Globally, hepatitis E virus (HEV) causes significant morbidity and mortality each year. Despite this burden, there are no specific antivirals available to treat HEV patients, and the only licensed vaccine is not available outside China.
KEYWORDS: broad-spectrum antivirals, direct-acting antivirals, hepatitis E virus, hepatitis therapy development
ABSTRACT
Globally, hepatitis E virus (HEV) causes significant morbidity and mortality each year. Despite this burden, there are no specific antivirals available to treat HEV patients, and the only licensed vaccine is not available outside China. Ribavirin and alpha interferon are used to treat chronic HEV infections; however, severe side effects and treatment failure are commonly reported. Therefore, this study aimed to identify potential antivirals for further development to combat HEV infection. We selected 16 compounds from the nucleoside and nonnucleoside antiviral classes that range in developmental status from late preclinical to FDA approved and evaluated them as potential antivirals for HEV infection, using genotype 1 replicon luminescence studies and replicon RNA quantification. Two potent inhibitors of HEV replication included NITD008 (half-maximal effective concentration [EC50], 0.03 μM; half-maximal cytotoxic concentration [CC50], >100 μM) and GPC-N114 (EC50, 1.07 μM, CC50, >100 μM), and both drugs reduced replicon RNA levels in cell culture (>50% reduction with either 10 μM GPC-N114 or 2.50 μM NITD008). Furthermore, GPC-N114 and NITD008 were synergistic in combinational treatment (combination index, 0.4) against HEV replication, allowing for dose reduction indices of 20.42 and 8.82 at 50% inhibition, respectively. Sofosbuvir has previously exhibited mixed results against HEV as an antiviral, both in vitro and in a few clinical applications; however, in this study it was effective against the HEV genotype 1 replicon (EC50, 1.97 μM; CC50, >100 μM) and reduced replicon RNA levels (47.2% reduction at 10 μM). Together these studies indicate drug repurposing may be a promising pathway for development of antivirals against HEV infection.
INTRODUCTION
Hepatitis E virus (HEV) is a clinically important emerging virus, causing significant global morbidity and approximately 20 million infections, 44,000 deaths, and 3,000 stillbirths each year (1, 2). Despite this substantial disease burden, currently no specific antivirals exist for treating HEV infections, and the sole licensed vaccine (Hecolin) is only available in China (3). Ribavirin and pegylated alpha interferon (PEG-IFN-α) are often used as therapies for chronic HEV infections (4–6); however, treatment failure is common (7–9) and accompanied by severe side effects (6, 10). Therefore, further research is needed to identify safe and effective antivirals to treat HEV patients.
Recovery from acute HEV infection is often protracted, with typical hepatitis symptoms persisting for approximately 4 to 6 weeks (11). Fulminant hepatitis from HEV infection can prove fatal, with mortality rates reported between 0.5% and 4%, depending on the geographical region (12–14). Higher mortality rates of up to 27% have been reported in pregnant women (14, 15), with HEV infection being particularly severe in the third trimester and fulminant hepatitis often resulting in stillbirth and maternal death (16, 17). Although HEV infection is usually self-limiting, persistent infections have been frequently reported in immunosuppressed and immunocompromised patients (18–22).
HEV is classified within the Hepeviridae family, which is thought to have arisen from an ancient recombination event between viruses from different positive-sense RNA superfamilies (23). HEV comprises a single-stranded RNA genome, encapsidated within a 27- to 34-nm icosahedral, nonenveloped virion (24). The HEV genome is around 7.2 kb, with a 5′ cap and a 3′ polyadenylated tail (25, 26). The genome consists of three open reading frames (ORFs), with ORF1 encoding the nonstructural proteins and ORF2 the capsid protein, while ORF3 is a phosphoprotein thought to act as a viroporin to facilitate viral release from the host cell (27, 28).
Every stage of the HEV replication cycle can be exploited for antiviral design, as many of the nonstructural proteins are essential for viral replication, including the RNA-dependent RNA polymerase (RdRp), which makes them ideal antiviral targets. Therapies targeting the RdRp have proven highly successful, with several hepatitis C virus (HCV) antivirals being FDA approved for treatment (reviewed in reference 29).
The RdRp is highly conserved across all RNA viral families, forming the canonical protein structure resembling a closed right hand, with finger, palm, and thumb domains (30, 31). Two classes of antivirals are employed to target the RdRp, nucleoside analogs (NAs) and nonnucleoside inhibitors (NNIs).
HEV is a largely understudied virus, and antiviral development has been hampered by poor viral replication levels in cell culture systems (32) and difficulties in purifying the viral polymerase in its active form (25, 33). As such, several HEV replicons have been constructed from various infectious clones (34–38), which have allowed for effective preclinical screening of antiviral candidates (39–41).
As many antivirals have been successfully developed against other viruses, such as HCV, repurposing these compounds as potential therapies against emerging infections such as HEV should be considered. This study aimed to identify broad-spectrum antiviral candidates to combat HEV infection. All compounds examined in this study were previously developed against other viruses (Table 1) but have not been reported against HEV before, with the exception of sofosbuvir. We employed a subgenomic replicon approach to screen 16 compounds belonging to the NA or NNI classes of antivirals and identified three potent compounds that were profiled for dose-responsiveness and cytotoxicity, two of which were further examined for combinational synergism. The potent compounds identified in this work provide a promising platform for the development of antivirals to treat HEV infections.
TABLE 1.
Antiviral compounds examined in this study
| Compound class/name | Chemical structure | Molecular mass (g/mol) | Original target virus | RdRp binding site | Developmental stage | Reference |
|---|---|---|---|---|---|---|
| Nonnucleoside inhibitors | ||||||
| Beclabuvir (BMS-791325) | ![]() |
659.8 | Hepatitis C virus | Thumb I | Phase II clinical trials | 81 |
| Dasabuvir (ABT-333) | ![]() |
493.6 | Hepatitis C virus | Palm I | FDA approved | 82 |
| Filibuvir (PF868554) | ![]() |
503.6 | Hepatitis C virus | Thumb II | Halted after phase II trials | 83 |
| GPC-N114 | ![]() |
436.8 | Picornaviruses | RNA channel | Preclinical | 46 |
| JTK-109 | ![]() |
638.1 | Hepatitis C virus | Thumb I | Halted after phase II trials | 84 |
| Lomibuvir (VX-222) | ![]() |
445.6 | Hepatitis C virus | Thumb II | Halted after phase II trials | 85 |
| Nesbuvir (HCV-796) | ![]() |
446.5 | Hepatitis C virus | Palm II | Halted after phase II trials | 86 |
|
Setrobuvir
(ANA-598) |
![]() |
560.6 | Hepatitis C virus | Palm I | Halted after phase II trials | 87 |
| Tegobuvir (GS-9190) | ![]() |
517.4 | Hepatitis C virus | Palm β | Halted after phase II trials | 88 |
| TMC-647055 | ![]() |
606.7 | Hepatitis C virus | Thumb I | Halted after phase II trials | 59 |
| Triazavirin | ![]() |
228.2 | Influenza | NDb | Preclinical | 89 |
| Nucleoside analogs | ||||||
| 2’-C-Methylcytidinea (2CMC) | ![]() |
257.2 | Hepatitis C virus | Active site | Halted after phase II clinical trials | 90 |
| 7-deaza-2′-C-methyladenosine (7DMA) | ![]() |
280.3 | Hepatitis C virus | Active site | Preclinical | 91 |
|
Favipiravir
(T705) |
![]() |
157.1 | Influenza | Active site | FDA approved | 92 |
| NITD008 | ![]() |
290.28 | Dengue virus | Active site | Preclinical | 45 |
|
Sofosbuvir
(PSI-7977) |
![]() |
529.5 | Hepatitis C virus | Active site | FDA approved | 93 |
| Other | ||||||
| Quercetagetin | ![]() |
318.23 | Herpesviruses | NDb | Preclinical | 94 |
Positive control compound.
ND, not determined.
RESULTS
Polymerase-targeted antivirals are examined for anti-HEV activity.
Sixteen compounds (Table 1) were selected for examination of HEV replication inhibition and screened at a fixed concentration of 10 μM against the HEV human genotype 1 (G1) replicon pSK-HEV-2-Luc (Fig. 1A). The NA 2′-C-methylcytidine (2CMC) was used as a positive control to demonstrate inhibition of HEV replication (Fig. 1B). Antiviral efficacy was quantified by the relative luminescence of treated cells (test compounds at 10 μM) compared to that of mock-treated cells (vehicle control; 0.5%, vol/vol, dimethyl sulfoxide [DMSO]). Four antivirals, including NITD008, GPC-N114, sofosbuvir, and dasabuvir, demonstrated more than 50% inhibition at 10 μM (Fig. 1B). These four antivirals were selected for further examination to ascertain effective dose and cytotoxicity profiles, while the other 12 compounds did not reach the threshold of >50% inhibition at 10 μM and were dropped from further investigation in this study.
FIG 1.
Screening of broad-spectrum antivirals against HEV G1 replication. (A) A schematic representation of the HEV genome (top) is shown for comparison with the subgenomic replicon (bottom), with nonstructural proteins encoded by ORF1 and the structural proteins and phosphoproteins encoded by ORF2 and ORF3, respectively. Nucleotide positions, 5′ untranslated regions (UTR), and 3′ polyadenylated tails [poly(A)] are indicated. The HEV G1 subgenomic replicon pSK-HEV-2-Luc (bottom) (38) shows the disruption of the ORF2 capsid gene (nucleotides 5148 to 5816), with the firefly luciferase gene as described in reference 38. (B) An initial screen of 16 broad-spectrum antivirals (Table 1) for inhibitory activities against the human HEV G1 subgenomic replicon pSK-HEV-2-Luc through quantitation of luminescence is shown. All compounds were examined at a fixed concentration of 10 μM, and the percentages of mock-treated HEV replication (compound vehicle only, 0.5%, vol/vol, DMSO) for each compound are plotted. The black horizontal dotted line represents 100% HEV replication (0% inhibition), while the red dotted line represents 50% HEV replication (50% inhibition). The NA positive control 2CMC is used to demonstrate effective inhibition of HEV replication. Mean values ± SEM are shown.
Three antivirals exhibit dose-dependent inhibition of HEV replication without cytotoxic effects.
As NITD008, GPC-N114, sofosbuvir, and dasabuvir were identified as potential HEV antivirals in the initial screen (Fig. 1B), these compounds were further examined for their effects on cell viability and dose-dependent inhibition of HEV replicon replication.
NITD008 was examined over a concentration range of 0.02 to 2.50 μM, while GPC-N114, sofosbuvir, and dasabuvir were all examined at 0.16 to 25.00 μM (Fig. 2). HEV replication was assessed using relative luminescence production compared to that of mock-treated cells (0.5%, vol/vol, DMSO), while cytotoxicity was quantified using a metabolic activity conversion assay.
FIG 2.
Dose-response curves and cytotoxicity profiles of lead inhibitory compounds against HEV replication. The HEV inhibitory and cytotoxicity effects of four compounds identified in Fig. 1B are shown. Dose-response graphs were generated by quantification of luminescence (red bars, left y axis), and effects on cell viability were examined using a fluorescent resazurin-to-resorufin assay (blue lines, right y axis). The black dotted horizontal lines represent 50% inhibition. The EC50 and CC50 values are shown on the graphs and in Table 2 for compounds NITD008 (0.019 μM to 2.5 μM) (A), GPC-N114 (0.16 μM to 25.0 μM) (B), dasabuvir (0.16 μM to 25.0 μM) (C), and sofosbuvir (0.16 μM to 25.0 μM) (D). The synthesis of HEV replicon RNA was reduced by three broad-spectrum antivirals compared to the mock-treated control (0.5%, vol/vol, DMSO), as quantified by qRT-PCR, using primers to detect the HEV RdRp. Sofosbuvir and GPC-N114 were examined at 10 μM (E), and NITD008 was examined at concentrations of 0.04 to 2.50 μM (F). The NA 2CMC (10 μM) was used as a positive control, and RNA levels were normalized to the housekeeping gene β-actin, while the relative fold expression was calculated using the ΔΔCT method. P < 0.001 (***); P < 0.01 (**); P < 0.05 (*). Mean values ± SEM are shown.
NITD008 was a potent inhibitor of HEV replication, with a half-maximal effective concentration (EC50) of 0.03 μM (Fig. 2A and Table 2). The half-maximal cytotoxic concentration (CC50) was not reached when examined up to 100 μM, indicating a therapeutic index for NITD008 of >3,333 (Table 2).
TABLE 2.
Potency and cytotoxicity of antiviral compounds with efficacy against the HEV G1 replicon
| Test compound | EC50
(μM) (95% CIb ) |
CC50
(μM) (95% CIb ) |
Therapeutic index (CC50/EC50) |
|---|---|---|---|
| Dasabuvir | 1.79 (1.38–2.32) | 12.28 (11.13–13.59) | 6.86 |
| GPC-N114 | 1.07 (0.13–1.35) | >100 | >93 |
| NITD008 | 0.03 (0.02–0.04) | >100 | >3,333 |
| Sofosbuvir | 1.97 (1.58–2.45) | >100 | >51 |
| 2CMCa | 3.04 (1.52–4.51) | >100 | >33 |
Positive control compound.
CI, confidence intervals.
GPC-N114 was also an effective inhibitor of HEV in vitro, giving an EC50 of 1.07 μM (Fig. 2B and Table 2). Huh7 cell viability started to drop at the higher compound concentrations (77.2% of mock-treated cells at 25 μM); however, the CC50 was not reached with concentrations up to 100 μM, giving a therapeutic index of >93.
Dasabuvir demonstrated dose-responsive inhibition of HEV replication with an EC50 of 1.79 μM (Fig. 2C and Table 2); however, evaluation of cell viability indicated that the compound was toxic to Huh7 cells at concentrations over 2.5 μM, with a CC50 of 12.28 μM. This resulted in a poor therapeutic index of 6.86; therefore, dasabuvir was dropped from further investigation in this study.
Sofosbuvir efficacy against HEV G1 and G3 replicons in vitro has already been published previously, with an EC50 of 1.20 μM against G3 and >10 μM against G1 (39, 42). These previously reported results for G1 HEV are in contrast to the findings in this study, as sofosbuvir inhibited the HEV G1 replicon in a dose-dependent fashion with an EC50 of 1.97 μM (Fig. 2D and Table 2), demonstrating results similar to those for the G3 HEV replicon published previously (39). No cytotoxic effects on Huh7 cells were observed up to 100 μM, indicating a therapeutic index of >51 (Table 2).
The positive control 2CMC gave an EC50 value of 3.04 μM (Table 2) against the HEV G1 replicon, compared to an EC50 of 1.60 μM previously reported for the G3 replicon (43), with no effects on cell viability up to 100 μM (therapeutic index, >33) (Table 2).
HEV RNA levels are reduced by NITD008, GPC-N114, and sofosbuvir.
NITD008, GPC-N114, and sofosbuvir all exhibited dose-dependent inhibition of HEV replicon-derived luminescence production without cytotoxic effects (Fig. 2A, B, and D). As such, these three antivirals were examined further for their abilities to reduce HEV replicon RNA levels in vitro. The positive control 2CMC was assessed at 10 μM and compared to mock-treated RNA levels (0.5% DMSO) to demonstrate a reduction in HEV replicon RNA (Fig. 2E and F). 2CMC reduced RNA levels to 43.6% of that of the mock-treated samples, from 7.7 × 105 replicon copies per well to 3.4 × 105 copies per well (Fig. 2E and F). Sofosbuvir (10 μM) reduced HEV RNA levels to 38.5% of that of the mock-treated wells, from 7.7 × 105 replicon copies per well to 3.0 × 105 copies per well, while the NNI GPC-N114 (10 μM) also reduced HEV RNA levels to 42.9% of that of the mock-treated wells, down to 3.3 × 105 copies per well (Fig. 2E).
NITD008 was the most potent inhibitor of HEV replication identified in Fig. 1 and 2 and therefore was investigated at a lower concentration range, from 0.04 to 2.50 μM. NITD008 reduced HEV replicon RNA levels in a dose-dependent fashion, from 7.7 × 105 replicon copies per well in mock-treated samples to 5.3 × 105 per well (69.5% of mock) at 0.04 μM, 4.3 × 105 replicon copies per well (56.1% of mock) at 0.16 μM, and 2.8 × 105 copies per well (36.7% of mock) at 2.50 μM (Fig. 2F).
NITD008 and GPC-N114 exhibited synergism in combination against HEV replication.
As GPC-N114 and NITD008 were the most potent HEV replication inhibitors identified in the replicon luciferase assays (Fig. 2A and B) and also effectively reduced HEV replicon RNA levels (Fig. 2E and F), these two antivirals were selected for combinational studies against HEV replication.
NITD008 is a broad-spectrum chain-terminating adenosine NA, initially developed as an antiviral against dengue virus (DENV), for which the reported EC50 range is 0.64 to 1.60 μM (44, 45). In contrast, GPC-N114 is a picornavirus NNI with an EC50 range of 0.13 to 5.44 μM, demonstrating broad-spectrum antiviral activity against multiple genera from within the Picornaviridae (46). GPC-N114 binds in the RNA channel of picornavirus polymerases (46), while NAs such as NITD008 bind in the polymerase active site for viruses such as DENV (45); therefore, they occupy different binding sites in their original target viruses. For these reasons we assessed combinational synergy against HEV replicon replication levels.
The effects of GPC-N114 (0.16 to 5.00 μM) and NITD008 (0.005 to 0.16 μM) were examined at a 32:1 ratio using the G1 HEV replicon. The Chou-Talalay method (47) was used to analyze the inhibition data, and Compusyn software was employed to generate the combinational isobologram (Fig. 3A). The mean of the combination indices at 50%, 75%, and 90% inhibition of HEV replicon replication was 0.4, indicating synergism of NITD008 and GPC-N114 in combination (Fig. 3A). This allowed for dose reduction indices at 50% inhibition of 20.42 for GPC-N114 and 8.82 for NITD008. The cytotoxic effects of the two drugs were also examined in combination, with no effect on Huh7 cell viability up to 100 μM (Fig. 3B).
FIG 3.
GPC-N114 and NITD008 exhibited synergistic inhibition of HEV replication. GPC-N114 and NITD008 were examined in combination against HEV replication in vitro as quantified by relative luminescence. GPC-N114 (0.16 to 5.00 μM) and NITD008 (0.005 to 0.16 μM) were evaluated in a 32:1 concentration ratio. (A) Isobologram of GPC-N114 and NITD008 in combination. Data were analyzed using the Chou-Talalay method (Compusyn software) with an average combination index of 0.4 over 50% (blue dots), 75% (red squares), and 90% inhibition (green triangles), indicating synergism. (B) Cytotoxicity effects of the two drugs in combination over the same concentration range as that described for panel A were assessed using a fluorescent resazurin-to-resorufin assay. Mean values ± SEM are shown.
DISCUSSION
HEV is a major cause of liver disease across the globe, resulting in significant morbidity and mortality (1, 2, 22). Despite this burden, HEV is a largely understudied virus, and as such, we currently lack specific therapies to combat HEV infection.
The development of safe and effective antivirals to combat HEV is needed to treat both chronically infected patients and also for acute infections, to reduce the lengthy recovery period, and to prevent fulminant hepatitis. Additionally, antivirals could be useful as a prophylactic measure in the case of outbreaks and epidemics, which are often reported in refugee camps (48, 49) and military troops (50–52). Furthermore, if safety and efficacy could be achieved without teratogenic side effects, then treating HEV-infected pregnant women to prevent the significant mortality rates for both mother and child may also be possible.
Current HEV treatment options include the reduction of immunosuppressants for HEV-infected organ transplant patients as the first choice of action, followed by courses of PEG-IFN-α and/or ribavirin (53). The reduction of immunosuppressants is effective in clearing the virus in around 30% of solid-organ transplant cases (22), leaving around 70% of patients requiring further action. PEG-IFN-α cannot be used in most organ donor recipients due to the risk of transplant rejection; therefore, ribavirin is prescribed in the majority of cases (9). Ribavirin monotherapy is usually required for at least 3 months and can result in significant and undesirable side effects, including severe anemia (4, 6). Furthermore, ribavirin and PEG-IFN-α are both contraindicated during pregnancy (54).
Clinical resistance to ribavirin resulting in treatment failure has been associated with several HEV G3 polymerase mutations, including Y1320H, K1383N, and G1634R, and an insertion into the HEV hypervariable region (8, 9, 55). The effects of these RdRp mutations have been studied in vitro using mutant replicons, cell culture of HEV isolates, and deep sequencing, and it was found that HEVs bearing Y1320H and G1634R mutations were still sensitive to ribavirin but were associated with enhanced replicative capacities, while the effects of K1383N mutations could not be elucidated in vitro (9). Further studies using deep sequencing of HEV G3 isolates from chronically infected patients revealed that viral heterogeneity was increased following ribavirin treatment and that the RdRp C-terminal mutation G1634R was particularly associated with ribavirin treatment failure (56).
As the current HEV therapies have several reported issues, ranging from drug resistance to severe side effects, further research is warranted to identify safe and efficacious antivirals to treat HEV patients.
G1 HEV infection results in the highest number of fetal and maternal mortalities, while G3 causes the majority of chronic infections worldwide (22). In addition, G1, G4, and G7 human HEVs have also caused chronic infections in immunocompromised and immunosuppressed patients (18–21), so effective, broad-spectrum therapeutics to combat all human HEV genotypes would be beneficial.
This study evaluated 16 antivirals (Table 1), previously developed against other viruses, for inhibition of HEV replicon replication in vitro. Four of the 16 antivirals examined inhibited the HEV G1 replicon with more than 50% inhibition at 10 μM (Fig. 1B). Of these four antivirals, three exhibited potent, dose-dependent, and nontoxic antiviral activity against HEV replicon replication in Huh7 cells (Table 2), with EC50 values of 0.03 μM for NITD008 (Fig. 2A), 1.10 μM for GPC-N114 (Fig. 2B), and 1.97 μM for sofosbuvir (Fig. 2D). Moreover, the same three antivirals effectively reduced HEV replicon RNA levels (Fig. 2E and F), albeit not as efficiently as was observed in the luciferase assays. However, discrepancies in antiviral efficacy examined by RNA quantification compared to that of luciferase assays are widely reported (57–61), and the trend of inhibition of HEV replication was the same across the two methodologies.
Two HCV NAs, 2CMC and sofosbuvir, have already been evaluated for antiviral efficacy against HEV (43). The HCV developmental drug 2CMC is a chain-terminating NA and has been shown to inhibit the G3 HEV replicon in vitro, with an EC50 of 1.6 μM (43). However, development of the oral prodrug for 2CMC (valopicitabine) was halted following reports of undesirable side effects, and further development of this NA as an antiviral is now unlikely.
The HCV NA sofosbuvir has also been previously evaluated against HEV G1 and G3 replicons in vitro, with inconsistent reports of antiviral efficacy (39, 42). One study reported efficacy of sofosbuvir against the HEV G3 replicon with an EC50 of 1.2 μM, but it was unexpectedly ineffective against the G1 replicon at concentrations of up to 10 μM (39). However, another study reported sofosbuvir to be ineffective against both G1 and G3 replicons in vitro, even at concentrations as high as 10 μM (42). In both these studies where sofosbuvir had no effect on G1 HEV, the Sar55/S17/luc replicon, which harbors an insertion from the human ribosomal S17 protein within the hypervariable region, was utilized (37, 62). This S17 insertion dramatically enhances replication levels (37) and, as such, likely renders it less susceptible to sofosbuvir inhibition than wild-type viruses and other replicons which lack this insertion, as previously observed with G1634R-associated ribavirin failure (63, 64).
In contrast to previous reports, in the present study using the pSK-HEV2-Luc replicon, we show that sofosbuvir can inhibit HEV G1 replication in vitro, and further investigation as a potential HEV antiviral may be warranted if known mutations associated with enhanced replication are absent. The discrepancies between the observed effects of sofosbuvir against HEV G1 in this study compared to data from previous studies may also be attributed with the use of a stable replicon cell line versus the transient replicon. However, previous studies using both stable and transient HEV replicons to assess the antiviral effects of IFN-α or ribavirin have revealed very similar results, indicating that these systems are comparable for screening potential antivirals (4, 39). Additionally, sofosbuvir may demonstrate variable antiviral efficacy across the different HEV genotypes, and further preclinical work would be required to ascertain its cross-genotypic activities.
Clinically, sofosbuvir has been used in a few individual cases with or without ribavirin to treat HEV G3-infected patients with variable success (63, 65–68), ranging from a reduction of HEV RNA to undetectable levels (63, 66) to treatment failure or relapse following treatment (65, 67, 68). These mixed reports of efficacy have raised questions around the pursuit of sofosbuvir as an HEV antiviral (69), indicating that further work is required to ascertain its suitability as an anti-HEV therapeutic. Clinical treatment failure of sofosbuvir has been attributed to the HEV phylogenetic subtype, patient immune status (63), and presence of HEV RdRp mutations known to confer increased replication levels that cause ribavirin treatment failure, particularly G1634R (63, 64).
In contrast to sofosbuvir, the antiviral efficacy of GPC-N114 has not been evaluated against HEV replication before. Under preclinical development as a picornavirus antiviral, GPC-N114 has broad-spectrum activity against multiple viruses from the Picornaviridae, with potency ranging from 0.13 μM against human enterovirus 71 to 5.44 μM against mengovirus (46). It has been reported to affect the viral RNA-template duplex binding to the picornavirus RdRp template channel, thereby inhibiting replication (46).
Similarly, NITD008 has also not been previously evaluated as an HEV antiviral. A broad-spectrum NA, NITD008, effectively inhibits the replication of a number of viruses, including the Norwalk (human norovirus) replicon (unpublished data), enterovirus 71 (70, 71), and several flaviviruses, such as DENV (44, 45), HCV (72), tick-borne encephalitis (73), Zika virus (57), and Japanese encephalitis virus (74). Reported potency ranges between 8.7 nM against HCV G2a (72) and 3.31 μM in the tick-borne flavivirus Alkhurma hemorrhagic fever virus (73).
During development of NITD008 against DENV, toxic side effects were observed after 2 weeks of in vivo treatment in rats and dogs (45). This reported NITD008 toxicity might not be clinically significant for a short-term therapeutic course to treat human HEV infection; however, reduced treatment doses may in turn reduce the risk of toxic side effects. As such, NITD008 and GPC-N114 were evaluated together for combinational synergy.
NITD008 and GPC-N114 were synergistic in combinational treatment against HEV G1 replicon-derived luminescence levels (mean combination index of 0.4) (Fig. 3A). This suggests that the two compounds occupy different HEV binding sites, as they do in their original target viruses, and allowed for dose reductions to achieve the same antiviral effect in vitro. Combinational therapy is of great importance to overcome viral resistance to direct-acting antivirals in addition to allowing dose reductions of drugs to reduce side effects. Combinations of at least two therapeutics have been used to successfully treat many HCV and HIV patients (reviewed in references 75–77) and should be considered for HEV antivirals to safeguard against viral evolution that can confer drug resistance.
In this study, we have identified two novel HEV antiviral candidates, NITD008 and GPC-N114, that demonstrated potent antiviral activity and combinational synergy against G1 HEV in vitro. These compounds could provide useful scaffolds for further antiviral development against HEV infection. Additionally, we have shown that in the absence of known polymerase mutations that confer increased replication levels, sofosbuvir demonstrates antiviral efficacy against G1 HEV replicon replication.
Further preclinical evaluation is required for these compounds as potential HEV antivirals before clinical assessment is undertaken. First, it would be prudent to test NITD008, GPC-N114, and sofosbuvir against other human-infecting HEV genotypes (G3, G4, and G7) using replicons and live virus in culture, where available, to ascertain the cross-genotypic activities of these compounds. Second, structure-activity relationship studies for these antivirals may reveal structural moieties that confer antiviral activity, allowing for the development of more potent and less toxic derivatives, particularly in the case of NITD008 as a potential HEV therapeutic. Third, longer-term treatment could be evaluated in cell culture to ascertain toxicity over time and to reveal if any resistance mutations arise. Finally, further assessment of the potential synergy between NITD008 and GPC-N114 against HEV in humanized mice or another animal model, such as swine, may be able to confirm the potential to reduce the required dose for the same nontoxic antiviral effects as those we observed in this study. These compounds represent promising candidates for further HEV antiviral development to combat this pervasive virus.
MATERIALS AND METHODS
Test compounds.
Compounds examined in this study included dasabuvir, sofosbuvir, and favipiravir (MedChemExpress, Monmouth Junction, NJ), GPC-N114 (kind gift from Gerhard Pürstinger [46], formerly of University of Innsbruck, Innsbruck, Austria), JTK-109 (Dalton Pharma Services, Toronto, Canada), lomibuvir (Selleckchem, Houston, TX), nesbuvir and tegobuvir (Haoyuan Chemexpress, Shanghai, China), NITD008 (collaboration with Subhash Vasudevan, Duke-NUS Graduate Medical School, Singapore), quercetagetin and 7-deaza-2’-C-methyladenosine (Santa Cruz Biotechnology, Dallas, TX), filibuvir and setrobuvir (Acme Biosciences, Palo Alto, CA), beclabuvir and TMC-647055 (Taizhou Crene Biotechnology, Zhejiang, China), triazavirin (Mcule, Palo Alto, CA), and 2’-C-methylcytidine (2CMC; Sigma-Aldrich, St. Louis, MO). All compounds were >95% pure and dissolved in 100% dimethyl sulfoxide (DMSO), and they were freshly diluted on the day of the experiment. Details of the 16 compounds examined in this study are shown in Table 1.
HEV replicon.
The HEV pSK-HEV-2-Luc replicon plasmid was a kind gift from Sue Emerson (National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD). The pSK-HEV-2-Luc replicon is based on a human genotype 1 (G1) HEV infectious clone (Sar55; GenBank accession number AF444002.1). The ORF2 capsid gene (nucleotides 5148 to 5816) is disrupted by a luciferase reporter gene (38).
Transcription of HEV replicon.
The HEV replicon plasmid was linearized with BglII and RNA synthesized using mMessage in vitro transcription kits (Ambion, Austin, TX). Unless stated otherwise, 100 μl capping transcription reaction mixtures contained approximately 7.5 μg linearized template, 10 μl of 10× transcription buffer, 50 μl of 2× capping deoxynucleoside triphosphate mix (containing 15 mM ATP, CTP, and UTP, 3 mM GTP, and 12 mM cap analog), 5 μl of 30 mM GTP, and 10 μl of the T7 RNA polymerase mix and were incubated for 2 h at 37°C. Reaction mixtures were then DNase treated for 15 min at 37°C and RNA purified using RNeasy kits (Qiagen, Hilden, Germany). RNA integrity was confirmed using agarose gel electrophoresis and quantified using spectrophotometry before transfection.
Cell culture.
The human hepatoma Huh7 cell line was a kind gift from Mark Douglas (Westmead Institute for Medical Research, Sydney, Australia). Cell culture was carried out as previously described (78). Briefly, Huh7 cells were maintained in Dulbecco’s modified Eagle’s medium (DMEM) (Life Technologies, Carlsbad, CA) supplemented with 10% fetal bovine serum (FBS) (Sigma-Aldrich), 1× GlutaMAX (Life Technologies), 10 mM HEPES buffer (ThermoFisher Scientific, Waltham, MA), and 100 U/ml penicillin-streptomycin (Life Technologies).
Transfection.
Huh7 cells were grown to 80% confluence in 96-well plates, and medium was changed 1 h prior to transfection. HEV RNA transcripts (75 ng/well) were chemically transfected into Huh7 cells using the TransIT-mRNA transfection kit (Mirus Bio LLC, Madison, WI) per the manufacturer’s instructions.
Cytotoxicity assays.
Compound cytotoxicity was assessed using monolayers of Huh7 cells, seeded into 96-well plates at a density of 5,000 cells/well. The next day, fresh medium (without antibiotics) was added 1 h prior to adding compounds. All compounds were freshly diluted in complete DMEM, added to cells, and then incubated for a total of 72 h. Cytotoxicity was quantified using a metabolic conversion assay (CellTiter-Blue; Promega, Madison, WI) per the manufacturer’s instructions, and fluorescence was measured on a FLUOstar Optima microplate reader (BMG Labtech, Ortenberg, Germany). CC50 values were determined by nonlinear regression in GraphPad Prism, version 7.02. Mean values were calculated from quadruplicate data sets from at least two independent experiments.
Luciferase assays.
Antiviral activities of test compounds were examined by adding a fixed concentration (10 μM) or increasing concentrations of each drug to replicon-transfected cells: 0.02 to 2.50 μM for NITD008 and 0.16 to 25.00 μM for GPC-N114, dasabuvir, and sofosbuvir. The NA 2CMC was used as a positive control at 10 μM to demonstrate inhibition of replicon replication (43). Huh7 cells were seeded into 96-well plates at a density of 5,000 cells/well. The next day, fresh antibiotic-free DMEM was added to the cells 1 h prior to transfection. Compounds were freshly diluted in complete DMEM, added to the cells 4 h posttransfection, and incubated for 72 h. Mock-treated cells were incubated with 0.5% (vol/vol) DMSO, the compound vehicle. Replication of the pSK-HEV-2-Luc replicon was determined by luciferase-derived luminescence using Luciferase assay system kits (Promega) per the manufacturer’s instructions. Luminescence was measured on a FLUOstar Optima microplate reader (BMG Labtech). HEV replicon luminescence levels in treated cells were compared to those from mock-treated cells (0.5%, vol/vol, DMSO) to calculate the percentage of HEV replication. All compounds were also tested in the absence of the HEV replicon to ensure that they did not interfere with the luciferase signal. EC50 values were determined by nonlinear regression in GraphPad Prism, version 7.02. Mean values were calculated from quadruplicate data sets from at least two independent experiments.
Combinational compound treatment.
Synergism was calculated using Compsyn software V1.0, which employs the Chou-Talalay method (47). GPC-N114 (concentration range, 0.16 to 5.00 μM) and NITD008 (concentration range, 0.005 to 0.16 μM) were examined alone and together at a 32:1 ratio against the HEV replicon, with luminescence quantified as described above. Data were generated from quadruplicate data sets.
RNA extraction.
Viral and cellular RNA was extracted from transfected Huh7 cell monolayers 72 h posttransfection with TRIzol LS (Invitrogen, Carlsbad, CA, USA) using phase separation per the manufacturer’s instructions. RNA was further purified using the RNeasy Minikit (Qiagen), which included DNA removal using RNase-free DNase (Qiagen). RNA was quantified using spectrophotometry, and RNA integrity was assessed by agarose gel electrophoresis.
HEV RNA level quantitation.
HEV replicon RNA levels were measured from transfected Huh7 cells by quantitative reverse transcriptase PCR (qRT-PCR). Briefly, cDNA was synthesized using a SuperScript VILO cDNA synthesis kit (Life Technologies). Replicon RNA was measured using an iTaq universal SYBR green supermix (Bio-Rad, Hercules, CA) as described previously (79). Samples were normalized to the housekeeping gene β-actin, and the fold change was analyzed using the ΔΔCT method, as described previously (80). HEV-specific primers were used for quantitation targeting the region encoding RdRp and included the forward primer (HEV replicon Fwd, 5′-TGTCCTGATTGCTGGCTGTG-3′) and the reverse primer (HEV replicon Rev, 5′-GAGAAGAATTGGGGCCCTGG-3′). Mean values were calculated from triplicate data sets, and two independent experiments were performed. All statistical calculations were performed using GraphPad Prism software (v7.02). Data were analyzed using an unpaired t test: P > 0.05, P ≤ 0.05 (*), P ≤ 0.01 (**), and P ≤ 0.001 (***).
ACKNOWLEDGMENTS
We thank Suzanne Emerson (National Institute of Allergy and Infectious Diseases, NIH) and X. J. Meng (Virginia-Maryland College of Veterinary Medicine) for generously providing the plasmid to generate the G1 HEV replicon used in this study. We also thank Mark Douglas (Westmead Institute for Medical Research, Sydney, Australia) for providing the Huh7 cell line used in this study. We thank Kitti Wing-Ki Chan (Emerging Infectious Diseases Program, Duke-NUS Medical School, Singapore) for her assistance during our collaboration. Finally, we thank Gerhard Pürstinger (formerly of Institute of Pharmacy, University of Innsbruck, Austria) for providing the compound GPC-N114. This work was funded by the National Health and Medical Research Council, grant/award numbers APP1083139 and APP1123135. N.E.N. and P.A.W. conceived the study, D.E.T. assisted N.E.N. with experimental design, and N.E.N. designed and performed the experiments under the supervision of P.A.W. S.G.V. and J.M.M. provided expertise and materials. N.E.N. and P.A.W. analyzed and interpreted the data. N.E.N. wrote the manuscript with the help of P.A.W., and it was edited by D.E.T., S.G.V., and J.M.M. We have no conflicts of interest to declare and nothing to disclose.
REFERENCES
- 1.Rein DB, Stevens GA, Theaker J, Wittenborn JS, Wiersma ST. 2012. The global burden of hepatitis E virus genotypes 1 and 2 in 2005. Hepatology 55:988–997. doi: 10.1002/hep.25505. [DOI] [PubMed] [Google Scholar]
- 2.World Health Organization. 19 September 2018. Hepatitis E fact sheet. http://www.who.int/news-room/fact-sheets/detail/hepatitis-e. World Health Organization, Geneva, Switzerland: Accessed 21 October 2018. [Google Scholar]
- 3.Park SB. 2012. Hepatitis E vaccine debuts. Nature 491:21–22. doi: 10.1038/491021a. [DOI] [PubMed] [Google Scholar]
- 4.Debing Y, Neyts J. 2014. Antiviral strategies for hepatitis E virus. Antiviral Res 102:106–118. doi: 10.1016/j.antiviral.2013.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Alric L, Bonnet D, Laurent G, Kamar N, Izopet J. 2010. Chronic hepatitis E virus infection: successful virologic response to pegylated interferon-alpha therapy. Ann Intern Med 153:135–136. doi: 10.7326/0003-4819-153-2-201007200-00256. [DOI] [PubMed] [Google Scholar]
- 6.Kamar N, Rostaing L, Abravanel F, Garrouste C, Lhomme S, Esposito L, Basse G, Cointault O, Ribes D, Nogier MB, Alric L, Peron JM, Izopet J. 2010. Ribavirin therapy inhibits viral replication on patients with chronic hepatitis E virus infection. Gastroenterol J 139:1612–1618. doi: 10.1053/j.gastro.2010.08.002. [DOI] [PubMed] [Google Scholar]
- 7.Ikram A, Hakim MS, Zhou JH, Wang W, Peppelenbosch MP, Pan Q. 2018. Genotype-specific acquisition, evolution and adaptation of characteristic mutations in hepatitis E virus. Virulence 9:121–132. doi: 10.1080/21505594.2017.1358349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Debing Y, Gisa A, Dallmeier K, Pischke S, Bremer B, Manns M, Wedemeyer H, Suneetha PV, Neyts J. 2014. A mutation in the hepatitis E virus RNA polymerase promotes its replication and associates with ribavirin treatment failure in organ transplant recipients. Gastroenterology 147:1008–1011 e7. quiz e15-6. doi: 10.1053/j.gastro.2014.08.040. [DOI] [PubMed] [Google Scholar]
- 9.Debing Y, Ramière C, Dallmeier K, Piorkowski G, Trabaud M-A, Lebossé F, Scholtès C, Roche M, Legras-Lachuer C, de Lamballerie X, André P, Neyts J. 2016. Hepatitis E virus mutations associated with ribavirin treatment failure result in altered viral fitness and ribavirin sensitivity. J Hepatol 65:499–508. doi: 10.1016/j.jhep.2016.05.002. [DOI] [PubMed] [Google Scholar]
- 10.Manns MP, Wedemeyer H, Cornberg M. 2006. Treating viral hepatitis C: efficacy, side effects, and complications. Gut 55:1350–1359. doi: 10.1136/gut.2005.076646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Kamar N, Bendall R, Legrand-Abravanel F, Xia N-S, Ijaz S, Izopet J, Dalton HR. 2012. Hepatitis E. Lancet 379:2477–2488. doi: 10.1016/S0140-6736(11)61849-7. [DOI] [PubMed] [Google Scholar]
- 12.Khuroo MS. 1980. Study of an epidemic of non-A, non-B hepatitis. Possibility of another human hepatitis virus distinct from post-transfusion non-A, non-B type. Am J Med 68:818–824. doi: 10.1016/0002-9343(80)90200-4. [DOI] [PubMed] [Google Scholar]
- 13.Aggarwal R, Krawczynski K. 2000. Hepatitis E: an overview and recent advances in clinical and laboratory research. J Gastroenterol Hepatol 15:9–20. doi: 10.1046/j.1440-1746.2000.02006.x. [DOI] [PubMed] [Google Scholar]
- 14.Teshale EH, Howard CM, Grytdal SP, Handzel TR, Barry V, Kamili S, Drobeniuc J, Okware S, Downing R, Tappero JW, Bakamutumaho B, Teo CG, Ward JW, Holmberg SD, Hu DJ. 2010. Hepatitis E epidemic, Uganda. Emerg Infect Dis 16:126–129. doi: 10.3201/eid1601.090764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Kumar A, Beniwal M, Kar P, Sharma JB, Murthy NS. 2004. Hepatitis E in pregnancy. Int J Gynecol Obstet 85:240–244. doi: 10.1016/j.ijgo.2003.11.018. [DOI] [PubMed] [Google Scholar]
- 16.Patra S, Kumar A, Trivedi SS, Puri M, Sarin SK. 2007. Maternal and fetal outcomes in pregnant women with acute hepatitis E virus infection. Ann Intern Med 147:28–33. doi: 10.7326/0003-4819-147-1-200707030-00005. [DOI] [PubMed] [Google Scholar]
- 17.Khuroo MS, Teli MR, Skidmore S, Sofi MA, Khuroo MI. 1981. Incidence and severity of viral hepatitis in pregnancy. Am J Med 70:252–255. doi: 10.1016/0002-9343(81)90758-0. [DOI] [PubMed] [Google Scholar]
- 18.Wang Y, Chen G, Pan Q, Zhao J. 2018. Chronic hepatitis E in a renal transplant recipient: the first report of genotype 4 hepatitis E virus caused chronic infection in organ recipient. Gastroenterology 154:1199–1201. doi: 10.1053/j.gastro.2017.12.028. [DOI] [PubMed] [Google Scholar]
- 19.Robins AEM, Bowden DJ, Gelson W. 2018. Chronic genotype 1 hepatitis E infection from immunosuppression for ileo-colonic Crohn's disease. Oxf Med Case Rep 2018:omy059. doi: 10.1093/omcr/omy059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Singh A, Seth R, Gupta A, Shalimar Nayak B, Acharya SK, Das P. 2016. Chronic hepatitis E–an emerging disease in an immunocompromised host. Gastroenterol Rep 16:152–155. doi: 10.1093/gastro/gow024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Lee GH, Tan BH, Chi-Yuan Teo E, Lim SG, Dan YY, Wee A, Aw PP, Zhu Y, Hibberd ML, Tan CK, Purdy MA, Teo CG. 2016. Chronic infection with camelid hepatitis E virus in a liver transplant recipient who regularly consumes camel meat and milk. Gastroenterology 150:355–357. doi: 10.1053/j.gastro.2015.10.048. [DOI] [PubMed] [Google Scholar]
- 22.Kamar N, Garrouste C, Haagsma EB, Garrigue V, Pischke S, Chauvet C, Dumortier J, Cannesson A, Cassuto-Viguier E, Thervet E, Conti F, Lebray P, Dalton HR, Santella R, Kanaan N, Essig M, Mousson C, Radenne S, Roque-Afonso AM, Izopet J, Rostaing L. 2011. Factors associated with chronic hepatitis in patients with hepatitis E virus infection who have received solid organ transplants. Gastroenterol J 140:1481–1489. doi: 10.1053/j.gastro.2011.02.050. [DOI] [PubMed] [Google Scholar]
- 23.Kelly AG, Netzler NE, White PA. 2016. Ancient recombination events and the origins of hepatitis E virus. BMC Evol Biol 16:210. doi: 10.1186/s12862-016-0785-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Krawczynski K, Aggarwal R, Kamili S. 2000. Hepatitis E. Infect Dis Clin North Am 14:669–687. doi: 10.1016/S0891-5520(05)70126-4. [DOI] [PubMed] [Google Scholar]
- 25.Agrawal S, Gupta D, Panda SK. 2001. The 3' end of hepatitis E virus (HEV) genome binds specifically to the viral RNA-dependent RNA polymerase (RdRp). Virology 282:87–101. doi: 10.1006/viro.2000.0819. [DOI] [PubMed] [Google Scholar]
- 26.Reyes GR, Huang CC, Tam AW, Purdy MA. 1993. Molecular organization and replication of hepatitis E virus (HEV). Arch Virol Suppl 7:15–25. doi: 10.1007/978-3-7091-9300-6_2. [DOI] [PubMed] [Google Scholar]
- 27.Ding Q, Heller B, Capuccino JM, Song B, Nimgaonkar I, Hrebikova G, Contreras JE, Ploss A. 2017. Hepatitis E virus ORF3 is a functional ion channel required for release of infectious particles. Proc Natl Acad Sci U S A 114:1147–1152. doi: 10.1073/pnas.1614955114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Tam AW, Smith MM, Guerra ME, Huang CC, Bradley DW, Fry KE, Reyes GR. 1991. Hepatitis E virus (HEV): molecular cloning and sequencing of the full-length viral genome. Virology 185:120–131. doi: 10.1016/0042-6822(91)90760-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Eltahla AA, Luciani F, White PA, Lloyd AR, Bull RA. 2015. Inhibitors of the hepatitis C virus polymerase; mode of action and resistance. Viruses 7:5206–5224. doi: 10.3390/v7102868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Ferrer-Orta C, Arias A, Escarmís C, Verdaguer N. 2006. A comparison of viral RNA-dependent RNA polymerases. Curr Opin Struct Biol 16:27–34. doi: 10.1016/j.sbi.2005.12.002. [DOI] [PubMed] [Google Scholar]
- 31.Ng KK-S, Arnold JJ, Cameron CE. 2008. Structure-function relationships among RNA-dependent RNA polymerases, p 137–156. In Paddison PJ, Vogt PK (ed), RNA interference. Springer, Berlin, Germany. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Chandra V, Taneja S, Kalia M, Jameel S. 2008. Molecular biology and pathogenesis of hepatitis E virus. J Biosci 33:451–464. doi: 10.1007/s12038-008-0064-1. [DOI] [PubMed] [Google Scholar]
- 33.Mahilkar S, Paingankar MS, Lole KS. 2016. Hepatitis E virus RNA-dependent RNA polymerase: RNA template specificities, recruitment and synthesis. J Gen Virol 97:2231–2242. doi: 10.1099/jgv.0.000528. [DOI] [PubMed] [Google Scholar]
- 34.Torresi J, Meanger J, Lambert P, Li F, Locarnini SA, Anderson DA. 1997. High level expression of the capsid protein of hepatitis E virus in diverse eukaryotic cells using the Semliki Forest virus replicon. J Virol Methods 69:81–91. doi: 10.1016/S0166-0934(97)00142-0. [DOI] [PubMed] [Google Scholar]
- 35.Emerson SU, Nguyen H, Graff J, Stephany DA, Brockington A, Purcell RH. 2004. In vitro replication of hepatitis E virus (HEV) genomes and of an HEV replicon expressing green fluorescent protein. J Virol 78:4838–4846. doi: 10.1128/JVI.78.9.4838-4846.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Graff J, Nguyen H, Kasorndorkbua C, Halbur PG, St Claire M, Purcell RH, Emerson SU. 2005. In vitro and in vivo mutational analysis of the 3'-terminal regions of hepatitis e virus genomes and replicons. J Virol 79:1017–1026. doi: 10.1128/JVI.79.2.1017-1026.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Shukla P, Nguyen HT, Faulk K, Mather K, Torian U, Engle RE, Emerson SU. 2012. Adaptation of a genotype 3 hepatitis E virus to efficient growth in cell culture depends on an inserted human gene segment acquired by recombination. J Virol 86:5697–5707. doi: 10.1128/JVI.00146-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Emerson SU, Zhang M, Meng XJ, Nguyen H, St Claire M, Govindarajan S, Huang YK, Purcell RH. 2001. Recombinant hepatitis E virus genomes infectious for primates: importance of capping and discovery of a cis-reactive element. Proc Natl Acad Sci U S A 98:15270–15275. doi: 10.1073/pnas.251555098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Dao Thi VL, Debing Y, Wu X, Rice CM, Neyts J, Moradpour D, Gouttenoire J. 2016. Sofosbuvir inhibits hepatitis E virus replication in vitro and results in an additive effect when combined with ribavirin. Gastroenterology 150:82–85. doi: 10.1053/j.gastro.2015.09.011. [DOI] [PubMed] [Google Scholar]
- 40.Wang Y, Zhou X, Debing Y, Chen K, Van Der Laan LJ, Neyts J, Janssen HL, Metselaar HJ, Peppelenbosch MP, Pan Q. 2014. Calcineurin inhibitors stimulate and mycophenolic acid inhibits replication of hepatitis E virus. Gastroenterology 146:1775–1783. doi: 10.1053/j.gastro.2014.02.036. [DOI] [PubMed] [Google Scholar]
- 41.Todt D, Moeller N, Praditya D, Kinast V, Friesland M, Engelmann M, Verhoye L, Sayed IM, Behrendt P, Dao Thi VL, Meuleman P, Steinmann E. 2018. The natural compound silvestrol inhibits hepatitis E virus (HEV) replication in vitro and in vivo. Antiviral Res 157:151–158. doi: 10.1016/j.antiviral.2018.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Wang W, Hakim MS, Nair VP, de Ruiter PE, Huang F, Sprengers D, Van Der Laan LJ, Peppelenbosch MP, Surjit M, Pan Q. 2016. Distinct antiviral potency of sofosbuvir against hepatitis C and E viruses. Gastroenterology 151:1251–1253. doi: 10.1053/j.gastro.2016.09.061. [DOI] [PubMed] [Google Scholar]
- 43.Qu C, Xu L, Yin Y, Peppelenbosch MP, Pan Q, Wang W. 2017. Nucleoside analogue 2'-C-methylcytidine inhibits hepatitis E virus replication but antagonizes ribavirin. Arch Virol 162:2989–2996. doi: 10.1007/s00705-017-3444-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Wang QY, Dong H, Zou B, Karuna R, Wan KF, Zou J, Susila A, Yip A, Shan C, Yeo KL, Xu H, Ding M, Chan WL, Gu F, Seah PG, Liu W, Lakshminarayana SB, Kang C, Lescar J, Blasco F, Smith PW, Shi PY. 2015. Discovery of dengue virus NS4B inhibitors. J Virol 89:8233–8244. doi: 10.1128/JVI.00855-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Yin Z, Chen YL, Schul W, Wang QY, Gu F, Duraiswamy J, Kondreddi RR, Niyomrattanakit P, Lakshminarayana SB, Goh A, Xu HY, Liu W, Liu B, Lim JY, Ng CY, Qing M, Lim CC, Yip A, Wang G, Chan WL, Tan HP, Lin K, Zhang B, Zou G, Bernard KA, Garrett C, Beltz K, Dong M, Weaver M, He H, Pichota A, Dartois V, Keller TH, Shi PY. 2009. An adenosine nucleoside inhibitor of dengue virus. Proc Natl Acad Sci U S A 106:20435–20439. doi: 10.1073/pnas.0907010106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.van der Linden L, Vives-Adrián L, Selisko B, Ferrer-Orta C, Liu X, Lanke K, Ulferts R, De Palma AM, Tanchis F, Goris N, Lefebvre D, De Clercq K, Leyssen P, Lacroix C, Pürstinger G, Coutard B, Canard B, Boehr DD, Arnold JJ, Cameron CE, Verdaguer N, Neyts J, van Kuppeveld FJM. 2015. The RNA template channel of the RNA-dependent RNA polymerase as a target for development of antiviral therapy of multiple genera within a virus family. PLoS Pathog 11:e1004733. doi: 10.1371/journal.ppat.1004733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Chou T-C. 2010. Drug combination studies and their synergy quantification using the Chou-Talalay method. Cancer Res 70:440–446. doi: 10.1158/0008-5472.CAN-09-1947. [DOI] [PubMed] [Google Scholar]
- 48.Ahmed JA, Moturi E, Spiegel P, Schilperoord M, Burton W, Kassim NH, Mohamed A, Ochieng M, Nderitu L, Navarro-Colorado C, Burke H, Cookson S, Handzel T, Waiboci LW, Montgomery JM, Teshale E, Marano N. 2013. Hepatitis E outbreak, Dadaab refugee camp, Kenya, 2012. Emerg Infect Dis 19:1010–1012. doi: 10.3201/eid1906.130275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Howard CM, Handzel T, Hill VR, Grytdal SP, Blanton C, Kamili S, Drobeniuc J, Hu D, Teshale E. 2010. Novel risk factors associated with hepatitis E virus infection in a large outbreak in northern Uganda: results from a case-control study and environmental analysis. Am J Trop Med Hyg 83:1170–1173. doi: 10.4269/ajtmh.2010.10-0384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Tsega E, Krawczynski K, Hansson BG, Nordenfelt E, Negusse Y, Alemu W, Bahru Y. 1991. Outbreak of acute hepatitis E virus infection among military personnel in northern Ethiopia. J Med Virol 34:232–236. doi: 10.1002/jmv.1890340407. [DOI] [PubMed] [Google Scholar]
- 51.Labrique A, Nelson K. 2010. Hepatitis E virus infections among US military personnel deployed to Afghanistan. J Infect Dis 202:1297–1299. doi: 10.1086/656599. [DOI] [PubMed] [Google Scholar]
- 52.Clayson ET, Vaughn DW, Innis BL, Shrestha MP, Pandey R, Malla DB. 1998. Association of hepatitis E virus with an outbreak of hepatitis at a military training camp in Nepal. J Med Virol 54:178–182. doi:. [DOI] [PubMed] [Google Scholar]
- 53.Kamar N, Lhomme S, Abravanel F, Marion O, Peron JM, Alric L, Izopet J. 2016. Treatment of HEV infection in patients with a solid-organ transplant and chronic hepatitis. Viruses 8:222. doi: 10.3390/v8080222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Dienstag JL, McHutchison JG. 2006. American Gastroenterological Association medical position statement on the management of hepatitis C. Gastroenterology 130:225–230. doi: 10.1053/j.gastro.2005.11.011. [DOI] [PubMed] [Google Scholar]
- 55.Todt D, Gisa A, Radonic A, Nitsche A, Behrendt P, Suneetha PV, Pischke S, Bremer B, Brown RJ, Manns MP, Cornberg M, Bock CT, Steinmann E, Wedemeyer H. 2016. In vivo evidence for ribavirin-induced mutagenesis of the hepatitis E virus genome. Gut 65:1733–1743. doi: 10.1136/gutjnl-2015-311000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Todt D, Francois C, Anggakusuma BP, Engelmann M, Knegendorf L, Vieyres G, Wedemeyer H, Hartmann R, Pietschmann T, Duverlie G, Steinmann E. 2016. Antiviral activities of different interferon types and subtypes against hepatitis E virus replication. Antimicrob Agents Chemother 60:2132–2139. doi: 10.1128/AAC.02427-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Deng YQ, Zhang NN, Li CF, Tian M, Hao JN, Xie XP, Shi PY, Qin CF. 2016. Adenosine analog NITD008 is a potent inhibitor of Zika virus. Open Forum Infect Dis 3:ofw175. doi: 10.1093/ofid/ofw175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Lee JC, Tseng CK, Wu YH, Kaushik-Basu N, Lin CK, Chen WC, Wu HN. 2015. Characterization of the activity of 2'-C-methylcytidine against dengue virus replication. Antiviral Res 116:1–9. doi: 10.1016/j.antiviral.2015.01.002. [DOI] [PubMed] [Google Scholar]
- 59.Devogelaere B, Berke JM, Vijgen L, Dehertogh P, Fransen E, Cleiren E, van der Helm L, Nyanguile O, Tahri A, Amssoms K, Lenz O, Cummings MD, Clayton RF, Vendeville S, Raboisson P, Simmen KA, Fanning GC, Lin TI. 2012. TMC647055, a potent nonnucleoside hepatitis C virus NS5B polymerase inhibitor with cross-genotypic coverage. Antimicrob Agents Chemother 56:4676–4684. doi: 10.1128/AAC.00245-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Klumpp K, Lévêque V, Le Pogam S, Ma H, Jiang W-R, Kang H, Granycome C, Singer M, Laxton C, Hang JQ, Sarma K, Smith DB, Heindl D, Hobbs CJ, Merrett JH, Symons J, Cammack N, Martin JA, Devos R, Nájera I. 2006. The novel nucleoside analog R1479 (4'-azidocytidine) is a potent inhibitor of NS5B-dependent RNA synthesis and hepatitis C virus replication in cell culture. J Biol Chem 281:3793–3799. doi: 10.1074/jbc.M510195200. [DOI] [PubMed] [Google Scholar]
- 61.Lam AM, Murakami E, Espiritu C, Steuer HM, Niu C, Keilman M, Bao H, Zennou V, Bourne N, Julander JG, Morrey JD, Smee DF, Frick DN, Heck JA, Wang P, Nagarathnam D, Ross BS, Sofia MJ, Otto MJ, Furman PA. 2010. PSI-7851, a pronucleotide of beta-D-2'-deoxy-2'-fluoro-2'-C-methyluridine monophosphate, is a potent and pan-genotype inhibitor of hepatitis C virus replication. Antimicrob Agents Chemother 54:3187–3196. doi: 10.1128/AAC.00399-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Shukla P, Nguyen HT, Torian U, Engle RE, Faulk K, Dalton HR, Bendall RP, Keane FE, Purcell RH, Emerson SU. 2011. Cross-species infections of cultured cells by hepatitis E virus and discovery of an infectious virus-host recombinant. Proc Natl Acad Sci U S A 108:2438–2443. doi: 10.1073/pnas.1018878108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Biliotti E, Franchi C, Spaziante M, Garbuglia AR, Volpicelli L, Palazzo D, De Angelis M, Esvan R, Taliani G. 2018. Autochthonous acute hepatitis E: treatment with sofosbuvir and ribavirin. Infection 46:725–727. doi: 10.1007/s15010-018-1168-7. [DOI] [PubMed] [Google Scholar]
- 64.Todesco E, Demeret S, Calin R, Roque-Afonso AM, Thibault V, Mallet V, Akhavan S, Jaspard M, Peytavin G, Poynard T, Katlama C, Pourcher V. 2017. Chronic hepatitis E in HIV/HBV coinfected patient: lack of power of sofosbuvir-ribavirin. AIDS 31:1346–1348. doi: 10.1097/QAD.0000000000001474. [DOI] [PubMed] [Google Scholar]
- 65.van der Valk M, Zaaijer HL, Kater AP, Schinkel J. 2017. Sofosbuvir shows antiviral activity in a patient with chronic hepatitis E virus infection. J Hepatol 66:242–243. doi: 10.1016/j.jhep.2016.09.014. [DOI] [PubMed] [Google Scholar]
- 66.De Martin E, Antonini TM, Coilly A, Pittau G, Vibert E, Duclos-Vallée J-C, Samuel D, Roque-Afonso A-M. 2017. HCV and HEV recurrence after liver transplantation: one antiviral therapy for two viruses. Transpl Int 30:318–319. doi: 10.1111/tri.12898. [DOI] [PubMed] [Google Scholar]
- 67.Todesco E, Mazzola A, Akhavan S, Abravanel F, Poynard T, Roque-Afonso AM, Peytavin G, Marcelin AG, Calmus Y, Lecuyer L, Guillemain R, Conti F. 2018. Chronic hepatitis E in a heart transplant patient: sofosbuvir and ribavirin regimen not fully effective. Antivir Ther doi: 10.3851/IMP3227. [DOI] [PubMed] [Google Scholar]
- 68.Donnelly MC, Imlach SN, Abravanel F, Ramalingam S, Johannessen I, Petrik J, Fraser AR, Campbell JD, Bramley P, Dalton HR, Hayes PC, Kamar N, Simpson KJ. 2017. Sofosbuvir and daclatasvir anti-viral therapy fails to clear HEV viremia and restore reactive T cells in a HEV/HCV co-infected liver transplant recipient. Gastroenterology 152:300–301. doi: 10.1053/j.gastro.2016.05.060. [DOI] [PubMed] [Google Scholar]
- 69.Wang W, Peppelenbosch MP, Pan Q. 2016. Targeting viral polymerase for treating hepatitis E infection: how far are we? Gastroenterology 150:1690. doi: 10.1053/j.gastro.2016.01.045. [DOI] [PubMed] [Google Scholar]
- 70.Deng CL, Yeo H, Ye HQ, Liu SQ, Shang BD, Gong P, Alonso S, Shi PY, Zhang B. 2014. Inhibition of enterovirus 71 by adenosine analog NITD008. J Virol 88:11915–11923. doi: 10.1128/JVI.01207-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Shang L, Wang Y, Qing J, Shu B, Cao L, Lou Z, Gong P, Sun Y, Yin Z. 2014. An adenosine nucleoside analogue NITD008 inhibits EV71 proliferation. Antiviral Res 112:47–58. doi: 10.1016/j.antiviral.2014.10.009. [DOI] [PubMed] [Google Scholar]
- 72.Qing J, Luo R, Wang Y, Nong J, Wu M, Shao Y, Tang R, Yu X, Yin Z, Sun Y. 2016. Resistance analysis and characterization of NITD008 as an adenosine analog inhibitor against hepatitis C virus. Antiviral Res 126:43–54. doi: 10.1016/j.antiviral.2015.12.010. [DOI] [PubMed] [Google Scholar]
- 73.Lo MK, Shi PY, Chen YL, Flint M, Spiropoulou CF. 2016. In vitro antiviral activity of adenosine analog NITD008 against tick-borne flaviviruses. Antiviral Res 130:46–49. doi: 10.1016/j.antiviral.2016.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Zhang QY, Li XD, Liu SQ, Deng CL, Zhang B, Ye HQ. 2017. Development of a stable Japanese encephalitis virus replicon cell line for antiviral screening. Arch Virol 162:3417–3423. doi: 10.1007/s00705-017-3508-9. [DOI] [PubMed] [Google Scholar]
- 75.Koike K. 2006. Antiviral treatment of hepatitis C: present status and future prospects. J Infect Chemother 12:227–232. doi: 10.1007/s10156-006-0460-0. [DOI] [PubMed] [Google Scholar]
- 76.Rehman S, Ashfaq UA, Javed T. 2011. Antiviral drugs against hepatitis C virus. Genet Vaccines Ther 9:11. doi: 10.1186/1479-0556-9-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Freedberg KA, Losina E, Weinstein MC, Paltiel AD, Cohen CJ, Seage GR, Craven DE, Zhang H, Kimmel AD, Goldie SJ. 2001. The cost effectiveness of combination antiretroviral therapy for HIV disease. N Engl J Med 344:824–831. doi: 10.1056/NEJM200103153441108. [DOI] [PubMed] [Google Scholar]
- 78.Eltahla AA, Lim KL, Eden JS, Kelly AG, Mackenzie JM, White PA. 2014. Nonnucleoside inhibitors of norovirus RNA polymerase: scaffolds for rational drug design. Antimicrob Agents Chemother 58:3115–3123. doi: 10.1128/AAC.02799-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Tu ET, Bull RA, Kim MJ, McIver CJ, Heron L, Rawlinson WD, White PA. 2008. Norovirus excretion in an aged-care setting. J Clin Microbiol 46:2119–2121. doi: 10.1128/JCM.02198-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Livak KJ, Schmittgen TD. 2001. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods 25:402–408. doi: 10.1006/meth.2001.1262. [DOI] [PubMed] [Google Scholar]
- 81.Gentles RG, Ding M, Bender JA, Bergstrom CP, Grant-Young K, Hewawasam P, Hudyma T, Martin S, Nickel A, Regueiro-Ren A, Tu Y, Yang Z, Yeung KS, Zheng X, Chao S, Sun JH, Beno BR, Camac DM, Chang CH, Gao M, Morin PE, Sheriff S, Tredup J, Wan J, Witmer MR, Xie D, Hanumegowda U, Knipe J, Mosure K, Santone KS, Parker DD, Zhuo X, Lemm J, Liu M, Pelosi L, Rigat K, Voss S, Wang Y, Wang YK, Colonno RJ, Gao M, Roberts SB, Gao Q, Ng A, Meanwell NA, Kadow JF. 2014. Discovery and preclinical characterization of the cyclopropylindolobenzazepine BMS-791325, a potent allosteric inhibitor of the hepatitis C virus NS5B polymerase. J Med Chem 57:1855–1879. doi: 10.1021/jm4016894. [DOI] [PubMed] [Google Scholar]
- 82.Feld JJ, Kowdley KV, Coakley E, Sigal S, Nelson DR, Crawford D, Weiland O, Aguilar H, Xiong J, Pilot-Matias T, DaSilva-Tillmann B, Larsen L, Podsadecki T, Bernstein B. 2014. Treatment of HCV with ABT-450/r-ombitasvir and dasabuvir with ribavirin. N Engl J Med 370:1594–1603. doi: 10.1056/NEJMoa1315722. [DOI] [PubMed] [Google Scholar]
- 83.Li H, Tatlock J, Linton A, Gonzalez J, Jewell T, Patel L, Ludlum S, Drowns M, Rahavendran SV, Skor H, Hunter R, Shi ST, Herlihy KJ, Parge H, Hickey M, Yu X, Chau F, Nonomiya J, Lewis C. 2009. Discovery of (R)-6-cyclopentyl-6-(2-(2,6-diethylpyridin-4-yl)ethyl)-3-((5,7-dimethyl-[1,2,4]tr iazolo[1,5-a]pyrimidin-2-yl)methyl)-4-hydroxy-5,6-dihydropyran-2-one (PF-00868554) as a potent and orally available hepatitis C virus polymerase inhibitor. J Med Chem 52:1255–1258. doi: 10.1021/jm8014537. [DOI] [PubMed] [Google Scholar]
- 84.Hirashima S, Suzuki T, Ishida T, Noji S, Yata S, Ando I, Komatsu M, Ikeda S, Hashimoto H. 2006. Benzimidazole derivatives bearing substituted biphenyls as hepatitis C virus NS5B RNA-dependent RNA polymerase inhibitors: structure-activity relationship studies and identification of a potent and highly selective inhibitor JTK-109. J Med Chem 49:4721–4736. doi: 10.1021/jm060269e. [DOI] [PubMed] [Google Scholar]
- 85.Chan L, Das SK, Reddy TJ, Poisson C, Proulx M, Pereira O, Courchesne M, Roy C, Wang W, Siddiqui A, Yannopoulos CG, Nguyen-Ba N, Labrecque D, Bethell R, Hamel M, Courtemanche-Asselin P, L'Heureux L, David M, Nicolas O, Brunette S, Bilimoria D, Bédard J. 2004. Discovery of thiophene-2-carboxylic acids as potent inhibitors of HCV NS5B polymerase and HCV subgenomic RNA replication. Part 1: sulfonamides. Bioorg Med Chem Lett 14:793–796. doi: 10.1016/j.bmcl.2003.10.067. [DOI] [PubMed] [Google Scholar]
- 86.Kneteman NM, Howe AY, Gao T, Lewis J, Pevear D, Lund G, Douglas D, Mercer DF, Tyrrell DL, Immermann F, Chaudhary I, Speth J, Villano SA, O'Connell J, Collett M. 2009. HCV796: a selective nonstructural protein 5B polymerase inhibitor with potent anti-hepatitis C virus activity in vitro, in mice with chimeric human livers, and in humans infected with hepatitis C virus. Hepatology 49:745–752. doi: 10.1002/hep.22717. [DOI] [PubMed] [Google Scholar]
- 87.Gopalsamy A, Chopra R, Lim K, Ciszewski G, Shi M, Curran KJ, Sukits SF, Svenson K, Bard J, Ellingboe JW, Agarwal A, Krishnamurthy G, Howe AY, Orlowski M, Feld B, O'Connell J, Mansour TS. 2006. Discovery of proline sulfonamides as potent and selective hepatitis C virus NS5b polymerase inhibitors. Evidence for a new NS5b polymerase binding site. J Med Chem 49:3052–3055. doi: 10.1021/jm060168g. [DOI] [PubMed] [Google Scholar]
- 88.Shih IH, Vliegen I, Peng B, Yang H, Hebner C, Paeshuyse J, Purstinger G, Fenaux M, Tian Y, Mabery E, Qi X, Bahador G, Paulson M, Lehman LS, Bondy S, Tse W, Reiser H, Lee WA, Schmitz U, Neyts J, Zhong W. 2011. Mechanistic characterization of GS-9190 (Tegobuvir), a novel nonnucleoside inhibitor of hepatitis C virus NS5B polymerase. Antimicrob Agents Chemother 55:4196–4203. doi: 10.1128/AAC.00307-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Rusinov VL, Sapozhnikova IM, Ulomskii EN, Medvedeva NR, Egorov VV, Kiselev OI, Deeva EG, Vasin AV, Chupakhin ON. 2015. Nucleophilic substitution of nitro group in nitrotriazolotriazines as a model of potential interaction with cysteine-containing proteins. Chem Heterocycl Comp 51:275–280. doi: 10.1007/s10593-015-1695-4. [DOI] [Google Scholar]
- 90.Pockros PJ. 2013. Nucleoside/nucleotide analogue polymerase inhibitors in development. Clin Liver Dis 17:105–110. doi: 10.1016/j.cld.2012.09.007. [DOI] [PubMed] [Google Scholar]
- 91.Olsen DB, Eldrup AB, Bartholomew L, Bhat B, Bosserman MR, Ceccacci A, Colwell LF, Fay JF, Flores OA, Getty KL, Grobler JA, LaFemina RL, Markel EJ, Migliaccio G, Prhavc M, Stahlhut MW, Tomassini JE, MacCoss M, Hazuda DJ, Carroll SS. 2004. A 7-deaza-adenosine analog is a potent and selective inhibitor of hepatitis C virus replication with excellent pharmacokinetic properties. Antimicrob Agents Chemother 48:3944–3953. doi: 10.1128/AAC.48.10.3944-3953.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Furuta Y, Takahashi K, Fukuda Y, Kuno M, Kamiyama T, Kozaki K, Nomura N, Egawa H, Minami S, Watanabe Y, Narita H, Shiraki K. 2002. In vitro and in vivo activities of anti-influenza virus compound T-705. Antimicrob Agents Chemother 46:977–981. doi: 10.1128/AAC.46.4.977-981.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Sofia MJ, Bao D, Chang W, Du J, Nagarathnam D, Rachakonda S, Reddy PG, Ross BS, Wang P, Zhang HR, Bansal S, Espiritu C, Keilman M, Lam AM, Steuer HM, Niu C, Otto MJ, Furman PA. 2010. Discovery of a beta-d-2'-deoxy-2'-alpha-fluoro-2'-beta-C-methyluridine nucleotide prodrug (PSI-7977) for the treatment of hepatitis C virus. J Med Chem 53:7202–7218. doi: 10.1021/jm100863x. [DOI] [PubMed] [Google Scholar]
- 94.Konig B, Dustmann JH. 1985. The caffeoylics as a new family of natural antiviral compounds. Naturwissenschaften 72:659–661. doi: 10.1007/BF00497441. [DOI] [PubMed] [Google Scholar]




















