Skip to main content
Springer Nature - PMC COVID-19 Collection logoLink to Springer Nature - PMC COVID-19 Collection
. 2014 Mar 2;508(7496):402–405. doi: 10.1038/nature13027

Protection against filovirus diseases by a novel broad-spectrum nucleoside analogue BCX4430

Travis K Warren 1, Jay Wells 1, Rekha G Panchal 1, Kelly S Stuthman 1, Nicole L Garza 1, Sean A Van Tongeren 1, Lian Dong 1, Cary J Retterer 1, Brett P Eaton 1, Gianluca Pegoraro 1, Shelley Honnold 1, Shanta Bantia 2, Pravin Kotian 2, Xilin Chen 2, Brian R Taubenheim 2,4, Lisa S Welch 1, Dena M Minning 3, Yarlagadda S Babu 2, William P Sheridan 2, Sina Bavari 1,
PMCID: PMC7095208  PMID: 24590073

A broad-spectrum antiviral small molecule is reported to act as an inhibitor of viral polymerase activity and is shown to be effective in protecting non-human primates from lethal filovirus infection when administered after exposure.

Supplementary information

The online version of this article (doi:10.1038/nature13027) contains supplementary material, which is available to authorized users.

Subject terms: Pharmaceutics, Antivirals, Marburg virus, Ebola virus

An antiviral active against Ebola virus

Viruses of the Filoviridae family can cause severe haemorrhagic fever in humans and non-human primates. Mortality rates are extremely high and no vaccines or drugs are currently licensed for the treatment of filovirus diseases. Here Sina Bavari and colleagues report the discovery of a small-molecule viral polymerase inhibitor with in vitro and in vivo antiviral activity against highly pathogenic viruses, including filoviruses such as Ebola virus and Sudan virus. The compound, BCX4430, is an adenosine analogue that acts as a non-obligate chain terminator. Administered either orally or intramuscularly, it can completely protect cynomolgus macaques from Marburg virus, even when administered as late as 48 hours after infection.

Supplementary information

The online version of this article (doi:10.1038/nature13027) contains supplementary material, which is available to authorized users.

Abstract

Filoviruses are emerging pathogens and causative agents of viral haemorrhagic fever. Case fatality rates of filovirus disease outbreaks are among the highest reported for any human pathogen, exceeding 90% (ref. 1). Licensed therapeutic or vaccine products are not available to treat filovirus diseases. Candidate therapeutics previously shown to be efficacious in non-human primate disease models are based on virus-specific designs and have limited broad-spectrum antiviral potential. Here we show that BCX4430, a novel synthetic adenosine analogue, inhibits infection of distinct filoviruses in human cells. Biochemical, reporter-based and primer-extension assays indicate that BCX4430 inhibits viral RNA polymerase function, acting as a non-obligate RNA chain terminator. Post-exposure intramuscular administration of BCX4430 protects against Ebola virus and Marburg virus disease in rodent models. Most importantly, BCX4430 completely protects cynomolgus macaques from Marburg virus infection when administered as late as 48 hours after infection. In addition, BCX4430 exhibits broad-spectrum antiviral activity against numerous viruses, including bunyaviruses, arenaviruses, paramyxoviruses, coronaviruses and flaviviruses. This is the first report, to our knowledge, of non-human primate protection from filovirus disease by a synthetic drug-like small molecule. We provide additional pharmacological characterizations supporting the potential development of BCX4430 as a countermeasure against human filovirus diseases and other viral diseases representing major public health threats.

Supplementary information

The online version of this article (doi:10.1038/nature13027) contains supplementary material, which is available to authorized users.

Main

Members of the family Filoviridae include Ebola virus (EBOV), Marburg virus (MARV), Ravn virus (RAVV), Sudan virus (SUDV) and Bundibugyo virus (BDBV), all of which cause severe viral haemorrhagic fevers in humans. In nature, filoviruses are transmitted by physical contact between infected individuals, presumably via infected bodily fluids2. Initial filovirus disease manifestations include fever, headache, vomiting and diarrhoea3. Fatal cases are characterized by viraemia, elevated liver-associated enzyme levels, coagulopathy and haemorrhage. Filovirus disease outbreaks occur sporadically, most frequently in sub-Saharan Africa, with reported case fatality rates exceeding 90% (ref. 1). In 2012, simultaneous outbreaks involved MARV, SUDV and BDBV, an emergent filovirus isolated in 20074. The historical occurrence of independent and simultaneous emergence of distinct filoviruses highlights the need for the identification and development of an efficacious broad-spectrum antiviral product.

Although no licensed antiviral preventative or therapeutic agents are currently available to combat filovirus disease in humans, a number of candidates are being developed and have been evaluated in non-human primate filovirus disease models. These models closely reproduce the known clinical and pathophysiological aspects of fatal human infection. Nucleic-acid-based products, antibody therapies and therapeutic vaccines have successfully protected non-human primates from filovirus disease5,6,7,8,9,10, but these approaches rely on virus-specific designs that inherently limit the spectrum of activity and potential utility of individual treatments. The development of a single therapeutic agent active against multiple filoviruses would provide a key, cost-effective component of public-health preparedness plans in outbreak-prone regions.

The broad-spectrum antiviral agent ribavirin, a trizole nucleoside effective against multiple pathogenic RNA viruses, is not active against filoviruses11. Other small molecules—including the adenosine analogue 3-deazaneplanocin A (c3-Npc A) and T-705 (favipiravir), a substituted pyrazine compound —have conferred a high-degree of protection against filoviruses in rodents but have not been reported to protect non-human primates12,13,14,15,16,17,18.

BCX4430, a novel nucleoside analogue (Fig. 1a), was synthesized (Supplementary Information) as part of a small-molecule library designed as inhibitors of viral RNA polymerase activity. BCX4430 is designed to inhibit viral RNA polymerase activity indirectly through non-obligate RNA chain termination, a mechanism requiring anabolism of the parent compound to BCX4430-triphosphate (BCX4430-TP). Then, after pyrophosphate cleavage, incorporation of BCX4430-monophosphate (BCX4430-MP) into nascent viral RNA strands would be expected to cause premature termination of transcription and replication of viral RNA. In support of this proposed mechanism, BCX4430 is rapidly phosphorylated to BCX4430-TP in cultured cell lines and primary hepatocytes, similar to the natural adenosine nucleoside (Fig. 1b and Extended Data Fig. 1a). Addition of BCX4430 reduces expression of green fluorescent protein (GFP) in an artificial EBOV minigenome replicon assay (Fig. 1c and Extended Data Fig. 1b), in which virion structural proteins comprising the viral ribonucleoprotein complex mediate transcription and replication of an RNA replicon template containing a GFP-reporter cassette. BCX4430-TP inhibits hepatitis C virus (HCV) RNA polymerase transcriptional activity in a cell-free, isolated enzyme transcription assay (Fig. 1d) (an isolated filovirus RNA polymerase enzyme assay has yet to be reported) and induces premature termination of RNA chain synthesis by HCV RNA polymerase during template-directed primer-extension assays (Fig. 1e and Extended Data Fig. 1c). In virus-infected cells, BCX4430 reduces surface-expressed MARV and EBOV glycoprotein and reduces the production of intracellular and extracellular MARV RNA (Fig. 1f–h and Extended Data Fig. 1d). Additionally, HeLa cells incubated with ≥25 µM BCX4430 produce no detectable infectious MARV virus (concentration providing 90% inhibition (IC90) = 5.4 µM) (Fig. 1h).

Figure 1. Pharmacological characterization of BCX4430.

Figure 1

a, BCX4430 chemical structure and properties. LogP, log of partition coefficient. b, Conversion of 3H-BCX4430 or 3H-adenosine to diphosphate (DP) or triphosphate (TP) forms in Huh-7 cells (n = 1). c, Effect of BCX4430 on replication of an EBOV minigenome RNA replicon in cultured cells (n = 6). No L, l-polymerase encoding plasmid omitted from reaction. d, BCX4430-TP suppresses HCV NS5B (NS5B-1b-Δ21) RNA polymerase activity (n = 2). e, RNA products synthesized by purified HCV polymerase, in a template-directed primer (32P-5′-GG) extension assay. nt, nucleotides. f, Expression of EBOV and MARV glycoprotein in infected HeLa cells (n = 5–6). g, MARV-infected HeLa cells. Green, α-MARV glycoprotein; blue, Hoechst dye. Scale bar, 100 µm. h, Production of intracellular MARV RNA (n = 4) and infectious virus in MARV-infected HeLa cells (n = 2 technical replicates). Data in c, d, and h are expressed as mean + standard deviation (s.d.). Data in f are expressed as mean + standard error of the mean (s.e.m.).

PowerPoint slide

Source data

Extended Data Figure 1. Phosphorylation and antiviral mechanism of action of BCX4430.

Extended Data Figure 1

a, Conversion of BCX4430 to the active triphosphate (TP) form in cultured cell lines and fresh primary hepatocytes (n = 3–6). Right axis, values normalized to mean 24 h value for human hepatocytes. b, Expression of EBOV NP and VP35 in an EBOV minigenome RNA replicon assay, in BHK-21-derived cells (n = 6). Right three lanes, plasmids expressing the indicated viral protein were omitted from the transfection mix. Gel image cropped for clarity. c, RNA products synthesized by purified HCV polymerase, in a template-directed primer (32P-5′-GG) extension assay. d, Production of intra- and extracellular MARV RNA and cell-surface expression of viral glycoprotein in MARV-infected HeLa cells (n = 4) treated with BCX4430 either 18 h before infection, or 1, 12 or 24 h after infection. e, Expression of EBOV glycoprotein in monocyte-derived primary human macrophages (n = 4). f, Incorporation of 3H-BCX4430 (3H-4430) or 3H-adenosine (3H-AD) in human Huh-7 cells after 24 h incubation (n = 1). CPM, counts per min. Percentage inhibition assessed against the average of medium-only infection-control wells. Data in a are expressed as mean ± s.d. Data in d are expressed as mean + s.e.m. Data in e are expressed as mean ± s.e.m.

Source data

Taken together, these assessments strongly support our hypothesis that BCX4430 inhibits viral RNA polymerase function by inducing RNA chain termination. Findings from primer-extension reactions suggest that termination occurs two bases after incorporation of BCX4430-MP, perhaps as a result of inhibitory stereochemical distortions of the nascent RNA chain. We observed no evidence of BCX4430-MP incorporation into human RNA or DNA on exposing human Huh-7 cells to concentrations of BCX4430 exceeding the MARV IC50 by more than tenfold (Extended Data Fig. 1f). The basis of the selectivity of BCX4430 for viral polymerases is not yet known.

To assess the spectrum of antiviral activity of BCX4430, we conducted additional high-content image (HCI)-based and neutral-red uptake antiviral assays using an array of other human viruses. BCX4430 exhibited antiviral activity against negative-sense RNA viruses representing the Filoviridae, Arenaviridae, Bunyaviridae, Orthomyxoviridae, Picornaviridae and Paramyxoviridae families, and positive-sense RNA viruses of the Flaviviridae and Coronaviridae families (Table 1 and Extended Data Fig. 2). Antiviral effects were specific and did not result from cellular toxicity or anti-proliferative effects, as assessed by enumeration of live cells during all HCI assays and by the lactate dehydrogenase (LDH) release assay. In HeLa cells, the 50% cytotoxic concentration (CC50) of BCX4430 exceeded 500 µM as determined by the LDH assay, and consistently exceeded 100 µM using the HCI-based assay (Table 1). An antiviral effect was confirmed in infected human macrophages cultured from normal peripheral blood monocytes, using EBOV (Extended Data Fig. 1e). It is important to note that immortalized cell lines exhibit a substantially attenuated capacity to convert BCX4430 to its active TP form, compared with primary hepatocytes (Extended Data Fig. 1a). Thus, the results obtained from antiviral assessments relying on immortalized cells may underrepresent the antiviral potency of BCX4430 achievable in vivo during experimental or clinical infections.

Table 1.

BCX4430 antiviral activity

Virus family Virus Strain/variant EC50 (µM) EC90 (µM) CC50 (µM)
Filoviridae MARV Musoke* 4.4 10.5 242
MARV Ci67* 6.7 16.1 255
MARV Angola* 5.0 12.3 242
EBOV Kikwit* 11.8 25.4 >100
SUDV Boniface* 3.4 10.3 >100
Togaviridae VEEV SH3* >100 >100 >100
EEEV FL93-939† 43.2 >100 >100
WEEV California† 21.3 >30 >100
CHIKV AF 15561* >100 >100 >100
Bunyaviridae RVFV ZH5018* 41.6 98.0 >100
LACV Wisconsin 1960† 13.4 65.0 >100
MPRLV HV97021050† 40.1 95.0 >250
Arenaviridae LASV Josiah* 43.0 >100 >100
JUNV Romero* 42.2 >100 >100
Paramyxoviridae NiV Malaysia* 41.9 >100 >100
RSV 2305† 11.0 25.7 >89
MeV Chicago† 6.19 34.4 >296
Coronaviridae MERS-CoV Jordan N3* 68.4 >100 >100
SARS-CoV Urbani† 57.7 >95 >296
Orthomyxoviridae Influenza pH1N1† 10.7 17.0 >296
Picornaviridae HRV2 HGP† 3.4 45.2 >296
Flaviviridae YFV 17D† 14.1 46.8 >100
JEV SA14† 43.6 93.4 >100
DENV2 New Guinea C† 32.8 89.3 >296

EC50, 50% effective concentration; EC90, 90% effective concentration. Definitions for virus abbreviations are provided in Methods.

*Antiviral activity assessed by high-content image analysis.

†Antiviral activity assessed by neutral-red uptake assay.

Extended Data Figure 2. Broad-spectrum antiviral activity of BCX4430.

Extended Data Figure 2

Antiviral activity was assessed in cell-based assays (n = 3–5; n = 2, MERS-CoV), either using high-content image-based analysis or neutral-red uptake, using cell lines described in Methods. Cells were pre-treated with BCX4430 for ∼18 h before infection. Definitions of virus abbreviations are provided in Methods. Except for the top row, viruses are arranged in rows by taxonomic family. Percentage inhibition of BCX4430-treatment wells was assessed against the average of medium-only infection-control wells. Negative inhibition values were transformed to zero for curve fit analysis and display. Data are expressed as mean + s.e.m.

Source data

In preparation for in vivo pharmacokinetics and efficacy testing, we sought to further characterize potential adverse pharmacological features of BCX4430 that could limit its utility in vivo or clinically. BCX4430 exhibited no mutagenicity when tested using the Ames assay at 5 mg per plate, produced no detectable chromosomal aberrations in human lymphocytes at a concentration of 787 µM, and did not inhibit the hERG ion channel current at 30 µM (data not shown). The compound is metabolically stable (half-life (t1/2) > 54 min) when incubated with S9 liver fractions obtained from various animal species (Extended Data Table 1). In mouse, rat, guinea pig and cynomolgus macaque, BCX4430 pharmacokinetics were characterized by rapid clearance from the plasma with a half-life of <5 min (Fig. 2b, Extended Data Fig. 4c and Extended Data Table 2). By contrast, the half-life of the active BCX4430-TP form in the liver in rats was substantially longer at 6.2 h (Fig. 2b).

Extended Data Table 1.

In vitro metabolic stability of BCX4430 in liver S9 fractions

graphic file with name 41586_2014_Article_BFnature13027_Figa_ESM.jpg

*Percentage remaining of test compound was calculated based on the peak area ratio of the test compound to the internal standard by LC-HRMS. Performance of positive biocontrol reagents testosterone and 7-hydroxycoumarin was assessed in parallel and met assay acceptability specifications.

†Half-life was calculated as t1/2 = 0.693/k, where k is the elimination rate constant in the equation describing first-order decay (Ct = C0 × ekt), and Ct and C0 are the peak area ratios at time t and time 0, respectively. Data points were fitted to a first-order decay model by nonlinear regression, using GraphPad Prism (version 5.04 or higher) without weighting or any user intervention.

‡Intrinsic clearance (CLint) was calculated based on CLint = k/P, where k is the elimination rate constant and P is the protein concentration in the incubation.

§Expected metabolite(s) detected.

Figure 2. BCX4430 in vivo efficacy and pharmacokinetics characterization.

Figure 2

a, Survival of RAVV-infected mice (n = 9–10 per group). BCX4430 treatments (Tx) were administered i.m. twice daily at 150 mg kg−1 doses for 9 days beginning at the indicated time before infection (BI) or post-infection (PI). b, Pharmacokinetics of BCX4430 and BCX4430-TP in Sprague–Dawley rats (n = 3) after single-dose i.m. administration of BCX4430. *P < 0.05 treatment versus vehicle survival curves by log-rank (Mantel–Cox) test. Data in b are expressed as mean ± s.e.m.

PowerPoint slide

Extended Data Figure 4. In vivo activity of BCX4430 in guinea pigs and cynomolgus macaques.

Extended Data Figure 4

a, b, Survival of guinea pigs (n = 8 per group) infected by i.p. injection with MARV-Musoke (a) or by exposure to aerosolized MARV-Angola (b). BCX4430 (i.m., 50 mg kg−1 twice daily) treatments (Tx) began at the indicated times before infection (BI) or post-infection (PI). c, Pharmacokinetics of BCX4430 in guinea pigs and cynomolgus macaques (n = 3) after single-dose i.m. administration. d, Individual animal maximal values of interferon-α2a in MARV-infected cynomolgus macaques. *P < 0.05 for comparison of treatment versus vehicle survival curves by log-rank (Mantel–Cox) test. Data in c are expressed as mean ± s.e.m. Horizontal bars in d represent group means.

Extended Data Table 2.

BCX4430 and BCX4430-TP pharmacokinetics

graphic file with name 41586_2014_Article_BFnature13027_Figb_ESM.jpg

Pharmacokinetics parameters were calculated from plasma BCX4430 and liver BCX4430-TP levels. Liver BCX4430-TP Cmax and area under the curve (AUC) parameters are expressed in parent drug equivalents.

Rodent models of filovirus disease do not always reliably reproduce the pathophysiology observed in humans and non-human primates. Nevertheless, rodent systems can provide proof-of-concept efficacy and inform dose and treatment-regimen selection for non-human primate efficacy trials. In RAVV-infected mice, intramuscular (i.m.) administration of BCX4430 conferred a high degree of protection (75–100%) in groups treated prophylactically at doses ≥3.3 mg kg−1 (Extended Data Fig. 3a, b). Notably, significant protection (P < 0.05) was also observed with doses of 150 mg kg−1 initiated at times as long as 96 h after infection (Fig. 2a). Additionally, we demonstrated that mice administered BCX4430 either i.m. or orally were protected against lethal EBOV challenge and that mice receiving i.m. treatments were protected against lethal Rift Valley fever virus challenge (Extended Data Fig. 3c, d).

Extended Data Figure 3. Efficacy of BCX4430 in mouse disease models.

Extended Data Figure 3

a, b, BCX4430 dose versus survival of RAVV-infected mice (a, n = 9–10). BCX4430 treatments (Tx) were administered for 9 days beginning ∼4 h before infection. Numbers in panel a indicate mg kg−1 doses. c, Survival of mice (n = 10) infected with EBOV. BCX4430 was administered twice daily i.m. or orally at a dose of 150 mg kg−1. d, Survival of mice (n = 10) infected with RVFV. BCX4430 was administered twice daily at doses of 5–150 mg kg−1 by i.m. injection. *P < 0.05 for comparison of treatment versus vehicle survival curves by log-rank (Mantel–Cox) test.

The in vivo post-exposure efficacy of BCX4430 was further verified in two guinea pig models of MARV disease19 in which animals were challenged either by intraperitoneal (i.p.) injection, or, because filoviruses can be transmitted via aerosolized particles20, by exposure to aerosolized virus. In both the i.p. and aerosolized-virus challenge models, BCX4430 conferred significant post-exposure protection when treatment was initiated within 48 h of virus exposure (Extended Data Fig. 4a). In the respiratory exposure model, BCX4430 protected guinea pigs when treatment initiation was delayed to 72 h after infection, the longest delay tested (Extended Data Fig. 4b).

We further explored the post-exposure efficacy of BCX4430 using the cynomolgus macaque MARV disease model, which accurately reproduces filovirus disease manifestations observed in fatal human cases21. Cynomolgus macaques were experimentally infected with a lethal dose of wild-type MARV (Musoke variant), derived from a human clinical isolate22, and animals were administered with 15 mg kg−1 BCX4430 twice daily by i.m. injection beginning 1–48 h after infection and continuing for 14 days. The six infection-control subjects succumbed by day 12 (Fig. 3a), after having developed viraemia (Fig. 3b) and characteristic signs of filovirus disease, including behavioural inactivity, maculopapular rash, increases in liver injury markers such as aspartate aminotransferase and bilirubin (Fig. 3c, d), and prolongations of prothrombin time (PT) and activated partial thromboplastin time (aPTT) (Fig. 3e, f). All animals treated with BCX4430 beginning 24 or 48 h after infection survived. Five out of six (83%) animals treated with BCX4430 beginning 1 h after infection survived (Fig. 3a). Consistent with the proposed antiviral mechanism of action, BCX4430 significantly and substantially reduced serum MARV burden (Fig. 3b), without inducing type I interferon responses (Extended Data Fig. 4d), as has been observed for c3-Npc A in mice15. Additionally, BCX4430 ameliorated haemorrhagic disease manifestations, as evidenced by shorter PT and aPTT times and reduced laboratory indices of liver damage, such as serum AST and bilirubin concentrations, compared with vehicle treatment (Fig. 3c–f).

Figure 3. Post-exposure protection of MARV-infected cynomolgus macaques by BCX4430.

Figure 3

af, Animals (n = 6) were challenged with MARV by subcutaneous injection, and BCX4430 (Tx) (15 mg kg−1 twice daily) or vehicle was administered i.m. beginning at the indicated times after challenge. a, Kaplan–Meier survival curves. PI, post-infection. b, Serum viral RNA load. GE, genomic equivalents. cf, Individual animal maximal values of serum aspartate aminotransferase concentrations (c), conjugated bilirubin concentrations (d), PT (e) and aPTT (f). Non-survivors are represented by open symbols. *P < 0.05 for comparison of treatment versus vehicle by log-rank (Mantel–Cox) test (a), two-tailed analyses using the Holm–Sidak method (b), or two-tailed Kruskal–Wallis test followed by Dunn’s post-test comparison (cf). Data in b are expressed as geometric mean + s.d. Horizontal bars in cf represent group means.

PowerPoint slide

Outbreaks in 2012 involving MARV, SUDV and BDBV reported in Uganda and the Democratic Republic of the Congo highlight the urgent need for development of an effective antiviral product to counter filovirus disease. For the first time, to our knowledge, we report the identification of a small molecule with efficacy against filovirus disease in non-human primates. Furthermore, we have provided evidence that BCX4430 exhibits broad-spectrum antiviral activity against other highly virulent RNA viruses (Table 1 and Extended Data Fig. 2). Additional evaluations are in progress to assess the in vivo efficacy of BCX4430 against EBOV and other highly virulent pathogens in non-human primates or other disease models that most closely recapitulate human disease.

BCX4430 was well tolerated, producing no overt signs of systemic toxicity or adverse local reactions in any of the efficacy studies. The substantial efficacy of BCX4430 observed with the i.m. route, which provides high bioavailability and rapid absorption, is conducive for use during outbreaks, potentially enabling administration by individuals lacking advanced medical training. In MARV-infected cynomolgus macaques, we did not observe any diminution of protective effects with delay of BCX4430 treatment initiation up to 48 h after infection, suggesting that even greater delays of treatment, yet to be tested, may also yield significant protective benefit. In preparation for advancing BCX4430 into human phase 1 clinical trials in the United States, additional studies are in progress to support filing of an Investigational New Drug Application.

Methods Summary

BCX4430 was synthesized by BioCryst Pharmaceuticals. Cell-based infection assays were conducted using HCI-based analyses as described previously23 for filoviruses and Rift Valley fever virus. For other viruses, inhibition of virus-induced cytopathic effect (CPE) was assessed in cell-based assays using neutral-red uptake, as described previously24. Replication of EBOV minigenome RNA constructs was assayed by using a plasmid-based reconstituted replication/transcription system25. Inhibition of RNA transcriptional activity was assessed in an isolated HCV polymerase assay26 and inhibition of HCV polymerase RNA synthesis activity was evaluated using a template-driven 32P-GG primer extension assay. Mouse models of EBOV, MARV and RAVV disease have been described previously27,28. Experiments were conducted using mouse-adapted strains of EBOV (Mayinga variant) and RAVV (Ravn variant). A guinea pig MARV-Musoke parenteral challenge model and virus aerosolization methods used for guinea pig exposures have been previously described19,29. BCX4430 was administered in a vehicle of sterile 0.9% saline for injection for all in vivo applications. Animal infection experiments were performed in biosafety level 4 containment facilities at the United States Army Medical Research Institute of Infectious Diseases (USAMRIID). All experimental treatment replication was conducted using biological replication, except for the virus-yield reduction assay (Fig. 1h), which relied on technical replication.

Research was conducted under an Institutional Animal Care and Use Committee approved protocol in compliance with the Animal Welfare Act, PHS Policy and other federal statutes and regulations relating to animals and experiments involving animals. The facility where this research was conducted is accredited by the Association for Assessment and Accreditation of Laboratory Animal Care, International and adheres to principles stated in the Guide for the Care and Use of Laboratory Animals, National Research Council, 2011.

Supplementary information

Supplementary Information (6.6MB, pdf)

This file contains Supplementary Methods describing the synthesis of BCX4430 and the analytical data that support the synthetic steps, Supplementary Figures 1-29 and additional references. (PDF 6744 kb)

Acknowledgements

J. Kuhn and J. Huggins provided insightful discussions and critically reviewed the manuscript. R. Kincaid and G. Feuerstein provided advice and guidance for BCX4430 development efforts. These studies were in part supported by The Joint Science and Technology Office for Chemical and Biological Defense of the Defense Threat Reduction Agency (proposal #TMTI0048_09_RD_T and CB3675) to S. Bavari. S. Radoshitzky assisted with the EBOV minigenome replicon assay. J. Reifman was essential to algorithm development of HCI image assessments. C. Basler provided the BHK-21-derived cell line constitutively expressing the T7 RNA polymerase. Plasmids encoding viral products and the EBOV minigenome replicon were provided by P. Kranzusch and S. Whelan. Neutral-red uptake antiviral assays were conducted by: D. L. Barnard, G. W. Day, B. Gowan, J. G. Julander, B. Tarbet, D. F. Smee and J. D. Morrey of Utah State University under National Institute of Allergy and Infectious Diseases (NIAID) contract HHSN272201100019I, BioQual Inc. under NIAID contract HHSN27220110005I, and at the University of Alabama Birmingham under NIAID contract HHSN272201100016I. Cell-based metabolism studies were conducted by C. Parker, X. Cheng, R. Upshaw and Y. Luo. A. Nalca, E. E. Zumbrun, H. Bloomfield, D. Dyer and J. Yeager assisted with virus aerosolization. C. Cooper provided assistance with the culture of human macrophage cell culture and R. Zamani provided assistance with high-content image assessments. S. Tritsch assisted with Nipah virus antiviral assays. Opinions, interpretations, conclusions and recommendations are those of the authors and are not necessarily endorsed by the US Army.

Extended data figures and tables

PowerPoint slides

Source data

Source data to Fig. 1 (15.8KB, xlsx)

Author Contributions

Y.S.B. and P.K. were responsible for the synthesis of BCX4430 and other small molecules. T.K.W. designed and supervised activities associated with rodent and non-human primate efficacy evaluations, evaluated study results, and wrote the manuscript. J.W., K.S.D., N.L.G. and S.A.V.T. conducted the rodent and non-human primate efficacy studies and performed sample analyses. R.G.P., G.P., C.J.R. and B.P.E. designed and executed cell-based filovirus assays and analysed these data. S. Bantia, Y.S.B., D.M.M., W.P.S., B.R.T. and others designed and analysed data from cell-based antiviral assays. L.D. conducted quantitative PCR analysis. B.R.T. conducted statistical evaluations of in vivo study results. S.H. performed post-mortem analysis of all non-human primate subjects. Y.S.B. supervised the pharmacokinetics studies of BCX4430 and W.P.S. conducted pharmacokinetics data analysis. S. Bantia conducted assessments of BCX4430 metabolite analysis and incorporation into host nucleic acids. X.C. conducted chain termination experiments. T.K.W., D.M.M., L.S.W., B.R.T., Y.S.B., W.P.S. and S. Bavari designed experiments, evaluated results and provided project oversight.

Competing interests

S. Bantia, P.K., Y.S.B. and BioCryst Pharmaceuticals, Inc. claim intellectual property regarding BCX4430 for treatment of viral infections. S. Bantia, X.C., P.K., Y.S.B. and W.P.S. are employees of BioCryst Pharmaceuticals, Inc.

References

  • 1.Kuhn JH. Filoviruses: A Compendium of 40 Years of Epidemiological, Clinical, and Laboratory Studies. 2008. [PubMed] [Google Scholar]
  • 2.Bausch DG, et al. Assessment of the risk of Ebola virus transmission from bodily fluids and fomites. J. Infect. Dis. 2007;196:S142–S147. doi: 10.1086/520545. [DOI] [PubMed] [Google Scholar]
  • 3.Kortepeter MG, Bausch DG, Bray M. Basic clinical and laboratory features of filoviral hemorrhagic fever. J. Infect. Dis. 2011;204:S810–S816. doi: 10.1093/infdis/jir299. [DOI] [PubMed] [Google Scholar]
  • 4.Towner JS, et al. Newly discovered Ebola virus associated with hemorrhagic fever outbreak in Uganda. PLoS Pathog. 2008;4:e1000212. doi: 10.1371/journal.ppat.1000212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Geisbert TW, et al. Vesicular stomatitis virus-based vaccines protect nonhuman primates against aerosol challenge with Ebola and Marburg viruses. Vaccine. 2008;26:6894–6900. doi: 10.1016/j.vaccine.2008.09.082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Geisbert TW, et al. Postexposure protection of non-human primates against a lethal Ebola virus challenge with RNA interference: a proof-of-concept study. Lancet. 2010;375:1896–1905. doi: 10.1016/S0140-6736(10)60357-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Warfield KL, et al. Ebola virus-like particle-based vaccine protects nonhuman primates against lethal Ebola virus challenge. J. Infect. Dis. 2007;196:S430–S437. doi: 10.1086/520583. [DOI] [PubMed] [Google Scholar]
  • 8.Warren TK, et al. Advanced antisense therapies for postexposure protection against lethal filovirus infections. Nature Med. 2010;16:991–994. doi: 10.1038/nm.2202. [DOI] [PubMed] [Google Scholar]
  • 9.Dye JM, et al. Postexposure antibody prophylaxis protects nonhuman primates from filovirus disease. Proc. Natl Acad. Sci. USA. 2012;109:5034–5039. doi: 10.1073/pnas.1200409109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Sullivan NJ, Sanchez A, Rollin PE, Yang ZY, Nabel GJ. Development of a preventive vaccine for Ebola virus infection in primates. Nature. 2000;408:605–609. doi: 10.1038/35046108. [DOI] [PubMed] [Google Scholar]
  • 11.Huggins JW. Prospects for treatment of viral hemorrhagic fevers with ribavirin, a broad-spectrum antiviral drug. Rev. Infect. Dis. 1989;11:S750–S761. doi: 10.1093/clinids/11.Supplement_4.S750. [DOI] [PubMed] [Google Scholar]
  • 12.Smither, S. J. et al. Post-exposure efficacy of Oral T-705 (Favipiravir) against inhalational Ebola virus infection in a mouse model. Antiviral Res. 10.1016/j.antiviral.2014.01.012 (2014) [DOI] [PubMed]
  • 13.Aman MJ, et al. Development of a broad-spectrum antiviral with activity against Ebola virus. Antiviral Res. 2009;83:245–251. doi: 10.1016/j.antiviral.2009.06.001. [DOI] [PubMed] [Google Scholar]
  • 14.Bray M, Driscoll J, Huggins JW. Treatment of lethal Ebola virus infection in mice with a single dose of an S-adenosyl-l-homocysteine hydrolase inhibitor. Antiviral Res. 2000;45:135–147. doi: 10.1016/S0166-3542(00)00066-8. [DOI] [PubMed] [Google Scholar]
  • 15.Bray M, Raymond JL, Geisbert T, Baker RO. 3-Deazaneplanocin A induces massively increased interferon-α production in Ebola virus-infected mice. Antiviral Res. 2002;55:151–159. doi: 10.1016/S0166-3542(02)00018-9. [DOI] [PubMed] [Google Scholar]
  • 16.Kinch MS, et al. FGI-104: a broad-spectrum small molecule inhibitor of viral infection. Am. J. Transl. Res. 2009;1:87–98. [PMC free article] [PubMed] [Google Scholar]
  • 17.Panchal RG, et al. Identification of an antioxidant small-molecule with broad-spectrum antiviral activity. Antiviral Res. 2012;93:23–29. doi: 10.1016/j.antiviral.2011.10.011. [DOI] [PubMed] [Google Scholar]
  • 18.Warren TK, et al. Antiviral activity of a small-molecule inhibitor of filovirus infection. Antimicrob. Agents Chemother. 2010;54:2152–2159. doi: 10.1128/AAC.01315-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Hevey M, Negley D, Pushko P, Smith J, Schmaljohn A. Marburg virus vaccines based upon alphavirus replicons protect guinea pigs and nonhuman primates. Virology. 1998;251:28–37. doi: 10.1006/viro.1998.9367. [DOI] [PubMed] [Google Scholar]
  • 20.Kobinger GP, et al. Replication, pathogenicity, shedding, and transmission of Zaire ebolavirus in pigs. J. Infect. Dis. 2011;204:200–208. doi: 10.1093/infdis/jir077. [DOI] [PubMed] [Google Scholar]
  • 21.Bente D, Gren J, Strong JE, Feldmann H. Disease modeling for Ebola and Marburg viruses. Dis. Model. Mech. 2009;2:12–17. doi: 10.1242/dmm.000471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Smith DH, et al. Marburg-virus disease in Kenya. Lancet. 1982;319:816–820. doi: 10.1016/S0140-6736(82)91871-2. [DOI] [PubMed] [Google Scholar]
  • 23.Panchal RG, et al. Development of high-content imaging assays for lethal viral pathogens. J. Biomol. Screen. 2010;15:755–765. doi: 10.1177/1087057110374357. [DOI] [PubMed] [Google Scholar]
  • 24.Barnard DL, et al. Inhibition of measles virus replication by 5′-nor carbocyclic adenosine analogues. Antivir. Chem. Chemother. 2001;12:241–250. doi: 10.1177/095632020101200405. [DOI] [PubMed] [Google Scholar]
  • 25.Mühlberger E, Weik M, Volchkov VE, Klenk HD, Becker S. Comparison of the transcription and replication strategies of Marburg virus and Ebola virus by using artificial replication systems. J. Virol. 1999;73:2333–2342. doi: 10.1128/jvi.73.3.2333-2342.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Lou H, Choi YH, LaVoy JE, Major ME, Hagedorn CH. Analysis of mutant NS5B proteins encoded by isolates from chimpanzees chronically infected following clonal HCV RNA inoculation. Virology. 2003;317:65–72. doi: 10.1016/j.virol.2003.08.038. [DOI] [PubMed] [Google Scholar]
  • 27.Bray M, Davis K, Geisbert T, Schmaljohn C, Huggins J. A mouse model for evaluation of prophylaxis and therapy of Ebola hemorrhagic fever. J. Infect. Dis. 1998;178:651–661. doi: 10.1086/515386. [DOI] [PubMed] [Google Scholar]
  • 28.Warfield KL, et al. Development and characterization of a mouse model for Marburg hemorrhagic fever. J. Virol. 2009;83:6404–6415. doi: 10.1128/JVI.00126-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Alves DA, et al. Aerosol exposure to the Angola strain of Marburg virus causes lethal viral hemorrhagic fever in cynomolgus macaques. Vet. Pathol. 2010;47:831–851. doi: 10.1177/0300985810378597. [DOI] [PubMed] [Google Scholar]

Associated Data

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

Supplementary Materials

Supplementary Information (6.6MB, pdf)

This file contains Supplementary Methods describing the synthesis of BCX4430 and the analytical data that support the synthetic steps, Supplementary Figures 1-29 and additional references. (PDF 6744 kb)


Articles from Nature are provided here courtesy of Nature Publishing Group

RESOURCES