CLINICAL PHARMACOLOGY
Mechanism Of Action
Daclatasvir is a direct-acting
antiviral agent (DAA) against the hepatitis C virus [see Microbiology].
Pharmacodynamics
Cardiac Electrophysiology
At a dose 3 times the maximum
recommended dose, daclatasvir did not prolong the QT interval to any clinically
relevant extent.
Pharmacokinetics
The pharmacokinetic properties
of daclatasvir were evaluated in healthy adult subjects and in subjects with
chronic HCV. Administration of daclatasvir tablets in HCV-infected subjects
resulted in approximately dose-proportional increases in Cmax, AUC, and Cmin up
to 60 mg once daily. Steady state is anticipated after approximately 4 days of
once-daily daclatasvir administration. Exposure of daclatasvir was similar
between healthy and HCV-infected subjects.
Population pharmacokinetic estimates for daclatasvir 60
mg once daily in chronic HCV-infected subjects are shown in Table 8.
Table 8: Population Pharmacokinetic Estimates for
Daclatasvir in Chronic HCV-Infected Subjects Receiving Daclatasvir 60 mg Once
Daily and Sofosbuvir 400 mg Once Daily
Parameters |
Daclatasvir 60 mg once daily (n=152) |
AUC0-24h (ng•h/mL) |
Mean ± standard deviation |
10973 ± 5288 |
Median (range) |
9680 (3807-41243) |
C24h (ng/mL) |
Mean ± standard deviation |
182±137 |
Median (range) |
148 (21-1050) |
Absorption And Bioavailability
In HCV-infected subjects
following multiple oral doses of daclatasvir tablet ranging from 1 mg to 100 mg
once daily, peak plasma concentrations occurred within 2 hours post dose.
In vitro studies with human Caco-2 cells indicated that daclatasvir
is a substrate of P-gp. The absolute bioavailability of the tablet formulation
is 67%.
Effect Of Food On Oral
Absorption
In healthy subjects,
administration of a daclatasvir 60 mg tablet after a high-fat, high-caloric
meal (approximately 951 total kcal, 492 kcal from fat, 312 kcal from
carbohydrates, 144 kcal from protein) decreased daclatasvir Cmax and AUC(0-inf)
by 28% and 23%, respectively, compared with fasted conditions. A food effect
was not observed with administration of a daclatasvir 60 mg tablet after a low-fat,
low-caloric meal (approximately 277 total kcal, 41 kcal from fat, 190 kcal from
carbohydrates, 44 kcal from protein) compared with fasted conditions [see DOSAGE
AND ADMINISTRATION].
Distribution
With multiple dosing, protein binding of daclatasvir in
HCV-infected subjects was approximately 99% and independent of dose at the dose
range studied (1-100 mg). In subjects who received daclatasvir 60 mg tablet
orally followed by 100 μg [13C,15N]-daclatasvir
intravenous dose, estimated volume of distribution at steady state was 47 L.
Metabolism
Daclatasvir is a substrate of CYP3A, with CYP3A4 being
the primary CYP isoform responsible for metabolism. Following single-dose oral
administration of 25 mg 14C-daclatasvir in healthy subjects, the
majority of radioactivity in plasma was predominately attributed to parent drug
(97% or greater).
Elimination
Following single-dose oral administration of 25 mg 14C-daclatasvir
in healthy subjects, 88% of total radioactivity was recovered in feces (53% of
the dose as unchanged daclatasvir) and 6.6% of the dose was excreted in the
urine (primarily as unchanged daclatasvir). Following multiple-dose
administration of daclatasvir in HCV-infected subjects, with doses ranging from
1 mg to 100 mg once daily, the terminal elimination half-life of daclatasvir
ranged from approximately 12 to 15 hours. In subjects who received daclatasvir
60 mg tablet orally followed by 100 μg [13C,15N]daclatasvir
intravenous dose, the total clearance was 4.2 L/h.
Specific Populations
Renal Impairment
The pharmacokinetics of daclatasvir following a single 60
mg oral dose was studied in nonâ⬓HCVinfected subjects with renal impairment.
Using a regression analysis, the predicted AUC(0-inf) of daclatasvir was
estimated to be 26%, 60%, and 80% higher in subjects with creatinine clearance (CLcr)
values of 60, 30, and 15 mL/min, respectively, relative to subjects with normal
renal function (CLcr of 90 mL/min, defined using the Cockcroft-Gault CLcr
formula), and daclatasvir unbound AUC(0-inf) was predicted to be 18%, 39%, and
51% higher for subjects with CLcr values of 60, 30, and 15 mL/min,
respectively, relative to subjects with normal renal function. Using observed
data, subjects with end-stage renal disease requiring hemodialysis had a 27%
increase in daclatasvir AUC(0-inf) and a 20% increase in unbound AUC(0-inf) compared
to subjects with normal renal function as defined using the Cockcroft-Gault CLcr
formula [see Use In Specific Populations].
Daclatasvir is highly protein bound to plasma proteins
and is unlikely to be removed by dialysis.
Hepatic Impairment
The pharmacokinetics of daclatasvir following a single 30
mg oral dose was studied in nonâ⬓HCVinfected subjects with mild (Child-Pugh A),
moderate (Child-Pugh B), and severe (Child-Pugh C) hepatic impairment compared
to a corresponding matched control group. The Cmax and AUC(0-inf) of total
daclatasvir (free and protein-bound drug) were lower by 46% and 43%,
respectively, in Child-Pugh A subjects; by 45% and 38%, respectively, in
Child-Pugh B subjects; and by 55% and 36%, respectively, in Child-Pugh C
subjects. The Cmax and AUC(0-inf) of unbound daclatasvir were lower by 43% and
40%, respectively, in Child-Pugh A subjects; by 14% and 2%, respectively, in
Child-Pugh B subjects; and by 33% and 5%, respectively, in Child-Pugh C
subjects [see Use In Specific Populations].
Pediatric Patients
The pharmacokinetics of daclatasvir in pediatric patients
has not been evaluated.
Geriatric Patients
Population pharmacokinetic analysis in HCV-infected
subjects showed that within the age range (18-79 years) analyzed, age did not
have a clinically relevant effect on the pharmacokinetics of daclatasvir [see
Use In Specific Populations].
Gender
Population pharmacokinetic analyses in HCV-infected
subjects estimated that female subjects have a 30% higher daclatasvir AUC
compared to male subjects. This difference in daclatasvir AUC is not considered
clinically relevant.
Race
Population pharmacokinetic analyses in HCV-infected
subjects indicated that race had no clinically relevant effect on daclatasvir
exposure.
Drug Interactions
Cytochrome P450 (CYP) Enzymes
Daclatasvir is a substrate of CYP3A. In vitro,
daclatasvir did not inhibit (IC50 greater than 40 microM) CYP enzymes 1A2, 2B6,
2C8, 2C9, 2C19, or 2D6. Daclatasvir did not have a clinically relevant effect
on the exposure of midazolam, a sensitive CYP3A substrate.
Transporters
Daclatasvir is a substrate of P-gp. However,
cyclosporine, which inhibits multiple transporters including P-gp, did not have
a clinically relevant effect on the pharmacokinetics of daclatasvir.
Daclatasvir, in vitro, did not inhibit OCT2 and did not have a clinically
relevant effect on the pharmacokinetics of tenofovir, an OAT substrate.
Daclatasvir demonstrated inhibitory effects on digoxin (a P-gp substrate) and
rosuvastatin (an OATP 1B1, OATP 1B3, and BCRP substrate) in drug-drug interaction
trials.
Drug interaction studies were conducted with daclatasvir
and other drugs likely to be coadministered or drugs used as probes to evaluate
potential drug-drug interactions. The effects of daclatasvir on the Cmax, AUC,
and Cmin of the coadministered drug are summarized in Table 9, and the effects
of the coadministered drug on the Cmax, AUC, and Cmin of daclatasvir are
summarized in Table 10. For information regarding clinical recommendations, see
CONTRAINDICATIONS and DRUG INTERACTIONS. Drug interaction studies
were conducted in healthy adults unless otherwise noted.
Table 9: Effect of DAKLINZA on the Pharmacokinetics of
Concomitant Drugs
Concomitant Drug |
Coadministered Drug Dose |
DAKLINZA Dose |
Ratio of Pharmacokinetic Parameters of Coadministered Drug Combination/No Combination (90% CI) |
Cmax |
AUC |
Cmina |
Buprenorphine /Naloxone |
Stable maintenance 8/2 mg to 24/6 mg QD |
60 mg QD |
Buprenorphineb 1.30 (1.03, 1.64)
Norbuprenorphineb 1.65 (1.38, 1.99) |
Buprenorphineb 1.37 (1.24, 1.52)
Norbuprenorphineb 1.62 (1.30, 2.02) |
Buprenorphineb 1.17 (1.03, 1.32)
Norbuprenorphineb 1.46 (1.12, 1.89) |
Darunavirc |
600 mg BID with ritonavir 100 mg BID |
30 mg QD |
0.97 (0.80, 1.17) |
0.90 (0.73, 1.11) |
0.98 (0.67, 1.44) |
Digoxin |
0.125 mg QD |
60 mg QD |
1.65 (1.52, 1.80) |
1.27 (1.20, 1.34) |
1.18 (1.09, 1.28) |
Dolutegravir |
50 mg QD |
60 mg QD |
1.29 (1.07, 1.57) |
1.33 (1.11, 1.59) |
1.45 (1.25, 1.68) |
Lopinavirc |
400 mg BID with ritonavir 100 mg BID |
30 mg QD |
1.22 (1.06, 1.41) |
1.15 (0.77, 1.72) |
1.54 (0.46, 5.07) |
Methadone |
Stable maintenance 40120 mg QD |
60 mg QD |
Total methadoned: 1.09 (0.99, 1.21)
R-methadoned: 1.07 (0.97, 1.18) |
Total methadoned: 1.11 (0.97, 1.26)
R-methadoned: 1.08 (0.94, 1.24) |
Total methadoned: 1.12 (0.96, 1.29)
R-methadoned: 1.08 (0.93, 1.26) |
Rosuvastatin |
10 mg single dose |
60 mg QD |
2.04 (1.83, 2.26) |
1.58 (1.44, 1.74) |
NA |
Simeprevir |
150 mg QD |
60 mg QD |
1.39 (1.27, 1.52) |
1.44 (1.32, 1.56) |
1.49 (1.33, 1.67) |
Note: In Table 9, for the concomitant
medication, drug-drug interaction data were not included if 90% CIs for Cmax,
AUC, and Cmin (if applicable for Cmin) were within 80% to 125%. These
concomitant medications include cyclosporine, escitalopram, ethinyl
estradiol/norgestimate, midazolam, tacrolimus, and tenofovir disoproxil
fumarate.
a Cmin was defined as either the Ctau or the Ctrough concentration
value.
b The buprenorphine and norbuprenorphine pharmacokinetic parameters
were dose normalized to 8 mg.
c Samples up to 6 hours collected; C0h substituted for C12h concentration
value.
d The methadone pharmacokinetic parameters were dose normalized to
40 mg.
NA = Not available. |
Table 10: Effect of Coadministered Drugs on DAKLINZA
Pharmacokinetics
Concomitant Drug |
Coadministered Drug Dose |
DAKLINZA Dose |
Ratio of Pharmacokinetic Parameters of Daclatasvir Combination/No Combination (90% CI) |
Cmax |
AUC |
C mina |
Atazanavir/ ritonavir |
300 mg/100 mg QD |
20 mg QD (test arm) |
0.45 (0.41, 0.49)b |
0.70 (0.65, 0.75)b |
1.22 (1.08, 1.37)b |
Cyclosporine |
400 mg single dose |
60 mg QD |
1.04 (0.94, 1.15) |
1.40 (1.29, 1.53) |
1.56 (1.41, 1.71) |
Darunavir/ ritonavir |
800 mg/100 mg QD |
30 mg QD (test arm) |
0.38 (0.35, 0.42)b |
0.70 (0.66, 0.75)b |
NA |
Dolutegravir |
50 mg QD |
60 mg QD |
1.03 (0.84, 1.25) |
0.98 (0.83, 1.15) |
1.06 (0.88, 1.29) |
Efavirenz |
600 mg QD |
120 mg QD (test arm) |
1.67 (1.51, 1.84)b |
1.37 (1.21, 1.55)b |
0.83 (0.69, 1.00)b |
Escitalopram |
10 mg QD |
60 mg QD |
1.14 (0.98, 1.32) |
1.12 (1.01, 1.26) |
1.23 (1.09, 1.38) |
Famotidine |
40 mg single dose |
60 mg single dose (2 hours after famotidine administration) |
0.56 (0.46, 0.67) |
0.82 (0.70, 0.96) |
0.89 (0.75, 1.06) |
Ketoconazole |
400 mg QD |
10 mg single dose |
1.57 (1.31, 1.88) |
3.00 (2.62, 3.44) |
NA |
Lopinavir/ ritonavir |
400 mg/100 mg BID |
30 mg QD (test arm) |
0.34 (0.31, 0.37)b |
0.58 (0.54, 0.62)b |
NA |
Omeprazole |
40 mg single dose |
60 mg single dose |
0.64 (0.54, 0.77) |
0.84 (0.73, 0.96) |
0.92 (0.80, 1.05) |
Rifampin |
600 mg QD |
60 mg single dose |
0.44 (0.40, 0.48) |
0.21 (0.19, 0.23) |
NA |
Simeprevir |
150 mg QD |
60 mg QD |
1.50 (1.39, 1.62) |
1.96 (1.84, 2.10) |
2.68 (2.42, 2.98) |
Tenofovir disoproxil fumarate |
300 mg QD |
60 mg QD |
1.06 (0.98, 1.15) |
1.10 (1.01, 1.21) |
1.15 (1.02, 1.30) |
Note: In Table 10, drug-drug interaction data for
daclatasvir were not included for a study with tacrolimus because the 90% CIs
for Cmax, AUC, and Cmin were within 80% to 125%.
a Cmin was defined as either the Ctau or the Ctrough daclatasvir
concentration value.
b Observed, non-dose normalized data. For the reference arm, a 60 mg
QD dose of daclatasvir was administered without the HIV comedications (boosted
protease inhibitors, efavirenz) in order to compare the effect on daclatasvir
exposures.
NA = Not available. |
No clinically relevant interaction is anticipated for
daclatasvir or the following concomitant medications: peginterferon alfa,
ribavirin, or antacids. No clinically relevant interaction is anticipated for
daclatasvir with concomitant use of rilpivirine.
Microbiology
Mechanism Of Action
Daclatasvir is an inhibitor of NS5A, a nonstructural
protein encoded by HCV. Daclatasvir binds to the N-terminus of NS5A and
inhibits both viral RNA replication and virion assembly. Characterization of
daclatasvir-resistant viruses, biochemical studies, and computer modeling data
indicate that daclatasvir interacts with the N-terminus within Domain 1 of the
protein, which may cause structural distortions that interfere with NS5A
functions.
Antiviral Activity
Daclatasvir had median EC50 values of 0.008 nM (range,
0.002-0.03 nM; n=35), 0.002 nM (range, 0.0007-0.006 nM; n=30), and 0.2 nM
(range, 0.006-3.2 nM; n=17) against hybrid replicons containing genotypes 1a,
1b, and 3a subject-derived NS5A sequences, respectively, without detectable
daclatasvir resistance-associated polymorphisms at NS5A amino acid positions
28, 30, 31, or 93. Daclatasvir activity was reduced against genotypes 1a, 1b,
and 3a subject-derived replicons with resistance-associated polymorphisms at
positions 28, 30, 31, or 93, with median EC50 values of 76 nM (range, 4.6-2409
nM; n=5), 0.05 nM (range, 0.002-10 nM; n=12), and 13.5 nM (range, 1.3-50 nM;
n=4), respectively. Similarly, the EC50 values of daclatasvir against 3
genotype 3b and 1 genotype 3i subject-derived NS5A sequences with polymorphisms
(relative to a genotype 3a reference) at positions 30+31 (genotype 3b) or 30+62
(genotype 3i) were ≥3620 nM.
Daclatasvir was not antagonistic with interferon alfa,
HCV NS3/4A protease inhibitors, HCV NS5B nucleoside analog inhibitors, and HCV
NS5B non-nucleoside inhibitors in cell culture combination antiviral activity
studies using the cell-based HCV replicon system.
Resistance
In Cell Culture
HCV genotype 1a, 1b, and 3a replicon variants with
reduced susceptibility to daclatasvir were selected in cell culture, and the
genotype and phenotype of daclatasvir-resistant NS5A amino acid variants were
characterized. Phenotypic analysis of genotype 1a replicons expressing single
NS5A M28T, Q30E, Q30H, Q30R, L31V, Y93C, Y93H, and Y93N substitutions exhibited
500-, 18500-, 1083-, 900-, 2500-, 1367-, 8500-, and 34833-fold reduced
susceptibility to daclatasvir, respectively. For genotype 1b, L31V and Y93H
single substitutions and L31M/Y93H and L31V/Y93H combinations exhibited 33-,
30-, 16000-, and 33667-fold reduced susceptibility to daclatasvir,
respectively. A P32-deletion (P32X) in genotype 1b reduced daclatasvir
susceptibility by >1,000,000-fold. For genotype 3a, single A30K, L31F, L31I,
and Y93H substitutions exhibited 117-, 320-, 240-, and 3733-fold reduced susceptibility
to daclatasvir, respectively.
In Clinical Studies
Among subjects with HCV genotype 1 or genotype 3
infection and treated in the ALLY-1, -2, and -3 trials with DAKLINZA and
sofosbuvir with or without ribavirin for 12 weeks, 31 subjects (11 with
genotype 1a, 1 with genotype 1b, and 19 with genotype 3) qualified for
resistance analysis due to virologic failure. Post-baseline NS5A and NS5B
population-based nucleotide sequence analysis results were available for 31 and
28 subjects, respectively.
Virus from all 31 subjects at the time of virologic
failure harbored one or more of the following NS5A resistance-associated
substitutions (including pre-existing amino acid polymorphisms or
treatment-emergent substitutions): M28T, Q30H/K/R, L31M/V, H54R, H58D/P, or
Y93C/N for genotype 1a subjects, P32-deletion (P32X) for the genotype 1b
subject, and A30K/S, L31I, S62A/L/P/R/T, or Y93H for genotype 3 subjects. Among
HCV genotype 1a virologic failure subjects, the most common NS5A amino acid
substitutions occurred at position Q30 (Q30H/K/R; 73% [8/11], all
treatment-emergent). Among HCV genotype 3 virologic failure subjects, the most
common NS5A amino acid polymorphism or treatment-emergent substitution was Y93H
(89% [17/19], treatment-emergent in 11 of 17 subjects).
For NS5B, 6 of 28 subjects at the time of virologic
failure had virus with NS5B substitutions possibly associated with sofosbuvir
resistance or exposure: A112T, L159F, E237G, or Q355H (genotype 1a subjects),
or S282T+Q355H (genotype 3 subject).
Persistence Of Resistance-Associated Substitutions
In a long-term follow-up study that included HCV genotype
1-and genotype 3-infected subjects treated with daclatasvir-containing regimens
in phase 2/3 clinical trials, viral populations with treatment-emergent NS5A
resistance-associated substitutions persisted at detectable levels for more
than 1 year in most subjects.
Effect Of Baseline HCV Amino Acid Polymorphisms On Treatment
Response
Genotype 1a NS5A Polymorphisms
In HCV genotype 1a-infected
subjects with cirrhosis, the presence of an NS5A amino acid polymorphism at
position M28, Q30, L31, or Y93 (defined as any change from reference identified
by population-based nucleotide sequencing) was associated with reduced efficacy
of DAKLINZA and sofosbuvir with or without ribavirin for 12 weeks in the ALLY-1
and ALLY-2 trials (see Table 11). Due to the limited sample size, insufficient
data are available to determine the impact of specific NS5A polymorphisms at
these positions on SVR12 rates in subjects with cirrhosis. Six of 54 subjects
(11%) with cirrhosis had one of the following specific NS5A polymorphisms at
baseline: M28V/T (n=2), Q30R (n=1), L31M (n=2), or Y93N (n=1); 2 subjects with
M28V or Q30R achieved SVR12 while 4 subjects with M28T, L31M, or Y93N did not
achieve SVR. Eleven of 112 subjects (10%) without cirrhosis had one or more of
the following specific NS5A polymorphisms at baseline: M28T/V (n=3), Q30H/L/R
(n=5), L31M (n=1), and Y93C/H/S (n=4); all noncirrhotic subjects with these
baseline NS5A polymorphisms achieved SVR12. Based on an analysis of 1026 HCV
genotype 1a NS5A amino acid sequences from pooled clinical trials, the
prevalence of polymorphisms at these positions was 11% overall, and 11% in the
U.S.
Genotype 1b NS5A Polymorphisms
In a pooled analysis of 43 subjects infected with HCV
genotype 1b with available baseline nucleotide sequence data in ALLY-1 and -2,
virus from 21% (n=9) of subjects receiving DAKLINZA and sofosbuvir with or
without ribavirin had one of the following baseline NS5A amino acid
polymorphisms: R30K/M/Q (n=4), L31M (n=2), or Y93H (n=3). All 9 subjects with
NS5A polymorphisms achieved SVR12, including 5 who were noncirrhotic and 4 who
were in the post-transplant period.
Genotype 3 NS5A Polymorphisms
In the ALLY-3 trial in which HCV genotype 3-infected
subjects received DAKLINZA and sofosbuvir for 12 weeks, the presence of an NS5A
Y93H polymorphism was associated with a reduced SVR12 rate (see Table 11). In a
pooled analysis of 175 subjects infected with HCV genotype 3 with available
baseline nucleotide sequence data in the ALLY-1, -2, and -3 trials, virus from
7% (13/175) of subjects had the NS5A Y93H polymorphism, and all 13 of these
subjects were in the ALLY-3 trial. Phylogenetic analysis of NS5A sequences
indicated that all genotype 3 subjects with available data in the ALLY-1, -2,
and -3 trials (n=175) were infected with HCV subtype 3a.
Table 11: Impact of NS5A Amino Acid Polymorphisms on
SVR12 Rates in Subjects with HCV Genotype 1a or Genotype 3 Infection in Phase 3
Trials of DAKLINZA + Sofosbuvir ± Ribavirin
NS5A Polymorphisms |
SVR12 Rates after 12 Weeks of Treatment with DAKLINZA + Sofosbuvir ± Ribavirina |
With NS5A Polymorphism(s) % (n/N) |
Without NS5A Polymorphism(s) % (n/N)b |
HCV genotype 1a-infected subjects: M28,c Q30,cL31,c or Y93c |
76% (13/17) |
95% (142/149) |
Without cirrhosisd |
100% (11/11) |
99% (100/101) |
With cirrhosis (Child-Pugh A, B, or C) |
33% (2/6) |
88% (42/48) |
HCV genotype 3-infected subjects: Y93H |
54% (7/13) |
92% (149/162) |
Without cirrhosisd |
67% (6/9) |
98% (125/128) |
With cirrhosis (Child-Pugh A, B, or C) |
25% (1/4) |
71% (24/34) |
a HCV genotype 1a-infected subjects received
DAKLINZA + sofosbuvir ± ribavirin for 12 weeks in the ALLY-1 and ALLY-2 trials.
HCV genotype 3-infected subjects received DAKLINZA + sofosbuvir for 12 weeks in
the ALLY-3 trial; no data on the impact of Y93H are available for HCV genotype
3-infected subjects treated with DAKLINZA + sofosbuvir ± ribavirin in ALLY-1
and ALLY-2 trials.
b None of the 11 subjects with Child-Pugh C cirrhosis had an
indicated NS5A polymorphism; 5 achieved SVR (genotype 1a: 4/9; genotype 3a:
½).
c Any change from genotype 1a reference.
d Includes subjects who were post-transplant with undefined
cirrhosis status. |
Cross-Resistance
Based on resistance patterns
observed in cell culture replicon studies and HCV-infected subjects,
cross-resistance between daclatasvir and other NS5A inhibitors is expected.
Cross-resistance between daclatasvir and other classes of direct-acting
antivirals is not expected. The impact of prior daclatasvir treatment
experience on the efficacy of other NS5A inhibitors has not been studied.
Conversely, the efficacy of DAKLINZA in combination with sofosbuvir has not
been studied in subjects who have previously failed treatment with regimens
that include an NS5A inhibitor.
Clinical Studies
Description Of Clinical Trials
The efficacy of DAKLINZA in combination with sofosbuvir
and with or without ribavirin was evaluated in three phase 3 clinical trials,
as summarized in Table 12 [see Clinical Studies]. HCV RNA levels were
measured during these clinical trials using the COBAS® TaqMan® HCV test
(version 2.0), for use with the High Pure System. The assay had a lower limit
of quantification (LLOQ) of 25 IU per mL. Sustained virologic response was the
primary endpoint and was defined as HCV RNA below the LLOQ at post-treatment
week 12 (SVR12).
Table 12: Genotype 1 and 3 Patient Populations from
DAKLINZA Trials
Trial |
Population |
Study Arms and Duration (Number of Subjects Treated) |
ALLY-3 (AI444218) |
Genotype 3, treatment-naive and treatment-experienced, with or without cirrhosis |
DAKLINZA and sofosbuvir for 12 weeks (N=152) |
ALLY-2 (AI444216) |
Genotype 1 and 3, treatment-naive and treatment-experienced, with or without cirrhosis, HCV/HIV-1 coinfection |
DAKLINZA and sofosbuvir for 12 weeks (N=137) |
ALLY-1 (AI444215) |
Genotype 1 and 3, treatment-naive or treatment-experienced, with or without cirrhosis, including decompensated cirrhosis and post-transplant |
DAKLINZA and sofosbuvir and ribavirin for 12 weeks (N=103) |
Clinical Trials In HCV Genotype 3 (ALLY-3)
ALLY-3 was an open-label trial
that included 152 subjects with chronic HCV genotype 3 infection and
compensated liver disease who were treatment naive (n=101) or treatment
experienced (n=51). Most treatment-experienced subjects had failed prior
treatment with peginterferon/ribavirin, but 7 subjects had been treated
previously with a sofosbuvir regimen and 2 subjects with a regimen containing
an investigational agent. Previous exposure to NS5A inhibitors was prohibited.
Subjects received DAKLINZA 60 mg plus sofosbuvir 400 mg once daily for 12 weeks
and were monitored for 24 weeks post treatment.
The 152 treated subjects in
ALLY-3 had a median age of 55 years (range, 24-73); 59% of the subjects were
male; 90% were white, 5% were Asian, and 4% were black. Most subjects (76%) had
baseline HCV RNA levels greater than or equal to 800,000 IU per mL; 21% of the
subjects had compensated cirrhosis, and 40% had the IL28B rs12979860 CC
genotype.
SVR12 and outcomes in subjects
without SVR12 in ALLY-3 are shown by patient population in Table 13. SVR12
rates were comparable regardless of HCV treatment history, age, gender, IL28B
allele status, or baseline HCV RNA level. For SVR outcomes related to baseline NS5A
amino acid polymorphisms, see Microbiology.
Table 13: ALLY-3: SVR12 in Treatment-Naive and
Treatment-Experienced Subjects with or without Cirrhosis with Genotype 3 HCV
Treated with DAKLINZA in Combination with Sofosbuvir for 12 Weeks
Treatment Outcomes |
Total n=152 |
SVR12 |
All |
89% (135/152) |
No cirrhosisa |
96% (115/120) |
With cirrhosis |
63% (20/32) |
Outcomes for subjects without SVR12 |
On-treatment virologic failureb |
0.7% (1/152) |
Relapsec |
11% (16/151) |
a Includes 11 subjects with missing or
inconclusive cirrhosis status.
b One subject had quantifiable HCV RNA at end of treatment.
c Relapse rates are calculated with a denominator of subjects with
HCV RNA not detected at the end of treatment. |
Clinical Trials In HCV/HIV Coinfected Subjects (ALLY-2)
ALLY-2 was an open-label trial
that included 153 subjects with chronic hepatitis C and HIV coinfection who
received DAKLINZA and sofosbuvir for 12 weeks. Subjects with HCV genotype 1, 2,
3, 4, 5, or 6 infection were eligible to enroll. Subjects were HCV
treatment-naive (n=101) or HCV treatment-experienced (n=52). Prior exposure to
NS5A inhibitors was prohibited. The dose of DAKLINZA was 60 mg once daily
(dose-adjusted for concomitant antiretroviral use) and the dose of sofosbuvir
was 400 mg once daily [see DRUG INTERACTIONS].
The 153 treated subjects had a
median age of 53 years (range, 24-71); 88% of subjects were male; 63% were
white, 33% were black, and 1% were Asian. Sixty-eight percent of subjects had
HCV genotype 1a, 15% had HCV genotype 1b, 8% had genotype 2, 7% had genotype 3,
and 2% had genotype 4. Most subjects (80%) had baseline HCV RNA levels greater
than or equal to 800,000 IU per mL; 16% of the subjects had compensated
cirrhosis, and 73% had IL28B rs12979860 non-CC genotype. Concomitant HIV
therapy included PI-based regimens (darunavir + ritonavir, atazanavir +
ritonavir, or lopinavir/ritonavir) for 46% of subjects, NNRTI-based regimens
(efavirenz, nevirapine, or rilpivirine) for 26%, integrase-based regimens
(raltegravir or dolutegravir) for 26%, and nucleoside-only regimens (abacavir +
emtricitabine + zidovudine) for 1%. Two patients were not receiving treatment
for HIV.
SVR and outcomes in subjects
with HCV genotype 1 without SVR12 in ALLY-2 are shown by patient population in
Table 14. Available data on subjects with HCV genotype 2, 4, 5, or 6 infection
are insufficient to provide recommendations for these genotypes; therefore,
these results are not presented in Table 14. SVR12 rates were comparable
regardless of antiretroviral therapy, HCV treatment history, age, race, gender,
IL28B allele status, HCV genotype 1 subtype, or baseline HCV RNA level. For SVR
outcomes related to baseline NS5A amino acid polymorphisms, see Microbiology.
No subjects switched their
antiretroviral therapy regimen due to loss of plasma HIV-1 RNA suppression.
There was no change in absolute CD4+ T-cell counts at the end of 12 weeks of
treatment.
Table 14: ALLY-2: SVR12 in
Subjects with Genotype 1 and 3 HCV/HIV Coinfection Treated with DAKLINZA in
Combination with Sofosbuvir for 12 Weeks
Treatment Outcomes |
Total
n=137 |
SVR12 |
Genotype 1 |
97% (123/127) |
No cirrhosisa |
98% (103/105) |
With cirrhosis |
91% (20/22) |
Genotype 3b |
100% (10/10) |
Outcomes for genotype 1 subjects without SVR12 |
On-treatment virologic failurec |
0.8% (1/127) |
Relapsed |
1.6% (2/126) |
Missing post-treatment data |
0.8% (1/126) |
a Includes 5 subjects with inconclusive
cirrhosis status.
b One subject with cirrhosis.
c One subject had detectable HCV RNA at end of treatment.
d Relapse rates are calculated with a denominator of subjects with
HCV RNA not detected at the end of treatment. |
Clinical Trials In Subjects With Child-Pugh A, B, Or C
Cirrhosis Or With HCV Recurrence After Liver Transplantation (ALLY-1)
ALLY-1 was an open-label trial
of DAKLINZA, sofosbuvir, and ribavirin that included 113 subjects with chronic
HCV infection and Child-Pugh A, B, or C cirrhosis (n=60) or HCV recurrence
after liver transplantation (n=53). Subjects with HCV genotype 1, 2, 3, 4, 5,
or 6 infection were eligible to enroll. Subjects could be HCV treatment-naive
or treatment-experienced, although prior exposure to NS5A inhibitors was
prohibited. Subjects received DAKLINZA 60 mg once daily, sofosbuvir 400 mg once
daily, and ribavirin for 12 weeks and were monitored for 24 weeks post
treatment. Subjects received an initial ribavirin dose of 600 mg or less daily
with food; the initial and on-treatment dosing of ribavirin was modified based
on hemoglobin and creatinine clearance measurements. If tolerated, the
ribavirin dose was titrated up to 1000 mg per day. A high proportion of
reductions in ribavirin dosing occurred in the trial. By week 6, approximately half
of the subjects received 400 mg per day or less of ribavirin. In total, 16
subjects (15%) completed less than 12 weeks and 11 subjects (10%) completed
less than 6 weeks of ribavirin therapy, respectively. For the cohort of
patients with cirrhosis (Child-Pugh A, B, or C), the median time to
discontinuation of ribavirin was 43 days (range, 8-82, n=9). For the
post-transplant cohort, the median time to discontinuation of ribavirin was 20
days (range, 3-57, n=7).
The 113 treated subjects in ALLY-1 had a median age of 59
years (range, 19-82); 67% of the subjects were male; 96% were white, 4% were
black, and 1% Asian. Most subjects (59%) were treatment-experienced, and most
(71%) had baseline HCV RNA levels greater than or equal to 800,000 IU per mL.
Fifty-eight percent of subjects had HCV genotype 1a, 19% had HCV genotype 1b,
4% had genotype 2, 15% had genotype 3, 4% had genotype 4, and 1% had genotype
6, 77% had IL28B rs12979860 non-CC genotype. Among the 60 subjects in the cirrhosis
cohort, 20% were Child-Pugh A, 53% were Child-Pugh B, and 27% were Child-Pugh
C, and 35% had a Baseline Model for End-Stage Liver Disease (MELD) score of 15
or greater. Most (55%) of the 53 subjects in the post-transplant cohort had F3
or F4 fibrosis (based on FibroSURE® results).
SVR12 and outcomes in subjects without SVR12 in ALLY-1
are shown for subjects with HCV genotype 1 by patient population in Table 15.
Available data on subjects with HCV genotype 2, 4, 5, or 6 infection are
insufficient to provide recommendations; therefore, these results are not
presented in Table 15.
SVR12 rates were comparable regardless of age, gender,
IL28B allele status, or baseline HCV RNA level. For SVR12 outcomes related to
baseline NS5A amino acid polymorphisms, see Microbiology. No HCV
genotype 1 or genotype 3 subjects with Child-Pugh C cirrhosis had baseline
resistance-associated NS5A amino acid polymorphisms. SVR12 rates were
comparable between genotype 3 (5/6 with Child-Pugh B or C cirrhosis and 10/11
post-liver transplant) and genotype 1 subjects with or without decompensated
cirrhosis.
Table 15: ALLY-1: SVR12 in
Genotype 1 Subjects with Child-Pugh A, B, or C Cirrhosis or with HCV Genotype 1
Recurrence after Liver Transplantation Treated with DAKLINZA in Combination
with Sofosbuvir and Ribavirin for 12 Weeks
Treatment Outcomes |
Child-Pugh A, B, or C Cirrhosis
n=45 |
Post-Liver Transplant
n=41 |
SVR12 |
Genotype 1 |
82% (37/45) |
95% (39/41) |
Child-Pugh A |
91% (10/11) |
- |
Child-Pugh B |
92% (22/24) |
- |
Child-Pugh C |
50% (5/10) |
- |
Genotype 1a |
76% (26/34) |
97% (30/31) |
Genotype 1b |
100% (11/11) |
90% (9/10) |
Outcomes for subjects without SVR12 |
On-treatment virologic failure |
2% (1/45)a |
0 |
Relapseb |
16% (7/44) |
5% (2/41) |
a One subject had detectable HCV RNA at end of
treatment.
b Relapse rates are calculated with a denominator of subjects with
HCV RNA not detected at end of treatment. |