OBJECTIVES

Remdesivir decreases the risk of SARS-CoV-2 infection progressing to severe disease in adults. This study evaluated remdesivir safety and pharmacokinetics in infants and children.

METHODS

This was a phase 2/3, open-label trial in children aged 28 days to 17 years hospitalized for polymerase chain reaction–confirmed SARS-CoV-2 infection. Participants received for ≤10 days once-daily intravenous remdesivir doses defined using physiologically based pharmacokinetic modeling (for ≥40 kg, 200 mg day 1, then 100 mg/day; for age ≥28 days and ≥3 to <40 kg, 5 mg/kg day 1, then 2.5 mg/kg/day). Sparse pharmacokinetic samples were analyzed using population–pharmacokinetic approaches for remdesivir and metabolites GS-704277 and GS-441524.

RESULTS

Among 53 participants, at enrollment the median (Q1, Q3) number of days of COVID-19 symptoms was 5 (3, 7) and hospitalization was 1 (1, 3). Underlying conditions included obesity in 19 (37%), asthma in 11 (21%), and cardiac disorders in 11 (21%). Median duration of remdesivir treatment was 5 days (range, 1–10). Remdesivir treatment had no new apparent safety trends. Two participants discontinued treatment because of adverse events including elevated transaminases; both had elevated transaminases at baseline. Three deaths occurred during treatment (and 1 after). When compared with phase 3 adult data, estimated mean pediatric parameters (area under the concentration-time curve over 1 dosing interval, AUCτ, Cmax, and Cτ) were largely overlapping but modestly increased (remdesivir, 33%–129%; GS-704277, 37%–124%; GS-441524, 0%–60%). Recovery occurred for 62% of participants on day 10 and 83% at last assessment.

CONCLUSIONS

In infants and children with COVID-19, the doses of remdesivir evaluated provided drug exposure similar to adult dosing. In this study with a small sample size, no new safety concerns were observed.

What’s Known on This Subject:

Remdesivir shortens time to recovery in adults with coronavirus disease. In infants and children <12 years, limited data have been available on remdesivir safety and efficacy.

What This Study Adds:

Our results support remdesivir as a treatment option starting from age 28 days in infants and children with COVID-19.

Children and adolescents account for a substantial burden of SARS-CoV-2 infections, with those aged <20 years accounting for ∼21% of global COVID-19 cases.1 Although children, including infants, generally have a mild course of COVID-19, a small proportion develop severe disease requiring ICU admission and prolonged ventilation.2 The hospitalization rate among children with COVID-19 is 0.1% to 1.5%, with mortality up to 0.02%.3 Approximately two thirds of children (68%) hospitalized for COVID-19 have 1 or more underlying medical conditions, with obesity being most commonly reported (32%), followed by asthma or reactive airway disease (16%).4 

Remdesivir (RDV) is a nucleotide analog prodrug that selectively inhibits the RNA-dependent RNA polymerase of several viruses, including SARS-CoV-2.5,6 It is the first antiviral to be approved for treatment of children with COVID-19 in the United States.7,8 In adults, RDV decreases the risk of coronavirus infection progressing to severe disease and shortens time to recovery.9,13 The only data available in children are derived from a compassionate-use program, in which 77 children aged 2 months to 17 years receiving RDV for severe COVID-19 had a high rate of clinical recovery.14 Clinical Administration of Remdesivir After COVID-19 Diagnosis in Children (CARAVAN) is a phase 2/3, open-label study that evaluated the safety, pharmacokinetics, and clinical and virologic effects of RDV in infants and children ages 28 days to 17 years and weighing ≥3 kg.

Children and adolescents ≥28 days to 17 years who had SARS-CoV-2 infection confirmed by polymerase chain reaction (PCR) and were hospitalized and required medical care for COVID-19 were eligible. Cohort 1 participants were aged ≥12 to 17 years and weighed ≥40 kg (Fig 1). Cohort 2 through 4 participants were aged ≥28 days to 17 years and weighed ≥3 kg to <40 kg. Cohorts 5 through 7 (term and preterm neonates) are not included in this manuscript. Cohort 8 participants were aged <12 years and weighed ≥40 kg. Exclusion criteria included concurrent treatment with agents with known or possible direct antiviral activity against SARS-CoV-2 within 24 hours before study drug dosing; alanine aminotransferase (ALT) or aspartate aminotransferase (AST) >5X the upper limit of normal; estimated glomerular filtration rate <30 mL/min/1.73 m2 using the Schwartz formula for participants aged ≥1 year; and creatinine values exceeding the 97.5th percentile15 or upper limit for age16 in those <1 year.

FIGURE 1

Patient disposition throughout the study. Per protocol, treatment duration was up to 10 days and could be shortened for those who demonstrated clinical improvement. Of the 13 participants who received treatment of less than 10 days according to investigator discretion, 12/13 were due to clinical improvement.

FIGURE 1

Patient disposition throughout the study. Per protocol, treatment duration was up to 10 days and could be shortened for those who demonstrated clinical improvement. Of the 13 participants who received treatment of less than 10 days according to investigator discretion, 12/13 were due to clinical improvement.

Close modal

This was a phase 2/3, multicenter, open-label study. A total of 6 cohorts were enrolled in parallel between July 2020 and May 2021. RDV was dosed intravenously once daily for up to 10 days. Discontinuation was allowed before 10 days in cases of clinical improvement as judged by the investigator or clinical team. The weight-based dosing regimen tested in this study was derived using physiologically based pharmacokinetic (PK) modeling.17 For participants aged ≥12 years and weighing ≥40 kg, RDV 200 mg on day 1 was followed by 100 mg daily. For those aged ≥28 days and weighing ≥3 kg to <40 kg, RDV 5 mg/kg on day 1 was followed by 2.5 mg/kg daily.

The study protocol (https://clinicaltrials.gov/ProvidedDocs/81/NCT03022981/Prot_000.pdf) was approved by the institutional review board or ethics committee of each institution or by a centralized institutional review board, as applicable. Parents or guardians provided informed consent, and older children and youth provided assent when able, per local regulations. The study was conducted in accordance with the International Conference on Harmonization Good Clinical Practice Guidelines and the Declaration of Helsinki.

Twelve subjects for each cohort (cohorts 1–4) were to be enrolled in this study. Pharmacokinetic data from these participants were to provide >99% power for each cohort to conclude exposure equivalence of RDV AUCτ in adolescents and children versus 25 healthy adult subjects in the study GS-US-399-5505 using two 1-sided tests with each performed at an α level of 0.05. In this power analysis, it was assumed that the expected geometric mean ratio of AUCτ between the adolescents and children group versus the adult group is equal to 1, the intersubject SD (natural log scale) of AUCτ is 0.18, and the equivalency boundary is 70% to 143%.

Twelve subjects from each cohort (cohorts 1–4) were also to provide >99% power to conclude exposure equivalence of RDV Cmax in adolescents and children, compared with 26 healthy adult subjects in study GS-US-399-5505, assuming the expected geometric mean ratio of Cmax between the adolescents and children group and the adult group is equal to 1, the intersubject SD (natural log scale) of Cmax is 0.19, and the equivalency boundary is 70% to 143%.

The primary objectives of this study were to evaluate the safety and PK of RDV among participants aged 28 days to 17 years with laboratory-confirmed COVID-19. The primary outcomes were the proportions of participants with treatment-emergent adverse events or laboratory abnormalities, as well as the plasma pharmacokinetics of RDV and its metabolites.

Secondary objectives were to determine clinical improvement among participants and the antiviral activity of RDV. Endpoints were change from baseline in SARS-CoV-2 viral load as well as clinical improvement as based on the 7-point ordinal scale and the Pediatric Early Warning Score (PEWS) Scale (Supplemental Appendix). When possible, plasma concentrations of sulfobutylether β-cyclodextrin (SBECD) sodium were evaluated.

Safety

Safety assessments conducted at screening and every day of treatment administration included monitoring of adverse events and concomitant medications, vital signs measurements, respiratory status documentation, weight measurement, and physical examinations. The Medical Dictionary for Regulatory Activities, version 24.0, was used to code treatment-emergent adverse events. Severity grades were defined by the Division of AIDS Table for Grading the Severity of Adult and Pediatric Adverse Events, version 2.1, July 2017. Electrocardiograms were done during screening. Clinical laboratory analyses were done at screening and days 2, 5, 8, and 10. Documentation of respiratory status was measured using either respiratory rate (if not on a ventilator), oxygen supplementation, or oxygenation.

Pharmacokinetics

Plasma samples for measuring concentrations of RDV, RDV metabolites (GS-704277 and GS-441524), and SBECD sodium (where possible) were obtained on days 2 and 3, with 5 being an optional collection day. A newly developed liquid chromatography tandem mass spectrometry method was used to analyze SBECD sodium plasma concentrations (lower limit of quantification [LLOQ] = 10 µg/mL and %Bias = 0.4–1.7 µg/mL),18 and previously described methods were used to analyze RDV and metabolites concentrations.19 Cohort 1 and 8 participants receiving RDV 100 mg had an estimated SBECD dose of ∼3000 mg. Cohorts 2 through 4 receiving RDV 2.5 mg/kg had an estimated SBECD dose of ∼75 mg/kg. SBECD exposures in this study were compared with 36 adults without COVID-19 with normal renal function (n = 16) or mild (n = 10) or moderate (n = 10) renal impairment.

Sparse PK samples were analyzed using population-PK (Pop-PK) models for RDV and its metabolites GS-704277 and GS-44152417 (data on file). PK data used to develop the Pop-PK models for RDV and its metabolites were obtained from the following: (1) sparse sampling PK data from the majority of pediatric participants enrolled in CARAVAN (n = 51); (2) intensive sampling PK data from 3 phase 1 healthy volunteer studies: GS-US-399-1812 (n = 78), GS-US-399-1954 (n = 16), and GS-US-399-5505 (n = 29); and (3) PK data from 2 phase 3 studies in adult COVID-19: sparse/intensive sampling from GS-US-540-9012, a randomized, double-blind, placebo-controlled study that evaluated RDV in adults with COVID-19 in the outpatient setting (n = 148), and sparse sampling from CO-US-540-5844, a randomized, double-blind study that compared the combination of RDV and tocilizumab to RDV alone in hospitalized adults with severe COVID-19 pneumonia (n = 289). Pediatric exposures were compared with adult phase 3 exposures, and geometric mean ratios and 90% confidence intervals were estimated. Correlation of pharmacokinetic exposures with adverse events (7 most common adverse events: constipation, pyrexia, ALT increased, hyperglycemia, hypertension, hypomagnesemia, and acute kidney injury) was assessed. The exposure-response relationship was explored for recovery by day 10 as well as last available assessment based on the 7-point ordinal scale; the PEWS scale (behavior, cardiovascular, respiratory domains) on day 10; and time to discharge alive from hospital. The PK-pharmacodynamic analysis set included all participants aged 28 days to 17 years with COVID-19 who received at least 1 dose of remdesivir and had evaluable Pop-PK–based PK parameter estimates (n = 50) and the pharmacodynamic measurement available.

Serology and Virology

For participants weighing ≥12 kg, SARS-CoV-2 serology (IgG, IgM, and IgA) was completed at screening, day 5, and day 10. Samples were collected for SARS-CoV-2 reverse transcriptase quantitative PCR (RT-qPCR) testing and viral resistance testing on days 1, 3, 5, 7, and 10 as applicable (collection described further in the Supplemental Appendix). Viral load was analyzed via the central laboratory, except for fecal and rectal swab data, which are not included in this manuscript because assays are being developed. The serology for SARS-CoV-2 and SARS-CoV-2 samples/swabs were collected on the day of discharge if before day 10. Thereafter, these samples could be collected by study staff at the participant’s home or as an outpatient on designated days.

Resistance testing (details in the Supplemental Appendix) was planned for all participants enrolled in the study who received at least 1 dose of remdesivir. Baseline and postbaseline (during and/or after treatment) samples with SARS-CoV-2 viral load above the LLOQ were sequenced. The whole genome of SARS-CoV-2 was amplified using the ARTIC version 3 primer set at 63 °C annealing temperature, and the nucleotide sequence was determined by next-generation sequencing at DDL Diagnostic Laboratory (Rijswijk, The Netherlands). Amino acid variants and indels were reported as a change from reference when present ≥15%. Sequencing analyses focused on amino acid substitutions in the SARS-CoV-2 nsp12 RdRp (target of RDV) or other parts of the replication-transcription complex (Nsp8, Nsp10, Nsp13, and Nsp14).

Phenotyping of emergent substitutions in Nsp12 was conducted using site-directed mutants (SDMs) of SARS-CoV-2 in a replicon system adapted from Xie et al 202020 and Zhang et al 202121 (Supplemental Appendix). Briefly, the 4 plasmids that encode SARS-CoV-2 genes for the nonstructural proteins and the nucleoprotein, with or without SDMs, were prepared. The 4 DNA fragments were isolated from SARS-CoV-2 SH01 (SARS-CoV-2/human/CHN/SH01/2020, GenBank MT121215). Huh7-1CN cells were mixed with RNA and subjected to immediate electroporation followed by luciferase assay. EC50 values were calculated using a nonlinear 4-parameter variable slope regression model as the concentration at which there was a 50% decrease in the luciferase reporter signal relative to dimethylsulfoxide vehicle alone (0% virus inhibition) and uninfected control culture (100% virus inhibition). The fold-change values were calculated by dividing the variant mean EC50 by the SH01 reference strain mean EC50.

Clinical Activity

Clinical status on the 7-point ordinal scale (1 = death and 7 = not hospitalized; Supplemental Appendix) was evaluated at baseline and daily for days 1 through 10 or until hospital discharge. Recovery was defined as an improvement from a baseline score of 2 through 5 to a score of 6 or 7 or an improvement from a baseline score of 6 to a score of 7. To measure clinical improvement, PEWS22,23 also was recorded at baseline and daily for days 1 through 10 or until hospital discharge (Supplemental Appendix).

From July 2020 to May 2021, 53 participants were enrolled and treated at 19 sites in Italy, Spain, the United States, and the United Kingdom. The median (Q1, Q3) age of participants was 7 (2, 12) years, with a range of 0.1 to 17 years (Table 1). Overall, 98% (52/53) of participants had a preexisting medical condition, with the most common being obesity (19/53, 37%), asthma (11/53, 21%), and cardiac disorders (11/53, 21%). Most participants (51 participants, 96%) received concomitant medications other than those used for treating COVID-19. Thirty-four participants (64%) received dexamethasone during the study.

TABLE 1

Participant Demographic Data

Cohort 1 12–17 y and ≥40 kg (n = 12)Cohort 2 28 d–17 y and 20–<40 kg (n = 12)Cohort 3 28–17 y and 12–<20 kg (n = 12)Cohort 4 28–17 y and 3–<12 kg (n = 12)Cohort 8 <12 y and ≥40 kg (n = 5)Total (N = 53)
Age, median (Q1, Q3), y 15 (14, 17) 9 (8, 12) 3.5 (2, 6) 0.5 (0.2, 0.7) 11 (11, 11) 7 (2, 12) 
Sex at birth, n (%)       
 Male 4 (33) 5 (42) 7 (58) 5 (42) 2 (40) 23 (43) 
 Female 8 (67) 7 (58) 5 (42) 7 (58) 3 (60) 30 (57) 
Race, n (%)a       
 Black 5 (42) 2 (18) 4 (40) 2 (20) 1 (25) 14 (30) 
 White 7 (58) 9 (82) 6 (60) 8 (80) 3 (75) 33 (70) 
 Other 
Hispanic or Latino ethnicity, n (%)a 3 (27) 7 (58) 7 (58) 3 (25) 3 (60) 23 (44) 
Weight, median (Q1, Q3), kg 84 (57, 107) 27 (25, 31) 15 (13, 18) 5 (4, 9) 73 (55, 80) 25 (13, 55) 
BMI, median (Q1, Q3), kg/m2 34 (22, 47) 18 (15, 20) 16 (16, 18) 16 (15, 20) 28 (27, 36) 19 (16, 25) 
Cohort 1 12–17 y and ≥40 kg (n = 12)Cohort 2 28 d–17 y and 20–<40 kg (n = 12)Cohort 3 28–17 y and 12–<20 kg (n = 12)Cohort 4 28–17 y and 3–<12 kg (n = 12)Cohort 8 <12 y and ≥40 kg (n = 5)Total (N = 53)
Age, median (Q1, Q3), y 15 (14, 17) 9 (8, 12) 3.5 (2, 6) 0.5 (0.2, 0.7) 11 (11, 11) 7 (2, 12) 
Sex at birth, n (%)       
 Male 4 (33) 5 (42) 7 (58) 5 (42) 2 (40) 23 (43) 
 Female 8 (67) 7 (58) 5 (42) 7 (58) 3 (60) 30 (57) 
Race, n (%)a       
 Black 5 (42) 2 (18) 4 (40) 2 (20) 1 (25) 14 (30) 
 White 7 (58) 9 (82) 6 (60) 8 (80) 3 (75) 33 (70) 
 Other 
Hispanic or Latino ethnicity, n (%)a 3 (27) 7 (58) 7 (58) 3 (25) 3 (60) 23 (44) 
Weight, median (Q1, Q3), kg 84 (57, 107) 27 (25, 31) 15 (13, 18) 5 (4, 9) 73 (55, 80) 25 (13, 55) 
BMI, median (Q1, Q3), kg/m2 34 (22, 47) 18 (15, 20) 16 (16, 18) 16 (15, 20) 28 (27, 36) 19 (16, 25) 

BMI, body mass index (kg/m2) = weight (kg)/height (cm2) × 10 000; Q1, first quartile; Q3, third quartile.

a

For race and ethnicity, “not permitted,” “missing,” and “other” were excluded from the percentage calculation. Not permitted meant local regulators did not allow collection of race/ethnicity information.

At baseline/enrollment, participants had a median (Q1, Q3) of 5 days (3, 7) of COVID-19 symptoms and 1 day (1, 3) on line of hospitalization for COVID-19 (Table 2). The most common COVID-19–related disease manifestations were respiratory (44 participants, 83%), followed by gastrointestinal (27 participants, 51%); 12 participants (23%) were receiving invasive mechanical ventilation. Of 21 participants with an abnormal baseline electrocardiogram, all but 3 had results that were considered not clinically significant.

TABLE 2

Participant Clinical Characteristics at Baseline

Cohort 1 12–17 y and ≥40 kg (n = 12)Cohort 2 28 d–17 y and 20–<40 kg (n = 12)Cohort 3 28 d–17 y and 12–<20 kg (n = 12)Cohort 4 28 d–17 y and 3–<12 kg (n = 12)Cohort 8 <12 y and ≥40 kg (n = 5)Total (N = 53)
Days before first RDV dose, median (Q1, Q3) 
 Symptoms 7 (3, 11) 5 (3, 7) 3 (3, 7) 5 (2, 8) 5 (5, 7) 5 (3, 7) 
 Hospitalization 1 (0, 3) 1 (1, 2) 2 (1, 3) 2 (1, 7) 1 (0, 1) 1 (1, 3) 
ALT, median (Q1, Q3), U/L 26 (19, 48) 19 (14, 37) 25 (18, 32) 19 (13, 31) 20 (20, 32) 23 (15, 33) 
AST, median (Q1, Q3), U/L 65 (32, 82) 32 (26, 54) 37 (29, 68) 40 (35, 60) 36 (26, 38) 38 (29, 66) 
 Oxygen support status, n (%)       
 Invasive mechanical ventilation 1 (8) 3 (25) 3 (25) 5 (42) 12 (23) 
 High-flow oxygen 6 (50) 4 (33) 3 (25) 3 (25) 2 (40) 18 (34) 
 Low-flow oxygen 2 (17) 3 (25) 3 (25) 2 (40) 10 (19) 
 Room air 3 (25) 2 (17) 6 (50) 1 (8) 1 (20) 13 (25) 
COVID-19–related disease manifestations, n (%)a 
 Circulatory 2 (17) 3 (25) 2 (17) 4 (33) 11 (21) 
 Gastrointestinal 6 (50) 6 (50) 7 (58) 4 (33) 4 (80) 27 (51) 
 Neurologic 5 (42) 3 (25) 3 (25) 1 (8) 12 (23) 
 Respiratory 9 (75) 12 (100) 6 (50) 12 (100) 5 (100) 44 (83) 
 Systemic inflammatory response (MIS-C) 3 (25) 4 (33) 5 (42) 2 (17) 14 (26) 
SARS-CoV-2 RNA viral load, median (Q1, Q3), log10 copies 
 From nasal swabs, n 18 
  Median (Q1, Q3) 5.7 (4.6, 6.4) 3.1 (2.7, 7.7) 2.7 (2.7, 3.7) 5.1 (3.4, 7.7) 6.2 (6.2, 6.2) 4.6 (2.7, 6.4) 
 From nasopharyngeal swabs, n 24 
  Median (Q1, Q3) 5.1 (5.0, 6.4) 5.6 (2.7, 6.8) 6.6 (4.0, 6.8) 6.4 (3.9, 7.2) 6.9 (5.2, 7.0) 6.4 (3.9, 6.9) 
 From endotracheal tube aspirates, n 
  Median (Q1, Q3) 5.4 (5.4, 5.4) 2.7 (2.7, 9.9) 6.1 (5.5, 6.7) 7.4 (4.9, 7.7) — 5.5 (4.9, 7.4) 
ECG interpretation, n (%) 
 Abnormal 4 (40) 7 (64) 5 (46) 5 (46) 21 (45) 
  Clinically significant 1 (9) 2 (18) 3 (6) 
 Creatinine, n 11 12 11 12 51 
  Median (Q1, Q3), mg/dL 0.7 (0.5, 0.9) 0.4 (0.3, 0.6) 0.2 (0.2, 0.4) 0.3 (0.2, 0.3) 0.5 (0.4, 0.6) 0.4 (0.3, 0.6) 
 eGFR, nb 11 12 36 
  Median (Q1, Q3), mL/min/1.73 m2 92 (79, 118) 122 (89, 182) 148 (120, 174) — 131 (98, 135) 119 (87, 148) 
Cohort 1 12–17 y and ≥40 kg (n = 12)Cohort 2 28 d–17 y and 20–<40 kg (n = 12)Cohort 3 28 d–17 y and 12–<20 kg (n = 12)Cohort 4 28 d–17 y and 3–<12 kg (n = 12)Cohort 8 <12 y and ≥40 kg (n = 5)Total (N = 53)
Days before first RDV dose, median (Q1, Q3) 
 Symptoms 7 (3, 11) 5 (3, 7) 3 (3, 7) 5 (2, 8) 5 (5, 7) 5 (3, 7) 
 Hospitalization 1 (0, 3) 1 (1, 2) 2 (1, 3) 2 (1, 7) 1 (0, 1) 1 (1, 3) 
ALT, median (Q1, Q3), U/L 26 (19, 48) 19 (14, 37) 25 (18, 32) 19 (13, 31) 20 (20, 32) 23 (15, 33) 
AST, median (Q1, Q3), U/L 65 (32, 82) 32 (26, 54) 37 (29, 68) 40 (35, 60) 36 (26, 38) 38 (29, 66) 
 Oxygen support status, n (%)       
 Invasive mechanical ventilation 1 (8) 3 (25) 3 (25) 5 (42) 12 (23) 
 High-flow oxygen 6 (50) 4 (33) 3 (25) 3 (25) 2 (40) 18 (34) 
 Low-flow oxygen 2 (17) 3 (25) 3 (25) 2 (40) 10 (19) 
 Room air 3 (25) 2 (17) 6 (50) 1 (8) 1 (20) 13 (25) 
COVID-19–related disease manifestations, n (%)a 
 Circulatory 2 (17) 3 (25) 2 (17) 4 (33) 11 (21) 
 Gastrointestinal 6 (50) 6 (50) 7 (58) 4 (33) 4 (80) 27 (51) 
 Neurologic 5 (42) 3 (25) 3 (25) 1 (8) 12 (23) 
 Respiratory 9 (75) 12 (100) 6 (50) 12 (100) 5 (100) 44 (83) 
 Systemic inflammatory response (MIS-C) 3 (25) 4 (33) 5 (42) 2 (17) 14 (26) 
SARS-CoV-2 RNA viral load, median (Q1, Q3), log10 copies 
 From nasal swabs, n 18 
  Median (Q1, Q3) 5.7 (4.6, 6.4) 3.1 (2.7, 7.7) 2.7 (2.7, 3.7) 5.1 (3.4, 7.7) 6.2 (6.2, 6.2) 4.6 (2.7, 6.4) 
 From nasopharyngeal swabs, n 24 
  Median (Q1, Q3) 5.1 (5.0, 6.4) 5.6 (2.7, 6.8) 6.6 (4.0, 6.8) 6.4 (3.9, 7.2) 6.9 (5.2, 7.0) 6.4 (3.9, 6.9) 
 From endotracheal tube aspirates, n 
  Median (Q1, Q3) 5.4 (5.4, 5.4) 2.7 (2.7, 9.9) 6.1 (5.5, 6.7) 7.4 (4.9, 7.7) — 5.5 (4.9, 7.4) 
ECG interpretation, n (%) 
 Abnormal 4 (40) 7 (64) 5 (46) 5 (46) 21 (45) 
  Clinically significant 1 (9) 2 (18) 3 (6) 
 Creatinine, n 11 12 11 12 51 
  Median (Q1, Q3), mg/dL 0.7 (0.5, 0.9) 0.4 (0.3, 0.6) 0.2 (0.2, 0.4) 0.3 (0.2, 0.3) 0.5 (0.4, 0.6) 0.4 (0.3, 0.6) 
 eGFR, nb 11 12 36 
  Median (Q1, Q3), mL/min/1.73 m2 92 (79, 118) 122 (89, 182) 148 (120, 174) — 131 (98, 135) 119 (87, 148) 

ALT, alanine aminotransferase; AST, aspartate aminotransferase; ECG, electrocardiogram; eGFR, estimated glomerular filtration rate; MIS-C, multisystem inflammatory syndrome in children; Q1, first quartile; Q3, third quartile; RDV, remdesivir (GS-5734).

a

Participants could have more than 1 COVID-19–related disease manifestation.

b

eGFR was calculated using bedside IDMS-traceable Schwartz formula (mL/min/1.73 m2) for children aged 1 to 17 y.

Of the 53 participants who received at least 1 dose of study drug, the median duration of treatment was 5 (4, 8) days with a range of 1 to 10 days; the median for each cohort was also 5 days (Fig 1). Reasons for discontinuation of study drug before 10 days were hospital discharge (22 participants, 42%), investigator discretion (13 participants, 25%), adverse event (2 participants, 4%), parent/guardian decision (2 participants, 4%), and patient decision (1 participant, 2%).

A total of 38 participants (72%) had at least 1 adverse event during the study, and 15 participants (28%) had a Grade 3 or higher adverse event (Table 3). Eleven participants had serious adverse events (Supplemental Table 4); none of which was deemed related to study drug, and all were consistent with either clinical manifestations of COVID-19 or underlying medical conditions. Grade 3 or 4 laboratory abnormalities were reported in 22 participants (42%) overall, including 9 participants (75%) in cohort 1, 2 participants (17%) in cohort 2, 4 participants (33%) in cohort 3, 4 participants (36%) in cohort 4, and 3 participants (60%) in cohort 8 (Supplemental Table 5). The most common grade 3–4 laboratory abnormalities were decreased hemoglobin (n = 9) and decreased estimated glomerular filtration rate (n = 7).

TABLE 3

Adverse Events

Cohort 1 12–17 y and ≥40 kg (n = 12)Cohort 2 28 d–17 y and 20–<40 kg (n = 12)Cohort 3 28 d–17 y and 12–<20 kg (n = 12)Cohort 4 28 d–17 y and 3–<12 kg (n = 12)Cohort 8 <12 y and ≥40 kg (n = 5)Total (N = 53)
Participants, n (%), with       
 Any adverse event 11 (92) 7 (58) 9 (75) 7 (58) 4 (80) 38 (72) 
 Adverse event grade 3 or higher 6 (50) 2 (17) 1 (8) 4 (33) 2 (40) 15 (28) 
 Drug-related adverse event 4 (33) 1 (8) 1 (8) 2 (40) 8 (15) 
 Drug-related adverse event grade 3 or higher 3 (25) 3 (6) 
 Serious adverse eventa 5 (42) 2 (17) 3 (25) 1 (20) 11 (21) 
 Drug-related serious adverse event 
 Adverse event leading to premature study drug discontinuation 2 (17) 2 (4) 
 Treatment-emergent deathb 1 (8) 1 (8) 1 (20) 3 (6) 
Adverse events in ≥2 participants       
 Constipation 3 (25) 1 (8) 1 (8) 1 (8) 3 (60) 9 (17) 
 Acute kidney injury 4 (33) 1 (8) 1 (20) 6 (11) 
 Hyperglycemia 1 (8) 1 (8) 1 (8) 2 (17) 5 (9) 
 Pyrexia 1 (8) 2 (17) 1 (8) 1 (8) 5 (9) 
 Alanine aminotransferase increased 2 (17) 1 (8) 1 (20) 4 (8) 
 Hypertension 2 (17) 1 (8) 1 (20) 4 (8) 
 Hypomagnesaemia 1 (8) 1 (8) 2 (40) 4 (8) 
 Agitation 1 (8) 2 (17) 3 (6) 
 Bradycardia 2 (17) 1 (8) 3 (6) 
 Infusion-site extravasation 2 (17) 2 (4) 
Cohort 1 12–17 y and ≥40 kg (n = 12)Cohort 2 28 d–17 y and 20–<40 kg (n = 12)Cohort 3 28 d–17 y and 12–<20 kg (n = 12)Cohort 4 28 d–17 y and 3–<12 kg (n = 12)Cohort 8 <12 y and ≥40 kg (n = 5)Total (N = 53)
Participants, n (%), with       
 Any adverse event 11 (92) 7 (58) 9 (75) 7 (58) 4 (80) 38 (72) 
 Adverse event grade 3 or higher 6 (50) 2 (17) 1 (8) 4 (33) 2 (40) 15 (28) 
 Drug-related adverse event 4 (33) 1 (8) 1 (8) 2 (40) 8 (15) 
 Drug-related adverse event grade 3 or higher 3 (25) 3 (6) 
 Serious adverse eventa 5 (42) 2 (17) 3 (25) 1 (20) 11 (21) 
 Drug-related serious adverse event 
 Adverse event leading to premature study drug discontinuation 2 (17) 2 (4) 
 Treatment-emergent deathb 1 (8) 1 (8) 1 (20) 3 (6) 
Adverse events in ≥2 participants       
 Constipation 3 (25) 1 (8) 1 (8) 1 (8) 3 (60) 9 (17) 
 Acute kidney injury 4 (33) 1 (8) 1 (20) 6 (11) 
 Hyperglycemia 1 (8) 1 (8) 1 (8) 2 (17) 5 (9) 
 Pyrexia 1 (8) 2 (17) 1 (8) 1 (8) 5 (9) 
 Alanine aminotransferase increased 2 (17) 1 (8) 1 (20) 4 (8) 
 Hypertension 2 (17) 1 (8) 1 (20) 4 (8) 
 Hypomagnesaemia 1 (8) 1 (8) 2 (40) 4 (8) 
 Agitation 1 (8) 2 (17) 3 (6) 
 Bradycardia 2 (17) 1 (8) 3 (6) 
 Infusion-site extravasation 2 (17) 2 (4) 
a

Serious adverse events occurring in 2 participants each: cardiorespiratory arrest, multiple organ dysfunction syndrome, pyrexia, respiratory distress, septic shock, and thrombosis.

b

One participant (cohort 1) died due to multisystem organ failure on day 14; this participant had preexisting elevated transaminases and hyperbilirubinemia and a serious adverse event of multiple organ dysfunction syndrome. One participant (cohort 2) had serious adverse events of hypotension, cardiorespiratory arrest, and respiratory failure; cause of death was reported as respiratory failure secondary to removal from life support (day 35). One participant (cohort 8) had serious adverse events of pneumoperitoneum, fatal respiratory distress, hemodynamic instability, gastrointestinal necrosis, cardiac failure, and multiple organ dysfunction syndrome; cause of death was reported as respiratory, cardiac, and kidney failure as well as acute blood loss from the abdomen (day 18).

Two participants (4%), both in cohort 1, prematurely discontinued study drug because of adverse events. One participant prematurely discontinued after 5 doses of RDV because of adverse events of grade 3 ALT increase, grade 3 AST increase, grade 4 blood sodium increase, and grade 4 hyperbilirubinemia; this participant had elevated transaminases and hyperbilirubinemia at study entry and died on day 14 as a result of multisystem organ failure. With the exception of blood sodium being increased, the laboratory abnormalities were considered related to study drug by the investigator. One participant, who had elevated ALT and AST at baseline, prematurely discontinued study drug after 3 doses of RDV because of grade 3 ALT increase, which was considered related to study drug by the investigator. None of the adverse events that resulted in premature study drug discontinuation were reported as serious adverse events.

Three treatment-emergent deaths occurred during the study. The cause of death for 1 participant (cohort 1), a 16-year-old female, was reported as multisystem organ failure on day 14; this participant (described previously) discontinued RDV after 5 doses because of elevated transaminases. At baseline, the participant was receiving oxygen through invasive mechanical ventilation. The cause of death for the second participant (cohort 2), who had serious adverse events of hypotension, cardiorespiratory arrest, and respiratory failure, was reported as respiratory failure secondary to removal from support on day 35 of hospitalization/study course. The participant was a 16-year-old female who was receiving oxygen via invasive mechanical ventilation at study baseline. For a third participant (cohort 8), who had serious adverse events of pneumoperitoneum, fatal respiratory distress, hemodynamic instability, gastrointestinal necrosis, cardiac failure, and multiple organ dysfunction syndrome, the cause of death was reported as respiratory, cardiac, and kidney failure as well as acute blood loss from the abdomen (day 18). This participant was an 8-year-old male receiving high-flow oxygen at baseline.

Thirty-two days after the last dose of RDV, a fourth participant, a 13-year-old female, died. The participant (in cohort 1) was receiving high-flow oxygen at baseline and had serious adverse events of Gram-negative septic shock as well as pulmonary hemorrhage after completion of treatment. The cause of death was hypoxemic and hypercarbic respiratory failure secondary to SARS-CoV-2 acute respiratory distress syndrome and secondary cytomegalovirus pneumonitis in the setting of systemic lupus erythematosus and lupus nephritis.

The range of exposures of RDV and its metabolites were generally similar, though with high variability, across all evaluated cohorts (Fig 2; Supplemental Fig 4). No apparent trends in increasing PK exposures with decreasing weight or age were observed for RDV and its metabolites (Supplemental Tables 6 and 7).

FIGURE 2

Steady-state plasma AUCτ for (A) remdesivir, (B) GS-704277, and (C) GS-441524 following remdesivir intravenous administration in pediatric participants with COVID-19 compared with the adult phase 3 population. AUCτ, area under the concentration-time curve over 1 dosing interval; IV, intravenous; RDV, remdesivir; TCZ, tocilizumab. Lines are medians and interquartile ranges; dots are individual values. In study GS-US-540-5823, participants assigned to cohort 1 (aged ≥12–17 years, weight ≥40 kg) received 200 mg RDV IV on the first day (loading dose) followed by 100 mg once daily for up to 10 days. Cohorts 2 to 4 (weight 3 kg to <40 kg) received RDV 5 mg/kg IV on the first day (loading dose) followed by RDV 2.5 mg/kg once daily for up to 10 days (steady state). Participants in cohort 8 (<12 years of age, weight ≥40 kg) received RDV 200 mg IV on the first day (loading dose) followed by 100 mg once daily for up to 10 days (steady state). In study CO-US-540-5844, adult participants assigned to RDV + placebo received a 200-mg RDV IV loading dose and placebo infusion on day 1 followed by 100 mg RDV IV on days 2 to 10 (steady state). Participants in study CO-US-540-5844 assigned to RDV + TCZ received a 200-mg RDV IV loading dose and a TCZ 8 mg/kg IV infusion on day 1 followed by 100 mg RDV IV on days 2 to 10 (steady state).

FIGURE 2

Steady-state plasma AUCτ for (A) remdesivir, (B) GS-704277, and (C) GS-441524 following remdesivir intravenous administration in pediatric participants with COVID-19 compared with the adult phase 3 population. AUCτ, area under the concentration-time curve over 1 dosing interval; IV, intravenous; RDV, remdesivir; TCZ, tocilizumab. Lines are medians and interquartile ranges; dots are individual values. In study GS-US-540-5823, participants assigned to cohort 1 (aged ≥12–17 years, weight ≥40 kg) received 200 mg RDV IV on the first day (loading dose) followed by 100 mg once daily for up to 10 days. Cohorts 2 to 4 (weight 3 kg to <40 kg) received RDV 5 mg/kg IV on the first day (loading dose) followed by RDV 2.5 mg/kg once daily for up to 10 days (steady state). Participants in cohort 8 (<12 years of age, weight ≥40 kg) received RDV 200 mg IV on the first day (loading dose) followed by 100 mg once daily for up to 10 days (steady state). In study CO-US-540-5844, adult participants assigned to RDV + placebo received a 200-mg RDV IV loading dose and placebo infusion on day 1 followed by 100 mg RDV IV on days 2 to 10 (steady state). Participants in study CO-US-540-5844 assigned to RDV + TCZ received a 200-mg RDV IV loading dose and a TCZ 8 mg/kg IV infusion on day 1 followed by 100 mg RDV IV on days 2 to 10 (steady state).

Close modal

PK exposures for RDV and its metabolites were largely overlapping, although modestly increased relative to corresponding PK exposures in adults; the percent increases varied depending on which analyte and which PK parameter, with increases in estimated mean AUCτ, Cmax, and Cτ ranging from 33% to 129% for RDV PK parameters, 37% to 124% for GS-704277 PK parameters, and 0% to 60% for GS-441524 PK parameters (Fig 2). No trends were identified between PK exposures (RDV, GS-704277, and GS-441524) and the 7 most commonly reported adverse events (ie, constipation, acute kidney injury, hyperglycemia, pyrexia, ALT increased, hypertension, and hypomagnesemia), except for increased levels of GS-441524 in participants with acute kidney injury (Supplemental Fig 5). No trends were identified between PK parameters and recovery by day 10 on the 7-point ordinal scale (Supplemental Fig 6) or other efficacy endpoints evaluated in this study (data on file). Also, no trends were observed between common COVID-19–related disease manifestations at baseline such as systemic inflammatory response or circulatory manifestation or baseline oxygen support status (data on file). Altogether these analyses demonstrated that sufficient exposures were achieved in all participants regardless of baseline demographics or disease characteristics.

Among 47 participants, no trends were identified in SBECD exposures across age and weight bands (Supplemental Fig 7). SBECD PK exposures (AUC0-4h, Cmax) after administration of RDV appear similar when comparing pediatric participants with COVID-19 to adults with mild to moderate renal impairment or healthy matched controls. The half-life and clearance parameters were also consistent between the adult and pediatric participants receiving remdesivir.

Confirmed negative SARS-CoV-2 PCR results on day 2 through day 10 were reported for 42% (8/19) participants with nasal/oropharyngeal samples, 21% (6/28) participants with nasopharyngeal/oropharyngeal samples, and 22% (2/9) participants with endotracheal tube aspirates. The mean (SD) SARS-CoV-2 viral load changes from baseline up to day 10 were −1.59 (1.514) log10 copies/mL for nasal/oropharyngeal samples (3/53 participants), −1.59 (1.697) log10 copies/mL for nasopharyngeal/oropharyngeal samples (5/53 participants), and −5.94 log10 copies/mL for endotracheal tube aspirates (1/53 participants).

Among the 23 participants with both baseline and postbaseline sequencing data available, 1 participant had 2 amino acid substitutions in RNA-dependent RNA polymerase Nsp12 (A656A/P and G670G/V) observed at day 3 (Supplemental Table 8). Based on analysis of previous cryo-EM structures of the SARS-CoV-2 polymerase complex, A656P and G670V have no direct interaction with the RNA or the incoming nucleotide. Phenotypic analysis of G670V showed no impact on susceptibility to RDV (≤0.96-fold change in EC50; Supplemental Table 9). For the A656P substitution, transfection of the mutant replicon was attempted twice; however, phenotypic results were unable to be generated because of lack of replication. In this participant, viral load declined rapidly, and days 5, 7, and 10 were negative for SARS-CoV-2 RNA. At baseline, this participant had a clinical status of 3 (hospitalized, on noninvasive ventilation or high-flow oxygen devices) and experienced a ≥2-point improvement and recovery based on ordinal score and was released from the hospital on day 15.

Phenotypic analyses were also conducted on amino acid substitutions observed in other parts of the replication-transcription complex (Nsp8, Nsp10, Nsp13, and Nsp14). Substitutions were observed in Nsp10 and Nsp13 (Nsp10 D64D/Y, T101T/I, Nsp13 R248R/I, S259S/L, V266V/F) in 3 participants (Supplemental Table 8). No substitutions were observed in Nsp8 or Nsp14. The substitutions observed either did not impact RDV susceptibility in vitro (Nsp10 T101I, Nsp13 S259L, and V266F showed ≤0.74-fold change in EC50) or could not be determined because of a lack of replication (Supplemental Table 9).

Recovery was reported for 62% of participants (95% CI, 48–75) on day 10 and 83% of participants (95% CI, 70–92) at the time of the last assessment. The median (Q1, Q3) time to recovery was 7 (5, 16) days. For participants who were discharged alive by day 30, the median (Q1, Q3) duration of hospitalization from day 1 was 7 (5, 12) days. The proportion of participants who were discharged from the hospital was 60% by day 10 and 83% by day 30. Among the participants who were not discharged by day 30, 7 were still hospitalized and 2 had died.

Participants showed improvement in clinical status based on the 7-point ordinal scale, discharge from hospitalization, viral load, oxygen requirement, and PEWS scale with treatment with RDV. For all cohorts, there was a trend in improvement of clinical status on the 7-point ordinal scale over time, as indicated by an increasing score (Fig 3). The proportion of participants with an ordinal score of ≤5 points at baseline who had a ≥2-point improvement in clinical status was 75% on day 10 and 85% at the time of the last assessment. The median (Q1, Q3) time to ≥2-point improvement for participants with an ordinal score of ≤5 points at baseline was 7 (5, 10) days. By day 10 or the time of the last assessment, the proportion of the total population with decreases in the PEWS total score (indicating improvement) was 38% of participants (20/53) by 1 to 3 points; 15% or participants (8/53) by 4 to 6 points; and 6% of participants (3/53) by 7 to 9 points.

FIGURE 3

Percentages of participants at each ordinal clinical status by study day.

FIGURE 3

Percentages of participants at each ordinal clinical status by study day.

Close modal

We provide results of a phase 2/3 study of RDV treatment of infants and children aged ≥28 days hospitalized for COVID-19 infection. The study population included 98% who had a preexisting medical condition. In this group, RDV had a favorable tolerability profile with no new apparent safety trends. Pharmacokinetic exposures of RDV in this population of children were similar to those reported in adults. Participants had a high rate (85%) of clinical improvement.

Safety findings were consistent with participants having COVID-19 and/or underlying medical conditions. The most common adverse events were constipation (17%) and acute kidney injury (11%). In clinical studies of RDV in adults, the most common adverse event has been nausea. In clinical studies in healthy adults, no evidence of nephrotoxicity has been observed with single doses of RDV up to 225 mg or multiple once-daily doses of RDV 150 mg for up to 14 days (studies GS-US-399-1812 and GS-US-399-1954). The amount of SBECD administered to children was well within established safe doses for existing commercial products. Based on a review by the European Medicines Agency,24 doses up to 250 mg/kg/day of SBECD are considered safe for children aged >2 years, and no significant toxicity has been seen in a small number of neonates treated with SBECD-containing products corresponding with up to 336 mg/kg/day for 18 to 24 days.

Given the high rates of recovery and absence of new safety concerns observed, the modest increases in RDV exposure observed in hospitalized pediatric participants were not deemed clinically relevant and support up to 10 days of dosing with an adult regimen (200 mg intravenous loading dose on day 1 followed by 100 mg intravenous maintenance doses) in children and adolescents weighing ≥40 kg. Weight-based dosing (5 mg/kg intravenous loading dose on day 1 followed by 2.5 mg/kg intravenous maintenance doses) is supported in children ≥28 days of age weighing ≥3 kg to <40 kg. No substitutions associated with resistance to RDV were observed in this study, supporting a high barrier to RDV resistance development in COVID-19 patients.

This was a single-arm study with small numbers of participants in each cohort, and comparisons between cohorts and extrapolation to larger populations are limited. The preexisting medical histories may also limit generalizability, although they are representative of populations at risk for severe COVID-19 outcomes.

RDV is the only approved COVID-19 treatment in children. This open-label study supports its use in the pediatric population.

The authors thank Leila Bianchi, Elizabeth Lang, and all site investigators and site staff for their contributions to the study. They also thank the participants and their families. Jennifer King, PhD, of August Editorial provided writing assistance. Deqing Xiao and Shalini Saxena supported sample bioanalysis. Eileen Kim and Luzelena Caro critically reviewed the manuscript. Sean Regan and Emma Hughes helped prepare figures.

*Investigators are listed in the Supplemental Appendix.

Drs Humeniuk and Guo contributed to the conception and design of the study; Drs Muller and Rojo contributed to the conception and design of the study and collection of data; Drs Ahmed, Munoz, Agwu, Kimberlin, Galli, Deville, Sue, and Mendez-Echevarria contributed to the collection of data; Drs Rodriguez and Hedskog and Mr Han contributed to resistance analyses; all authors contributed to data analysis and interpretation and manuscript drafting or revising; and all authors approved the final version of the manuscript and agree to be accountable for all aspects of the work.

This trial has been registered at www.clinicaltrials.gov (identifier NCT04431453) and www.eudract.ema.europa.eu (identifier 2020-001803-17).

FUNDING: Funding for this study was provided by Gilead Sciences, Inc.

CONFLICT OF INTEREST DISCLOSURES: Dr Ahmed received research support to the institution from Gilead. Dr Munoz received research support from Gilead, Pfizer, the Centers for Disease Control and Prevention, and National Institutes of Health, has served on data safety monitoring boards for Pfizer, Moderna, Meissa, and Virometix, and serves as a consultant to Moderna, Merck, Sanofi, AstraZeneca, and Seqirus. Dr Muller received research support to institution from Ansun, Astellas, AstraZeneca, Eli Lilly, Enanta Pharmaceuticals, Genentech, Gilead, Janssen, Karius, Melinta, Merck, Moderna, Nabriva, Paratek, Pfizer, and Tetraphase, is on the advisory board of AstraZeneca and Invivyd, and is a consultant for DiaSorin, Seqirus, and ProventionBio. Dr Agwu is a site investigator for a multisite study (Gilead, Merck) and serves on the expert scientific advisory board for Gilead and ViiV. Dr Galli is a site investigator for the current study. Dr Sue is a site investigator for sponsored clinical trials (Gilead, Merck, Allovir). Dr Rojo is on the expert scientific advisory board for Gilead, ViiV, and Merck. The following authors are employees of Gilead Sciences and hold stock interest in the company: Drs Humeniuk, Guo, and Rodriguez, Mr Han, and Dr Hedskog, Ms Maxwell, Dr Palaparthy, and Ms Kersey. The remaining authors have no relevant conflicts of interest to disclose.

Data Sharing Statement: Gilead Sciences shares anonymized individual patient data on request or as required by law or regulation with qualified external researchers based on submitted curriculum vitae and reflecting non conflict of interest. The request proposal must also include a statistician. Approval of such requests is at Gilead Science’s discretion and is dependent on the nature of the request, the merit of the research proposed, the availability of the data, and the intended use of the data. Data requests should be sent to [email protected].

ALT

alanine aminotransferase

AST

aspartate aminotransferase

CARAVAN

Clinical Administration of Remdesivir After COVID-19 Diagnosis in Children Study

LLOQ

lower limit of quantification

PCR

polymerase chain reaction

PEWS

Pediatric Early Warning Score

Pop-PK

population-pharmacokinetics

PK

pharmacokinetics

RDV

remdesivir

RT-qPCR

reverse transcription quantitative polymerase chain reaction

SBECD

sulfobutylether β-cyclodextrin

SDM

site-directed mutant

1
UNICEF
.
COVID-19 confirmed cases and deaths
. Available at: https://data.unicef.org/resources/covid-19-confirmed-cases-and-deaths-dashboard/. Accessed December 19, 2022
2
Götzinger
F
,
Santiago-García
B
,
Noguera-Julián
A
, et al
;
ptbnet COVID-19 Study Group
.
COVID-19 in children and adolescents in Europe: a multinational, multicentre cohort study
.
Lancet Child Adolesc Health
.
2020
;
4
(
9
):
653
661
3
American Academy of Pediatrics
.
Children and COVID-19: state-level data report
. Available at: https://www.aap.org/en/pages/2019-novel-coronavirus-covid-19-infections/children-and-covid-19-state-level-data-report/. Accessed June 7, 2022
4
Wanga
V
,
Gerdes
ME
,
Shi
DS
, et al
;
BMBS1
.
Characteristics and clinical outcomes of children and adolescents aged <18 years hospitalized with COVID-19 - six hospitals, United States, July-August 2021
.
MMWR Morb Mortal Wkly Rep
.
2021
;
70
(
5152
):
1766
1772
5
Wang
M
,
Cao
R
,
Zhang
L
, et al
.
Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro
.
Cell Res
.
2020
;
30
(
3
):
269
271
6
Gordon
CJ
,
Tchesnokov
EP
,
Woolner
E
, et al
.
Remdesivir is a direct-acting antiviral that inhibits RNA-dependent RNA polymerase from severe acute respiratory syndrome coronavirus 2 with high potency
.
J Biol Chem
.
2020
;
295
(
20
):
6785
6797
7
U.S. Food and Drug Administration
.
FDA approves first treatment for COVID-19
. Available at: https://www.fda.gov/news-events/press-announcements/fda-approves-first-treatment-covid-19. Accessed June 14, 2022
8
U.S. Food & Drug Administration
.
Coronavirus (COVID-19) update: FDA approves first COVID-19 treatment for young children
. Available at: https://www.fda.gov/news-events/press-announcements/coronavirus-covid-19-update-fda-approves-first-covid-19-treatment-young-children. Accessed June 14, 2022
9
Beigel
JH
,
Tomashek
KM
,
Dodd
LE
, et al
;
ACTT-1 Study Group Members
.
Remdesivir for the treatment of Covid-19 - final report
.
N Engl J Med
.
2020
;
383
(
19
):
1813
1826
10
Spinner
CD
,
Gottlieb
RL
,
Criner
GJ
, et al
;
GS-US-540-5774 Investigators
.
Effect of remdesivir vs standard care on clinical status at 11 days in patients with moderate COVID-19: a randomized clinical trial
.
JAMA
.
2020
;
324
(
11
):
1048
1057
11
Goldman
JD
,
Lye
DCB
,
Hui
DS
, et al
;
GS-US-540-5773 Investigators
.
Remdesivir for 5 or 10 days in patients with severe Covid-19
.
N Engl J Med
.
2020
;
383
(
19
):
1827
1837
12
Olender
SA
,
Perez
KK
,
Go
AS
, et al
;
GS-US-540-5773 and GS-US-540-5807 Investigators
.
Remdesivir for severe coronavirus disease 2019 (COVID-19) versus a cohort receiving standard of care
.
Clin Infect Dis
.
2021
;
73
(
11
):
e4166
e4174
13
Gottlieb
RL
,
Vaca
CE
,
Paredes
R
, et al
;
GS-US-540-9012 (PINETREE) Investigators
.
Early remdesivir to prevent progression to severe Covid-19 in outpatients
.
N Engl J Med
.
2022
;
386
(
4
):
305
315
14
Goldman
DL
,
Aldrich
ML
,
Hagmann
SHF
, et al
.
Compassionate use of remdesivir in children with severe COVID-19
.
Pediatrics
.
2021
;
147
(
5
):
147
15
Vieux
R
,
Hascoet
JM
,
Merdariu
D
,
Fresson
J
,
Guillemin
F
.
Glomerular filtration rate reference values in very preterm infants
.
Pediatrics
.
2010
;
125
(
5
):
e1186
e1192
16
Colantonio
DA
,
Kyriakopoulou
L
,
Chan
MK
, et al
.
Closing the gaps in pediatric laboratory reference intervals: a CALIPER database of 40 biochemical markers in a healthy and multiethnic population of children
.
Clin Chem
.
2012
;
58
(
5
):
854
868
17
Lutz
JD
,
Mathias
A
,
German
P
,
Pikora
C
,
Reddy
S
,
Kirby
BJ
.
Physiologically-based pharmacokinetic modeling of remdesivir and its metabolites to support dose selection for the treatment of pediatric patients with COVID-19
.
Clin Pharmacol Ther
.
2021
;
109
(
4
):
1116
1124
18
Yang
Z
,
Xiao
D
,
Ling
KHJ
, et al
.
The determination of sulfobutylether β-cyclodextrin sodium (SBECD) by LC-MS/MS and its application in remdesivir pharmacokinetics study for pediatric patients
.
J Pharm Biomed Anal
.
2022
;
212
:
114646
19
Humeniuk
R
,
Mathias
A
,
Cao
H
, et al
.
Safety, tolerability, and pharmacokinetics of remdesivir, an antiviral for treatment of COVID-19, in healthy subjects
.
Clin Transl Sci
.
2020
;
13
(
5
):
896
906
20
He
X
,
Quan
S
,
Xu
M
, et al
.
Generation of SARS-CoV-2 reporter replicon for high-throughput antiviral screening and testing
.
Proc Natl Acad Sci USA
.
2021
;
118
(
15
):
118
21
Zhang
QY
,
Deng
CL
,
Liu
J
, et al
.
SARS-CoV-2 replicon for high-throughput antiviral screening
.
J Gen Virol
.
2021
;
102
(
5
):
102
22
Akre
M
,
Finkelstein
M
,
Erickson
M
,
Liu
M
,
Vanderbilt
L
,
Billman
G
.
Sensitivity of the pediatric early warning score to identify patient deterioration
.
Pediatrics
.
2010
;
125
(
4
):
e763
e769
23
Monaghan
A
.
Detecting and managing deterioration in children
.
Paediatr Nurs
.
2005
;
17
(
1
):
32
35
24
European Medicines Agency
.
Background review for cyclodextrins used as excipients
. Available at: https://www.ema.europa.eu/en/documents/report/background-review-cyclodextrins-used-excipients-context-revision-guideline-excipients-label-package_en.pdf. Accessed July 13, 2023

Supplementary data