Next Article in Journal
Metal-Free Air Oxidation in a Convenient Cascade Approach for the Access to Isoquinoline-1,3,4(2H)-triones
Next Article in Special Issue
Design, Synthesis, and Bioactivity Evaluation of Novel Isoxazole-Amide Derivatives Containing an Acylhydrazone Moiety as New Active Antiviral Agents
Previous Article in Journal
Catechins Controlled Bioavailability of Benzo[a]pyrene (B[α]P) from the Gastrointestinal Tract to the Brain towards Reducing Brain Toxicity Using the In Vitro Bio-Mimic System Coupled with Sequential Co-Cultures
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Synthesis and Biological Evaluation of NH2-Sulfonyl Oseltamivir Analogues as Influenza Neuraminidase Inhibitors

1
Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China
2
School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China
*
Authors to whom correspondence should be addressed.
Molecules 2019, 24(11), 2176; https://doi.org/10.3390/molecules24112176
Submission received: 13 May 2019 / Revised: 4 June 2019 / Accepted: 6 June 2019 / Published: 10 June 2019
(This article belongs to the Special Issue Antiviral Agents)

Abstract

:
A series of NH2-sulfonyl oseltamivir analogues were designed, synthesized, and their inhibitory activities against neuraminidase from H5N1 subtype evaluated. The results indicated that the IC50 value of compound 4a, an oseltamivir analogue via methyl sulfonylation of C5-NH2, was 3.50 μM. Molecular docking simulations suggested that 4a retained most of the interactions formed by oseltamivir carboxylate moieties and formed an additional hydrogen bond with the methylsulfonyl group. Meanwhile, 4a showed high stability towards human liver microsomes. More importantly, 4a without basic moieties is not a zwitterion as reported on the general structure of neuraminidase inhibitors. This research will provide valuable reference for the research of new types of neuraminidase inhibitors.

Graphical Abstract

1. Introduction

Influenza, an infectious disease caused by influenza viruses, has a high morbidity and mortality. In the 20th century, there were three pandemic outbreaks of influenza, for example, the “Spanish” influenza resulting in approximately 50 million deaths in 1918 and 1919 [1]. The pandemic nature of influenza presents a serious public health concern [2,3]. At present, there are two types of agents for influenza, adamantane-based M2 ion channel protein inhibitors and neuraminidase inhibitors (NAIs). The application of the former is limited due to adverse side effects and rapidly developed drug resistance. Thus the latter is widely used [4,5,6,7]. So far, four neuraminidase inhibitors, oseltamivir phosphate [6] and zanamivir hydrate [8] approved around the world, peramivir hydrate [9,10], and laninamivir octanoate [11] marketed only in a few countries, are available. Because of its convenient oral administration, oseltamivir has the largest market share [12,13].
Neuraminidase (NA) existing as a tetramer is responsible for removing sialic acid residues from receptors and facilitates virus release and spread to infect host cells [3,8,14]. The active site of NA is divided into five subsites (S1–S5) (Figure 1). S1, composed of three basic amino acids (Arg118, Arg292, and Arg371), has affinity for the acidic moieties of the inhibitors to form a salt bridge beneficial for potent inhibitory activity. S2, formed by three acidic amino acids (Glu119, Asp151, and Glu227), has affinity for basic moieties to form a salt bridge [7,15]. Therefore, powerful inhibitors are generally zwitterions with poor lipophilicity and oseltamivir carboxylate (OC, the active ingredient of oseltamivir as a prodrug) is no exception [16]. The non-zwitterionic NAIs have potential advantages for several aspects such as lipophilicity.
The 150-cavity close to S2, formed by residues of 147–152, offered new opportunities for the research of a new type of NAIs [7,14]. Guided by the 150-cavity, several oseltamivir derivatives substituted on the C5-amine have been reported. Xie and his colleagues revealed compounds A and B (Figure 2) with IC50 values of 1.9 nM and 2.1 nM against NAs from H5N1 (A/Chicken/China/1220/2012) respectively, which preliminarily showed that the 150-cavity was an advantage for enhancing inhibitory activity [17]. In 2010, Mohan et al. discovered compound C containing triazole with a Ki value of 72 nM. Saturation transfer difference (STD) NMR spectroscopic studies suggested that compound C interacted with the 150-cavity successfully [12]. Our laboratory discovered compound D (Figure 2) which showed powerful inhibitory activities against wild-type NAs with IC50 values similar to those of OC and enhanced inhibitory activities against mutant NAs in the process of searching for inhibitors targeting the 150-cavity. What is more, compound D without basic moieties was not consistent with the traditional structure–activity relationships in that a basic moiety is essential for powerful activity [18]. In consideration that sulfonamido (R1SO2NHR2) can act both as hydrogen bond donor and hydrogen bond acceptor, which may be beneficial for biological activity [19,20], we continue to search for NAIs without basic moieties. A series of oseltamivir analogues bearing sulfonamido were designed with the aim of the sulfonamido generating hydrogen bonds with the S2 and R (Figure 3) group interacting with the 150-cavity.

2. Results and Discussion

2.1. Synthesis

The synthetic approaches to oseltamivir were investigated, and several approaches were found to be innovative or interesting [21,22,23,24], however, we followed our previous semi-synthetic procedure for oseltamivir (2) [18]. The synthetic route of the target compounds is depicted in Scheme 1. Oseltamivir was reacted with the corresponding sulfonyl chlorides to obtain 3a3k [18,25,26]. The nitro fragment of 3i3k was reduced to amino by iron powder to afford intermediates 5i5k. Finally, 3a3k and 5i5k were treated with NaOH in aq. methanol to yield target compounds 4a4k and 6i6k via saponification. All compounds were confirmed by 1H-NMR, 13C-NMR, and HRMS (ESI).

2.2. Neuraminidase Enzyme Inhibitory Assay

Compounds 4a4k and 6i6k were evaluated as inhibitors of the NA from the H5N1 subtype of influenza A. The inhibition rates at 10 μM and 100 μM are shown in Table 1 [18]. The relatively good compounds such as compound 4a, 4h, 4i, and 6i were further screened to give IC50 values. The inhibitory activities varied from the sulfonyl fragments substituted on the amine of OC. Compounds 4a4e showed high to weak inhibitory activities. The length of the sulfonyl moieties played a crucial role in the inhibitory activities. The increased length of the substituents led to decreased inhibitions against NA as suggested by 4a4e. Compound 4a, possessing the shortest substituent, exhibited the most powerful neuraminidase inhibitory activity with an IC50 value of 3.50 μM (Table 2). Meanwhile, 4f as a fluoro-substituted congener of 4a did not exhibit good inhibitory activity, indicating fluorine substitution was harmful for interaction with amino acids near or belonging to S2. Compounds 4g4k and 6i6k bearing aromatic rings exhibited moderate to weak inhibitory activities. Compound 4g containing 4-acetylamido phenyl exerted little inhibition even at 100 μM. Compared with 4f, the inhibitory activity of compound 4h bearing trifluoromethyl was enhanced, and the IC50 value was 12.00 μM. Among the compounds 4i4k and 6i6k, ortho-substituted 4i and 6i showed more potent inhibitory activities than the meta-substituted and para-substituted counterparts. Inhibitory activities decreased in the order of ortho-, meta-, and para-position. Compared to the nitro, the amino group was more beneficial for inhibitory activity.
Compound 4a exerted the most powerful inhibitory activity. The inhibition of 4a was weaker than that of OC, but 4a without basic moieties is not a zwitterion, which is not consistent with traditional structure–activity relationships of NAIs.

2.3. Molecular Docking Model Analysis

As shown in Figure 4B, due to the added methylsulfonyl group the OC fragment of compound 4a did not overlap well with OC, so the interactions formed by the OC moiety with NA were discounted to a certain extent. It is a pity that on account of the short length of the methylsulfonyl compound 4a could not access the 150 cavity as expected. The oxygen of the sulfonyl of compound 4a generated a new hydrogen bond with Arg152 (Figure 4A). The new interactions generated by methylsulfonyl were not enough to make up for the lost or decreased ones formed by the C5 amine of the OC fragment with S2, which explained the weaker inhibitory activity of 4a compared to that of OC.

2.4. Metabolic Stability in Human Liver Microsomes In Vitro

Compound 4a with the most powerful inhibition was selected to test the metabolic stability in the presence of human liver microsomes in vitro and the control compounds of testosterone, diclofenac, and propafenone validated the assay [27,28,29,30]. The results are shown in Table 3. After incubating with human liver microsomes for 1 h, almost of all of 4a (101.6%) was detected and the T1/2 (half time) was more than 145 min, which indicated that 4a showed high stability towards human liver microsomes in vitro.

3. Materials and Methods

3.1. Chemistry

All of the raw materials and solvents were purchased from commercial suppliers. Melting points were determined in slides on a WRX-4 Micro melting point apparatus (Yice). High resolution mass spectra (HRMS) were recorded on an Agilent 6530 ultrahigh definition (UHD) accurate mass Q-TOF MS by ESI mode. The 1H-nuclear magnetic resonance (NMR) and 13C-NMR spectra were recorded on a Bruker ARX 600 MHz using tetramethylsilane as the internal standard. The reaction process was monitored by thin-layer chromatography (TLC) on silica gel GF254. Plates were visualized using UV light (254 nm). The purity of target compound was determined by HPLC. The detailed method is as follows: a Shimadzu (Kyoto, Japan) HPLC; Column: DIAMONSIL® C18, 250 mm × 4.6mm, 5 μm; mobile phase (isocratic elution): 50% acetonitrile (0.1% trifluoroacetic acid) for method A; 35% acetonitrile (0.1% trifluoroacetic acid) for method B; 25% acetonitrile (0.1% trifluoroacetic acid) for method C. Flow rate: 1 mL/min; Detector: UV (254 nm).

General Procedure for the Preparation of Compounds 4a4k and 6i6k

Oseltamivir (2, 312.4 mg, 1.0 mmol), TEA (208 μL, 1.5 mmol) and 10 mL CH2Cl2 were charged in a 50 mL round bottom flask, then the corresponding sulfonyl chloride (1.2 mmol) was added dropwise. The mixture was stirred at 0 °C until oseltamivir was completely consumed, as indicated by TLC analysis. The organic layer was washed successively with 1N HCl aqueous solution, saturated aq.Na2CO3, and brine. The organic layer was concentrated in vacuo. The crude product was purified by column chromatography to obtain one of the intermediates (3a3k).
One of intermediates 3a3k (1.0 mmol), 1N NaOH aqueous solution (2.5 mmol), methanol (15 mL) and deionized water (V (methanol): V (water) = 5:1) were added to a round bottom flask. The mixture was stirred at room temperature. Then the methanol was evaporated in vacuo and the residual solution was acidified with 1N HCl aqueous solution to pH 1 to 2. The precipitate was separated and filtered. Finally, one of the title compounds (4a4k) was obtained.
A mixture of one of compounds (3i3k) (1 mmol), iron powder (8 mmol) and NH4Cl (10 mmol) in 90% ethanol aqueous solution (30 mL) was stirred under reflux until the starting material was consumed completely, as indicated by TLC analysis. The precipitate was filtered, and the filtrate was evaporated in vacuo. The residue was extracted by dichloromethane until the new dichloromethane did not contain one of intermediates (5i5k) any more, and the organic layer was concentrated in vacuo to obtain one of the crude compounds (5i5k). Following the procedure for compounds 4a4k, the compounds (6i6k) were obtained. More detailed information can be found in the supplementary materials.
(3R, 4R, 5S)-4-acetamido-5-(methylsulfonyl)amino-3-(pentan-3-yloxy) cyclohex-1-enecarboxylic acid (4a). White solid, m.p. 212.9–215.2 °C, yield, 45%; 1H-NMR (600MHz, DMSO-d6): δ 12.61 (s, 1H), 7.86 (d, J = 9.2 Hz, 1H), 7.04 (d, J = 9.0 Hz, 1H), 6.60 (s, 1H), 4.11 (d, J = 8.7 Hz, 1H), 3.63 (dd, J = 20.2, 9.1 Hz, 1H), 3.36 (dt, J = 10.9, 5.6 Hz, 2H), 2.90 (s, 3H), 2.64 (dd, J = 17.5, 5.4 Hz, 1H), 2.28–2.22 (m, 1H), 1.84 (s, 3H), 1.45–1.35 (m, 4H), 0.84 (t, J = 7.4 Hz, 3H), 0.79 (t, J = 7.4 Hz, 3H); 13C-NMR (150 MHz, DMSO-d6): δ 170.10, 167.58, 138.09, 129.36, 81.45, 75.66, 54.20, 52.46, 41.88, 32.88, 26.17, 25.59, 23.46, 9.96, 9.38; ESI-HRMS (m/z), Calcd. for C15H26N2O6S[M − H]: 361.1439, found: 361.1452. Method C: retention time: 8.64 min, 96.7% purity.
(3R, 4R, 5S)-4-acetamido-5-(ethylsulfonyl)amino-3-(pentan-3-yloxy)cyclohex-1-enecarboxylic acid (4b). White solid, m.p. 187.7–190.0 °C, yield, 50%; 1H-NMR (600 MHz, DMSO-d6): δ 12.58 (s, 1H), 7.84 (d, J = 9.2 Hz, 1H), 7.00 (d, J = 9.2 Hz, 1H), 6.59 (s, 1H), 4.09 (d, J = 8.7 Hz, 1H), 3.63 (dd, J = 20.0, 9.2 Hz, 1H), 3.37–3.34 (m, 1H), 2.98 (q, J = 7.1 Hz, 2H), 2.63 (dd, J = 18.2, 5.8 Hz, 1H), 2.30–2.23 (m, 1H), 2.02–1.95 (m, 1H), 1.83 (s, 3H), 1.41 (ddd, J = 20.5, 13.0, 6.7 Hz, 4H), 1.17 (t, J = 7.3 Hz, 3H), 0.84 (t, J = 7.4 Hz, 3H), 0.79 (t, J = 7.4 Hz, 3H); 13C-NMR (150 MHz, DMSO-d6): δ 170.00, 167.58, 138.09, 129.34, 81.41, 75.73, 54.24, 52.29, 47.36, 33.11, 26.14, 25.55, 23.53, 9.97, 9.34, 8.65; ESI-HRMS (m/z), Calcd. for C16H28N2O6S[M − H]: 375.1590, found: 375.1618. Method B: retention time: 5.05 min, 99.4% purity
(3R, 4R, 5S)-4-acetamido-5-(n-propylsulfonyl)amino-3-(pentan-3-yloxy)cyclohex-1-enecarboxylic acid (4c). White solid, m.p. 196.3–198.6 °C, yield, 70%; 1H-NMR (600 MHz, DMSO-d6): δ 12.64 (s, 1H), 7.84 (d, J = 9.2 Hz, 1H), 6.99 (d, J = 9.2 Hz, 1H), 6.58 (s, 1H), 4.08 (d, J = 8.5 Hz, 1H), 3.63 (dd, J = 20.2, 9.1 Hz, 1H), 3.39–3.34 (m, 1H), 2.95 (tt, J = 7.3, 3.7 Hz, 2H), 2.63 (dd, J = 17.7, 5.4 Hz, 1H), 2.26 (ddt, J = 16.5, 10.2, 3.0 Hz, 1H), 1.82 (s, 3H), 1.68–1.60 (m, 2H), 1.49–1.34 (m, 4H), 0.97 (t, J = 7.5 Hz, 3H), 0.83 (t, J = 7.4 Hz, 3H), 0.79 (t, J = 7.4 Hz, 3H); 13C-NMR (150 MHz, DMSO-d6): δ 169.96, 167.77, 137.78, 129.64, 81.41, 75.78, 54.75, 54.21, 52.38, 33.19, 26.15, 25.57, 23.48, 17.51, 13.18, 9.97, 9.37; ESI-HRMS (m/z), Calcd. for C17H30N2O6S[M − H]: 389.1746, found: 389.1737. Method B: retention time: 7.05 min, 98.5% purity.
(3R, 4R, 5S)-4-acetamido-5-(n-butylsulfonyl)amino-3-(pentan-3-yloxy) cyclohex-1-enecarboxylic acid (4d). White solid, m.p. 183.3–184.4 °C, yield, 55%; 1H-NMR (600 MHz, DMSO-d6): δ 12.60 (s, 1H), 7.88 (d, J = 9.2 Hz, 1H), 6.99 (d, J = 9.2 Hz, 1H), 6.59 (s, 1H), 4.09 (d, J = 8.6 Hz, 1H), 3.64 (dd, J = 20.1, 9.0 Hz, 1H), 3.36 (dt, J = 10.8, 5.4 Hz, 2H), 3.03–2.91 (m, 2H), 2.64 (dd, J = 17.6, 5.3 Hz, 1H), 2.30–2.21 (m, 1H), 1.82 (s, 3H), 1.63–1.56 (m, 2H), 1.48–1.41 (m, 2H), 1.41–1.33 (m, 4H), 0.91–0.86 (m, 3H), 0.83 (t, J = 7.4 Hz, 3H), 0.79 (t, J = 7.4 Hz, 3H); 13C-NMR (150 MHz, DMSO-d6): δ 169.97, 167.55, 138.06, 129.33, 81.41, 75.71, 54.13, 52.77, 52.34, 33.18, 26.15, 25.87, 25.57, 23.45, 21.37, 14.05, 9.95, 9.36; ESI-HRMS (m/z), Calcd. for C18H32N2O6S[M − H]: 403.1903, found: 403.1932. Method B: retention time: 10.67 min, 99.5% purity.
(3R, 4R, 5S)-4-acetamido-5-(n-pentylsulfonyl)amino-3-(pentan-3-yloxy)cyclohex-1-enecarboxylic acid (4e). White solid, m.p. 179.1–182.8 °C, yield, 55%; 1H-NMR (600 MHz, DMSO-d6): δ 12.60 (s, 1H), 7.84 (d, J = 9.2 Hz, 1H), 6.99 (d, J = 9.2 Hz, 1H), 6.59 (s, 1H), 4.08 (d, J = 8.7 Hz, 1H), 3.63 (dd, J = 20.1, 9.1 Hz, 1H), 3.39–3.34 (m, 1H), 3.02–2.90 (m, 2H), 2.63 (dd, J = 17.7, 5.4 Hz, 1H), 2.29–2.22 (m, 1H), 1.82 (s, 3H), 1.66–1.58 (m, 2H), 1.49–1.36 (m, 4H), 1.33 (dd, J = 13.9, 7.1 Hz, 2H), 1.31–1.26 (m, 2H), 0.87 (t, J = 7.1 Hz, 3H), 0.83 (t, J = 7.4 Hz, 3H), 0.79 (t, J = 7.4 Hz, 3H); 13C-NMR (150 MHz, DMSO-d6): δ 169.94, 167.58, 138.05, 129.35, 81.43, 75.72, 54.19, 53.08, 52.30, 33.08, 30.29, 26.14, 25.57, 23.52, 23.46, 22.23, 14.21, 9.94, 9.36; ESI-HRMS (m/z), Calcd. for C19H34N2O6S[M − H]: 417.2059, found: 417.2032. Method A: retention time: 7.12 min, 95.7% purity.
(3R, 4R, 5S)-4-acetamido-5-((trifluoromethyl)sulfonyl)amino-3-(pentan-3-yloxy)cyclohex-1-enecarboxylic acid (4f). White solid, m.p. 193.1–195.4 °C, yield, 69%; 1H-NMR (600 MHz, DMSO-d6): δ 12.72 (s, 1H), 9.57 (d, J = 9.0 Hz, 1H), 7.93 (d, J = 9.4 Hz, 1H), 6.60 (s, 1H), 4.16 (d, J = 8.8 Hz, 1H), 3.76 (dd, J = 20.4, 9.2 Hz, 1H), 3.45–3.40 (m, 1H), 3.38 (dd, J = 11.1, 5.6 Hz, 1H), 2.59 (dd, J = 17.5, 5.6 Hz, 1H), 2.39–2.32 (m, 1H), 1.84 (s, 3H), 1.49–1.33 (m, 4H), 0.83 (t, J = 7.4 Hz, 3H), 0.77 (t, J = 7.4 Hz, 3H); 13C-NMR (150 MHz, DMSO-d6): δ 169.88, 167.31, 138.49, 128.48, 119.97 (q, J = 321.5 Hz), 81.71, 75.15, 54.54, 54.10, 31.64, 26.18, 25.59, 23.29, 9.87, 9.41; ESI-HRMS (m/z), Calcd. for C15H23F3N2O6S[M − H]: 415.1151, found: 415.1172. Method B: retention time: 16.50 min, 96.0% purity.
(3R, 4R, 5S)-4-acetamido-5-((4-(acetamido)pheny)sulfonyl)amino-3-(pentan-3-yloxy)cyclohex-1-enecarboxylic acid (4g). White solid, m.p. 236.7–238.6 °C, yield, 60%; 1H-NMR (600 MHz, DMSO-d6): δ 12.51 (s, 1H), 10.32 (s, 1H), 7.74 (d, J = 8.8 Hz, 2H), 7.71–7.66 (m, 3H), 7.49 (d, J = 8.6 Hz, 1H), 6.53 (s, 1H), 4.06 (d, J = 8.4 Hz, 1H), 3.62 (dd, J = 19.9, 9.0 Hz, 1H), 3.25–3.18 (m, 1H), 2.25 (dd, J = 17.8, 5.4 Hz, 1H), 2.12 (dd, J = 10.4, 7.5 Hz, 1H), 2.08 (s, 3H), 1.69 (s, 3H), 1.46–1.29 (m, 4H), 0.81 (t, J = 7.4 Hz, 3H), 0.75 (t, J = 7.4 Hz, 3H); 13C-NMR (150 MHz, DMSO-d6): δ 170.13, 169.44, 167.43, 143.01, 138.15, 136.25, 129.00, 127.87 (2C), 118.93 (2C), 81.49, 75.28, 54.12, 52.79, 31.71, 26.18, 25.61, 24.63, 23.36, 9.87, 9.46; ESI-HRMS (m/z), Calcd. for C22H31N3O7S[M − H]: 480.1804, found: 480.1838. Method B: retention time: 6.23 min, 99.2% purity.
(3R, 4R, 5S)-4-acetamido-5-((4-(trifluoromethyl)phenyl)sulfonyl)amino-3-(pentan-3-yloxy)cyclohex-1-enecarboxylic acid (4h). White solid, m.p. 211.9–213.6 °C, yield, 77%; 1H-NMR (600 MHz, DMSO-d6): δ 12.56 (s, 1H), 7.99 (d, J = 9.0 Hz, 4H), 7.66 (d, J = 9.1 Hz, 1H), 6.50 (s, 1H), 4.06 (d, J = 8.4 Hz, 1H), 3.62 (dd, J = 20.0, 9.0 Hz, 1H), 3.33 (dd, 1H), 2.27 (dd, J = 17.5, 5.5 Hz, 1H), 2.17–2.09 (m, 1H), 1.90 (s, 1H), 1.60 (s, 3H), 1.41–1.32 (m, 4H), 0.81 (t, J = 7.4 Hz, 3H), 0.75 (t, J = 7.4 Hz, 3H); 13C-NMR (150 MHz, DMSO-d6): δ 169.95, 146.75, 132.27 (q, J = 32.3 Hz,), 127.64 (2C), 126.86 (d, J = 3.6 Hz, 2C), 126.76, 124.95, 123.15, 121.34, 81.41, 75.45, 54.29, 53.20, 32.34, 26.19, 25.62, 23.14, 9.86, 9.42; ESI-HRMS (m/z), Calcd. for C21H27F3N2O6S[M − H]: 491.1464, found: 491.1464. Method A: retention time: 8.21 min, 98.6% purity.
(3R, 4R, 5S)-4-acetamido-5-((2-nitrophenyl)sulfonyl)amino-3-(pentan-3-yloxy)cyclohex-1-enecarboxylic acid (4i). White solid, m.p. 183.6–185.1 °C, yield, 70%; 1H-NMR (600 MHz, DMSO-d6): δ 12.58 (s, 1H), 8.03–8.00 (m, 1H), 7.97–7.94 (m, 2H), 7.89–7.83 (m, 2H), 7.73 (d, J = 9.1 Hz, 1H), 6.58 (s, 1H), 4.10 (d, J = 8.4 Hz, 1H), 3.71 (dd, J = 19.8, 9.0 Hz, 1H), 3.46 (ddd, J = 16.5, 10.4, 5.7 Hz, 1H), 3.38–3.34 (m, 1H), 2.40 (dd, J = 17.5, 5.5 Hz, 1H), 2.28–2.21 (m, 2H), 1.64 (s, 3H), 1.43–1.35 (m, 4H), 0.82 (t, J = 7.4 Hz, 3H), 0.75 (t, J = 7.4 Hz, 3H); 13C-NMR (150 MHz, DMSO-d6): δ 170.00, 167.40, 147.59, 138.11, 134.58, 134.33, 133.15, 130.23, 128.94, 124.66, 81.54, 75.17, 53.91, 53.19, 31.61, 26.16, 25.60, 23.18, 9.88, 9.42. ESI-HRMS (m/z), Calcd. for C20H27N3O8S[M − H]: 468.1441, found: 468.1458. Method A: Retention time: 6.45 min, 99.4% purity.
(3R, 4R, 5S)-4-acetamido-5-((3-nitrophenyl)sulfonyl)amino-3-(pentan-3-yloxy)cyclohex-1-enecarboxylic acid (4j). Faint yellow solid, m.p. 114.3–115.9 °C, yield, 60%; 1H-NMR (600 MHz, DMSO-d6) δ 8.50 (t, J = 1.8 Hz, 1H), 8.47 (dd, J = 8.2, 1.5 Hz, 1H), 8.19 (d, J = 7.9 Hz, 1H), 8.11 (s, 1H), 7.90 (t, J = 8.0 Hz, 1H), 7.66 (d, J = 9.1 Hz, 1H), 6.48 (s, 1H), 4.04 (d, J = 8.2 Hz, 1H), 3.62 (dd, J = 19.9, 9.2 Hz, 1H), 3.33–3.30 (m, 1H), 2.64 (dd, J = 13.8, 6.7 Hz, 1H), 2.30 (dd, J = 17.7, 5.2 Hz, 1H), 2.16–2.09 (m, 1H), 1.44–1.29 (m, 4H), 1.01 (t, J = 7.2 Hz, 3H), 0.81 (t, J = 7.4 Hz, 3H), 0.74 (t, J = 7.4 Hz, 3H); 13C-NMR (150 MHz, DMSO-d6): δ 169.94, 167.35, 148.24, 144.36, 138.15, 132.77, 131.81, 128.88, 127.27, 121.60, 81.51, 75.32, 54.07, 52.85, 31.94, 26.13, 25.59, 23.19, 9.84, 9.41; ESI-HRMS (m/z), Calcd. for C20H27N3O8S[M − H]: 468.1441, found: 468.1470. Method A: retention time: 7.00 min, 99.3% purity.
(3R, 4R, 5S)-4-acetamido-5-((4-nitrophenyl)sulfonyl)amino-3-(pentan-3-yloxy)cyclohex-1-enecarboxylic acid (4k). White solid, m.p. 187.4–189.2 °C, yield, 55%; 1H-NMR (600 MHz, DMSO-d6): δ 12.57 (s, 1H), 8.41 (d, J = 8.8 Hz, 2H), 8.12 (d, J = 8.8 Hz, 1H), 8.03 (d, J = 8.8 Hz, 2H), 7.69 (d, J = 9.1 Hz, 1H), 6.54 (s, 1H), 4.07 (d, J = 8.5 Hz, 1H), 3.64 (dd, J = 20.0, 9.0 Hz, 1H), 3.39–3.34 (m, 1H), 2.29 (dd, J = 17.6, 5.4 Hz, 1H), 2.19–2.11 (m, 1H), 1.64 (s, 3H), 1.46–1.29 (m, 4H), 0.81 (t, J = 7.4 Hz, 3H), 0.75 (t, J = 7.4 Hz, 3H); 13C-NMR (150 MHz, DMSO-d6): δ 170.03, 167.34, 149.74, 148.26, 138.15, 128.89, 128.35, 125.00, 81.48, 75.22, 54.15, 53.02, 31.95, 26.16, 25.61, 23.24, 9.85, 9.42; ESI-HRMS (m/z), Calcd. for C20H27N3O8S[M − H]: 468.1441, found: 468.1456. Method A: retention time: 7.22 min, 99.8% purity.
(3R, 4R, 5S)-4-acetamido-5-((2-aminophenyl)sulfonyl)amino-3-(pentan-3-yloxy)cyclohex-1-enecarboxylic acid (6i). White solid, m.p. 161.8–164.0 °C, yield, 58%; 1H-NMR (600 MHz, DMSO-d6): δ 12.28 (s, 1H), 7.65 (d, J = 9.2 Hz, 1H), 7.54–7.44 (m, 2H), 7.27–7.20 (m, 1H), 6.79 (d, J = 8.2 Hz, 1H), 6.58 (t, J = 7.5 Hz, 1H), 5.88 (s, 2H), 4.06 (d, J = 8.2 Hz, 1H), 3.62 (dd, J = 19.9, 9.1 Hz, 1H), 3.34 (s, 1H), 3.11 (s, 1H), 2.29 (dd, J = 17.7, 5.2 Hz, 1H), 2.15–2.05 (m, 1H), 1.90 (s, 3H), 1.78 (s, 3H), 1.46–1.30 (m, 4H), 0.81 (t, J = 7.4 Hz, 3H), 0.76 (t, J = 7.4 Hz, 3H); 13C-NMR (150 MHz, DMSO-d6): δ 170.20, 167.43, 146.52, 138.30, 133.85, 129.34, 129.02, 121.39, 117.21, 115.37, 81.51, 75.30, 54.13, 52.20, 31.43, 26.16, 25.61, 23.47, 9.87, 9.45. ESI-HRMS (m/z), Calcd. for C20H29N3O6S[M − H]: 438.1699, found: 438.1711. Method A: retention time: 5.99 min, 98.9% purity.
(3R, 4R, 5S)-4-acetamido-5-((3-aminophenyl)sulfonyl)amino-3-(pentan-3-yloxy)cyclohex-1-enecarboxylic acid (6j). White solid, m.p. 177.4–178.1 °C, yield, 40%; 1H-NMR (600 MHz, DMSO-d6): δ 7.25 (d, J = 7.7 Hz, 1H), 7.16 (t, J = 7.9 Hz, 1H), 6.94 (t, J = 2.0 Hz, 1H), 6.86 (dd, J = 4.6, 3.8 Hz, 1H), 6.72 (ddd, J = 8.1, 2.2, 0.8 Hz, 1H), 6.30 (s, 1H), 5.52 (s, 1H), 4.01 (d, J = 8.1 Hz, 2H), 3.59 (dd, J = 19.9, 8.9 Hz, 1H), 3.46–3.40 (m, 1H), 3.13 (s, 1H), 2.31 (dd, J = 17.7, 5.2 Hz, 1H), 2.10–2.03 (m, 1H), 1.74 (s,3H), 1.46–1.28 (m, 4H), 0.81 (t, J = 7.4 Hz, 3H), 0.75 (t, J = 7.4 Hz, 3H).; 13C-NMR (150 MHz, DMSO-d6): δ 170.43, 168.63, 149.70, 143.05, 135.11, 131.90, 129.89, 117.47, 113.54, 111.29, 81.32, 75.61, 54.52, 53.38, 32.39, 26.29, 25.68, 23.35, 9.86, 9.51. ESI-HRMS (m/z), Calcd. for C20H29N3O6S[M − H]: 438.1699, found: 438.1713. Retention time: 4.0 min, 99.2% purity.
(3R, 4R, 5S)-4-acetamido-5-((4-aminophenyl)sulfonyl)amino-3-(pentan-3-yloxy)cyclohex-1-enecarboxylic acid (6k). Faint yellow solid, m.p. 176.7–181.4 °C, yield, 40%; 1H-NMR (600 MHz, DMSO-d6): δ 7.63 (d, J = 9.1 Hz, 1H), 7.38 (d, J = 8.7 Hz, 2H), 6.97 (d, J = 8.2 Hz, 1H), 6.58 (d, J = 8.7 Hz, 2H), 6.37 (s, 1H), 5.89 (s, 2H), 4.01 (d, J = 8.1 Hz, 1H), 3.62–3.54 (m, 1H), 3.33 (dt, J = 11.2, 5.6 Hz, 1H), 3.10–3.03 (m, 1H), 2.32 (dd, J = 17.8, 5.5 Hz, 1H), 2.06 (ddd, J = 10.2, 8.7, 5.0 Hz, 1H), 1.74 (s, 3H), 1.46–1.28 (m, 4H), 0.81 (t, J = 7.4 Hz, 3H), 0.75 (t, J = 7.4 Hz, 3H); 13C-NMR (150 MHz, DMSO-d6): δ 170.25, 167.50, 152.78, 138.07, 129.15, 128.69 (2C), 127.45, 113.08 (2C), 81.47, 75.26, 52.60, 46.09, 31.67, 26.20, 25.63, 23.41, 9.86, 9.48; ESI-HRMS (m/z), Calcd. for C20H29N3O6S[M − H]: 438.1699, found: 438.1714. Method B: retention time: 5.54 min, 97.9% purity.

3.2. Biological Evaluation

3.2.1. Neuraminidase Enzyme Inhibitory Assay

The H5N1 neuraminidase (A/Anhui/2005) was purchased from Sino Biological Inc (Beijing, China). OC as a positive control was purchased from MedChemExpress (Monmouth Junction, NJ, USA). Fluorogenic substrate MUNANA (2′-(4-methylumbelliferyl)-α-N-acetylneuraminic acid) was purchased from Sigma-Aldrich (Munich, Germany). The procedure followed previous method [19].

3.2.2. Metabolic Stability Assay

The human liver microsomes were purchased from BD (San Jose, USA). NADPH was purchased from Sigma-Aldrich (Munich, Germany). Two parallel assays with and without NADPH regenerating system were determined.
The compound 4a and control compounds (testosterone, diclofenac, or propafenone) were prepared by dilution of reaction buffer, and the final concentration of human liver microsomes was 0.5 mg/mL. Incubation was carried out in a thermostat at 37 °C and started by the addition of the appropriate compound. The samples were taken at 0, 5, 10, 20, 30, and 60 min. The reaction was terminated by the addition of stop solution. After collection, samples were centrifuged (20 min, 4000 rpm), then the centrifuged supernatant was directly analyzed by using LC-MS/MS analysis. Metabolic half-time (T1/2) was calculated by using the equation of first order kinetics.

4. Conclusions

In summary, a series of oseltamivir analogues bearing the sulfonamido group were designed based on our previous work on potent neuraminidase inhibitors without basic moieties and with the opportunities offered by the 150-cavity. Among these analogues, compound 4a showed the most potent inhibition against NA from H5N1 subtype with an IC50 value of 3500 nM. Molecular docking simulations revealed that 4a retained most of the interactions formed by the OC fragment and generated a new hydrogen bond. Due to the short length of the methylsulfonyl group, 4a did not get access to the 150-cavity as expected. Meanwhile, 4a exhibited high metabolic stability against human liver microsomes in vitro. What is more, compound 4a without basic moieties is not consistent with traditional inhibitors as zwitterions. Thus this study has enriched the structure types of NAIs and may provide valuable reference for the discovery of new types of NAIs.

Supplementary Materials

The supplementary materials (Figures S1–S28) are available online: the spectrum of compounds 4a4k and 6i6k.

Author Contributions

Investigation, Y.H., Z.L. and H.Z. (Hongqian Zhao); Methodology, B.C.; Supervision, P.Q. and Y.T.; Validation, H.Z. (Hongxi Zhu).

Funding

This work was financially supported by the Foundation of Shenyang Science and Technology Bureau (NO. F13-196-9-00).

Acknowledgments

We gratefully acknowledge the financial support from the Foundation of Shenyang Science and Technology Bureau (NO. F13-196-9-00).

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

NAneuraminidase
NAsneuraminidases
NAIsneuraminidase inhibitors
OCoseltamivir carboxylate
TEAtriethylamine

References

  1. Taubenberger, J.K.; Morens, D.M. 1918 Influenza: The Mother of all Pandemics. Emerg. Infect. Dis. 2006, 12, 15–22. [Google Scholar] [CrossRef] [PubMed]
  2. Salomon, R.; Webster, R.G. The Influenza Virus Enigma. Cell 2009, 136, 402–410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Adabala, P.J.P.; LeGresley, E.B.; Bance, N.; Niikura, M.; Pinto, B.M. Exploitation of the Catalytic Site and 150 Cavity for Design of Influenza A Neuraminidase Inhibitors. J. Org. Chem. 2013, 78, 10867–10877. [Google Scholar] [CrossRef] [PubMed]
  4. Clercq, E.D. Antiviral agents active against influenza a viruse. Nat. Rev. Drug Discov. 2006, 5, 1015–1025. [Google Scholar] [CrossRef] [PubMed]
  5. Kotthaus, J.; Riebling, L.; Kotthaus, J.; Müller-Fielitz, H.; Seidel, N.; Clement, B.; Schade, D.; Raasch, W.; Koch, O.; Schmidtke, M. Development of Novel Potent Orally Bioavailable Oseltamivir Derivatives Active against Resistant Influenza A. J. Med. Chem. 2014, 57, 759–769. [Google Scholar]
  6. Lew, W.; Chen, X.; Kim, C. Discovery and Development of GS 4104 (oseltamivir) An Orally Active Influenza Neuraminidase Inhibitor. Curr. Med. Chem. 2000, 7, 663–672. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Das, K. Antivirals Targeting Influenza A Virus. J. Med. Chem. 2012, 55, 6263–6277. [Google Scholar] [CrossRef]
  8. Ye, D.; Shin, W.-J.; Li, N.; Tang, W.; Feng, E.; Li, J.; He, P.-L.; Zuo, J.-P.; Kim, H.; Nam, K.-Y.; et al. Synthesis of C-4-modified zanamivir analogs as neuraminidase inhibitors and their anti-AIV activities. Eur. J. Med. Chem. 2012, 54, 764–770. [Google Scholar] [CrossRef]
  9. McLaughlin, M.M.; Skoglund, E.W.; Ison, M.G. Peramivir: An intravenous neuraminidase inhibitor. Expert Opin. Pharmacother. 2015, 16, 1889–1900. [Google Scholar] [CrossRef]
  10. Li, Y.; Wang, Z.Y.; Xin, L.; Wei, G.; Xie, X.; Yang, Y.; Zhong, W.; Zheng, A. In Vitro Evaluation of Absorption Characteristics of Peramivir for Oral Delivery. Eur. J. Drug. Metab. Pharmacokinet. 2017, 42, 757–765. [Google Scholar] [CrossRef]
  11. Toyama, K.; Furuie, H.; Ishizuka, H. Safety and Pharmacokinetics of Nebulized Laninamivir Octanoate, A Long Acting Neuraminidase Inhibitor, In Healthy Subjects. Clin. Ther. 2017, 39, e25–e26. [Google Scholar] [CrossRef]
  12. Mohan, S.; McAtamney, S.; Haselhorst, T.; Von Itzstein, M.; Pinto, B.M. Carbocycles Related to Oseltamivir as Influenza Virus Group-1-Specific Neuraminidase Inhibitors. Binding to N1 Enzymes in the Context of Virus-like Particles. J. Med. Chem. 2010, 53, 7377–7391. [Google Scholar] [CrossRef] [PubMed]
  13. Cheng, T.-J.R.; Weinheimer, S.; Tarbet, E.B.; Jan, J.-T.; Cheng, Y.-S.E.; Shie, J.-J.; Chen, C.-L.; Chen, C.-A.; Hsieh, W.-C.; Huang, P.-W.; et al. Development of Oseltamivir Phosphonate Congeners as Anti-Influenza Agents. J. Med. Chem. 2012, 55, 8657–8670. [Google Scholar] [CrossRef] [PubMed]
  14. Russell, R.J.; Haire, L.F.; Stevens, D.J.; Collins, P.J.; Lin, Y.P.; Blackburn, G.M.; Hay, A.J.; Gamblin, S.J.; Skehel, J.J. The structure of H5N1 avian influenza neuraminidase suggests new opportunities for drug design. Nature 2006, 443, 45–49. [Google Scholar] [CrossRef]
  15. Maring, C.J.; Stoll, V.S.; Zhao, C.; Sun, M.; Krueger, A.C.; Stewart, K.D.; Madigan, D.L.; Kati, W.M.; Xu, Y.; Carrick, R.J.; et al. Structure-Based Characterization and Optimization of Novel Hydrophobic Binding Interactions in a Series of Pyrrolidine Influenza Neuraminidase Inhibitors. J. Med. Chem. 2005, 48, 3980–3990. [Google Scholar] [CrossRef] [PubMed]
  16. Proudfoot, J.R. The Evolution of Synthetic Oral Drug Properties. Bioorg. Med. Chem. Lett. 2005, 15, 1087–1090. [Google Scholar] [CrossRef] [PubMed]
  17. Xie, Y.; Xu, D.; Huang, B.; Ma, X.; Qi, W.; Shi, F.; Liu, X.; Zhang, Y.; Xu, W. Discovery of N-Substituted Oseltamivir Derivatives as Potent and Selective Inhibitors of H5N1 Influenza Neuraminidase. J. Med. Chem. 2014, 57, 8445–8458. [Google Scholar] [CrossRef] [PubMed]
  18. Wang, K.; Yang, F.; Wang, L.; Liu, K.; Sun, L.; Lin, B.; Hu, Y.; Wang, B.; Cheng, M.; Tian, Y. Synthesis and biological evaluation of NH 2 -acyl oseltamivir analogues as potent neuraminidase inhibitors. Eur. J. Med. Chem. 2017, 141, 648–656. [Google Scholar] [CrossRef] [PubMed]
  19. Gulçin, I.; Taslimi, P.; Gülçın, I. Sulfonamide inhibitors: A patent review 2013-present. Expert Opin. Ther. Pat. 2018, 28, 541–549. [Google Scholar] [CrossRef]
  20. Rakesh, K.P.; Wang, S.-M.; Leng, J.; Ravindar, L.; Asiri, A.M.; Marwani, H.M.; Qin, H.-L. Recent development of sulfonyl or sulfonamide hydrids as potential anticancer agents: A key review. Anti-Cancer Agents Med. Chem. 2018, 18, 488–505. [Google Scholar] [CrossRef]
  21. Magano, J. Synthetic Approaches to the Neuraminidase Inhibitors Zanamivir (Relenza) and Oseltamivir Phosphate (Tamiflu) for the Treatment of Influenza. Chem. Rev. 2009, 109, 4398–4438. [Google Scholar] [CrossRef] [PubMed]
  22. Shie, J.-J.; Fang, J.-M.; Wang, S.-Y.; Tsai, K.-C.; Cheng, Y.-S.E.; Yang, A.-S.; Hsiao, S.-C.; Su, C.-Y.; Wong, C.-H. Synthesis of Tamiflu and its Phosphonate Congeners Possessing Potent Anti-Influenza Activity. J. Am. Chem. Soc. 2007, 129, 11892–11893. [Google Scholar] [CrossRef] [PubMed]
  23. Silva, S.; Maycock, C.D. Formal enantioselective syntheses of oseltamivir and tamiphosphor. Org. Chem. Front. 2017, 4, 236–240. [Google Scholar] [CrossRef]
  24. Hajzer, V.; Fišera, R.; Latika, A.; Durmis, J.; Kollár, J.; Frecer, V.; Tučeková, Z.; Miertuš, S.; Kostolanský, F.; Varečková, E.; et al. Stereoisomers of oseltamivir—Synthesis, in silico prediction and biological evaluation. Org. Biomol. Chem. 2017, 15, 1828–1841. [Google Scholar] [CrossRef] [PubMed]
  25. Wang, Y.-J.; Zhou, D.-G.; He, F.-C.; Chen, J.-X.; Chen, Y.-Z.; Gan, X.-H.; Hu, D.-Y.; Song, B.-A. Synthesis and antiviral bioactivity of novel chalcone derivatives containing purine moiety. Chin. Chem. Lett. 2018, 29, 127–130. [Google Scholar] [CrossRef]
  26. Wan, Y.; Auberger, N.; Thétiot-Laurent, S.; Bouillère, F.; Zulauf, A.; He, J.; Courtiol-Legourd, S.; Guillot, R.; Kouklovsky, C.; Combes, S.C.D.; et al. Constrained Cyclic β,γ-Diamino Acids from Glutamic Acid: Synthesis of Both Diastereomers and Unexpected Kinetic Resolution. Eur. J. Org. Chem. 2018, 2018, 329–340. [Google Scholar] [CrossRef]
  27. Shi, D.; Yang, J.; Yang, D.; Lecluyse, E.L.; Black, C.; You, L.; Akhlaghi, F.; Yan, B. Anti-Influenza Prodrug Oseltamivir Is Activated by Carboxylesterase Human Carboxylesterase 1, and the Activation Is Inhibited by Antiplatelet Agent Clopidogrel. J. Pharmacol. Exp. Ther. 2006, 319, 1477–1484. [Google Scholar] [CrossRef] [PubMed]
  28. Sweeny, D.J.; Lynch, G.; Bidgood, A.M.; Lew, W.; Wang, K.Y.; Cundy, K.C. Metabolism of the influenza neuraminidase inhibitor prodrug oseltamivir in the rat. Drug Metab. Dispos. 2000, 28, 737–741. [Google Scholar] [PubMed]
  29. Sławiński, J.; Szafrański, K.; Pogorzelska, A.; Żołnowska, B.; Kawiak, A.; Macur, K.; Belka, M.; Bączek, T. Novel 2-benzylthio-5-(1,3,4-oxadiazol-2-yl)benzenesulfonamides with anticancer activity: Synthesis, QSAR study, and metabolic stability. Eur. J. Med. Chem. 2017, 132, 236–248. [Google Scholar] [CrossRef] [PubMed]
  30. Zhao, Y.; Bai, L.; Liu, L.; McEachern, D.; Stuckey, J.A.; Meagher, J.L.; Yang, C.-Y.; Ran, X.; Zhou, B.; Hu, Y.; et al. Structure-Based Discovery of 4-(6-Methoxy-2-methyl-4-(quinolin-4-yl)-9 H -pyrimido[4,5- b ]indol-7-yl)-3,5-dimethylisoxazole (CD161) as a Potent and Orally Bioavailable BET Bromodomain Inhibitor. J. Med. Chem. 2017, 60, 3887–3901. [Google Scholar] [CrossRef] [PubMed]
Sample Availability: Samples of the compounds 4a4k, 6i6k are available from the authors.
Figure 1. 2D diagram of S1–S5 of the active site with oseltamivir carboxylate (OC).
Figure 1. 2D diagram of S1–S5 of the active site with oseltamivir carboxylate (OC).
Molecules 24 02176 g001
Figure 2. Chemical structures of compound A, B, C, and D, OC.
Figure 2. Chemical structures of compound A, B, C, and D, OC.
Molecules 24 02176 g002
Figure 3. The general structure of designed compounds.
Figure 3. The general structure of designed compounds.
Molecules 24 02176 g003
Scheme 1. Synthetic route of target compounds (4a4k and 6i6k): Reagents and conditions: (a) corresponding sulfonyl chloride, TEA, CH2Cl2, 0 °C; (b) NaOH, CH3OH/H2O; (c) Fe, NH4Cl, 90% CH3CH2OH/H2O; (d) NaOH, CH3OH/H2O.
Scheme 1. Synthetic route of target compounds (4a4k and 6i6k): Reagents and conditions: (a) corresponding sulfonyl chloride, TEA, CH2Cl2, 0 °C; (b) NaOH, CH3OH/H2O; (c) Fe, NH4Cl, 90% CH3CH2OH/H2O; (d) NaOH, CH3OH/H2O.
Molecules 24 02176 sch001
Figure 4. (A) The docking modes of compound 4a (purple) with NA from H5N1 (PDB code 2HU4, downloaded from RCSB PDB (http://www.rcsb.org/). This figure was done by Glide program in MAESTRO software. (B) Molecular surface representation of the neuraminidase active site with OC (green) and 4a (purple), and the 150-cavity indicated. This figure was carried out by Chimera.
Figure 4. (A) The docking modes of compound 4a (purple) with NA from H5N1 (PDB code 2HU4, downloaded from RCSB PDB (http://www.rcsb.org/). This figure was done by Glide program in MAESTRO software. (B) Molecular surface representation of the neuraminidase active site with OC (green) and 4a (purple), and the 150-cavity indicated. This figure was carried out by Chimera.
Molecules 24 02176 g004
Table 1. The inhibition rates of synthesized compounds against NA from H5N1a subtype.
Table 1. The inhibition rates of synthesized compounds against NA from H5N1a subtype.
Compounds10 μM100 μMCompounds10 μM100 μM
4a73.9%91.8%4h40.4%79.2%
4b50.3%85.1%4i32.0%70.1%
4c43.2%56.5%4j20.3%33.8%
4d30.3%53.5%4k0.5%38.9%
4e28.5%50.5%6i63.8%86.6%
4f24.8%49.7%6j37.3%52.7%
4gNDbND6kND12.5%
OC91.0%95.0%
a A/Anhui/2005(H5N1). b Not Determined.
Table 2. The IC50 valuesa of several compounds against neuraminidase from H5N1b subtype.
Table 2. The IC50 valuesa of several compounds against neuraminidase from H5N1b subtype.
CompoundsOC4a4h4i6i
IC50/μM0.21 ± 0.0213.50 ± 0.1712.00 ± 2.4920.74 ± 1.148.50 ± 0.63
a IC50 is compound concentration causing 50% inhibition of NA based on the inhibition rates of 7 different concentrations, values are the mean of three independent experiments. b A/Anhui/2005(H5N1).
Table 3. Human liver microsomal stability of compound 4a.
Table 3. Human liver microsomal stability of compound 4a.
CompoundsMicrosomal Stability T1/2 (min)Remaining (T = 60 min)
4a>145101.6%
Testosterone12.53.8%
Diclofenac9.21.1%
Propafenone5.70.1%

Share and Cite

MDPI and ACS Style

Hu, Y.; Chen, B.; Lei, Z.; Zhao, H.; Zhu, H.; Quan, P.; Tian, Y. Synthesis and Biological Evaluation of NH2-Sulfonyl Oseltamivir Analogues as Influenza Neuraminidase Inhibitors. Molecules 2019, 24, 2176. https://doi.org/10.3390/molecules24112176

AMA Style

Hu Y, Chen B, Lei Z, Zhao H, Zhu H, Quan P, Tian Y. Synthesis and Biological Evaluation of NH2-Sulfonyl Oseltamivir Analogues as Influenza Neuraminidase Inhibitors. Molecules. 2019; 24(11):2176. https://doi.org/10.3390/molecules24112176

Chicago/Turabian Style

Hu, Yaping, Binfeng Chen, Zaiqiang Lei, Hongqian Zhao, Hongxi Zhu, Peng Quan, and Yongshou Tian. 2019. "Synthesis and Biological Evaluation of NH2-Sulfonyl Oseltamivir Analogues as Influenza Neuraminidase Inhibitors" Molecules 24, no. 11: 2176. https://doi.org/10.3390/molecules24112176

Article Metrics

Back to TopTop