvir 1.30 (1.17, 1.45) 1.50 (1.35, 1.66) 1.60 (1.44, 1.78) sofosbuvir 1.01 (0.85, 1.19) 1.24 (1.13, 1.37) NA GS-331007 1.13 (1.07, 1.18) 1.35 (1.30, 1.40) 1.45 (1.38, 1.52) velpatasvir 1.05 (0.93, 1.19) 1.19 (1.07, 1.34) 1.37 (1.22, 1.54) sofosbuvir 0.92 (0.82, 1.05) 0.82 (0.74, 0.91) NA GS-331007 0.84 (0.78, 0.89) 0.94 (0.91, 0.98) NA 19 12 29 14 23 24 60 Gilead Sciences Reference ID: 3951919 17 40 single dose 12 hours prior to EPCLUSA Ketoconazole Lopinavir/ ritonavir + emtricitabine/ tenofovir DF Methadone 200 twice daily 4x200/50 + 200/300 once daily 30 to 130 daily 20 once daily simultaneously with EPCLUSA 20 once daily 12 hours prior to EPCLUSA Omeprazole 20 once daily 2 hours prior to EPCLUSA 20 once daily 4 hours after EPCLUSA 40 once daily 4 hours after EPCLUSA 60 ND 400 once daily 100 single dose 100 once daily 400 once daily ND 400 single dose fasted 100 single dose fasted 400 single dose fasted 400 single dose e fed 400 single dose e fed 400 single dose e fed 100 single dose fasted 100 single dose e fed 100 single dose e fed 100 single dose e fed velpatasvir 0.80 (0.70, 0.91) 0.81 (0.71, 0.91) NA sofosbuvir 0.77 (0.68, 0.87) 0.80 (0.73, 0.88) NA GS-331007 1.20 (1.13, 1.28) 1.04 (1.01, 1.08) NA velpatasvir 0.87 (0.76, 1.00) 0.85 (0.74, 0.97) NA velpatasvir 1.29 (1.02, 1.64) 1.71 (1.35, 2.18) NA sofosbuvir 0.59 (0.49, 0.71) 0.71 (0.64, 0.78) NA GS-331007 1.01 (0.98, 1.05) 1.15 (1.09, 1.21) 1.15 (1.07, 1.25) velpatasvir 0.70 (0.59, 0.83) 1.02 (0.89, 1.17) 1.63 (1.43, 1.85) sofosbuvir 0.95 (0.68, 1.33) 1.30 (1.00, 1.69) NA GS-331007 0.73 (0.65, 0.83) 1.04 (0.89, 1.22) NA sofosbuvir 0.66 (0.55, 0.78) 0.71 (0.60, 0.83) NA GS-331007 1.18 (1.10, 1.26) 1.00 (0.95, 1.05) NA velpatasvir 0.63 (0.50, 0.78) 0.64 (0.52, 0.79) NA sofosbuvir 0.55 (0.47, 0.64) 0.56 (0.49, 0.65) NA GS-331007 1.26 (1.18, 1.34) 0.97 (0.94, 1.01) NA velpatasvir 0.43 (0.35, 0.54) 0.45 (0.37, 0.55) NA sofosbuvir 0.84 (0.68, 1.03) 1.08 (0.94, 1.25) NA GS-331007 0.94 (0.88, 1.02) 0.99 (0.96, 1.03) NA velpatasvir 0.52 (0.43, 0.64) 0.62 (0.51, 0.75) NA sofosbuvir 0.79 (0.68, 0.92) 1.05 (0.94, 1.16) NA GS-331007 0.91 (0.85, 0.98) 0.99 (0.95, 1.02) NA velpatasvir 0.67 (0.58, 0.78) 0.74 (0.63, 0.86) NA sofosbuvir 0.70 (0.57, 0.87) 0.91 (0.76, 1.08) NA GS-331007 1.01 (0.96, 1.07) 0.99 (0.94, 1.03) NA velpatasvir 0.44 0.47 NA 12 24 14 60 60 40 38 40 Gilead Sciences Reference ID: 3951919 18 (0.37, 0.60) sofosbuvir 0.23 (0.19, 0.29) 0.28 (0.24, 0.32) NA GS-331007 1.23 (1.14, 1.34) 0.95 (0.88, 1.03) NA 400 single dose ND ND 100 single dose 12 velpatasvir 0.29 (0.23, 0.37) 0.18 (0.15, 0.22) NA 600 single dose ND 100 single dose 12 velpatasvir 1.28 (1.05, 1.56) 1.46 (1.17, 1.83) NA sofosbuvir 0.97 (0.65, 1.43) 1.13 (0.81, 1.57) NA 5 single dose 400 single dose GS-331007 0.97 (0.83, 1.14) 1.00 (0.87, 1.13) NA 600 once daily Rifampin Tacrolimus (0.34, 0.57) ND 17 16 NA = not available/not applicable, ND = not dosed, DF = disoproxil fumarate. a. All interaction studies conducted in healthy volunteers. b. Administered as ATRIPLA (efavirenz, emtricitabine and tenofovir DF fixed-dose combination). c. Administered as GENVOYA (elvitegravir, cobicistat, emtricitabine and tenofovir alafenamide fixed-dose combination). d. Administered as STRIBILD (elvitegravir, cobicistat, emtricitabine and tenofovir DF fixed-dose combination). e. EPCLUSA was administered under fasted conditions in the reference arms. No effect on the pharmacokinetic parameters of sofosbuvir, GS-331007 or velpatasvir was observed with dolutegravir, the combination of emtricitabine, rilpivirine, and tenofovir DF, emtricitabine, raltegravir, or tenofovir DF. Table 6 Co­ administered Drug Changes in Pharmacokinetic Parameters for Coadministered Drug in the Presence of Sofosbuvir, Velpatasvir, or EPCLUSAa Dose of Co­ administered Drug (mg) SOF Dose (mg) VEL Dose (mg) Mean Ratio (90% CI) of Coadministered Drug PK With/Without Sofosbuvir, Velpatasvir or EPCLUSA No Effect=1.00 C max AUC C min 1.09 (1.00, 1.19) 1.20 (1.10, 1.31) 1.39 (1.20, 1.61) 0.89 (0.82, 0.97) 0.97 (0.89, 1.05) 1.29 (1.15, 1.44) 1.01 (0.96, 1.06) 1.02 (0.99, 1.04) 1.06 (1.02, 1.11) tenofovir DF 300 once daily 1.55 (1.43, 1.68) 1.30 (1.24, 1.36) 1.39 (1.31, 1.48) darunavir 800 once daily 0.90 (0.86, 0.95) 0.92 (0.87, 0.98) 0.87 (0.79, 0.95) 1.07 (0.97, 1.17) 1.12 (1.05, 1.19) 1.09 (1.02, 1.15) 1.05 (1.01, 1.08) 1.05 (1.02, 1.08) 1.04 (0.98, 1.09) 1.55 (1.45, 1.66) 1.39 (1.33, 1.44) 1.52 (1.45, 1.59) N atazanavir 300 once daily Atazanavir/ ritonavir + emtricitabine/ b tenofovir DF Darunavir/ ritonavir + emtricitabine/ c tenofovir DF ritonavir 100 once daily emtricitabine 200 once daily ritonavir 100 once daily emtricitabine 200 once daily 400 once daily 400 once daily 100 once daily 100 once daily 24 29 tenofovir DF 300 once daily Gilead Sciences Reference ID: 3951919 19 Digoxin 1.88 (1.71, 2.08) 1.34 (1.13, 1.60) NA 0.81 (0.74, 0.89) 0.85 (0.80, 0.91) 0.90 (0.85, 0.95) 1.07 (0.98, 1.18) 1.07 (1.00, 1.14) 1.10 (0.97, 1.25) tenofovir DF 300 once daily 1.77 (1.53, 2.04) 1.81 (1.68, 1.94) 2.21 (2.00, 2.43) elvitegravir 150 once daily 0.87 (0.80, 0.94) 0.94 (0.88, 1.00) 1.08 (0.97, 1.20) 1.16 (1.09, 1.23) 1.30 (1.23, 1.38) 2.03 (1.67, 2.48) 1.02 (0.97, 1.06) 1.01 (0.98, 1.04) 1.02 (0.97, 1.07) tenofovir alafenamide 10 once daily 0.80 (0.68, 0.94) 0.87 (0.81, 0.94) NA elvitegravir 150 once daily 0.93 (0.86, 1.00) 0.93 (0.87, 0.99) 0.97 (0.91, 1.04) 1.11 (1.06, 1.17) 1.23 (1.17, 1.29) 1.71 (1.54, 1.90) 1.02 (0.97, 1.08) 1.01 (0.98, 1.04) 1.06 (1.01, 1.11) tenofovir DF 300 once daily 1.36 (1.25, 1.47) 1.35 (1.29, 1.42) 1.45 (1.39, 1.51) emtricitabine 200 once daily 0.95 (0.90, 1.00) 0.99 (0.97, 1.02) 1.05 (0.99, 1.11) 0.93 (0.88, 0.98) 0.95 (0.90, 1.00) 0.96 (0.90, 1.03) 1.44 (1.33, 1.55) 1.40 (1.34, 1.46) 1.84 (1.76, 1.92) 0.25 single dose ND 100 21 efavirenz 600 once daily Efavirenz/ emtricitabine/ d tenofovir DF Elvitegravir/ cobicistat/ emtricitabine/ tenofovir e alafenamide Elvitegravir/ cobicistat/ emtricitabine/ f tenofovir DF Emtricitabine/ rilpivirine/ g tenofovir DF emtricitabine 200 once daily cobicistat 150 once daily emtricitabine 200 once daily cobicistat 150 once daily emtricitabine 200 once daily rilpivirine 25 once daily 400 once daily 400 once daily 400 once daily 400 once daily 100 once daily 100 once daily 100 once daily 100 once daily 15 24 24 24 tenofovir DF 300 once daily Norelgestromin Norgestrel norgestimate 0.180/0.215/0.25/ ethinyl estradiol 0.025 once daily Ethinyl estradiol Pravastatin pravastatin 40 single dose ND 100 once daily 13 0.97 (0.88, 1.07) 0.90 (0.82, 0.98) 0.92 (0.83, 1.03) 400 once daily ND 15 1.07 (0.94, 1.22) 1.06 (0.92, 1.21) 1.07 (0.89, 1.28) ND 100 once daily 13 0.96 (0.78, 1.19) 0.91 (0.73, 1.15) 0.92 (0.73, 1.18) 400 once daily ND 15 1.18 (0.99, 1.41) 1.19 (0.98, 1.45) 1.23 (1.00, 1.51) ND 100 once daily 12 1.39 (1.17, 1.66) 1.04 (0.87, 1.24) 0.83 (0.65, 1.06) 400 once daily ND 15 1.15 (0.97, 1.36) 1.09 (0.94, 1.26) 0.99 (0.80, 1.23) ND 100 once daily 18 1.28 (1.08, 1.52) 1.35 (1.18, 1.54) NA Gilead Sciences Reference ID: 3951919 20 Rosuvastatin rosuvastatin 10 single dose ND 100 once daily 18 emtricitabine 200 once daily Raltegravir + emtricitabine/ tenofovir DF tenofovir DF 300 once daily 400 once daily 100 once daily 30 raltegravir 400 twice daily 2.61 (2.32, 2.92) 2.69 (2.46, 2.94) NA 1.08 (1.04, 1.12) 1.05 (1.03, 1.07) 1.02 (0.97, 1.08) 1.46 (1.39, 1.54) 1.40 (1.34, 1.45) 1.70 (1.61, 1.79) 1.03 (0.74, 1.43) 0.97 (0.73, 1.28) 0.79 (0.42, 1.48) 400 0.73 1.09 ND 16 NA single (0.59, 0.90) (0.84, 1.40) dose NA = not available/not applicable, ND = not dosed, DF = disoproxil fumarate. a. All interaction studies conducted in healthy volunteers. b. Comparison based on exposures when administered as atazanavir/ritonavir + emtricitabine/tenofovir DF. c. Comparison based on exposures when administered as darunavir/ritonavir + emtricitabine/tenofovir DF. d. Administered as ATRIPLA (efavirenz, emtricitabine and tenofovir DF fixed-dose combination). e. Administered as GENVOYA (elvitegravir, cobicistat, emtricitabine and tenofovir alafenamide fixed-dose combination). f. Administered as STRIBILD (elvitegravir, cobicistat, emtricitabine and tenofovir DF fixed-dose combination). g. Administered as COMPLERA (emtricitabine, rilpivirine and tenofovir DF fixed-dose combination). Tacrolimus 5 single dose No effect on the pharmacokinetic parameters of the following coadministered drugs was observed with EPCLUSA (dolutegravir or lopinavir/ritonavir) or its components sofosbuvir (cyclosporine or methadone) or velpatasvir (cyclosporine). 12.4 Microbiology Mechanism of Action Sofosbuvir is an inhibitor of the HCV NS5B RNA-dependent RNA polymerase, which is required for viral replication. Sofosbuvir is a nucleotide prodrug that undergoes intracellular metabolism to form the pharmacologically active uridine analog triphosphate (GS-461203), which can be incorporated into HCV RNA by the NS5B polymerase and acts as a chain terminator. In a biochemical assay, GS-461203 inhibited the polymerase activity of the recombinant NS5B from HCV genotype 1b, 2a, 3a and 4a with an IC 50 value ranging from 0.36 to 3.3 micromolar. GS-461203 is neither an inhibitor of human DNA and RNA polymerases nor an inhibitor of mitochondrial RNA polymerase. Velpatasvir is an inhibitor of the HCV NS5A protein, which is required for viral replication. Resistance selection in cell culture and cross-resistance studies indicate velpatasvir targets NS5A as its mode of action. Antiviral Activity The EC 50 values of sofosbuvir and velpatasvir against full-length or chimeric replicons encoding NS5B and NS5A sequences from the laboratory strains are presented in Table 7. The EC 50 values of sofosbuvir and velpatasvir against clinical isolates are presented in Table 8. Gilead Sciences Reference ID: 3951919 21 Table 7 Activity of Sofosbuvir and Velpatasvir Against Full Length or Chimeric Laboratory Replicons a Replicon Genotype Sofosbuvir EC 50 , nM Velpatasvir EC 50 , nM 1a 40 0.014 1b 110 0.016 2a 50 0.005-0.016 2b 15 b 0.002-0.006 3a 50 0.004 4a 40 0.009 4d 33.4 0.004 5a b 15 6a 14-25 6e NA a c c d 0.021-0.054 b 0.006-0.009 d 0.130 NA = not available a. Mean value from multiple experiments of same laboratory replicon. b. Stable chimeric 1b replicons carrying NS5B genes from genotype 2b, 5a or 6a were used for testing. c. Data from various strains of full-length NS5A replicons or chimeric NS5A replicons carrying full-length NS5A genes that contain L31 or M31 polymorphisms. d. Data from a chimeric NS5A replicon carrying NS5A amino acids 9-184. Table 8 Activity of Sofosbuvir and Velpatasvir Against Transient Replicons Containing NS5A or NS5B from Clinical Isolates Replicons Containing NS5B from Clinical Isolates Replicons Containing NS5A from Clinical Isolates Replicon Genotype Number of clinical isolates Median sofosbuvir EC 50 , nM (range) Number of clinical isolates Median velpatasvir EC 50 , nM (range) 1a 67 62 (29-128) 23 0.019 (0.011-0.078) 1b 29 102 (45-170) 34 0.012 (0.005-0.500) 2a 1 28 8 0.011 (0.006-0.364) 2b 14 30 (14-81) 16 0.002 (0.0003-0.007) 3a 106 81 (24-181) 38 0.005 (0.002-1.871) 4a NA NA 5 0.002 (0.001-0.004) 4d NA NA 10 0.007 (0.004-0.011) 4r NA NA 7 0.003 (0.002-0.006) 5a NA NA 42 0.005 (0.001-0.019) 6a NA NA 26 0.007 (0.0005-0.113) 6e NA NA 15 0.024 (0.005-0.433) NA = not available Velpatasvir was not antagonistic in reducing HCV RNA levels in replicon cells when combined with sofosbuvir or interferon-α, ribavirin, an HCV NS3/4A protease inhibitor, or HCV NS5B non-nucleoside inhibitors. Gilead Sciences Reference ID: 3951919 22 Resistance In Cell Culture HCV replicons with reduced susceptibility to sofosbuvir have been selected in cell culture for multiple genotypes including 1b, 2a, 2b, 3a, 4a, 5a, and 6a. Reduced susceptibility to sofosbuvir was associated with the NS5B substitution S282T in all replicon genotypes examined. An M289L substitution developed along with the S282T substitution in genotype 2a, 5 and 6 replicons. Site-directed mutagenesis of the S282T substitution in replicons of genotypes 1 to 6 conferred 2- to 18-fold reduced susceptibility to sofosbuvir. HCV genotype 1a, 1b, 2a, 3a, 4a, 5a, and 6a replicon variants with reduced susceptibility to velpatasvir were selected in cell culture. Variants developed amino acid substitutions at NS5A resistance-associated positions 24, 28, 30, 31, 32, 58, 92, and 93. Phenotypic analysis of site-directed mutant replicons of the selected NS5A substitutions showed that single and double combinations of L31V and Y93H/N in genotype 1a, the combination of L31V + Y93H in genotype 1b, Y93H/S in genotype 3a, and L31V and P32A/L/Q/R in genotype 6 conferred greater than 100-fold reduction in velpatasvir susceptibility. In the genotype 2a replicon, the single mutants F28S and Y93H showed 91-fold and 46-fold reduced susceptibility to velpatasvir, respectively. The single mutant Y93H conferred 3-fold reduced susceptibility to velpatasvir in genotype 4a replicons. Combinations of these NS5A substitutions often showed greater reductions in susceptibility to velpatasvir than single substitutions alone. In Clinical Trials Studies in Subjects without Cirrhosis and Subjects with Compensated Cirrhosis In a pooled analysis of subjects without cirrhosis or with compensated cirrhosis who received EPCLUSA for 12 weeks in Phase 3 trials (ASTRAL-1, ASTRAL-2 and ASTRAL-3), 12 subjects (2 with genotype 1 [1a, 1c/h] and 10 with genotype 3a) qualified for resistance analysis due to virologic failure. No subjects with genotype 2, 4, 5, or 6 HCV infection experienced virologic failure. Of the 2 genotype 1 virologic failure subjects, one subject had virus with emergent NS5A resistance substitution Y93N and the other had virus with emergent NS5A resistance substitutions Y93H and low-level K24M/T and L31I/V at virologic failure. The latter subject had genotype 1c/h virus at baseline harboring NS5A resistance polymorphisms (Q30R, L31M, H58P) relative to genotype 1a. No sofosbuvir NS5B nucleoside analog resistance-associated substitutions were observed at failure in the 2 subjects. Of the 10 genotype 3a virologic failure subjects, NS5A resistance substitution Y93H was observed in all 10 subjects at failure (7 subjects had Y93H emerge post-treatment and 3 subjects had Y93H at baseline and post-treatment). Treatment-emergent sofosbuvir NS5B substitutions L314F (n=2) and L314I (n=1) were observed at high frequency (greater than or equal to 15%) in the NS5B polymerase in 3 genotype 3a subjects who relapsed: one in the EPCLUSA group and two in the sofosbuvir plus ribavirin 24-week group. In addition, low frequency (less than 4%) treatment-emergent Gilead Sciences Reference ID: 3951919 23 L314P was detected in 2 genotype 3a subjects who relapsed, including one subject in the sofosbuvir plus ribavirin 24-week group in ASTRAL-3 and one in the EPCLUSA group in ASTRAL-4. The clinical significance of this substitution is unknown. Studies in Subjects with Decompensated Cirrhosis In the ASTRAL-4 trial in subjects with decompensated cirrhosis who received EPCLUSA with ribavirin for 12 weeks, 3 subjects (1 with genotype 1a and 2 with genotype 3a) qualified for resistance analysis due to virologic failure. No subjects with genotype 2 or 4 HCV infection who received EPCLUSA with ribavirin for 12 weeks experienced virologic failure. The genotype 1 virologic failure subject had no NS5A or NS5B resistance substitutions at failure. The 2 genotype 3a virologic failure subjects had the NS5A resistance substitutions Y93H and either low-level M28V or S38P emerge at failure. One of these subjects also developed low levels (less than 5%) of NS5B nucleoside analog inhibitor resistance substitutions N142T and E237G at failure. In the ASTRAL-4 trial, 2 subjects treated with EPCLUSA for 12 or 24 weeks without ribavirin had emergent sofosbuvir NS5B resistance-associated substitutions S282T at low levels (less than 5%) along with L159F. EPCLUSA for 12 or 24 weeks without ribavirin is not recommended in patients with decompensated cirrhosis. Persistence of Resistance-Associated Substitutions No data are available on the persistence of sofosbuvir or velpatasvir resistanceassociated substitutions. NS5A resistance-associated substitutions observed with administration of other NS5A inhibitors have been found to persist for longer than 1 year in most patients. The long-term clinical impact of the emergence or persistence of virus containing sofosbuvir or velpatasvir resistance-associated substitutions is unknown. Effect of Baseline HCV Polymorphisms on Treatment Response Analyses were conducted to explore the association between relapse rates and pre­ existing baseline NS5A resistance-associated polymorphisms (RAPs) (any change from reference at NS5A amino acid positions 24, 28, 30, 31, 58, 92, or 93) identified by population or deep sequencing analysis at a sensitivity threshold of 15% or higher for subjects without cirrhosis or with compensated cirrhosis in ASTRAL-1, ASTRAL-2 and ASTRAL-3 and subjects with decompensated cirrhosis in ASTRAL-4. Studies in Subjects without Cirrhosis and Subjects with Compensated Cirrhosis Among the subjects who received treatment with EPCLUSA for 12 weeks, 18% (37/209), 32% (38/117), 64% (149/232), 20% (56/274), 63% (73/115), 9% (3/34) and 83% (35/42) of subjects with genotype 1a, 1b, 2, 3, 4, 5 and 6 HCV, respectively, had baseline virus with NS5A RAPs. Gilead Sciences Reference ID: 3951919 24 Genotype 1: Among the 75 genotype 1 subjects who had baseline NS5A RAPs, one subject (1%) with Q30R, L31M and H58P polymorphisms at baseline and compensated cirrhosis relapsed. Genotype 3: Among the 56 genotype 3 subjects who had baseline NS5A RAPs, 4 subjects (7%) relapsed (3 with baseline Y93H and 1 with baseline A30K). Overall, 20% (3/15) of genotype 3 subjects with the Y93H polymorphism at baseline relapsed. For genotype 3 subjects with compensated cirrhosis, relapse rates were 33% (3/9) for subjects with baseline NS5A RAPs compared to 6% (4/71) for subjects without baseline NS5A RAPs. Genotypes 2, 4, 5, and 6: The presence of baseline NS5A RAPs did not affect relapse rates for subjects with genotypes 2, 4, 5 and 6, because all achieved SVR12. SVR12 was achieved in all 77 subjects who had baseline NS5B nucleoside analog inhibitor resistance polymorphisms including N142T, L159F, E/N237G, C/M289L/I, L320F/I/V, V321A/I, and S282G + V321I. The sofosbuvir NS5B nucleoside analog inhibitor resistance substitution S282T was not detected in the baseline NS5B sequence of any subject using 1% deep sequencing cutoff in Phase 3 trials. Studies in Subjects with Decompensated Cirrhosis In ASTRAL-4, the prevalence of NS5A RAPs at baseline was 24% (48/198), 60% (6/10), 11% (4/37), and 63% (5/8) in GT1, GT2, GT3, and GT4 HCV subjects, respectively. No subjects with genotypes 2, 4 and 6 relapsed. There were no subjects with genotype 5 in this trial. For genotype 1 subjects, the overall relapse rates were numerically lower for the 12­ week EPCLUSA with ribavirin group (2%; 1/66) compared to EPCLUSA 12-week and 24-week treatment groups. For subjects with NS5A RAPs, relapse rates were 0% (0/17) compared to 2% (1/49) for subjects without NS5A RAPs in the 12-week EPCLUSA with ribavirin containing group. For genotype 3 subjects, overall virologic failure rates were numerically lower for the 12­ week EPCLUSA with ribavirin group (15%; 2/13) compared to EPCLUSA 12-week and 24-week treatment groups. There are insufficient data to determine the impact of HCV NS5A RAPs in genotype 3 subjects with decompensated cirrhosis. Three subjects in the EPCLUSA with ribavirin 12-week group had baseline NS5B nucleoside analog inhibitor polymorphisms (N142T and L159F) using 1% deep sequencing cutoff and all 3 subjects achieved SVR12. Cross Resistance Both sofosbuvir and velpatasvir were fully active against substitutions associated with resistance to other classes of direct-acting antivirals with different mechanisms of action, such as NS5B non-nucleoside inhibitors and NS3 protease inhibitors. The Gilead Sciences Reference ID: 3951919 25 efficacy of EPCLUSA has not been established in patients who have previously failed treatment with other regimens that include an NS5A inhibitor. 13 NONCLINICAL TOXICOLOGY 13.1 Carcinogenesis, Mutagenesis, Impairment of Fertility Carcinogenesis and Mutagenesis Sofosbuvir: Sofosbuvir was not genotoxic in a battery of in vitro or in vivo assays, including bacterial mutagenicity, chromosome aberration using human peripheral blood lymphocytes and in vivo mouse micronucleus assays. Sofosbuvir was not carcinogenic in a 2-year mouse study (up to 200 mg/kg/day in males and 600 mg/kg/day in females) and in a 2-year rat study (up to 750 mg/kg/day), resulting in exposures of the predominant circulating metabolite GS-331007 of approximately 3 and 15 times (in mice) and 7 and 9 times (in rats), in males and females, respectively, the exposure in humans at the recommended human dose (RHD). Velpatasvir: Velpatasvir was not genotoxic in a battery of in vitro or in vivo assays, including bacterial mutagenicity, chromosome aberration using human peripheral blood lymphocytes, and in vivo rat micronucleus assays. Carcinogenicity studies of velpatasvir in mice and rats are ongoing. Impairment of Fertility Sofosbuvir: Sofosbuvir had no effects on embryo-fetal viability or on fertility when evaluated in rats. At the highest dose tested, AUC exposure to the predominant circulating metabolite GS-331007 was approximately 4 times the exposure in humans at the RHD. Velpatasvir: Velpatasvir had no effects on embryo-fetal viability or on fertility when evaluated in rats. At the highest dose tested, velpatasvir exposure was approximately 6 times the exposure in humans at the RHD. 14 CLINICAL STUDIES 14.1 Description of Clinical Trials Table 9 presents the clinical trial design including different treatment groups that were conducted with EPCLUSA with and without ribavirin in subjects with chronic hepatitis C (HCV) genotype 1, 2, 3, 4, 5 and 6 infection. For detailed description of trial design and recommended regimen and duration [see Dosage and Administration (2) and Clinical Studies (14.2, 14.3)]. Gilead Sciences Reference ID: 3951919 26 Table 9 Trials Conducted with EPCLUSA in Subjects with Genotype 1, 2, 3, 4, 5 or 6 HCV Infection Population EPCLUSA and Comparator Groups (Number of Subjects Treated) ASTRAL-1 Genotype 1, 2, 4, 5, and 6 TN and TE, without cirrhosis or with compensated cirrhosis EPCLUSA 12 weeks (624) Placebo 12 weeks (116) ASTRAL-2 Genotype 2 TN and TE, without cirrhosis or with compensated cirrhosis EPCLUSA 12 weeks (134) SOF + RBV 12 weeks (132) ASTRAL-3 Genotype 3 TN and TE, without cirrhosis or with compensated cirrhosis EPCLUSA 12 weeks (277) SOF + RBV 24 weeks (275) ASTRAL-4 Genotype 1, 2, 3, 4, 5, and 6 TN and TE, with CP class B decompensated cirrhosis EPCLUSA 12 weeks (90) EPCLUSA + RBV 12 weeks (87) EPCLUSA 24 weeks (90) Trial TN = treatment-naïve subjects; TE = treatment-experienced subjects (including those who have failed a peginterferon alfa + ribavirin based regimen with or without an HCV protease inhibitor); SOF = sofosbuvir, RBV = ribavirin; CP = Child-Pugh. The ribavirin dosage was weight-based (1000 mg daily administered in two divided doses for subjects less than 75 kg and 1200 mg for those greater than or equal to 75 kg) and administered in two divided doses when used in combination with sofosbuvir in the ASTRAL-2 and ASTRAL-3 trials or in combination with EPCLUSA in the ASTRAL-4 trial. Ribavirin dosage adjustments were performed according to the ribavirin prescribing information. Serum HCV RNA values were measured during the clinical trials using the COBAS AmpliPrep/COBAS Taqman HCV test (version 2.0) with a lower limit of quantification (LLOQ) of 15 IU/mL. Sustained virologic response (SVR12), defined as HCV RNA less than LLOQ at 12 weeks after the cessation of treatment, was the primary endpoint in all the trials. Relapse is defined as HCV RNA greater than or equal to LLOQ during the post-treatment period after having achieved HCV RNA less than LLOQ at the end of treatment. On-treatment virologic failure is defined as breakthrough, rebound, or non-response. 14.2 Clinical Trials in Subjects without Cirrhosis and Subjects with Compensated Cirrhosis Genotype 1, 2, 4, 5, and 6 HCV Infected Adults (ASTRAL-1) ASTRAL-1 was a randomized, double-blind, placebo-controlled trial that evaluated 12 weeks of treatment with EPCLUSA compared with 12 weeks of placebo in subjects with genotype 1, 2, 4, 5, or 6 HCV infection without cirrhosis or with compensated cirrhosis. Subjects with genotype 1, 2, 4 or 6 HCV infection were randomized in a 5:1 ratio to treatment with EPCLUSA or placebo for 12 weeks. Subjects with genotype 5 HCV infection were enrolled to the EPCLUSA group. Randomization was stratified by HCV genotype (1, 2, 4, 6, and indeterminate) and the presence or absence of compensated cirrhosis. Demographics and baseline characteristics were balanced between the EPCLUSA and placebo group. Of the 740 treated subjects, the median age was 56 years (range: 18 to Gilead Sciences Reference ID: 3951919 27 82); 60% of the subjects were male; 79% were White, 9% were Black; 21% had a baseline body mass index at least 30 kg/m2; the proportions of subjects with genotype 1, 2, 4, 5, or 6 HCV infection were 53%, 17%, 19%, 5% and 7%, respectively; 69% had non-CC IL28B alleles (CT or TT); 74% had baseline HCV RNA levels at least 800,000 IU/mL; 19% had compensated cirrhosis; and 32% were treatment-experienced. Table 10 presents SVR12 and other virologic outcomes in EPCLUSA-treated subjects in the ASTRAL-1 trial by HCV genotype. No subjects in the placebo group achieved SVR12. Table 10 Study ASTRAL-1: Virologic Outcomes by HCV Genotype in EPCLUSA-Treated Subjects without Cirrhosis or with Compensated Cirrhosis (12 Weeks After Treatment) EPCLUSA 12 Weeks (N=624) Total (all GTs) (N=624) GT-1 GT-1a (N=210) GT-1b (N=118) Total (N=328) GT-2 (N=104) GT-4 (N=116) 99% 98% 99% 98% 100% 100% (618/624) (206/210) (117/118) (323/328) (104/104) (116/116) SVR12 GT-5 (N=35) GT-6 (N=41) 97% (34/35) 100% (41/41) Outcome for Subjects without SVR On-Treatment Virologic Failure a Relapse Other b 0/624 0/210 0/118 0/328 0/104 0/116 0/35 0/41 <1% (2/623) <1% (1/209) 1% (1/118) 1% (2/327) 0/104 0/116 0/35 0/41 1% (4/624) 1% (3/210) 0/118 1% (3/328) 0/104 0/116 3% (1/35) 0/41 GT = genotype; no subjects in the placebo group achieved SVR12. a. The denominator for relapse is the number of subjects with HCV RNA