Skip to main content
Microbiology Spectrum logoLink to Microbiology Spectrum
. 2021 Nov 24;9(3):e01537-21. doi: 10.1128/Spectrum.01537-21

Broad-Spectrum In Vitro Antiviral Activity of ODBG-P-RVn: An Orally-Available, Lipid-Modified Monophosphate Prodrug of Remdesivir Parent Nucleoside (GS-441524)

Michael K Lo a,, Punya Shrivastava-Ranjan a, Payel Chatterjee a, Mike Flint a, James R Beadle b, Nadejda Valiaeva b, Joyce Murphy b, Robert T Schooley b, Karl Y Hostetler b, Joel M Montgomery a, Christina F Spiropoulou a,
Editor: Manjula Kaliac
PMCID: PMC8612139  PMID: 34817209

ABSTRACT

The necessity for intravenous administration of remdesivir confines its utility for treatment of coronavirus disease 2019 (COVID-19) to hospitalized patients. We evaluated the broad-spectrum antiviral activity of ODBG-P-RVn, an orally available, lipid-modified monophosphate prodrug of the remdesivir parent nucleoside (GS-441524), against viruses that cause diseases of human public health concern, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). ODBG-P-RVn showed 20-fold greater antiviral activity than GS-441524 and had activity nearly equivalent to that of remdesivir in primary-like human small airway epithelial cells. Our results warrant in vivo efficacy evaluation of ODBG-P-RVn.

IMPORTANCE While remdesivir remains one of the few drugs approved by the FDA to treat coronavirus disease 2019 (COVID-19), its intravenous route of administration limits its use to hospital settings. Optimizing the stability and absorption of remdesivir may lead to a more accessible and clinically potent therapeutic. Here, we describe an orally available lipid-modified version of remdesivir with activity nearly equivalent to that of remdesivir against emerging viruses that cause significant disease, including Ebola and Nipah viruses. Our work highlights the importance of such modifications to optimize drug delivery to relevant and appropriate human tissues that are most affected by such diseases.

KEYWORDS: SARS-CoV-2, Ebola virus, Nipah virus, respiratory viruses, hemorrhagic fever virus, filovirus, paramyxovirus, henipavirus, remdesivir, GS-5734, remdesivir nucleoside, GS-441524, antiviral agents, lipid prodrugs, ODBG, Vero E6 cells, Huh7 cells, NCI-H358 cells, human telomerase reverse-transcriptase (hTERT)-immortalized microvascular endothelial cells (TIME), human small airway epithelial cells, HSAEC1-KT, ODBG-P-RVn

OBSERVATION

Remdesivir (RDV; Veklury, GS-5734) is an adenosine nucleotide analog phosphoramidate prodrug with broad-spectrum antiviral activity in vitro and in vivo (1) and is currently the only FDA-approved therapeutic for treating coronavirus 2019 disease (COVID-19) in hospitalized patients over the age of 12 (2). While RDV did not significantly reduce COVID-19 mortality, it shortened the time to recovery compared to the time for placebo controls (3). The short half-life of RDV in human and animal plasma (47), alongside the in vivo efficacy of the RDV parent nucleoside (RVn; GS-441524) against coronaviruses, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) (811), have driven proposals to utilize the RVn instead of RDV to treat COVID-19 (12). A recent comparative pharmacokinetic study in nonhuman primates, however, demonstrated higher levels of the active metabolite RVn-triphosphate (RVn-TP) in lower respiratory tract tissues of RDV-dosed animals than in RVn-dosed animals (6). A significant drawback of RDV is the requirement for intravenous administration, which limits its use to hospital contexts. To develop an orally bioavailable form of remdesivir, we recently synthesized a 1-O-octadecyl-2-O-benzyl-sn-glycerylester (ODBG) lipid-modified monophosphate prodrug of RVn (ODBG-P-RVn) (C40H62N5O9P) (Fig. 1A), which demonstrated more favorable in vitro antiviral activity than RVn and RDV against SARS-CoV-2 in Vero-E6 cells (13).

FIG 1.

FIG 1

Comparison of antiviral activities of RVn, RDV, and ODBG-P-RVn in African green monkey (Vero-E6), human hepatoma (Huh7), human bronchioalveolar carcinoma (NCI-H358), and primary-like human telomerase reverse transcriptase-immortalized small airway epithelial (HSAEC1-KT) cell lines using fluorescent-reporter-based, image-based, and cytopathic effect (CPE) assays. Values for representative dose-response inhibition of viral replication and induction of cellular cytotoxicity by RVn, RDV, and ODBG-P-RVn are shown. (A) The chemical structure of ODBG-P-RVn. (B) Direct measurement of reporter fluorescence intensities from recombinant Ebola virus (EBOV) expressing ZsGreen protein in Vero-E6 (left) and Huh7 (middle left) cells and recombinant Nipah virus (NiV) expressing ZsGreen protein in NCI-H358 (middle right) and HSAEC1-KT (right) cells. (C) Image-based counting of reporter fluorescence-positive cells infected with recombinant severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) expressing mNeonGreen protein (Vero-E6 and Huh7 cells) and recombinant respiratory syncytial virus (RSV) expressing enhanced green fluorescent protein (eGFP) (NCI-H358 and HSAEC1-KT cells). Values for infected cells treated with dimethyl sulfoxide (DMSO) were considered the 100% fluorescence intensity signals and 100% fluorescence-positive cell counts. (D) Compound-based inhibition of CPE induced by yellow fever virus (YFV) (Vero-E6 and Huh7 cells) and by Hendra virus (HeV) (NCI-H358 and HSAEC1-KT cells) determined by measuring cellular ATP levels (CellTiterGlo 2.0). ATP levels in uninfected cells treated with DMSO were considered 100% CPE inhibition. (E) Compound cytotoxicity/cell viability for the respective cell lines used measured by CellTiterGlo 2.0 assay. (F) Measurement of RVn-triphosphate (RVn-TP) levels in Vero E6 cells treated with RVn, RDV, or ODBG-P-RVn at various time points until 48 h posttreatment. (G) Reductions of infectious yields of EBOV-ZsG (left) and NiV-ZsG (right) by RDV and ODBG-P-RVn in HSAEC1-KT cells. y axis denotes 50% tissue culture infectious dose (TCID50) expressed in logarithmic scale. Dose-response curves for antiviral assays in panels B, C, and D were fitted to the mean values of experiments performed in biological triplicate for each concentration in the 8-point, 3-fold dilution series using a 4-parameter nonlinear logistic regression curve with variable slope. Data points and error bars indicate the mean values and standard deviations from 3 biological replicates; each colored shape/line in the legend represents an independent experiment performed in biological triplicate as indicated above panel B. Infectious yield reduction assays were conducted once in biological quadruplicates. RVn and RDV used in this study were obtained from MedChemExpress (Monmouth Junction, NJ, USA).

In this study, we extended our in vitro comparisons to include 14 viruses from 7 virus families responsible for causing diseases of significant human public health concern. These were Ebola virus (EBOV) and Marburg virus (MARV) from the family Filoviridae, Nipah virus (NiV), Hendra virus (HeV), human parainfluenza virus 3 (hPIV3), measles virus (MV), mumps virus (MuV), and Sosuga virus (SoSuV) from the family Paramyxoviridae, respiratory syncytial virus (RSV) from the family Pneumoviridae, yellow fever virus (YFV) from the family Flaviviridae, Lassa virus (LASV) from the family Arenaviridae, Crimean-Congo hemorrhagic fever virus (CCHFV) from the family Nairoviridae, and SARS-CoV-2 from the family Coronaviridae (1418). We utilized the following three previously described assays to compare the antiviral activities of RVn, RDV, and ODBG-P-RVn against this panel of viruses (15, 17): (i) directly measuring the fluorescence of a reporter protein expressed by recombinant viruses (REP) (Fig. 1B), (ii) quantitating focus-forming units (FFU) via fluorescent-reporter imaging (Fig. 1C), and (iii) indirectly measuring cytopathic effect (CPE) based on cellular ATP levels (CellTiterGlo 2.0; Promega) (Fig. 1D), which was also used to evaluate compound cytotoxicity (Fig. 1E). Assay conditions varied based on virus replication kinetics and on the specific assay used; the multiplicities of infection (MOIs) ranged from 0.01 to 0.25, and endpoint measurements were conducted between 72 and 144 h postinfection (hpi) (see Methods in the supplemental material). We conducted dose-response experiments using 8-point, 3-fold serial dilutions of RVn, RDV, and ODBG-P-RVn against our panel of viruses in Vero-E6 cells and showed that ODBG-P-RVn consistently had greater antiviral activity than RVn and RDV against all viruses susceptible to RVn/RDV inhibition, with 50% effective concentration (EC50) values ranging from 0.026 to 1.13 µM (Fig. 1B to D, left; Table 1; Fig. S1 in the supplemental material). RVn and ODBG-P-RVn induced partial cytotoxicity but only at the highest concentration tested (100 µM) and without reaching 50% cytotoxic concentration (CC50). To understand the comparatively greater potency of ODBG-P-RVn in Vero E6 cells, we measured the levels of RVn-TP in cells treated with RVn, RDV, or ODBG-P-RVn. We observed that RVn-TP levels indeed correlated with antiviral activity, with ODBG-P-RVn consistently accumulating to higher levels than both RVn and RDV across 3 time points (Fig. 1F). We then compared these antivirals in human hepatoma (Huh7) and bronchioalveolar carcinoma (NCI-H358) cell lines, which represent more-relevant cell types that are targeted by subsets of viruses used in our study. In both human cell lines, although ODBG-P-RVn showed EC50 values comparable to those observed in Vero-E6 cells and was 3- to 5-fold more active than RVn, it consistently showed 6- to 20-fold less activity than RDV (Fig. 1B to D, middle left and middle right; Table 1; Fig. S2 and S3). Whereas the CC50 values for RDV in Huh7 and NCI-H358 cells were 54.2 and 77.2 µM, respectively, ODBG-P-RVn was less cytotoxic in Huh7 cells (CC50 = 93.4 µM) and did not show measurable cytotoxicity in NCI-H358 cells even at the highest concentration tested (100 µM) (Fig. 1E, middle right; Table 1).

TABLE 1.

Mean antiviral activities of RVn, RDV, and ODBG-P-RVn in Vero E6, Huh7, and NCI-H358 cell lines

Virus family Virusa Species/variantb Assayc Mean value (µM) ± SD for indicated antiviral in indicated cellsd
Vero E6
Huh7/NCI-H358e
RVn (GS-441524)
RDV (GS-5734)
ODBG-P-RVn
RVn (GS-441524)
RDV (GS-5734)
ODBG-P-RVn
EC50 EC90 SI (CC50 > 100 µM) EC50 EC90 SI (CC50 > 100 µM) EC50 EC90 SI (CC50 > 100 µM) EC50 EC90 SI (CC50 > 100 µM/>100) EC50 EC90 SI (CC50: 54.2 ± 6.0/77.2 ± 5.3) EC50 EC90 SI (CC50: 93.4 ± 3.0/>100)
Filoviridae EBOV Rec. Makona-ZsG REP 2.03 ± 0.50 7.54 ± 1.09 49 5.15 ± 1.09 17.31 ± 0.89 >19 0.39 ± 0.10 1.71 ± 0.25 >258 1.84 ± 0.31 6.91 ± 1.79 >54 0.020 ± 0.003 0.16 ± 0.02 2,710 0.37 ± 0.06 2.13 ± 0.37 251
MARV Rec. Bat371-ZsG REP 0.96 ± 0.09 4.05 ± 1.42 104 2.16 ± 0.27 10.22 ± 2.02 >46 0.19 ± 0.04 0.81 ± 0.12 >521 1.92 ± 0.06 4.47 ± 0.48 >52 0.025 ± 0.002 0.075 ± 0.003 2,128 0.33 ± 0.02 0.99 ± 0.09 285
Paramyxoviridae NiV-M Rec. Malaysia-ZsG REP 1.10 ± 0.40 2.20 ± 1.05 73 5.87 ± 0.19 9.82 ± 0.43 >16 0.31 ± 0.04 0.78 ± 0.28 >196 2.43 ± 0.31 5.95 ± 1.10 >41 0.075 ± 0.001 0.31 ± 0.04 1,026 0.50 ± 0.06 2.83 ± 1.39 >198
CPE 0.48 ± 0.06 0.78 ± 0.19 207 3.34 ± 0.34 5.39 ± 0.29 >30 0.19 ± 0.01 0.30 ± 0.04 >522 ND ND NA ND ND NA ND ND NA
NiV-B Bangladesh CPE 0.52 ± 0.02 1.14 ± 0.02 192 2.84 ± 0.10 5.81 ± 0.44 >35 0.17 ± 0.01 0.38 ± 0.04 >599 3.42 ± 0.005 5.41 ± 0.29 >29 0.12 ± 0.0004 0.19 ± 0.01 661 0.82 ± 0.053 1.38 ± 0.05 >122
HeV 1996 CPE 1.43 ± 0.17 12.06 ± 3.14 70 4.56 ± 0.20 17.58 ± 3.91 >22 0.37 ± 0.04 3.93 ± 1.98 >270 3.68 ± 0.08 6.33 ± 0.18 >27 0.16 ± 0.02 0.25 ± 0.03 491 0.95 ± 0.12 1.42 ± 0.03 >105
MV Rec. rMVEZGFP(3) REP 0.58 ± 0.20 1.71 ± 0.07 172 4.97 ± 0.25 6.12 ± 0.3 >20 0.16 ± 0.03 0.21 ± 0.01 >609 0.88 ± 0.16 6.99 ± 1.90 >113 0.025 ± 0.007 0.13 ± 0.09 3,074 0.12 ± 0.003 0.86 ± 0.22 >803
hPIV3 Rec. JS-GFP FFU 0.14 ± 0.01 0.28 ± 0.02 70 0.43 ± 0.09 0.90 ± 0.03 >232 0.026 ± 0.002 0.050 ± 0.002 >3,896 1.43 ± 0.16 1.98 ± 0.05 >70 0.031 ± 0.002 0.052 ± 0.01 2,458 0.22 ± 0.01 0.43 ± 0.02 >457
MuV Rec. IA2006-eGFP FFU 5.11 ± 0.20 7.80 ± 0.64 18 16.81 ± 1.23 25.1 ± 1.97 >4.9 1.13 ± 0.04 2.53 ± 0.25 >56 9.3 ± 0.30 13.71 ± 0.24 >11 0.20 ± 0.003 0.24 ± 0.003 266 1.85 ± 0.11 2.24 ± 0.23 50
SoSuV Rec. 2012-ZsG REP 1.00 ± 0.10 2.72 ± 0.62 100 5.31 ± 1.8 19.10 ± 9.31 >19 0.31 ± 0.089 0.80 ± 0.06 >325 2.06 ± 0.09 7.76 ± 1.11 >48 0.052 ± 0.01 0.13 ± 0.02 1,042 0.52 ± 0.10 1.08 ± 0.15 180
Pneumoviridae RSV Rec. rgRSV0224 (A2) FFU 0.49 ± 0.05 0.62 ± 0.01 206 1.80 ± 0.08 2.40 ± 0.27 >55 0.10 ± 0.02 0.22 ± 0.03 >997 1.93 ± 0.02 2.36 ± 0.08 >51 0.078 ± 0.004 0.17 ± 0.02 991 0.55 ± 0.057 1.41 ± 0.09 >180
Coronaviridae SARS-CoV-2 Rec. icSARS-CoV-2 mNG (WA1) FFU 0.42 ± 0.09 0.60 ± 0.06 236 1.77 ± 0.13 2.81 ± 0.78 >56 0.10 ± 0.005 0.16 ± 0.01 >997 0.69 ± 0.01 1.50 ± 0.20 >144 0.011 ± 0.001 0.035 ± 0.002 5,073 0.12 ± 0.02 0.69 ± 0.07 778
Flaviviridae YFV 17D CPE 3.52 ± 0.24 30.25 ± 10.08 28 19.86 ± 1.73 >50 >5 0.87 ± 0.043 7.37 ± 1.59 >114 36.83 ± 2.85 >50 >2.7 0.88 ± 0.057 3.09 ± 1.47 62 14.11 ± 0.90 >50 6.6
Arenaviridae LASV Rec. Josiah-ZsG REP NI NI NA NI NI NA 31.14 ± 7.79 >50 >3 NI NI NA 2.87 ± 0.61 5.17 ± 0.33 19 NI NI NA
Nairoviridae CCHFV Rec. IbAr10200-ZsG REP NI NI NA NI NI NA NI NI NA NI NI NA NI NI NA NI NI NA
a

EBOV, Ebola virus; MARV, Marburg virus; NiV-M, Nipah virus Malaysia strain; NiV-B, Nipah virus Bangladesh strain; HeV, Hendra virus; MV, measles virus; hPIV3, human parainfluenza virus 3; MuV, mumps virus; SoSuV, Sosuga virus; RSV, respiratory syncytial virus; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; YFV, yellow fever virus; LASV, Lassa virus; CCHFV, Crimean-Congo hemorrhagic fever virus.

b

Rec., recombinant; ZsG, ZsGreen fluorescent protein; GFP, green fluorescent protein; eGFP, enhanced GFP; mNG, mNeonGreen.

c

REP, CPE, and FFU assays were conducted between 72 and 144 hpi. REP, fluorescent reporter; CPE, cytopathic effect; FFU, focus-forming units.

d

Values were derived from 3 independent experiments performed in biological triplicates, except for assays of NiV-B (NCI-H358 cells), HeV (NCI-H358 cells), and YFV (Vero E6 cells), which were performed twice in biological triplicates. EC50, EC90, and CC50 values were calculated using GraphPad Prism 9 software. EC50 and EC90, 50% and 90% effective concentrations; CC50, 50% cytotoxic concentration; SI, selective index (EC50/CC50); ND, not determined; NI, no inhibition; NA, not applicable. The CC50 values for each compound in the respective cell lines are indicated in parentheses above the column indicated for SI values.

e

Data in boldface were derived from Huh7 cells, and underlined data were derived from NCI-H358 cells.

To further evaluate cell type-specific effects on the antiviral activities of RVn, RDV, and ODBG-P-RVn, we tested them against a smaller subset of reporter viruses in primary-like human telomerase reverse transcriptase (hTERT)-immortalized human microvascular endothelial (TIME) cells (19, 20). In TIME cells, we observed a trend in antiviral activity similar to those in Huh7 and NCI-H358 cells, with ODBG-P-RVn showing 15- to 22-fold greater activity than RVn but 5- to 8-fold less activity than RDV in reporter-based assays (Table 2; Fig. S4A). We further compared the activities of RDV and ODBG-P-RVn by infectious yield assay and observed that both compounds equivalently reduced the infectious yield of EBOV expressing ZsGreen protein (EBOV-ZsG), by up to 4 log10, and that of of NiV-ZsG, by approximately 2 log10, in a dose-dependent manner, with EC50 values closely mirroring the values determined in reporter assays (Table 2; Fig. S4B). However, RDV was more cytotoxic (CC50 = 17.2 µM) than ODBG-P-RVn (CC50 > 50 µM), which is reflected in its biphasic inhibition of NiV-ZsG, with cytotoxic inhibition by RDV shown at 16.6 µM (Fig. S4C).

TABLE 2.

Mean antiviral activities of RVn, RDV, and ODBG-P-RVn in TIME and HSAEC1-KT cell lines

Virus family Virusa Species/variantb Assayc Mean value (µM) ± SD for indicated antiviral in indicated cellsd
HSAEC1-KT
TIME
RVn (GS-441524)
RDV (GS-5734)
ODBG-P-RVn
RVn (GS-441524)
RDV (GS-5734)
ODBG-P-RVn
EC50 EC90 SI (CC50 > 100 µM) EC50 EC90 SI (CC50 > 100 µM) EC50 EC90 SI (CC50 = 20.5 ± 0.29 µM) EC50 EC90 SI (CC50 > 100 µM) EC50 EC90 SI (CC50 = 17.2 ± 0.42 µM) EC50 EC90 SI (CC50 > 50 µM)
Filoviridae EBOV Rec. Makona-ZsG REP 10.7 ± 2.62 21.79 ± 3.16 >9.3 0.17 ± 0.02 0.41 ± 0.14 >587 0.21 ± 0.02 1.06 ± 0.18 98 14.88 ± 0.28 17.24 ± 0.16 >3.36 0.13 ± 0.04 0.2 ± 0.01 132 0.99 ± 0.063 1.96 ± 0.043 >50
VTR ND ND NA 0.11 0.82 >909 0.21 0.95 98 ND ND NA 0.032 0.064 530 0.15 0.39 >324
MARV Rec. Bat371-ZsG REP/FFU 35.53 ± 7.07 71.35 ± 1.28 >2.8 0.75 ± 0.19 2.92 ± 0.14 >133 0.71 ± 0.11 3.67 ± 0.49 29 5.2 ± 0.26 6.89 ± 0.86 >9.61 0.04 ± 0.003 0.086 ± 0.004 430 0.23 ± 0.036 0.66 ± 0.032 >213
Paramyxoviridae NiV-M Rec. Malaysia-ZsG REP 16.46 ± 0.04 19.12 ± 0.05 >6.1 0.23 ± 0.01 0.31 ± 0.06 >440 0.57 ± 0.013 0.97 ± 0.21 36 13.53 ± 2.44 17.52 ± 0.77 >3.70 0.10 ± 0.01 0.20 ± 0.01 172 0.75 ± 0.05 2.01 ± 0.30 >66
CPE 16.12 ± 4.21 78.1 ± 35.08 >6.2 0.31 ± 0.04 0.075 ± 0.004 >318 0.90 ± 0.07 10.22 ± 4.99 23 ND ND NA ND ND NA ND ND NA
VTR ND ND NA 0.26 0.36 >379 0.47 0.77 44 ND ND NA 0.054 0.07 319 0.26 0.77 >195
NiV-B Bangladesh CPE 11.23 ± 0.63 33.6 ± 1.58 >8.9 0.21 ± 0.063 0.62 ± 0.20 >379 0.41 ± 0.039 1.71 ± 0.66 50
HeV 1994 CPE 11.52 ± 1.49 26.11 ± 4.44 >8.7 0.22 ± 0.04 0.65 ± 0.11 >463 0.42 ± 0.023 1.19 ± 0.061 49
MV Rec. rMVEZGFP(3) REP 4.98 ± 0.37 12.02 ± 2.7 >20 0.063 ± 0.02 0.128 ± 0.016 >1,587 0.082 ± 0.026 0.29 ± 0.043 251
hPIV3 Rec. JS-GFP FFU 4.96 ± 0.05 5.77 ± 0.06 >20 0.063 ± 0.001 0.074 ± 0.002 >1,582 0.091 ± 0.009 0.20 ± 0.008 226
Pneumoviridae RSV Rec. rgRSV0224 (A2) FFU 4.92 ± 0.47 8.09 ± 0.68 >20 0.088 ± 0.026 0.21 ± 0.033 >,1134 0.12 ± 0.008 0.34 ± 0.047 176
a

EBOV, Ebola virus; MARV, Marburg virus; NiV-M, Nipah virus Malaysia strain; NiV-B, Nipah virus Bangladesh strain; HeV, Hendra virus; MV, measles virus; hPIV3, human parainfluenza virus 3; RSV, respiratory syncytial virus.

b

Rec., recombinant; ZsG, ZsGreen fluorescent protein; GFP, green fluorescent protein.

c

REP, FFU, CPE, and VTR assays were conducted at 72 hpi. REP, fluorescent reporter; VTR, virus titer reduction; CPE, cytopathic effect; FFU, focus-forming units.

d

Values were derived from 3 independent experiments performed in biological triplicates. TIME, primary-like human telomerase reverse-transcriptase (hTERT)-immortalized human microvascular endothelial cell line; HSAEC1-KT, hTERT-immortalized small airway epithelial cell line; EC50, EC90, and CC50 values were calculated using GraphPad Prism 9 software. EC50 and EC90, 50% and 90% effective concentrations; CC50, 50% cytotoxic concentration; SI, selective index (EC50/CC50); ND, not determined; NI, no inhibition; NA, not applicable. The CC50 values for each compound in the respective cell lines are indicated in parentheses above the column indicated for SI values.

Since ODBG lipid nucleoside modification enhances in vivo lung tissue distribution via the chylomicron pathway (21, 22), we compared the activities of the three compounds against filoviruses, paramyxoviruses, and RSV in another primary-like, hTERT-immortalized small airway epithelial cell line (HSAEC1-KT) (23). Notably, the dose-response curves of RDV and ODBG-P-RVn were strikingly similar, with EC50 values in the submicromolar range within a 3-fold range of each other; the EC50 values for some viruses were almost identical (Fig. 1B to D, right; Table 2; Fig. S5). Furthermore, RDV and ODBG-P-RVn reduced the infectious yields of EBOV-ZsG and NiV-ZsG in HSAEC1-KT cells equivalently, by 5 log10 and 3 log10, respectively, and their EC50 values reflected the limited differential in antiviral activities between them (Fig. 1G; Table 2). Although ODBG-P-RVn was more cytotoxic (CC50 = 20.5) than RDV (CC50 > 100) in HSAEC1-KT cells (Fig. 1D, right; Table 2), it also effectively reduced the virus yields at noncytotoxic concentrations. We also evaluated the antiviral activity of the ODBG lipid alone and observed no detectable antiviral activity against any of the viruses tested in HSAEC1-KT cells (data not shown).

Our results demonstrate that ODBG-P-RVn has greater antiviral activity than RVn and has cell type-dependent activity levels that range from moderately lower than to nearly equal to those of RDV. In vivo, RDV is converted rapidly to RVn (47), which has 0.5 to 2 log10 less activity than RDV against most of the viruses tested. In contrast, ODBG-P-RVn was stable in plasma for >24 h and reached therapeutic plasma levels (above the EC90 for SARS-CoV-2) after oral administration of 16.9 mg/kg of body weight to Syrian hamsters; it also did not produce virologically significant levels of RVn (13). Thus, one would predict sustained in vivo antiviral activity with ODBG-P-RVn, without substantial generation of RVn, the less active metabolite, in plasma. Taken together, our results support further optimization of ODBG-P-RVn and future in vivo evaluation of such monophosphate lipid-modified analogs of RVn for their efficacy against viruses significant to human health.

Supplementary Material

Reviewer comments
reviewer-comments.pdf (86.1KB, pdf)

ACKNOWLEDGMENTS

We thank Tatyana Klimova for helpful comments in reviewing the manuscript. We thank Pei-Yong Shi (University of Texas Medical Branch) for the kind gift of the SARS-CoV-2 reporter strain expressing mNeonGreen.

The findings and conclusions in this report are those of the authors and do not necessarily represent those of the Centers for Disease Control and Prevention. This work was supported by CDC core funding and by the National Institute of Allergy and Infectious Diseases (grant number RO1-AI131424).

Footnotes

Supplemental material is available online only.

SUPPLEMENTAL FILE 1
Supplemental material. Download SPECTRUM01537-21_Supp_1_seq6.pdf, PDF file, 6.8 MB (6.9MB, pdf)

Contributor Information

Michael K. Lo, Email: mko2@cdc.gov.

Christina F. Spiropoulou, Email: ccs8@cdc.gov.

Manjula Kalia, Regional Centre for Biotechnology.

REFERENCES

  • 1.Malin JJ, Suárez I, Priesner V, Fätkenheuer G, Rybniker J. 2020. Remdesivir against COVID-19 and other viral diseases. Clin Microbiol Rev 34:e00162-20. doi: 10.1128/CMR.00162-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Gilead. 2020. Veklury/remdesivir. FDA approved to treat COVID-19. Gilead, Foster City, CA. https://www.veklury.com/. Accessed 8 July 2021.
  • 3.Beigel JH, Tomashek KM, Dodd LE, Mehta AK, Zingman BS, Kalil AC, Hohmann E, Chu HY, Luetkemeyer A, Kline S, Lopez de Castilla D, Finberg RW, Dierberg K, Tapson V, Hsieh L, Patterson TF, Paredes R, Sweeney DA, Short WR, Touloumi G, Lye DC, Ohmagari N, Oh M-D, Ruiz-Palacios GM, Benfield T, Fätkenheuer G, Kortepeter MG, Atmar RL, Creech CB, Lundgren J, Babiker AG, Pett S, Neaton JD, Burgess TH, Bonnett T, Green M, Makowski M, Osinusi A, Nayak S, Lane HC. 2020. Remdesivir for the treatment of Covid-19—final report. N Engl J Med 383:1813–1826. doi: 10.1056/NEJMoa2007764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Warren TK, Jordan R, Lo MK, Ray AS, Mackman RL, Soloveva V, Siegel D, Perron M, Bannister R, Hui HC, Larson N, Strickley R, Wells J, Stuthman KS, Van Tongeren SA, Garza NL, Donnelly G, Shurtleff AC, Retterer CJ, Gharaibeh D, Zamani R, Kenny T, Eaton BP, Grimes E, Welch LS, Gomba L, Wilhelmsen CL, Nichols DK, Nuss JE, Nagle ER, Kugelman JR, Palacios G, Doerffler E, Neville S, Carra E, Clarke MO, Zhang L, Lew W, Ross B, Wang Q, Chun K, Wolfe L, Babusis D, Park Y, Stray KM, Trancheva I, Feng JY, Barauskas O, Xu Y, Wong P, et al. 2016. Therapeutic efficacy of the small molecule GS-5734 against Ebola virus in rhesus monkeys. Nature 531:381–385. doi: 10.1038/nature17180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Tempestilli M, Caputi P, Avataneo V, Notari S, Forini O, Scorzolini L, Marchioni L, Ascoli Bartoli T, Castilletti C, Lalle E, Capobianchi MR, Nicastri E, D’Avolio A, Ippolito G, Agrati C, COVID 19 INMI Study Group. 2020. Pharmacokinetics of remdesivir and GS-441524 in two critically ill patients who recovered from COVID-19. J Antimicrob Chemother 75:2977–2980. doi: 10.1093/jac/dkaa239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Mackman RL, Hui HC, Perron M, Murakami E, Palmiotti C, Lee G, Stray K, Zhang L, Goyal B, Chun K, Byun D, Siegel D, Simonovich S, Du Pont V, Pitts J, Babusis D, Vijjapurapu A, Lu X, Kim C, Zhao X, Chan J, Ma B, Lye D, Vandersteen A, Wortman S, Barrett KT, Toteva M, Jordan R, Subramanian R, Bilello JP, Cihlar T. 2021. Prodrugs of a 1′-CN-4-aza-7,9-dideazaadenosine C-nucleoside leading to the discovery of remdesivir (GS-5734) as a potent inhibitor of respiratory syncytial virus with efficacy in the African green monkey model of RSV. J Med Chem 64:5001–5017. doi: 10.1021/acs.jmedchem.1c00071. [DOI] [PubMed] [Google Scholar]
  • 7.Humeniuk R, Mathias A, Cao H, Osinusi A, Shen G, Chng E, Ling J, Vu A, German P. 2020. Safety, tolerability, and pharmacokinetics of remdesivir, an antiviral for treatment of COVID-19, in healthy subjects. Clin Transl Sci 13:896–906. doi: 10.1111/cts.12840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Shi Y, Shuai L, Wen Z, Wang C, Yan Y, Jiao Z, Guo F, Fu ZF, Chen H, Bu Z, Peng G. 2021. The preclinical inhibitor GS441524 in combination with GC376 efficaciously inhibited the proliferation of SARS-CoV-2 in the mouse respiratory tract. Emerg Microbes Infect 10:481–492. doi: 10.1080/22221751.2021.1899770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Li Y, Cao L, Li G, Cong F, Li Y, Sun J, Luo Y, Chen G, Li G, Wang P, Xing F, Ji Y, Zhao J, Zhang Y, Guo D, Zhang X. 1 February 2021. Remdesivir metabolite GS-441524 effectively inhibits SARS-CoV-2 infection in mouse models. J Med Chem. doi: 10.1021/acs.jmedchem.0c01929. [DOI] [PubMed] [Google Scholar]
  • 10.Murphy BG, Perron M, Murakami E, Bauer K, Park Y, Eckstrand C, Liepnieks M, Pedersen NC. 2018. The nucleoside analog GS-441524 strongly inhibits feline infectious peritonitis (FIP) virus in tissue culture and experimental cat infection studies. Vet Microbiol 219:226–233. doi: 10.1016/j.vetmic.2018.04.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Pedersen NC, Perron M, Bannasch M, Montgomery E, Murakami E, Liepnieks M, Liu H. 2019. Efficacy and safety of the nucleoside analog GS-441524 for treatment of cats with naturally occurring feline infectious peritonitis. J Feline Med Surg 21:271–281. doi: 10.1177/1098612X19825701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Yan VC, Muller F. 2020. Comprehensive summary supporting clinical investigation of GS-441524 for Covid-19 treatment. OSFPREPRINTS. doi: 10.31219/osf.io/mnhxu. [DOI] [Google Scholar]
  • 13.Schooley RT, Carlin AF, Beadle JR, Valiaeva N, Zhang X-Q, Clark AE, McMillan RE, Leibel SL, McVicar RN, Xie J, Garretson AF, Smith VI, Murphy J, Hostetler KY. 2021. Rethinking remdesivir: synthesis, antiviral activity and pharmacokinetics of oral lipid prodrugs. Antimicrob Agents Chemother 65:e01155-21. doi: 10.1128/AAC.01155-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Xie X, Muruato A, Lokugamage KG, Narayanan K, Zhang X, Zou J, Liu J, Schindewolf C, Bopp NE, Aguilar PV, Plante KS, Weaver SC, Makino S, LeDuc JW, Menachery VD, Shi PY. 2020. An infectious cDNA clone of SARS-CoV-2. Cell Host Microbe 27:841–848.e3. doi: 10.1016/j.chom.2020.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Lo MK, Jordan R, Arvey A, Sudhamsu J, Shrivastava-Ranjan P, Hotard AL, Flint M, McMullan LK, Siegel D, Clarke MO, Mackman RL, Hui HC, Perron M, Ray AS, Cihlar T, Nichol ST, Spiropoulou CF. 2017. GS-5734 and its parent nucleoside analog inhibit Filo-, Pneumo-, and Paramyxoviruses. Sci Rep 7:43395. doi: 10.1038/srep43395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Lo MK, Spengler JR, Krumpe LRH, Welch SR, Chattopadhyay A, Harmon JR, Coleman-McCray JD, Scholte FEM, Hotard AL, Fuqua JL, Rose JK, Nichol ST, Palmer KE, O’Keefe BR, Spiropoulou CF. 2020. Griffithsin inhibits Nipah virus entry and fusion and can protect Syrian golden hamsters from lethal Nipah virus challenge. J Infect Dis 221:S480–S492. doi: 10.1093/infdis/jiz630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Lo MK, Jordan PC, Stevens S, Tam Y, Deval J, Nichol ST, Spiropoulou CF. 2018. Susceptibility of paramyxoviruses and filoviruses to inhibition by 2′-monofluoro- and 2′-difluoro-4′-azidocytidine analogs. Antiviral Res 153:101–113. doi: 10.1016/j.antiviral.2018.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Welch SR, Chakrabarti AK, Wiggleton Guerrero L, Jenks HM, Lo MK, Nichol ST, Spiropoulou CF, Albariño CG. 2018. Development of a reverse genetics system for Sosuga virus allows rapid screening of antiviral compounds. PLoS Negl Trop Dis 12:e0006326. doi: 10.1371/journal.pntd.0006326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Siegel D, Hui HC, Doerffler E, Clarke MO, Chun K, Zhang L, Neville S, Carra E, Lew W, Ross B, Wang Q, Wolfe L, Jordan R, Soloveva V, Knox J, Perry J, Perron M, Stray KM, Barauskas O, Feng JY, Xu Y, Lee G, Rheingold AL, Ray AS, Bannister R, Strickley R, Swaminathan S, Lee WA, Bavari S, Cihlar T, Lo MK, Warren TK, Mackman RL. 2017. Discovery and synthesis of a phosphoramidate prodrug of a pyrrolo[2,1-f][triazin-4-amino] adenine C-nucleoside (GS-5734) for the treatment of Ebola and emerging viruses. J Med Chem 60:1648–1661. doi: 10.1021/acs.jmedchem.6b01594. [DOI] [PubMed] [Google Scholar]
  • 20.Venetsanakos E, Mirza A, Fanton C, Romanov SR, Tlsty T, McMahon M. 2002. Induction of tubulogenesis in telomerase-immortalized human microvascular endothelial cells by glioblastoma cells. Exp Cell Res 273:21–33. doi: 10.1006/excr.2001.5424. [DOI] [PubMed] [Google Scholar]
  • 21.Hostetler KY, Beadle JR, Trahan J, Aldern KA, Owens G, Schriewer J, Melman L, Buller RM. 2007. Oral 1-O-octadecyl-2-O-benzyl-sn-glycero-3-cidofovir targets the lung and is effective against a lethal respiratory challenge with ectromelia virus in mice. Antiviral Res 73:212–218. doi: 10.1016/j.antiviral.2006.10.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Hostetler KY. 2009. Alkoxyalkyl prodrugs of acyclic nucleoside phosphonates enhance oral antiviral activity and reduce toxicity: current state of the art. Antiviral Res 82:A84–A98. doi: 10.1016/j.antiviral.2009.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Ramirez RD, Sheridan S, Girard L, Sato M, Kim Y, Pollack J, Peyton M, Zou Y, Kurie JM, Dimaio JM, Milchgrub S, Smith AL, Souza RF, Gilbey L, Zhang X, Gandia K, Vaughan MB, Wright WE, Gazdar AF, Shay JW, Minna JD. 2004. Immortalization of human bronchial epithelial cells in the absence of viral oncoproteins. Cancer Res 64:9027–9034. doi: 10.1158/0008-5472.CAN-04-3703. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Reviewer comments
reviewer-comments.pdf (86.1KB, pdf)
SUPPLEMENTAL FILE 1

Supplemental material. Download SPECTRUM01537-21_Supp_1_seq6.pdf, PDF file, 6.8 MB (6.9MB, pdf)


Articles from Microbiology Spectrum are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES