Human respiratory syncytial virus (RSV) is a major cause of lower respiratory tract infections in early childhood. However, no vaccines have yet been approved for prevention of RSV infection, and the treatment options are limited. Therefore, development of effective and safe anti-RSV drugs is needed. In this study, we evaluated the antiviral activity and mechanism of action of a novel macrocyclic anti-RSV compound, TP0591816. TP0591816 showed significant antiviral activities against both subgroup A and subgroup B RSV, while exerting no cytotoxicity.
KEYWORDS: F protein, TP0591816, antiviral agents, fusion inhibitor, respiratory syncytial virus
ABSTRACT
Human respiratory syncytial virus (RSV) is a major cause of lower respiratory tract infections in early childhood. However, no vaccines have yet been approved for prevention of RSV infection, and the treatment options are limited. Therefore, development of effective and safe anti-RSV drugs is needed. In this study, we evaluated the antiviral activity and mechanism of action of a novel macrocyclic anti-RSV compound, TP0591816. TP0591816 showed significant antiviral activities against both subgroup A and subgroup B RSV, while exerting no cytotoxicity. Notably, the antiviral activity of TP0591816 was maintained against a known fusion inhibitor-resistant RSV strain with a mutation in the cysteine-rich region or in heptad repeat B. Results of a time-of-addition assay and a temperature shift assay indicated that TP0591816 inhibited fusion of RSV with the cell membrane during viral entry. In addition, TP0591816 added after cell infection also inhibited cell-cell fusion. A TP0591816-resistant virus strain selected by serial passage had an L141F mutation, but no mutation in the cysteine-rich region or in heptad repeat B in the fusion (F) protein. Treatment with TP0591816 reduced lung virus titers in a dose-dependent manner in a mouse model of RSV infection. Furthermore, the estimated effective dose of TP0591816 for use against F protein mutants was thought to be clinically realistic and potentially tolerable. Taken together, these findings suggest that TP0591816 is a promising novel candidate for the treatment of resistant RSV infection.
INTRODUCTION
Human respiratory syncytial virus (RSV) is a common cause of bronchiolitis and pneumonia in early childhood (1). An estimated 33.8 million children under 5 years old are reported to suffer from acute lower respiratory infection (ALRI) caused by RSV (accounting for 22% of all cases of ALRI); over 3.4 million of them need hospitalization, and 66,000 to 199,000 die annually (2). Moreover, development of severe RSV bronchiolitis under 1 year of age is associated with an increased risk of development of allergic asthma in early adulthood (3). In adults, advanced age, immunocompromised status, and the presence of cardiopulmonary disease are reported as high-risk factors for the development of severe RSV infection (4). However, no vaccines for prevention of RSV infection have been approved yet and the treatment options are limited. Palivizumab, which is a monoclonal antibody directed against the RSV fusion (F) protein, has been demonstrated as being effective for preventing hospitalization in children with RSV infection, but it is only used for high-risk patients because of the high cost (5, 6). Although a guanosine analogue, ribavirin, has also been demonstrated to show antiviral activity against RSV, it has relatively high toxicity, and its clinical efficacy remains controversial (7, 8). Therefore, new effective and safe drugs are needed to treat clinically significant RSV infections.
RSV attaches to the host cells via glycoprotein (G) and enters the cells by fusion of the viral envelope with the cell membrane mediated by the F protein (9). After invasion, the RNA-dependent RNA polymerase, which consists of a large (L) protein and phosphoprotein (P), carries out both transcription and replication using the genomic RNA-nucleoprotein (N) complex as the template (10), the viruses spread by cell-cell fusion mediated by the F protein, and a syncytium is formed (11). It is thought that for controlling RSV infection, these viral factors that are needed for productive viral infection need to be inhibited, and so far, various anti-RSV compounds targeting the F protein (presatovir [formerly GS-5806], JNJ-53718678, RV521, ziresovir [AK0529], BMS-433771, and VP-14637) (12–17), L protein (lumicitabine [ALS-8176] and PC786) (18, 19), and N protein (RSV604 and EDP-938) (20, 21) have been reported.
Among these classes of RSV inhibitors, fusion inhibitors have the advantage of being able to act without penetrating the cells—they inhibit membrane fusion outside the cells as the virus begins to invade. For this reason, fusion inhibitors are expected to have therapeutic potential, and RSV inhibitors are the test drugs in most clinical trials (12–15, 22, 23). However, it is known that this class of drugs often loses antiviral activity even with a single point mutation of the virus. Fusion inhibitor-resistant isolates most often show resistance-associated mutations in the following three regions: (i) the fusion peptide, 140 to 144 amino acids (aa) long; (ii) the cysteine-rich region, 392 to 401 aa long; and (iii) heptad repeat B, 486 to 489 aa long; the resistant mutants exhibit wide cross-resistance to other fusion inhibitors (24). Therefore, development of resistance is of critical potential concern when developing a new fusion inhibitor.
We have synthesized a number of small-molecule RSV inhibitors (25, 26), and identified some macrocyclic pyrazolo[1,5-a]pyrimidine derivatives that appear to have antiviral activity against RSV with the D486N mutation in the F protein that is resistant to known fusion inhibitors (15, 26–31). We further optimized these compounds with improved activities against the D486N mutant and obtained TP0591816 (Fig. 1). Here, we report the results of our evaluation of the antiviral activity against not only the D486N mutant, but also against known fusion inhibitor-resistant RSV with mutations other than D486N in the F protein and the mechanism of action of TP0591816. In addition, the in vivo efficacy of TP0591816 was investigated in a mouse model of RSV infection. Furthermore, analyses were conducted to predict the clinically effective dose of TP0591816 for cases of infection with the resistant virus.
FIG 1.
Chemical structure of TP0591816.
RESULTS
Antiviral activity and cytotoxicity of TP0591816.
The antiviral activities of TP591816 against wild-type and some RSV F protein mutants were evaluated by 2,3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide salt (XTT) assay using the HEp-2 cell line (Table 1). TP0591816 showed potent antiviral activity against RSV A2 (subgroup A) and RSV 18537 (subgroup B), with 50% effective concentrations (EC50) of 0.255 and 0.0824 nM, respectively. These results demonstrate that TP0591816 has more potent antiviral activity than the other known RSV inhibitors tested in this study. In addition, TP0591816 exhibited excellent antiviral activity against the D486N mutant RSV A2, with an EC50 of 0.549 nM. Moreover, it also showed antiviral activity against F488L and T400I mutants, which are resistant to the known fusion inhibitors, with EC50 values of 5.09 and 2.97 nM, respectively. TP0591816 showed no cytotoxicity in uninfected HEp-2 cells, with a 50% cytotoxic concentration (CC50) of >30,000 nM, similar to that of other RSV inhibitors.
TABLE 1.
Antiviral activities against wild-type and F protein mutant RSV and cytotoxicity of TP0591816 and known RSV inhibitors
| Compound | EC50 (nM)a for: |
CC50 (nM)a | ||||
|---|---|---|---|---|---|---|
| A2 (subgroup A) |
18537 (subgroup B) |
|||||
| WTb | D486N | F488L | T400I | WTb | ||
| TP0591816 | 0.255 | 0.549 | 5.09 | 2.97 | 0.0824 | >30,000 |
| BMS-433771 | 50.0 | >1,000 | >1,000 | >1,000 | 68.4 | >30,000 |
| VP-14637 | 0.467 | >1,000 | >1,000 | >1,000 | 0.458 | >30,000 |
| Ribavirin | 11,700 | 12,700 | 13,700 | 9,710 | 8,450 | 159,000 |
All 50% effective concentration (EC50) and 50% cytotoxic concentration (CC50) values are represented as the geometric means of the results of at least four independent experiments.
WT, wild type.
Inhibition of fusion of the viral envelope with the cell membrane by TP0591816.
In order to explore the step of the viral life cycle that is blocked by TP0591816, a time-of-addition experiment was performed. TP0591816 and other known fusion inhibitors were added to HEp-2 cells at 0, 6, or 24 h postinfection (hpi). Addition of TP0591816 to HEp-2 cells at 0 hpi resulted in a reduction of the virus titer in the supernatant by approximately 4 log compared to that of the untreated control. However, the antiviral activity of TP0591816 was much lower when it was added at 6 and 24 hpi (Fig. 2). The antiviral activities of the known fusion inhibitors BMS-433771 and VP-14637 were also attenuated when they were added at 6 and 24 hpi. These results suggest that TP0591816 inhibits viral entry into the cells, an early step in the viral life cycle.
FIG 2.
Effect of time of addition of TP0591816 and known fusion inhibitors on the antiviral efficacy. HEp-2 cells were infected with RSV A2, and 4.5 nM TP0591816, 900 nM BMS-433771, or 9 nM VP-14637 was added at 0, 6, or 24 h postinfection (hpi). The virus titers in the supernatants were determined at 3 days postinfection (dpi). Data are represented as the means ± standard errors of the means (SEM) from three independent experiments performed in triplicate.
A temperature shift assay was performed to investigate the detailed mechanism of inhibition of viral entry into the cells by TP0591816. RSV can attach to cells at 4°C, but fusion requires a temperature greater than 18°C (32). Therefore, temperature shift assays can clarify whether a test compound inhibits viral attachment to a cell or fusion of the viral envelope to the cell membrane. Addition of TP0591816 at 4°C did not reduce the virus titer compared to that observed in the untreated control, but addition after shift of the temperature to 37°C led to a reduction of the viral titer, as in the case of the presence of the drug throughout the experiment (Fig. 3). BMS-433771 and VP-14637 also inhibited viral infection only when added at 37°C. In contrast, heparin, which is known as an RSV attachment inhibitor (33), did not inhibit viral infection at 37°C but inhibited it at 4°C. Collectively, these data reveal that TP0591816 inhibits fusion of the viral envelope with the cell membrane at the time of cell entry.
FIG 3.
Temperature shift assay of TP0591816 and known fusion inhibitors. HEp-2 cells were infected with RSV A2 at 4°C for 1 h, and the temperature was shifted to 37°C; 4.5 nM TP0591816, 900 nM BMS-433771, or 9 nM VP-14637 was added only at 4°C, only at 37°C, or for the entire duration of the experiment (4°C and 37°C). The virus titers in the supernatants were determined at 3 dpi. Data are represented as the means ± SEM from three independent experiments performed in triplicate.
Inhibitory effect of TP0591816 on syncytium formation.
The mechanism of inhibition by TP0591816 of RSV fusion with the cell membrane was confirmed in a syncytium formation assay. TP0591816 was added at 24 hpi to ensure viral entry into the cells, and the cell-cell fusion was evaluated. The cell-cell fusion could be completely blocked by addition of 1.5 nM TP0591816 and partially blocked by addition of the drug at a dose of 0.75 nM (Fig. 4). BMS-433771 and VP-14637 also inhibited syncytium formation. On the other hand, ribavirin, which inhibits viral RNA synthesis at an early step of the replication phase, could not suppress syncytium formation. These results suggest that TP0591816 targets the RSV F protein and can inhibit F protein-mediated cell-cell fusion.
FIG 4.
Inhibitory effect of TP0591816 and known RSV inhibitors on syncytium formation. HEp-2 cells were infected with RSV A2, and 0.75 or 1.5 nM TP0591816, 300 nM BMS-433771, 3 nM VP-14637, or 100,000 nM ribavirin was added at 24 hpi. The inhibition of syncytium formation was evaluated at 3 dpi. Syncytium scores are shown as follows: −, no syncytium; +, <1/3; ++, ∼1/3 to 2/3; +++, ≥2/3 of syncytium compared to uninfected control. Each image is representative of two independent experiments.
In vitro selection of TP0591816-resistant viruses.
To support the proposed mechanism of action of TP0591816, resistant RSV was selected in HEp-2 cells in the presence of TP0591816. RSV was serially passaged with increasing inhibitor concentrations, and resistant viruses that could grow in the presence of an almost 300-fold higher EC50 of TP0591816 were obtained. Genotypic analysis of this TP0591816-resistant virus strain identified the mutation L141F in the F protein. L141F mutation is known to confer resistance to inhibitors of F protein (presatovir and RV521) (14, 22, 23).
The susceptibilities of the TP0591816-resistant L141F mutant virus to TP0591816 and other RSV inhibitors were evaluated. The L141F mutant exhibited a >4,720-fold reduction in the susceptibility to TP0591816 (Table 2). The L141F mutant also exhibited reduced susceptibility to BMS-433771 and VP-14637 while maintaining sensitivity to other class of inhibitors, such as ribavirin and palivizumab. These results confirmed that the target of TP0591816 was the F protein.
TABLE 2.
Susceptibility testing of TP0591816-resistant virus
| Compound | EC50 (nM)a for: |
EC50 fold difference (×)b for: |
|||
|---|---|---|---|---|---|
| Parent virus | Untreated control virus | TP0591816-resistant virus (L141F) | Untreated control virus | TP0591816-resistant virus (L141F) | |
| TP0591816 | 0.212 | 0.459 | >1,000 | 2.17 | >4,720 |
| BMS-433771 | 54.8 | 242 | >1,000 | 4.41 | >18.3 |
| VP-14637 | 0.502 | 0.846 | 5.57 | 1.69 | 11.1 |
| Ribavirin | 11,900 | 17,000 | 14,000 | 1.42 | 1.17 |
| Palivizumab | 0.317 | 0.657 | 0.398 | 2.07 | 1.26 |
All EC50 values are represented as the geometric means of the results of at least three independent experiments. Values are expressed as nM for all compounds except palivizumab (μg/ml).
Relative to the value for the parent virus.
Efficacy of TP0591816 in a mouse model.
After it was confirmed that TP0591816 showed significant activity in vitro, its in vivo efficacy was examined in a mouse model of RSV infection. TP0591816 was administered subcutaneously to mice at 1 h before RSV inoculation, and the lung virus titers were determined by plaque assay at 4 days postinfection (dpi). Treatment with TP0591816 at 1, 10, and 100 mg/kg reduced the lung virus titers in a dose-dependent manner by 0.72, 0.92, and 2.04 log, respectively, compared to the titers in the vehicle control (Fig. 5A). The reductions in the lung virus titers were statistically significant at all doses. In addition, lung weights of the infected mice were also reduced by TP0591816 (Fig. 5B).
FIG 5.
Efficacy of TP0591816 in a mouse model. TP0591816 was administered subcutaneously to mice at doses of 1, 10, and 100 mg/kg at 1 h before virus inoculation. The mice were then inoculated intranasally with 1 × 107 PFU of RSV A2, and the lung virus titers (A) and lung weights (B) were determined at 4 dpi. Data are represented as the means ± SEM (n = 8 mice per group). **, P < 0.01 versus vehicle (Steel’s test).
Prediction of the clinical efficacy of TP0591816.
Since, as shown above, TP0591816 has the potential to suppress infection by F protein mutants of RSV, we attempted to estimate the clinically effective dose of TP0591816. First, the pharmacokinetic parameters of TP0591816 were evaluated in vitro (Table 3). The apparent permeability (Papp) of TP0591816 at a pH of 6.2 was calculated by parallel artificial membrane permeability assay (PAMPA) to be 63.7 × 10−6 cm/sec. In the metabolic stability study using human liver microsomes, disappearance of TP0591816 followed the first-order reaction, and the hepatic intrinsic clearance (CLint) was calculated to be 182 μl/min/mg protein. The fraction unbound in incubated human liver microsomes (fu,mic) at 0.25 mg microsomal protein/ml was estimated to be 0.966. The percentage of plasma protein binding of TP0591816 in humans was determined to be 90.7%, and the unbound fraction (fu,p) was calculated to be 0.093. Then, the pharmacokinetics of TP0591816 were evaluated in vivo in the monkey (Table 4). The plasma concentration-time profiles of TP0591816 were analyzed, and the pharmacokinetic parameters were calculated. The steady-state volume of distribution (Vdss) and renal clearance (CLR) of TP0591816 were calculated to be 1,800 ml/kg and 241 ml/h/kg, respectively. Third, physiologically based pharmacokinetic (PBPK) modeling was performed using the physicochemical properties, in vitro data, and predicted pharmacokinetic parameters (Table 3), and the plasma concentration-time profile of TP0591816 in humans was simulated in this model. Finally, the clinically effective dose twice a day (BID) was calculated as the dose at which the trough plasma concentration yielded plasma-adjusted 95% effective concentrations (paEC95). The estimated oral BID doses of TP0591816 for treatment of infection by RSV A2 and the D486N mutant of RSV were 26.9 or 55.5 mg, respectively. It was also predicted that TP0591816 would also be effective for the treatment of infections caused by T400I or F488L mutants (Table 5).
TABLE 3.
Input parameters used for TP0591816 in a human PBPK model
| Parametera | Value or condition |
|---|---|
| Physicochemical properties and blood binding | |
| Mol wt | 539.03 |
| Compound type | Neutral |
| logD (pH 7.4) | 1.54 |
| fu,p | 0.093 |
| B/Pb | 0.7 |
| Absorption | |
| Absorption model | ADAM |
| Formulation | Solution |
| fu,gutb | 1 |
| Peff,man (10−4 cm/sec)c | 5.32 |
| Fac | 0.995 |
| ka (h−1)c | 2.32 |
| PAMPA Papp (10−6 cm/sec) | 63.7 |
| Distribution | |
| Distribution model | Full PBPK model |
| Vdss (liter/kg)d | 1.45 |
| Elimination | |
| Clearance type | Enzyme kinetics |
| CLint (μl/min/mg protein) | 182 |
| fm,CYP3A4b | 1 |
| fu,micd | 0.966 |
| CLR (liter/h)d | 7.88 |
D, distribution coefficient; fu,p, fraction unbound in plasma; B/P, blood-to-plasma ratio; ADAM, advanced dissolution, absorption, and metabolism; fu,gut, fraction unbound in the gut; Peff,man, effective permeability in humans; Fa, fraction absorbed; ka, absorption rate constant; PAMPA, parallel artificial membrane permeability assay; Papp, apparent permeability; PBPK, physiologically based pharmacokinetic; Vdss, steady-state volume of distribution; CLint, hepatic intrinsic clearance; fm,CYP3A4, fraction metabolized by CYP3A4; fu,mic, fraction unbound in fraction unbound in human liver microsomes; CLR, renal clearance.
Assumed.
Calculated from the PAMPA in Simcyp.
Predicted.
TABLE 4.
Pharmacokinetic parameters of TP0591816 in the male cynomolgus monkeyb
| Parametera | Value |
|---|---|
| CLtot (ml/h/kg) | 1,770 ± 170 |
| Vdss (ml/kg) | 1,800 ± 220 |
| t1/2 (h) | 1.3 ± 0.3 |
| AUC0–∞ (h · ng/ml) | 140 ± 13 |
| Urinary excretion (% of dose) | 13.6 ± 1.8 |
| CLR (ml/h/kg) | 241 |
CLtot, total clearance from plasma; Vdss, steady-state volume of distribution; t1/2, terminal-phase half-life; AUC0–∞, area under the concentration-time curve from time zero to infinity; CLR, renal clearance.
TP0591816 was administered intravenously to male cynomolgus monkeys at a dose of 0.25 mg/kg. Data are represented as the means ± SD (n = 3 monkeys per group).
TABLE 5.
paEC95 and predicted clinical effective dose of TP0591816
| Parameter | F protein type |
|||
|---|---|---|---|---|
| WTa | D486N | F488L | T400I | |
| paEC95 (ng/ml) | 3.23 | 6.67 | 115 | 84.6 |
| Predicted dose (mg BID)b | 26.9 | 55.5 | 955 | 705 |
WT, wild-type.
Dose required to achieve a trough plasma concentration greater than the paEC95.
DISCUSSION
While fusion inhibitors are expected to become effective agents for treating RSV infection, one of the important problems that need to be overcome is that they exhibit broad cross-resistance with each other (24). Recently, the binding modes of several fusion inhibitors were investigated based on the X-ray crystal structure, and it was revealed that all of the inhibitors bind to the same site of the 3-fold-symmetric pocket of the prefusion RSV F protein despite their dissimilar structures (34). For this reason, fusion inhibitors of different structural types induced the same mutations in the common regions of the F protein, and such mutations cause broad cross-resistance (24, 35). Among these mutations, D486N and F488L in heptad repeat B and T400I in the cysteine-rich region are some of the most commonly reported. The D486N mutation was induced by JNJ-2408068, ziresovir, and TMC353121 in in vitro experiments (15, 27, 28). Also, the F488L mutation was induced by presatovir and VP-14637 (29, 36), and the T400I mutation was induced by VP-14637 and P13 in in vitro experiments (36, 37). In addition, all of these mutations were selected by presatovir in clinical trials that included RSV-challenged volunteers or RSV-infected patients (22, 23). Our results suggested that the antiviral activity of TP0591816 was retained against RSV harboring the D486N, F488L, or T400I mutations. On the other hand, BMS-433771 and VP-14637 showed no activity against the resistant mutants. To the best of our knowledge, TP0591816 is the first fusion inhibitor identified that exhibits antiviral activity against these F protein mutants.
The results of the time-of-addition assay and the temperature shift assay indicate that TP0591816 inhibits the fusion of RSV with the cell membrane during viral cell entry. In addition, TP0591816 also inhibited cell-cell fusion when added after infection in the syncytium formation assay. These data suggest that TP0591816 inhibits the functions of the F protein. Genetic analysis of TP0591816-resistant mutants revealed that TP0591816 induced L141F mutation in the fusion peptide but no mutation in the cysteine-rich region or heptad repeat B, where the known fusion inhibitors are known to induce mutations. These results suggest that TP0591816 binds to a similar site of RSV F protein as the known fusion inhibitors, but that its manner of binding to this site is slightly different. While the binding conformation of TP0591816 was not investigated in this study, docking models of a macrocyclic pyrazolo[1,5-a]pyrimidine derivative, a prototype of TP0591816, with antiviral activity against the D486N mutant indicated that the compound was located in almost the same position in the wild-type and D486N F protein (26). From this information, it is supposed that TP0591816 can bind to the protein in the D486N, F488L, and T400I mutants in a similar manner to that in wild-type RSV. However, further studies are needed to elucidate the details of the binding mode of TP0591816 to understand the reason for its potent antiviral activity against these mutants.
TP0591816 not only showed antiviral activity in vitro, but also in a mouse infection model. TP0591816 reduced not only virus titers but also lung weights in RSV-infected mice. RSV infection induced inflammation in the lung (38), and it has been reported that lung weights of mice infected with RSV or influenza virus increase due to infiltrated inflammatory cells (39, 40). The results of our in vivo studies suggest that TP0591816 reduces RSV and, as a consequence, can suppress inflammation that causes damage to the respiratory system. The clinically effective doses of TP0591816 were predicted from the in vitro antiviral activities and the predicted plasma concentration-time profile, based on the paEC95. In the presatovir challenge study, the antiviral activity was found to be dependent on the trough plasma concentration (12). Although the trough plasma concentration of cohort 7 (10 mg as a loading dose, followed by 4 daily doses of 5 mg) in the challenge study did not achieve paEC95, the regimen significantly reduced both viral load and symptom score. Then, we set paEC95 as the effective trough plasma concentration. The predicted effective doses BID of TP0591816 were 26.9 mg for wild-type RSV and 55.5 to 955 mg for F protein mutants. Fusion inhibitors are well tolerated in general; for example, the leading compounds JNJ-53718678 and ziresovir showed no serious adverse events at doses of 1,000 and 1,200 mg in healthy adult subjects in a phase 1 study (41, 42). Also, the toxicity of TP0591816 is thought to be low because its cytotoxicity is as low as those of the other fusion inhibitors (CC50: >30,000 nM). Since TP0591816 is safe, similarly to the known fusion inhibitors, the predicted effective doses of TP0591816 are considered to be clinically realistic and tolerated.
In conclusion, we have identified a novel macrocyclic RSV fusion inhibitor, TP0591816, that appears to exert significant antiviral activity against not only wild-type RSV but also against a known fusion inhibitor-resistant RSV strain. Furthermore, TP0591816 also showed efficacy in a mouse model and the promise to treat patients with resistant RSV infection. Taken together, these findings suggest that TP0591816 is a promising new clinical candidate to overcome infection with fusion inhibitor-resistant RSV strains.
MATERIALS AND METHODS
Compounds.
TP0591816 was synthesized by the Department of Medicinal Chemistry, Taisho Pharmaceutical Co., Ltd. (Saitama, Japan). BMS-433771, VP-14637, ribavirin, and Synagis (palivizumab) were purchased from Sigma-Aldrich (St. Louis, MO), Cayman Chemical (Ann Arbor, MI), Tokyo Chemical Industry Co., Ltd. (Tokyo, Japan), and AbbVie GK (Tokyo, Japan), respectively.
Cells and virus.
HEp-2 cells were purchased from DS Pharma Biomedical Co., Ltd. (Osaka, Japan), and cultured in minimum essential medium (MEM) supplemented with 10% fetal bovine serum (FBS), 50 μg/ml gentamicin, and 600 μg/ml l-glutamine. RSV A2 (ATCC VR-1540, subgroup A) and RSV 18537 (ATCC VR-1580, subgroup B) were purchased from American Type Culture Collection (Manassas, VA). RSV A2 with the T400I, D486N, or F488L mutation in the F protein were selected in the presence of a pyrazolo[1,5-a]pyrimidine derivative 9c or 14e as described in our previous report (25).
Antiviral assay and cytotoxicity assay.
HEp-2 cells were seeded onto 96-well plates at 1 × 104 cells/well and cultured overnight, followed by washing with phosphate-buffered saline (PBS). Each test compound was diluted with MEM supplemented with 2% FBS, 100 U/ml penicillin, 100 μg/ml streptomycin, and 300 μg/ml l-glutamine (maintenance medium). The diluted compounds were added, and the cells were then infected with RSV A2 at a multiplicity of infection (MOI) of 0.5 or with the other strains at an MOI of 0.75. After incubation at 37°C in the presence of 5% CO2 for 4 days, the RSV-induced cytopathic effect (CPE) was determined by XTT assay using a cell proliferation kit II (Sigma-Aldrich). The cytotoxicity assay was performed in a similar manner but without the viral infection. The EC50 and EC95 values of the test compound were calculated by the least-squares method.
Time-of-addition assay.
HEp-2 cells were seeded onto 12-well plates and cultured until confluence and then were infected with RSV A2 at an MOI of 0.5. After 1 h of adsorption, the infected cells were washed three times with PBS, and each test compound was added at 0, 6, or 24 hpi. After incubation at 37°C in the presence of 5% CO2 for 3 days, the culture supernatants were collected, and the virus titers were determined by plaque assay. Serial 10-fold dilutions of the supernatants were added to the confluent HEp-2 cells in the 12-well plates. After 1 h of adsorption, culture medium containing 0.75% methylcellulose was added. After incubation at 37°C in the presence of 5% CO2 for 5 days, the cells were fixed with formalin and stained with hematoxylin and eosin. The virus titers were calculated by scoring the PFU.
Temperature shift assay.
HEp-2 cells were seeded on to 12-well plates and cultured until confluence and then infected with RSV A2 at an MOI of 0.5 in culture medium containing (for adding the test compound at 4°C) or not containing (for adding the test compound at 37°C) each test compound. After 1 h of adsorption at 4°C, the infected cells were washed three times with ice-cold PBS, and fresh medium containing (for adding the test compound at 37°C) or not containing (for adding the test compound at 4°C) each test compound was added. After incubation at 37°C in the presence of 5% CO2 for 3 days, the culture supernatants were collected and the virus titers were determined by plaque assay as described above.
Syncytium formation assay.
HEp-2 cells were seeded on to 96-well plates at 1 × 104 cells/well and cultured overnight, followed by washing with PBS. The cells were infected with RSV A2 at an MOI of 1 in maintenance medium. Each test compound was added to the wells at 24 hpi. The infected cells were incubated at 37°C in the presence of 5% CO2 for 3 days and then examined microscopically for syncytium formation.
Selection and genotypic characterization of the resistant RSV strains.
HEp-2 cells were seeded on to 12-well plates and cultured until confluence. The cells were washed with PBS and infected with RSV A2 at an MOI of 0.05 in the presence of 0.3 nM TP0591816. The infected cells were incubated at 37°C in the presence of 5% CO2 and harvested when a CPE was observed. The harvested cells were in a 10-fold volume of fresh medium containing either the same or a 3-fold higher concentration of TP0591816 and then transferred to newly cultured cells. The variants were passaged until the concentration reached 72.9 nM; control viruses were also passaged in parallel, but without the addition of TP0591816. The RNA of the wild-type and resistant viruses was reverse transcribed and amplified by PCR with primer pairs specific for the F gene. The PCR products were sequenced, and the mutation in the resistant virus was identified by comparing with the sequence of the wild-type virus (17).
Mouse model of RSV infection.
All of the in vivo experimental protocols used in this study were approved a priori by the Institutional Animal Care and Use Committee of Taisho Pharmaceutical Co., Ltd., and were in accordance with the Guidelines for Proper Conduct of Animal Experiments (Science Council of Japan, 2006). Female BALB/c mice (8 weeks of age) purchased from Charles River Laboratories Japan, Inc. (Kanagawa, Japan), were used. TP0591816 was dissolved in 10% hydroxypropyl-β-cyclodextrin and administered subcutaneously to the mice at the doses of 1, 10, and 100 mg/kg at 1 h prior to the virus inoculation. The mice were then anesthetized by intramuscular injection of ketamine (62.5 mg/kg) and xylazine (12.5 mg/kg) and inoculated intranasally with 1 × 107 PFU of RSV A2 in 100 μl of culture medium. On day 4 after the RSV challenge, the mice were euthanized, and the lungs were harvested and homogenized. The virus titers in the lung homogenates were determined by plaque assay as described above. The statistical significance of the difference versus the vehicle group was analyzed by Steel’s test.
PAMPA.
The permeability of TP0591816 was evaluated using the PAMPA Evolution instrument (Pion, Inc., Woburn, MA). The permeation of TP0591816 across an artificial membrane after a 4-h incubation at room temperature was quantified using a UV plate reader. The Papp at pH 6.2 was calculated using PAMPA Evolution software (Pion, Inc.).
In vitro metabolic stability study.
The metabolic stability of TP0591816 was determined using pooled human liver microsomes (XenoTech, LLC, Lenexa, KS). TP0591816 was incubated with a reaction mixture containing 0.25 mg/ml human liver microsomes and an NADPH generation system (1.3 mmol/liter β-NADP+, 3.3 mmol/liter glucose-6-phosphate, 3.3 mmol/liter MgCl2, and 0.4 U/ml glucose-6-phosphate dehydrogenase), in the presence of 100 mmol/liter potassium sodium phosphate buffer (pH 7.4). Reactions were initiated by the addition of the NADPH regenerating system. After incubation for 5, 10, 15, and 30 min at 37°C, reactions were terminated by the addition of acetonitrile/methanol (9:1, vol/vol), and the resultant mixtures were centrifuged at 3,974 × g for 10 min at 4°C. An aliquot of the supernatant was injected into a liquid chromatography-tandem mass spectrometry (LC-MS/MS) system. CLint was calculated as described by Obach (43). The fu,mic was estimated using the lipophilicity relationship algorithms described by Hallifax and Houston (44).
Plasma protein binding.
The plasma protein binding of TP0591816 in humans was evaluated by the equilibrium dialysis method. Equilibrium dialysis was conducted on a 96-well equilibrium dialysis device (HTDialysis, LLC, Gales Ferry, CT) with a 12- to 14-kDa-cutoff dialysis membrane. Blank plasma was spiked with TP0591816 dissolved in dimethyl sulfoxide (DMSO) at a final concentration of 1 μg/ml. The plasma samples were equilibrated with phosphate buffer (pH 7.4) at 37°C in 5% CO2 for 4 h. After the dialysis, the concentration of TP0591816 in the plasma (Cp) and phosphate buffer (Cu) were determined by LC-MS/MS. The fu,p was calculated as fu,P = Cu/Cp.
Pharmacokinetic evaluations in the monkey.
The pharmacokinetics and urinary excretion of TP0591816 in male cynomolgus monkeys were evaluated following intravenous administration of the drug at the dose of 0.25 mg/kg in a cocktail of compounds. TP0591816 was dissolved in 20% 2-hydroxypropyl-β-cyclodextrin and administered intravenously into the saphenous vein of fasting cynomolgus monkeys. Blood samples were obtained from the cephalic vein of the animals prior to the dosing, and at 0.0833, 0.25, 0.5, 1, 2, 4, 6, 8, and 24 h after the dosing; the samples were centrifuged to separate plasma. Urine was collected for 24 h after the dosing.
The concentration of TP0591816 in each plasma and urine sample was determined by liquid chromatography-tandem mass spectrometry (LC-MS/MS). The plasma concentration-time profiles of TP0591816 were analyzed by a noncompartmental analysis method using the pharmacokinetic analysis software Phoenix WinNonlin v8.0 (Certara, Princeton, NJ), and the systemic clearance (CL) and the apparent steady-state volume of distribution (Vdss) were calculated. The fraction excreted in urine was calculated by dividing the amount excreted in the urine by the administered dose. The CLR was calculated as the systemic clearance multiplied by the fraction excreted in the urine.
PBPK modeling.
Physiologically based pharmacokinetic (PBPK) modeling was performed using the Simcyp software v17 release 1 (Simcyp, Sheffield, UK) to predict the plasma concentration-time profile of TP0591816 in humans. The Simcyp model for TP0591816 was developed using the physicochemical properties, in vitro data, and predicted pharmacokinetic parameters. The Vdss (liter/kg) was predicted from simple allometric scaling of the data from monkeys using equation 1 (45).
| (1) |
The hepatic clearance was predicted from the intrinsic clearance in human liver microsomes. The renal clearance (CLR [liter/h/kg]) was predicted by simple allometric scaling of the data from monkeys using equation 2 (46).
| (2) |
Bioavailability was predicted using the PAMPA permeability and intrinsic clearance in human liver microsomes based on the assumption that TP0591816 was mainly metabolized by CYP3A4. The plasma concentration-time profile of TP0591816 after oral administration of a 200-mg dose was simulated for 100 subjects (10 trials × 10 subjects).
Determination of the effective clinical dose of TP0591816 for the treatment of RSV infection.
The effective clinical doses of TP0591816 for the treatment of infections caused by RSV A2 and D486N, F488L, or T400I mutants were predicted. The target trough plasma concentration for each virus strain was calculated as the paEC95 value using equation 3.
| (3) |
Plasma concentration of TP0591816 at 12 h after oral administration of 200 mg was simulated by PBPK modeling using Simcyp, as described previously, and the effective dose (mg/day) was predicted from equation 4, based on the assumption of linear PK.
| (4) |
ACKNOWLEDGMENTS
We thank S. Murakami and T. Aida for their technical support with the experiments.
No specific financial assistance from any funding agency in the public, commercial, or not-for-profit sectors was obtained for this research.
REFERENCES
- 1.Hall CB, Weinberg GA, Iwane MK, Blumkin AK, Edwards KM, Staat MA, Auinger P, Griffin MR, Poehling KA, Erdman D, Grijalva CG, Zhu Y, Szilagyi P. 2009. The burden of respiratory syncytial virus infection in young children. N Engl J Med 360:588–598. doi: 10.1056/NEJMoa0804877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Nair H, Nokes DJ, Gessner BD, Dherani M, Madhi SA, Singleton RJ, O’Brien KL, Roca A, Wright PF, Bruce N, Chandran A, Theodoratou E, Sutanto A, Sedyaningsih ER, Ngama M, Munywoki PK, Kartasasmita C, Simões EA, Rudan I, Weber MW, Campbell H. 2010. Global burden of acute lower respiratory infections due to respiratory syncytial virus in young children: a systematic review and meta-analysis. Lancet 375:1545–1555. doi: 10.1016/S0140-6736(10)60206-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Sigurs N, Aljassim F, Kjellman B, Robinson PD, Sigurbergsson F, Bjarnason R, Gustafsson PM. 2010. Asthma and allergy patterns over 18 years after severe RSV bronchiolitis in the first year of life. Thorax 65:1045–1052. doi: 10.1136/thx.2009.121582. [DOI] [PubMed] [Google Scholar]
- 4.Falsey AR, Walsh EE. 2000. Respiratory syncytial virus infection in adults. Clin Microbiol Rev 13:371–384. doi: 10.1128/cmr.13.3.371-384.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Homaira N, Rawlinson W, Snelling TL, Jaffe A. 2014. Effectiveness of palivizumab in preventing RSV hospitalization in high risk children: a real-world perspective. Int J Pediatr 2014:1–13. doi: 10.1155/2014/571609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Wegner S, Vann JJ, Liu G, Byrns P, Cypra C, Campbell W, Stiles A. 2004. Direct cost analyses of palivizumab treatment in a cohort of at-risk children: evidence from the North Carolina Medicaid Program. Pediatrics 114:1612–1619. doi: 10.1542/peds.2004-0959. [DOI] [PubMed] [Google Scholar]
- 7.Snell NJ. 2001. Ribavirin—current status of a broad spectrum antiviral agent. Expert Opin Pharmacother 2:1317–1324. doi: 10.1517/14656566.2.8.1317. [DOI] [PubMed] [Google Scholar]
- 8.Guerguerian AM, Gauthier M, Lebel MH, Farrell CA, Lacroix J. 1999. Ribavirin in ventilated respiratory syncytial virus bronchiolitis: a randomized, placebo-controlled trial. Am J Respir Crit Care Med 160:829–834. doi: 10.1164/ajrccm.160.3.9810013. [DOI] [PubMed] [Google Scholar]
- 9.Smith EC, Popa A, Chang A, Masante C, Dutch RE. 2009. Viral entry mechanisms: the increasing diversity of paramyxovirus entry. FEBS J 276:7217–7227. doi: 10.1111/j.1742-4658.2009.07401.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Grosfeld H, Hill MG, Collins PL. 1995. RNA replication by respiratory syncytial virus (RSV) is directed by the N, P, and L proteins; transcription also occurs under these conditions but requires RSV superinfection for efficient synthesis of full-length mRNA. J Virol 69:5677–5686. doi: 10.1128/JVI.69.9.5677-5686.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Merz DC, Scheid A, Choppin PW. 1980. Importance of antibodies to the fusion glycoprotein of paramyxoviruses in the prevention of spread of infection. J Exp Med 151:275–288. doi: 10.1084/jem.151.2.275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.DeVincenzo JP, Whitley RJ, Mackman RL, Scaglioni-Weinlich C, Harrison L, Farrell E, McBride S, Lambkin-Williams R, Jordan R, Xin Y, Ramanathan S, O’Riordan T, Lewis SA, Li X, Toback SL, Lin S-L, Chien JW. 2014. Oral GS-5806 activity in a respiratory syncytial virus challenge study. N Engl J Med 371:711–722. doi: 10.1056/NEJMoa1401184. [DOI] [PubMed] [Google Scholar]
- 13.Stevens M, Rusch S, DeVincenzo J, Kim YI, Harrison L, Meals EA, Boyers A, Fok-Seang J, Huntjens D, Lounis N, Mari N K, Remmerie B, Roymans D, Koul A, Verloes R. 2018. Antiviral Activity of oral JNJ-53718678 in healthy adult volunteers challenged with respiratory syncytial virus: a placebo-controlled study. J Infect Dis 218:748–756. doi: 10.1093/infdis/jiy227. [DOI] [PubMed] [Google Scholar]
- 14.DeVincenzo J, Tait D, Efthimiou J, Mori J, Kim YI, Thomas E, Wilson L, Harland R, Mathews N, Cockerill S, Powell K, Littler E. 2019. Human antiviral efficacy, safety and pharmacokinetics of RV521, an inhibitor of the RSV-F protein: a randomized, placebo-controlled, respiratory syncytial virus challenge study. Antimicrob Agents Chemother 64:e01884-19. doi: 10.1128/AAC.01884-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Zheng X, Gao L, Wang L, Liang C, Wang B, Liu Y, Feng S, Zhang B, Zhou M, Yu X, Xiang K, Chen L, Guo T, Shen HC, Zou G, Wu JZ, Yun H. 2019. Discovery of ziresovir as a potent, selective, and orally bioavailable respiratory syncytial virus fusion protein inhibitor. J Med Chem 62:6003–6014. doi: 10.1021/acs.jmedchem.9b00654. [DOI] [PubMed] [Google Scholar]
- 16.Cianci C, Yu KL, Combrink K, Sin N, Pearce B, Wang A, Civiello R, Voss S, Luo G, Kadow K, Genovesi EV, Venables B, Gulgeze H, Trehan A, James J, Lamb L, Medina I, Roach J, Yang Z, Zadjura L, Colonno R, Clark J, Meanwell N, Krystal M. 2004. Orally active fusion inhibitor of respiratory syncytial virus. Antimicrob Agents Chemother 48:413–422. doi: 10.1128/aac.48.2.413-422.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Douglas JL, Panis ML, Ho E, Lin KY, Krawczyk SH, Grant DM, Cai R, Swaminathan S, Cihlar T. 2003. Inhibition of respiratory syncytial virus fusion by the small molecule VP-14637 via specific interactions with F protein. JVI 77:5054–5064. doi: 10.1128/JVI.77.9.5054-5064.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.DeVincenzo JP, McClure MW, Symons JA, Fathi H, Westland C, Chanda S, Lambkin-Williams R, Smith P, Zhang Q, Beigelman L, Blatt LM, Fry J. 2015. Activity of oral ALS-008176 in a respiratory syncytial virus challenge study. N Engl J Med 373:2048–2058. doi: 10.1056/NEJMoa1413275. [DOI] [PubMed] [Google Scholar]
- 19.Coates M, Brookes D, Kim YI, Allen H, Fordyce EAF, Meals EA, Colley T, Ciana CL, Parra GF, Sherbukhin V, Stockwell JA, Thomas JC, Hunt SF, Anderson-Dring L, Onions ST, Cass L, Murray PJ, Ito K, Strong P, DeVincenzo JP, Rapeport G. 2017. Preclinical characterization of PC786, an inhaled small-molecule respiratory syncytial virus L protein polymerase inhibitor. Antimicrob Agents Chemother 61:e00737-17. doi: 10.1128/AAC.00737-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Chapman J, Abbott E, Alber DG, Baxter RC, Bithell SK, Henderson EA, Carter MC, Chambers P, Chubb A, Cockerill GS, Collins PL, Dowdell VC, Keegan SJ, Kelsey RD, Lockyer MJ, Luongo C, Najarro P, Pickles RJ, Simmonds M, Taylor D, Tyms S, Wilson LJ, Powell KL. 2007. RSV604, a novel inhibitor of respiratory syncytial virus replication. Antimicrob Agents Chemother 51:3346–3353. doi: 10.1128/AAC.00211-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Beigel JH, Nam HH, Adams PL, Krafft A, Ince WL, El-Kamary SS, Sims AC. 2019. Advances in respiratory virus therapeutics—a meeting report from the 6th ISIRV Antiviral Group conference. Antiviral Res 167:45–67. doi: 10.1016/j.antiviral.2019.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Chemaly RF, Dadwal SS, Bergeron A, Ljungman P, Kim YJ, Cheng GS, Pipavath SN, Limaye AP, Blanchard E, Winston DJ, Stiff PJ, Zuckerman T, Lachance S, Rahav G, Small CB, Mullane KM, Patron RL, Lee DG, Hirsch HH, Waghmare A, McKevitt M, Jordan R, Guo Y, German P, Porter DP, Gossage DL, Watkins TR, Marty FM, Chien JW, Boeckh M. 2019. A phase 2, randomized, double-blind, placebo-controlled trial of presatovir for the treatment of respiratory syncytial virus upper respiratory tract infection in hematopoietic-cell transplant recipients. Clin Infect Dis doi: 10.1093/cid/ciz1166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Marty FM, Chemaly RF, Mullane KM, Lee DG, Hirsch HH, Small CB, Bergeron A, Shoham S, Ljungman P, Waghmare A, Blanchard E, Kim YJ, McKevitt M, Porter DP, Jordan R, Guo Y, German P, Boeckh M, Watkins TR, Chien JW, Dadwal SS. 2019. A phase 2b, randomized, double-blind, placebo-controlled multicenter study evaluating antiviral effects, pharmacokinetics, safety, and tolerability of presatovir in hematopoietic cell transplant recipients with respiratory syncytial virus (RSV) infection of the lower respiratory tract. Clin Infect Dis doi: 10.1093/cid/ciz1167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Melero JA, Mas V. 2015. The Pneumovirinae fusion (F) protein: a common target for vaccines and antivirals. Virus Res 209:128–135. doi: 10.1016/j.virusres.2015.02.024. [DOI] [PubMed] [Google Scholar]
- 25.Yamaguchi-Sasaki T, Tamura Y, Ogata Y, Kawaguchi T, Kurosaka J, Sugaya Y, Iwakiri K, Busujima T, Takahashi R, Ueda-Yonemoto N, Tanigawa E, Abe-Kumasaka T, Sugiyama H, Kanuma K. 2020. Design and synthesis of 2-(1-alkylaminoalkyl)pyrazolo[1,5-a]pyrimidines as new respiratory syncytial virus fusion protein inhibitors. Chem Pharm Bull (Tokyo) 68:345–362. doi: 10.1248/cpb.c19-00895. [DOI] [PubMed] [Google Scholar]
- 26.Yamaguchi-Sasaki T, Tokura S, Ogata Y, Kawaguchi T, Sugaya Y, Takahashi R, Iwakiri K, Abe-Kumasaka T, Yoshida I, Arikawa K, Sugiyama H, Kanuma K. 2020. Discovery of a potent dual inhibitor of wild-type and mutant respiratory syncytial virus fusion proteins. ACS Med Chem Lett 11:1145–1151. doi: 10.1021/acsmedchemlett.0c00008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Andries K, Moeremans M, Gevers T, Willebrords R, Sommen C, Lacrampe J, Janssens F, Wyde PR. 2003. Substituted benzimidazoles with nanomolar activity against respiratory syncytial virus. Antiviral Res 60:209–219. doi: 10.1016/j.antiviral.2003.07.004. [DOI] [PubMed] [Google Scholar]
- 28.Roymans D, De Bondt HL, Arnoult E, Geluykens P, Gevers T, Van Ginderen M, Verheyen N, Kim H, Willebrords R, Bonfanti JF, Bruinzeel W, Cummings MD, van Vlijmen H, Andries K. 2010. Binding of a potent small-molecule inhibitor of six-helix bundle formation requires interactions with both heptad-repeats of the RSV fusion protein. Proc Natl Acad Sci U S A 107:308–313. doi: 10.1073/pnas.0910108106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Morton CJ, Cameron R, Lawrence LJ, Lin B, Lowe M, Luttick A, Mason A, McKimm-Breschkin J, Parker MW, Ryan J, Smout M, Sullivan J, Tucker SP, Young PR. 2003. Structural characterization of respiratory syncytial virus fusion inhibitor escape mutants: homology model of the F protein and a syncytium formation assay. Virology 311:275–288. doi: 10.1016/S0042-6822(03)00115-6. [DOI] [PubMed] [Google Scholar]
- 30.Stray K, Perron M, Porter DP, Anderson F, Lewis SA, Perry J, Miller M, Cihlar T, DeVincenzo J, Chien JW, Jordan R. 2020. Drug resistance assessment following administration of RSV fusion inhibitor presatovir to participants experimentally infected with respiratory syncytial virus. J Infect Dis 22:1468–1477. doi: 10.1093/infdis/jiaa028. [DOI] [PubMed] [Google Scholar]
- 31.Porter DP, Guo Y, Perry J, Gossage DL, Watkins TR, Chien JW, Jordan R. 2020. Assessment of drug resistance during phase 2b clinical trials of presatovir in adults naturally infected with respiratory syncytial virus. Antimicrob Agents Chemother 64:e02312-19. doi: 10.1128/AAC.02312-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Srinivasakumar N, Ogra PL, Flanagan TD. 1991. Characteristics of fusion of respiratory syncytial virus with HEp-2 cells as measured by R18 fluorescence dequenching assay. J Virol 65:4063–4069. doi: 10.1128/JVI.65.8.4063-4069.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Krusat T, Streckert HJ. 1997. Heparin-dependent attachment of respiratory syncytial virus (RSV) to host cells. Arch Virol 142:1247–1254. doi: 10.1007/s007050050156. [DOI] [PubMed] [Google Scholar]
- 34.Battles MB, Langedijk JP, Furmanova-Hollenstein P, Chaiwatpongsakorn S, Costello HM, Kwanten L, Vranckx L, Vink P, Jaensch S, Jonckers TH, Koul A, Arnoult E, Peeples ME, Roymans D, McLellan JS. 2016. Molecular mechanism of respiratory syncytial virus fusion inhibitors. Nat Chem Biol 12:87–93. doi: 10.1038/nchembio.1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Yan D, Lee S, Thakkar VD, Luo M, Moore ML, Plemper RK. 2014. Cross-resistance mechanism of respiratory syncytial virus against structurally diverse entry inhibitors. Proc Natl Acad Sci U S A 111:E3441–E3449. doi: 10.1073/pnas.1405198111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Perron M, Stray K, Kinkade A, Theodore D, Lee G, Eisenberg E, Sangi M, Gilbert BE, Jordan R, Piedra PA, Toms GL, Mackman R, Cihlar T. 2015. GS-5806 inhibits a broad range of respiratory syncytial virus clinical isolates by blocking the virus-cell fusion process. Antimicrob Agents Chemother 60:1264–1273. doi: 10.1128/AAC.01497-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Lundin A, Bergström T, Bendrioua L, Kann N, Adamiak B, Trybala E. 2010. Two novel fusion inhibitors of human respiratory syncytial virus. Antiviral Res 88:317–324. doi: 10.1016/j.antiviral.2010.10.004. [DOI] [PubMed] [Google Scholar]
- 38.Openshaw PJ, Tregoning JS. 2005. Immune responses and disease enhancement during respiratory syncytial virus infection. Clin Microbiol Rev 18:541–555. doi: 10.1128/CMR.18.3.541-555.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Shen C, Zhang Z, Xie T, Ji J, Xu J, Lin L, Yan J, Kang A, Dai Q, Dong Y, Shan J, Wang S, Zhao X. 2019. Rhein suppresses lung inflammatory injury induced by human respiratory syncytial virus through inhibiting NLRP3 inflammasome activation via NF-κB pathway in mice. Front Pharmacol 10:1600. doi: 10.3389/fphar.2019.01600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Fukao K, Noshi T, Yamamoto A, Kitano M, Ando Y, Noda T, Baba K, Matsumoto K, Higuchi N, Ikeda M, Shishido T, Akira A. 2019. Combination treatment with the cap-dependent endonuclease inhibitor baloxavir marboxil and a neuraminidase inhibitor in a mouse model of influenza A virus infection. J Antimicrob Chemother 74:654–662. doi: 10.1093/jac/dky462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Huntjens DRH, Ouwerkerk-Mahadevan S, Brochot A, Rusch S, Stevens M, Verloes R. 2017. Population pharmacokinetic modeling of JNJ-53718678, a novel fusion inhibitor for the treatment of respiratory syncytial virus: results from a phase I, double-blind, randomized, placebo-controlled first-in-human study in healthy adult subjects. Clin Pharmacokinet 56:1331–1342. doi: 10.1007/s40262-017-0522-8. [DOI] [PubMed] [Google Scholar]
- 42.McKimm-Breschkin JL, Jiang S, Hui DS, Beigel JH, Govorkova EA, Lee N. 2018. Prevention and treatment of respiratory viral infections: presentations on antivirals, traditional therapies and host-directed interventions at the 5th ISIRV Antiviral Group conference. Antiviral Res 149:118–1142. doi: 10.1016/j.antiviral.2017.11.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Obach RS. 1999. Prediction of human clearance of twenty-nine drugs from hepatic microsomal intrinsic clearance data: an examination of in vitro half-life approach and nonspecific binding to microsomes. Drug Metab Dispos 27:1350–1359. [PubMed] [Google Scholar]
- 44.Hallifax D, Houston JB. 2006. Binding of drugs to hepatic microsomes: comment and assessment of current prediction methodology with recommendation for improvement. Drug Metab Dispos 34:724–726; author reply 727. doi: 10.1124/dmd.105.007658. [DOI] [PubMed] [Google Scholar]
- 45.Lombardo F, Waters NJ, Argikar UA, Dennehy MK, Zhan J, Gunduz M, Harriman SP, Berellini G, Rajlic IL, Obach RS. 2013. Comprehensive assessment of human pharmacokinetic prediction based on in vivo animal pharmacokinetic data, part 1: volume of distribution at steady state. J Clin Pharmacol 53:167–177. doi: 10.1177/0091270012440281. [DOI] [PubMed] [Google Scholar]
- 46.Tang H, Hussain A, Leal M, Mayersohn M, Fluhler E. 2007. Interspecies prediction of human drug clearance based on scaling data from one or two animal species. Drug Metab Dispos 35:1886–1893. doi: 10.1124/dmd.107.016188. [DOI] [PubMed] [Google Scholar]





