Remdesivir Other names: GS-5734

Chemical formula: C₂₇H₃₅N₆O₈P  Molecular mass: 602.585 g/mol 

Mechanism of action

Remdesivir is an adenosine nucleotide prodrug that is metabolized within host cells to form the pharmacologically active nucleoside triphosphate metabolite. Remdesivir triphosphate acts as an analog of adenosine triphosphate (ATP) and competes with the natural ATP substrate for incorporation into nascent RNA chains by the SARS-CoV-2 RNA-dependent RNA polymerase, which results in delayed chain termination during replication of the viral RNA. As an additional mechanism, remdesivir triphosphate can also inhibit viral RNA synthesis following its incorporation into the template viral RNA as a result of read-through by the viral polymerase that may occur in the presence of higher nucleotide concentrations. When remdesivir nucleotide is present in the viral RNA template, the efficiency of incorporation of the complementary natural nucleotide is compromised, thereby inhibiting viral RNA synthesis.

Pharmacodynamic properties

Antiviral activity

Remdesivir exhibited in vitro activity against a clinical isolate of SARS-CoV-2 in primary human airway epithelial cells with a 50% effective concentration (EC50) of 9.9 nM after 48 hours of treatment. Remdesivir inhibited the replication of SARS-CoV-2 in the continuous human lung epithelial cell lines Calu-3 and A549-hACE2 with EC50 values of 280 nM after 72 hours of treatment and 115 nM after 48 hours of treatment, respectively. The EC50 values of remdesivir against SARS-CoV-2 in Vero cells were 137 nM at 24 hours and 750 nM at 48 hours post-treatment.

The antiviral activity of remdesivir was antagonised by chloroquine phosphate in a dose-dependent manner when the two drugs were co-incubated at clinically relevant concentrations in HEp-2 cells infected with respiratory syncytial virus (RSV). Higher remdesivir EC50 values were observed with increasing concentrations of chloroquine phosphate. Increasing concentrations of chloroquine phosphate reduced formation of remdesivir triphosphate in A549-hACE2, HEp-2 and normal human bronchial epithelial cells.

Based on in vitro testing, remdesivir retained similar antiviral activity (EC50 fold change values below the in vitro susceptibility change cutoff of 2.8-fold) against clinical isolates of SARS-CoV-2 variants compared to an earlier lineage SARS-CoV-2 (lineage A) isolate, including Alpha (B.1.1.7), Beta (B.1.351), Gamma (P.1), Delta (B.1.617.2), Epsilon (B.1.429), Zeta (P.2), Iota (B.1.526), Kappa (B.1.617.1), Lambda (C.37) and Omicron variants (including B.1.1.529/BA.1, BA.2, BA.2.12.1, BA.2.75, BA.4, BA.4.6, BA.5, BF.5, BF.7, BQ.1, BQ.1.1, CH.1.1, XBB and XBB.1.5). For these variants, the EC50 fold change values ranged between 0.2 to 2.3 compared to an earlier lineage SARSCoV-2 (lineage A) isolate.

Resistance in cell culture

SARS-CoV-2 isolates with reduced susceptibility to remdesivir have been selected in cell culture. In one selection with GS-441524, the parent nucleoside of remdesivir, virus pools emerged expressing combinations of amino acid substitutions at V166A, N198S, S759A, V792I, C799F, and C799R in the viral RNA-dependent RNA polymerase, conferring EC50 fold-changes of 2.7 up to 10.4. When individually introduced into a wild-type recombinant virus by site-directed mutagenesis, 1.7- to 3.5-fold reduced susceptibility to remdesivir was observed. In a second selection with remdesivir using a SARS-CoV-2 isolate containing the P323L substitution in the viral polymerase, a single amino acid substitution at V166L emerged. Recombinant viruses with substitutions at P323L alone or P323L+V166L in combination exhibited 1.3- and 1.5-fold changes in remdesivir susceptibility, respectively.

Cell culture resistance profiling of remdesivir using the rodent CoV murine hepatitis virus identified two substitutions (F476L and V553L) in the viral RNA-dependent RNA polymerase at residues conserved across CoVs that conferred 5.6-fold reduced susceptibility to remdesivir. Introduction of the corresponding substitutions (F480L and V557L) into SARS-CoV resulted in 6-fold reduced susceptibility to remdesivir in cell culture and attenuated SARS-CoV pathogenesis in a mouse model. When individually introduced into a SARS-CoV-2 recombinant virus, the corresponding substitutions at F480L and V557L each conferred 2-fold reduced susceptibility to remdesivir.

Pharmacokinetic properties

The pharmacokinetic properties of remdesivir have been investigated in healthy volunteers and patients with COVID-19.

Absorption

The pharmacokinetic properties of remdesivir and the predominant circulating metabolite GS-441524 have been evaluated in healthy adult subjects. Following intravenous administration of remdesivir adult dosage regimen, peak plasma concentration was observed at end of infusion, regardless of dose level, and declined rapidly thereafter with a half-life of approximately 1 hour. Peak plasma concentrations of GS-441524 were observed at 1.5 to 2.0 hours post start of a 30 minutes infusion.

Distribution

Remdesivir is approximately 93% bound to human plasma proteins (ex-vivo data) with free fraction ranging from 6.4% to 7.4%. The binding is independent of drug concentration over the range of 1 to 10 μM, with no evidence for saturation of remdesivir binding. After a single 150 mg dose of [14C]-remdesivir in healthy subjects, the blood to plasma ratio of [14C]-radioactivity was approximately 0.68 at 15 minutes from start of infusion, increased over time reaching ratio of 1.0 at 5 hours, indicating differential distribution of remdesivir and its metabolites to plasma or cellular components of blood.

Biotransformation

Remdesivir is extensively metabolized to the pharmacologically active nucleoside analog triphosphate GS-443902 (formed intracellularly). The metabolic activation pathway involves hydrolysis by esterases, which leads to the formation of the intermediate metabolite, GS-704277. In the liver, carboxylesterase 1 and cathepsin A are the esterases responsible for 80% and 10% of remdesivir metabolism, respectively. Phosphoramidate cleavage followed by phosphorylation forms the active triphosphate, GS-443902. Dephosphorylation of all phosphorylated metabolites can result in the formation of nucleoside metabolite GS-441524 that itself is not efficiently re-phosphorylated. Decyanation of remdesivir and/or its metabolites, followed by subsequent rhodanese mediated conversion generates thiocyanate anion. The levels of thiocyanate detected following administration of 100 mg and 200 mg remdesivir were observed to be significantly below endogenous levels in human plasma.

Elimination

Following a single 150 mg IV dose of [14C]-remdesivir, mean total recovery of the dose was 92%, consisting of approximately 74% and 18% recovered in urine and feces, respectively. The majority of the remdesivir dose recovered in urine was GS-441524 (49%), while 10% was recovered as remdesivir. These data indicate that renal clearance is the major elimination pathway for GS-441524. The median terminal half-lives of remdesivir and GS-441524 were approximately 1 and 27 hours, respectively.

Pharmacokinetics of remdesivir and metabolites in adults with COVID-19

Pharmacokinetic exposures for remdesivir and its metabolites in adults with COVID-19 are provided in Table 8.

Table 8. Multiple dose PK parametersa of remdesivir and metabolites (GS-441524 and GS-704277) following IV administration of remdesivir 100 mg to adults with COVID-19:

Parameters
Meanb (95% CI)
Remdesivir GS-441524 GS-704277
Cmax (ng/mL) 2700 (2440, 2990) 143 (135, 152) 198 (180, 218)
AUCtau (ng•h/mL) 1710 (1480, 1980) 2410 (2250, 2580) 392 (348, 442)
Ctau (ng/mL) ND 61.5 (56.5, 66.8) ND

CI = Confidence Interval; ND=Not detectable (at 24 hours post-dose)
a Population PK estimates for 30-minute IV infusion of remdesivir for 3 days (Study GS-US-540-9012, n=147).
b Geometric mean estimates

Other special populations

Gender, race and age

Based on gender, race and age, pharmacokinetic differences on the exposures of remdesivir were evaluated using population pharmacokinetic analysis. Gender and race did not affect the pharmacokinetics of remdesivir and its metabolites (GS-704277 and GS-441524). Pharmacokinetic exposures of the GS-441524 metabolite were modestly increased in hospitalised COVID-19 patients ≥60 years of age, however no dose adjustment is needed in these patients.

Pregnancy

In CO-US-540-5961 (IMPAACT 2032) study, mean exposures (AUCtau, Cmax, and Ctau) of remdesivir and its metabolites (GS-441524 and GS-704277) were comparable between pregnant and nonpregnant women of child-bearing potential.

Paediatric patients

Population pharmacokinetic models for remdesivir and its circulating metabolites (GS-704277 and GS-441524), developed using pooled data from studies in healthy subjects and in adult and paediatric patients with COVID-19, were used to predict pharmacokinetic exposures in 50 paediatric patients aged ≥28 days to <18 years and weighing ≥3 kg (Study GS-US-540-5823) (Table 9). Geometric mean exposures (AUCtau, Cmax and Ctau) for these patients at the doses administered were higher for remdesivir (44% to 147%), GS-441524 (-21% to 25%), and GS-704277 (7% to 91%) as compared to those in adult hospitalised patients with COVID-19. The increases were not considered clinically significant.

Table 9. Pharmacokinetic parametersa estimate of steady-state plasma remdesivir, GS-441524 and GS-704277 in paediatric and adult hospitalised COVID-19 patients:

Parameters
Meanb
Paediatric patients Adult
hospitalised
patients
(N=277)
Cohort 1 Cohort 8 Cohort 2 Cohort 3 Cohort 4
12 to <18
Years and
Weighing
≥40 kg
(N=12)
<12 Years
and
Weighing
≥40 kg
(N=5)
28 Days to
<18 Years
and
Weighing
20 to
<40 kg
(N=12)
28 Days to
<18 Years
and
Weighing
12 to
<20 kg
(N=11)
28 Days to
<18 Years
and
Weighing 3
to <12 kg
(N=10)
Remdesivir
Cmax (ng/mL) 3910 3920 568055304900 2650
AUCtau (h•ng/mL) 2470 22803500 3910 2930 1590
GS-441524
Cmax (ng/mL) 197 162 181 158 202 170
AUCtau (h•ng/mL) 3460 2640 2870 2400 2770 3060
Ctau (ng/mL) 98.376.273.8 69.478.478.4
GS-704277
Cmax (ng/mL) 307 278 423 444 390 233
AUCtau (h•ng/mL) 815537 754 734 691501

a PK parameters were simulated using PopPK modeling with 0.5 hour of duration for remdesivir infusions.
b Geometric mean estimates.
Paediatric hospitalised patients are from Study GS-US-540-5823; patients received 200 mg on Day 1 followed by remdesivir 100 mg once daily on subsequent days (Cohort 1 and 8), or 5 mg/kg on Day 1 followed by remdesivir 2.5 mg/kg once daily on subsequent days (Cohort 2-4) for a total treatment duration of up to 10 days.
Adult hospitalised patients are from Study CO-US-540-5844 (a phase 3 randomised study to evaluate the safety and antiviral activity of remdesivir in patients with severe COVID-19); patients received 200 mg on Day 1 followed by remdesivir 100 mg once daily on subsequent days (10 days total treatment duration).

Renal impairment

The pharmacokinetics of remdesivir and its metabolites (GS-441524 and GS-704277) and the excipient SBECD were evaluated in healthy subjects, those with mild (eGFR 60-89 mL/minute), moderate (eGFR 30-59 mL/minute), severe (eGFR 15-29 mL/minute) renal impairment, or with ESRD (eGFR <15 mL/minute) on haemodialysis or not on haemodialysis following a single dose of up to 100 mg of remdesivir (Table 10); and in a Phase 3 study in COVID-19 patients with severely reduced kidney function (eGFR <30 mL/minute) receiving remdesivir 200 mg on Day 1 followed by 100 mg from Day 2 to Day 5 (Table 11).

Pharmacokinetic exposures of remdesivir were not affected by renal function or timing of remdesivir administration around dialysis. Exposures of GS-704277, GS-441524, and SBECD were up to 2.8-fold, 7.9-fold and 26-fold higher, respectively, in those with renal impairment than those with normal renal function which is not considered clinically significant based on limited available safety data. No dose adjustment of remdesivir is required for patients with renal impairment, including those on dialysis.

Table 10. Statistical comparison of single-dose pharmacokinetic parametersa of remdesivir and metabolites (GS-441524 and GS-704277) between adult subjects with decreased renal functionb (mild, moderate, severe renal impairment and ESRD) and adult subjectsa with normal renal function:

GLSM Ratioc
(90% CI)
60-89 mL
per
minute
N=10
30-59 mL
per
minute
N=10
15-29 mL
per
minute
N=10
<15 mL per minute
Pre-haemodialysis
N=6
Posthaemodialysis
N=6
No dialysis
N=3
Remdesivir
Cmax
(ng/mL)
96.0
(70.5, 131)
120
(101, 142)
97.1
(83.3, 113)
89.1
(67.1, 118)
113
(79.4, 160)
93.9
(65.4, 135)
AUCinf
(h•ng/mL)
99.5
(75.3, 132)
122
(97.5, 152)
94
(83.0, 107)
79.6
(59.0, 108)
108
(71.5, 163)
88.9
(55.2, 143)
GS-441524
Cmax
(ng/mL)
107
(90, 126)
144
(113, 185)
168
(128, 220)
227
(172, 299)
307
(221, 426)
300
(263, 342)
AUCinfd
(h•ng/mL)
119
(97, 147)
202
(157, 262)
326
(239, 446)
497
(365, 677)
622
(444, 871)
787
(649, 953)
GS-704277
Cmax
(ng/mL)
225
(120, 420)
183
(134, 249)
127
(96.1, 168)
143
(100, 205)
123
(83.6, 180)
176
(119, 261)
AUCinf (h•ng/mL)139
(113, 171)
201
(148, 273)
178
(127, 249)
218
(161, 295)
206
(142, 297)
281
(179, 443)

CI = Confidence Interval; GLSM = geometric least-squares mean
a Exposures were estimated using noncompartmental analysis from a dedicated Phase 1 renal impairment study GS-US540-9015; single doses up to 100 mg were administered; each subject with renal impairment had a matched adult subject enrolled with normal renal function (eGFR ≥90 mL/min/1.73m²), same sex, and similar body mass index (BMI (± 20%)) and age (± 10 years)
Subjects with reduced renal function and matched adult subjects with normal renal function received the same remdesivir dose
b eGFR was calculated using Modification of Diet in Renal Disease equation and reported in mL/min/1.73 m²
c Ratio calculated for the comparison of PK parameters of test (subjects with reduced renal function) to reference (subjects with normal renal function)
d AUC0-72h for subjects on haemodialysis

Table 11. Pharmacokinetic parametersa of remdesivir and metabolites (GS-441524 and GS704277) following IV administration of remdesivir (200 mg on day 1 followed by 100 mg daily on days 2-5) to adults with COVID-19 and severely reduced kidney function (eGFR <30 mL/min/1.73 m²):

Parameter
Meanb (percentile, 5th, 95th)
Remdesivir GS-441524 GS-704277
Cmax
(ng/mL)
3850
(1530, 8720)
703
(343, 1250)
378
(127, 959)
AUCtau
(h•ng/mL)
2950
(1390, 8370)
15400
(7220, 27900)
1540
(767, 3880)

a Population PK estimates for 30-minute IV infusion of remdesivir for 5 days (Study GS-US-540-5912, n=90).
b Geometric mean estimates.

Hepatic impairment

The pharmacokinetics of remdesivir and its metabolites (GS-441524 and GS-704277) were evaluated in healthy subjects and those with moderate or severe hepatic impairment (Child-Pugh Class B or C) following a single dose of 100 mg of remdesivir. Relative to subjects with normal hepatic function, mean exposures (AUCinf, Cmax) of remdesivir and GS-704277 were comparable in moderate hepatic impairment and up to 2.4 fold higher in severe hepatic impairment; however, the increase was not considered clinically significant.

Hospitalisation

Pharmacokinetic exposures for remdesivir in hospitalised patients with severe COVID-19 pneumonia were generally within the range of the exposures in non-hospitalised patients. The GS-704277 and GS-441524 metabolite levels were modestly increased.

Interactions

Remdesivir inhibited CYP3A4 in vitro. At physiologically relevant concentrations (steady-state), remdesivir or its metabolites GS-441524 and GS-704277 did not inhibit CYP1A2, 2B6, 2C8, 2C9, 2C19, and 2D6 in vitro. Remdesivir is not a time-dependent inhibitor of CYP450 enzymes in vitro.

Remdesivir induced CYP1A2 and potentially CYP3A4, but not CYP2B6 in vitro.

In vitro data indicates no clinically relevant inhibition of UGT1A3, 1A4, 1A6, 1A9 or 2B7 by remdesivir or its metabolites GS-441524 and GS-704277. Remdesivir, but not its metabolites, inhibited UGT1A1 in vitro.

For GS-441524 and GS-704277, the only enzyme for which metabolism could be detected was UGT1A3.

Remdesivir inhibited OAT3, MATE1, OCT1, OATP1B1 and OATP1B3 in vitro. At physiologically relevant concentrations, remdesivir and its metabolites did not inhibit P-gp and BCRP in vitro.

Preclinical safety data

Toxicology

Following intravenous administration (slow bolus) of remdesivir to rhesus monkeys and rats, severe renal toxicity occurred after short treatment durations. In male rhesus monkeys at dosage levels of 5, 10, and 20 mg/kg/day for 7 days resulted, at all dose levels, in increased mean urea nitrogen and increased mean creatinine, renal tubular atrophy, and basophilia and casts, and an unscheduled death of one animal at the 20 mg/kg/day dose level. In rats, dosage levels of >3 mg/kg/day for up to 4 weeks resulted in findings indicative of kidney injury and/or dysfunction. Systemic exposures (AUC) of the predominant circulating metabolite of remdesivir (GS-441524) were 0.1 times (monkeys at 5 mg/kg/day) and 0.3 times (rats at 3 mg/kg/day) the exposure in humans following intravenous administration at the recommended human dose (RHD).

Carcinogenesis

Long-term animal studies to evaluate the carcinogenic potential of remdesivir have not been performed.

Mutagenesis

Remdesivir was not genotoxic in a battery of assays, including bacterial mutagenicity, chromosome aberration using human peripheral blood lymphocytes, and in vivo rat micronucleus assays.

Reproductive toxicity

In female rats, decreases in corpora lutea, numbers of implantation sites, and viable embryos, were seen when remdesivir was administered intravenously daily at a systemically toxic dose (10 mg/kg/day) 14 days prior to mating and during conception; exposures of the predominant circulating metabolite (GS-441524) were 1.3 times the exposure in humans at the RHD. There were no effects on female reproductive performance (mating, fertility, and conception) at this dose level.

In rats and rabbits, remdesivir demonstrated no adverse effect on embryofoetal development when administered to pregnant animals at systemic exposures (AUC) of the predominant circulating metabolite of remdesivir (GS-441524) that were up to 4 times the exposure in humans at the RHD.

In rats, there were no adverse effects on pre- and post-natal development at systemic exposures (AUC) of the predominant circulating metabolite of remdesivir (GS-441524) that were similar to the exposure in humans at the RHD.

Related medicines

© All content on this website, including data entry, data processing, decision support tools, "RxReasoner" logo and graphics, is the intellectual property of RxReasoner and is protected by copyright laws. Unauthorized reproduction or distribution of any part of this content without explicit written permission from RxReasoner is strictly prohibited. Any third-party content used on this site is acknowledged and utilized under fair use principles.