Chemical and Pharmaceutical Bulletin
Online ISSN : 1347-5223
Print ISSN : 0009-2363
ISSN-L : 0009-2363
Regular Articles
Chemical Hybridization of Vizantin and Lipid A to Generate a Novel LPS Antagonist
Hirofumi Yamamoto Masataka OdaMarina KannoShota TamashiroIkuko TamuraToshihiko YonedaNaoto YamasakiHisanori DomonMayo NakanoHironobu TakahashiYutaka TeraoYusuke KasaiHiroshi Imagawa
Author information
JOURNAL FREE ACCESS FULL-TEXT HTML
Supplementary material

2016 Volume 64 Issue 3 Pages 246-257

Details
Abstract

Lipopolysaccharide (LPS) antagonists have attracted considerable interest as promising candidates for the treatment of severe sepsis triggered by Gram-negative bacteria. In this article, we describe the development of a novel LPS antagonist based on chemical hybridization of vizantin and the hydrophobic molecular unit of LPS (lipid A). Vizantin, 6,6′-bis-O-(3-nonyldodecanoyl)-α,α′-trehalose, was designed as an immunostimulator from a structure–activity relationship (SAR) study with trehalose 6,6′-dicorynomycolate (TDCM). Our recent study indicated that vizantin displays adjuvant activity by specifically binding to the Toll-like receptor 4 (TLR4)/MD2 protein complex. Because lipid A unit (or LPS) is also known to trigger an inflammatory response via the same TLR4/MD2 complex as vizantin, we designed a hybrid compound of vizantin and lipid A with the aim of developing a novel biofunctional glycolipid. Focusing on the antagonism to Escherichia coli LPS in an in vitro model with human macrophages (THP-1 cells), we identified a potent LPS antagonist among the synthesized hybrid compounds. The novel LPS antagonist effectively inhibited LPS-induced release of tumor necrosis factor-alpha (TNF-α) in a dose-dependent manner with an IC50 value of 3.8 nM, making it a candidate for the treatment drug of Gram-negative sepsis and/or septic shock.

Gram-negative sepsis and septic shock are serious conditions caused by the release of bacterial endotoxin.14) Endotoxin, which is also called lipopolysaccharide (LPS), is a structural component of the outer membrane of Gram-negative bacteria that is released when the cells become disrupted.5) LPS is known to activate immune cells, such as macrophages and monocytes, by binding of the lipid A (1) molecular unit (i.e., the hydrophobic domain of LPS) to the complex protein of Toll-like receptor 4 (TLR4) and MD2. This binding event results in the release of various cytokines, including tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6), from the immune cells.68) While the immune response with lipid A brings about the onset of the host defense to Gram-negative infection, the sudden release of high levels of cytokines and cellular mediators can trigger many pathophysiological events including fever, sepsis, septic shock, disseminated intravascular coagulation (DIC) and organ failure.9) These symptoms, especially Gram-negative sepsis and DIC, are irremediable even after administering potent antibiotics to combat the infection. Hence, LPS antagonists have been developed by several groups as promising candidates for the prevention and/or treatment of severe sepsis and excessive inflammatory response.1014)

We are interested in designing a safe adjuvant that displays immunological activity with no associated immunotoxicity. During the course of these investigations we recently developed vizantin (3)1517) based on a structure–activity relationship (SAR) study of trehalose 6,6′-dicorynomycolate (TDCM), which was first characterized in 1963 as a cell surface glycolipid of Corynebacterium spp. by Ioneda et al.18) Vizantin also triggers the innate immune response through the specific binding to the same TLR4/MD2 protein complex as lipid A,19) and effectively activates some types of immune system. However, the finding that vizantin induces almost no TNF-α production is clearly different from that of lipid A (or LPS). TNF-α is known to have significant high proinflammatory activity, which may lead to serious complications such as cytokine storm.20) Thus, vizantin induces no TNF-α production; a factor which contributes to its very low immunotoxicity. Vizantin also stimulates human macrophages, making it a promising candidate as an adjuvant in clinical applications. Lipid A and its analogues have also attracted considerable attention as lead compounds for adjuvant development, although as yet with only limited success2124) (Fig. 1).

Fig. 1. Lipid A, TDCM, Vizantin and 3NDDA

Vizantin possesses 3-nonyldodecanoic acid (4) (hereafter 3NDDA) as the fatty acid side-chain, which is a key structure for stimulating human macrophages without TNF-α production.15) Thus, we reasoned that a new biofunctional glycolipid, which shows potent adjuvant activity without the associated production of TNF-α or antagonistic activity against LPS in human tissue, might be produced by introducing 3NDDA into the structure of lipid A in place of its acyl chains. Although the exact biological behavior of the hybrid compound was unclear, our approach might assist in developing the latent biological activity of 3NDDA.

Results and Discussion

Lipid A comprises two glucosamine units with attached acyl chains and normally contains one phosphate group on each sugar unit. A typical example is Escherichia coli lipid A (1) with four (R)-3-hydroxytetradecanoic acids directly attached to both the sugars, and C14- and C12-fatty acids further condensed with the β-hydroxyl group of one sugar unit.25) Because the number of acyl side-chains is known to affect the biological activity,26) we initially designed N,O-3NDDA-dicondensed 5, O-monocondensed 6 and N-monocondensed 7 by imitating the E. coli lipid A with six side-chains (Fig. 2).

Fig. 2. Hybrid Compounds Based on E. coli Lipid A

The synthesis of N,O-3NDDA-dicondensed 5 was effectively achieved through the coupling of sugar unit 10 and 14 (Chart 1A). Unit 10 was prepared from a glucosamine building block 8 and (R)-3-(benzyloxy)tetradecanoic acid (9) according to the reported procedures developed by the Kusumoto group.2729) Conveniently, unit 14 could also be prepared from a common 8. The C-3 hydroxy group of 8 was condensed with 3NDDA by the Keck procedure with 1-ethyl-(3-(3-dimethylamino)propyl)-carbodiimide hydrochloride (EDCI) and N,N-dimethyl-4-aminopyridine (DMAP) as condensing agents, and 11 was treated with Et3SiH and BF3·Et2O in CH2Cl2 at 0°C. The desired regioselective cleavage of the benzylidene acetal moiety in 11 proceeded smoothly, giving 12 as sole product in 90% yield. The resulting alcohol of 12 was phosphorylated with N,N-diethyl-1,5-dihydro-2,3,4-benzodioxaphosphepin-3-amine and 1H-tetrazole followed by in situ oxidation with m-chloroperbenzoic acid (mCPBA). Next, the allyl group of 13 was removed using a catalytic amount of [Ir(cod)(MePh2P)2]PF6 and I2 in tetrahydrofuran (THF) at room temperature, and then unit 14 was converted to the trichloroacetimidate derivative by treatment of CCl3CN and Cs2CO3 in order to couple with unit 10. Using a catalytic amount of trimethylsilyl trifluoromethanesulfonate (TMSOTf) in the presence of molecular sieves 4 Å at −20°C, the coupling reaction was completed within 0.5 h, giving the desired β-linked disaccharide 15 in 82% yield with an excellent stereoselectivity. Next, the N-Troc group of 15 was removed using Zn–Cu couple dust in AcOH, and the resulting amino group of 16 was acylated with 3NDDA using EDCI. Deprotection of the allyl groups in 17 was accomplished in the same manner as that of 13, and the phosphorylation of 18 was achieved by treatment with n-BuLi and then [(BnO)2PO]2O. Finally, hydrogenolysis over 100% (w/v) palladium black resulted in the removal of the benzyl-protecting groups to give 5 as the desired compound. The synthesis of O-3NDDA-monocondensed 6 was also achieved with 16 and (R)-3-(dodecanoyloxy)tetradecanoic acid (19), which was readily prepared from a synthetic intermediate of 9 (Chart 1B). N-3NDDA-monocondensed 7 could be derived from known 2228) by the established four-step sequence for 5 (Chart 1C).

Chart 1. Syntheses of Hybrid Compounds 5, 6 and 7

Reagents and conditions: (a) EDCI, DMAP, MS 4 Å, CH2Cl2, reflux, 4 h; (b) Et3SiH, BF3·Et2O, CH2Cl2, 0°C, 1 h; (c) C6H4(CH2O)2PNEt2, 1H-tetrazole, (CH2Cl)2, −20°C, 10 min, then mCPBA, −20°C, 10 min; (d) [Ir(cod)(MePh2P)2]PF6, H2, THF, rt, 1 h, then I2, rt, 10 min; (e) CCl3CN, Cs2CO3, CH2Cl2, rt, 30 min, then 10, TMSOTf, MS 4 Å, CH2Cl2, −20°C, 45 min; (f) Zn–Cu alloy, AcOH, rt, 2 h; (g) 3NDDA, EDCI, MS 4 Å, CH2Cl2, rt, 11 h; (h) n-BuLi, THF, −78°C, 5 min, then [(BnO)2PO]2O, rt, 1 h; Pd/C, H2, THF, rt, 4 h. (i) EDCI, MS 4 Å, CH2Cl2, rt, 12 h.

With the desired compounds in hand, our attention then shifted to exploring their biological properties. In the course of investigating the adjuvant properties of vizantin, we found that this compound effectively induced macrophage inflammatory protein-1β (MIP-1β) for human monocyte macrophage cells (THP-1 cells) without increasing TNF-α production.15) Thus synthesized 5, 6 and 7 were initially evaluated by assessing their effect on the release of TNF-α and MIP-1β from THP-1 cells. Each sample (100 µM) was added to 5.0×105 cells, and cell culture was then carried out at 37°C for 3 h. The presence of TNF-α and MIP-1β in the culture medium was measured using a commercial capture enzyme-linked immunosorbent assay (ELISA) kit.

Our assay results showed that the compounds 5, 6 and 7 did not stimulate the cells to release MIP-1β or TNF-α into the medium (Fig. 3). These findings suggested that the hybrid compounds might inhibit TNF-α and MIP-1β production triggered by LPS through blocking the binding site of the lipid A unit on the TLR4/MD2 complex. Thus, the antagonism of these hybrid compounds against E. coli LPS (10 ng/mL) was tested with THP-1 cells. Our group recently demonstrated that vizantin represses TNF-α production triggered by LPS in the same assay system.19) Among the synthesized compounds, 5 effectively inhibited LPS-induced release of TNF-α in a dose-dependent manner (Fig. 4A) with an IC50 value of 3.8 nM. This compound also displayed some inhibition of the release of MIP-1β triggered by LPS, albeit at a much greater IC50 value (IC50=0.6 µM). However, no such effect was observed for compounds 6 and 7 (Figs. 4B, C).

Fig. 3. TNF-α and MIP-1β Releasing Activities

The release of TNF-α and MIP-1β was determined using ELISA kits. E. coli LPS (10 ng/mL) was employed as a positive control.

Fig. 4. Effects of 5 (A), 6 (B) and 7 (C) on TNF-α and MIP-1β Production upon Treatment with LPS

Cells were treated with sample compound and LPS at the same time. The release of TNF-α and/or MIP-1β was determined using an ELISA kit. Values represent the mean±S.E.M.; n=5; * p<0.01, ** p<0.005 compared with cells treated with LPS (10 ng/mL) plus vehicle (0.125% BSA).

The observation that of the tested compounds only 5 suppressed TNF-α and MIP-1β release is of great interest, and suggests that 5 acts as a highly active LPS antagonist.14,3033) Thus, its analog compounds with a variety of chain-lengths 2528 were also prepared in order to elucidate the effect of the β-branched acyl chain (Fig. 5A). Compounds 2528 were readily obtained by using the established synthetic route for 5.34)

Fig. 5. Structures of 2528 (A), and the Effect of 27 (B) and 28 (C) on TNF-α and MIP-1β Production in THP-1 Cells Treated with LPS

The antagonistic activities of 2528 were evaluated in the same manner as described for 5. Surprisingly, short-chain analogues 25 and 26 showed significant cytotoxicity to THP-1 cells. In addition, long-chain analogues 27 and 28 were practically inactive in this assay (Figs. 5B, C). In the case of long-chain analogues, their low solubility may influence the biological activity. However, it is clear that chain length affects both cytotoxicity as well as the observed antagonism. Nonetheless, the introduction of 3NDDA is ideal for inducing antagonistic activity.

Docking simulation analysis of 5 and MD2 protein supports the suggestion that introduced 3NDDAs act as a key structure to induce its antagonistic activity. Figure 6 shows the most stable structure of 5 on the molecular electrostatic potential surface of MD2. Two introduced 3NDDA chains and one (R)-3-hydroxytetradecanoyl chain penetrate the cavity of MD2, forming a complex with stabilization energy of −43.17 kcal/mol. As indicated in the graphic abstract, the simulation of 5 with TLR4/MD2 complex also gave a similar result.

Fig. 6. Docking Simulation Analysis of 5 and MD2 Using MOE Software

The purple and green surfaces indicate the hydrophilic and hydrophobic surfaces, respectively. The gray, red, blue, and yellow in the compounds indicate carbon, oxygen, nitrogen, and phosphate, respectively.

Conclusion

Based on the known biological activity of lipid A and vizantin, we designed hybrid compounds of these glycolipids, and identified 5 as a potent endotoxin antagonist among a variety of the synthesized compounds. Specifically, 5 effectively inhibited the release of proinflammatory cytokines normally induced by E. coli LPS in an in vitro model with human immune cells (THP-1 cells). Thus, 5 may be useful in drug development for the treatment of Gram-negative sepsis and/or septic shock. Moreover, we anticipate a further enhancement in the antagonism efficiency of 5 by optimizing the moiety of the unmodified sugar unit. Experiments are currently underway to further enhance the remarkable biological activity of 5 for clinical utility by additional chemical modification combined with in vivo studies.

Experimental

General

Reactions were performed under argon unless otherwise stated. Commercially available chemicals were used as purchased. Special grade and dehydrated solvents were purchased from Kanto Chemical Co., Inc. (Tokyo, Japan). The progress of the reaction was monitored by TLC using precoated silica gel 60 F254 plates (Merck, Darmstadt, Germany). Spots were visualized under UV illumination (254 nm) after immersion in 2% p-anisaldehyde and 5% H2SO4 in EtOH, followed by heating on a hot plate. Solvents were removed under reduced pressure using a standard rotary evaporator. Flash column chromatography was performed using 63–210 mesh silica gel 60 (Kanto Chemical Co., Inc.). Specific rotations were determined on a JASCO P-1030 digital polarimeter using the sodium D line (λ=589 nm) at specific temperatures and are reported as follows: [α]Dtemp., concentration (c=g/100 mL), and solvent. Fourier transform (FT)-IR spectra were measured on a JASCO FT/IR-410 infrared spectrophotometer and reported as wavenumber (cm−1) of significant peaks. 1H- and 13C-NMR spectra were recorded on a Varian Mercury plus-300-4N spectrometer, a Varian 400-MR spectrometer, a Varian 500-MR spectrometer and a Varian Unity-600 spectrometer. The 1H chemical shifts are reported in parts per million (ppm) from an internal standard of tetramethylsilane (TMS) (0.00 ppm), residual CHCl3 (7.26 ppm), or C5H5N (7.19 ppm, right peak), and the 13C chemical shifts were reported using an internal standard of CDCl3 (77.03 ppm, central peak) or C5D5N (123.50 ppm, central peak in right-side triplet). The 1H-NMR spectra are reported as follows: chemical shift, multiplicity (s=singlet, d=doublet, t=triplet, q=quartet, qui=quintet and br=broad), relative integral, coupling constants J (Hz). High-resolution (HR)-MS were recorded on a JEOL the Mstation JMS-700.

Allyl 4,6-O-Benzylidene-2-deoxy-3-O-(3-nonyldodecanoyl)-2-(2,2,2-trichloroethoxycarbonylamino)-α-D-glucopyranoside (11)

To a solution of 827) (18.6 g, 39.0 mmol) in CH2Cl2 (130 mL) were added 3-nonyldodecanoic acid (16.6 g, 50.8 mmol), MS 4 Å (30.0 g), 4,4-dimethylaminopyridine (1.40 g, 11.7 mmol) and EDCI (11.2 g, 58.5 mmol) at room temperature. After the reaction mixture was stirred for 4 h at reflux, it was cooled to room temperature and then filtrated by Celite®. The filtrate was diluted with CH2Cl2 and quenched by H2O. The organic layer was separated, and the aqueous layer was extracted with CH2Cl2 three times. The combined organic layer was dried over MgSO4, and concentrated in vacuo. The residue was purified by silica gel chromatography (hexane–AcOEt=10 : 1) to give 11 as colorless syrup (28.6 g, 36.1 mmol, 93%). 11: [α]D21 +28.1 (c=1.0, CHCl3); FT-IR (neat) 3323, 3065, 3036, 3013, 2954, 2924, 2852, 1742, 1722, 1647 cm−1; 1H-NMR (300 MHz in CDCl3) δ: 0.88 (6H, m, CH3×2), 1.24 (32H, m, CH2×16), 1.80 (1H, m, β-CH), 2.23 (2H, t, J=6.6 Hz, α-CH2), 3.74 (2H, m, H-4 and H-6b), 4.00 (3H, m, H-2, H-5 and OCH2CH=CH2), 4.22 (1H, dd, J=12.9, 5.4 Hz, OCH2CH=CH2), 4.29 (1H, dd, J=10.2, 4.8 Hz, H-6a), 4.66 (1H, d, J=12.0 Hz, CH2 of Troc), 4.77 (1H, d, J=12.0 Hz, CH2 of Troc), 4.94 (1H, d, J=3.9 Hz, H-1), 5.28 (2H, m, OCH2CH=CH2), 5.41 (2H, m, H-3 and NH), 5.53 (1H, s, PhCH(CO)2), 5.89 (1H, m, OCH2CH=CH2), 7.39 (5H, m, PhCH(CO)2); 13C-NMR (75 MHz in CDCl3) δ: 14.16, 22.71, 26.39, 26.43, 29.37, 29.64, 29.67, 29.84, 29.87, 31.93, 33.40, 34.87, 38.95, 54.74, 63.03, 68.37, 69.79, 74.59, 79.26, 95.38, 97.02, 101.44, 118.55, 126.06, 128.15, 129.01, 133.03, 136.90, 154.31, 173.40; MS (FAB+) m/z 789 [M]+; HR-MS (FAB+) m/z Calcd for C40H62NO8Cl3: 789.3541. Found 789.3556.

Allyl 6-O-Benzyl-2-deoxy-3-O-(3-nonyldodecanoyl)-2-(2,2,2-trichloroethoxycarbonylamino)-α-D-glucopyranoside (12)

To a solution of 11 (10.0 g, 12.6 mmol) in CH2Cl2 (85 mL) were added triethylsilane (7.30 g, 63.0 mmol) and BF3·Et2O (8.90 g, 63.0 mmol) at 0°C. After the mixture was stirred for 1 h at 0°C, it was quenched with triethylamine and concentrated in vacuo. The residue was purified by silica gel chromatography (hexane–AcOEt=10 : 1) to give 12 as colorless syrup (9.00 g, 11.3 mmol, 90%). 12: [α]D21 +38.6 (c=1.0, CHCl3); FT-IR (neat) 3437, 3339, 3086, 3065, 3031, 2927, 2855, 1950, 1864, 1743, 1649 cm−1; 1H-NMR (300 MHz in CDCl3) δ: 0.88 (6H, m, CH3×2), 1.25 (32H, m, CH2×16), 1.82 (1H, m, β-CH), 2.27 (2H, t, J=6.0 Hz, α-CH2), 2.68 (1H, d, J=3.6 Hz, OH), 3.78 (4H, m, H-4, H-5 and H-6ab), 3.98 (2H, m, H-2 and OCH2CH=CH2), 4.19 (1H, dd, J=12.6, 5.1 Hz, OCH2CH=CH2), 4.57 (1H, d, J=12.0 Hz, CH2 of Troc), 4.63 (1H, d, J=12.0 Hz, CH2 of Troc), 4.65 (1H, d, J=12.0 Hz, PhCH2), 4.74 (1H, d, J=12.0 Hz, PhCH2), 4.91 (1H, d, J=3.6 Hz, H-1), 5.21 (4H, m, H-3, NH and OCH2CH=CH2), 5.87 (1H, m, OCH2CH=CH2), 7.34 (5H, m, PhCH2); 13C-NMR (75 MHz in CDCl3) δ: 14.11, 22.66, 26.35, 26.56, 29.32, 29.59, 29.63, 29.88, 29.89, 31.88, 33.51, 33.60, 34.82, 39.00, 53.79, 68.43, 69.59, 70.24, 70.46, 73.42, 73.64, 74.54, 95.37, 96.24, 118.21, 127.59, 127.75, 128.39, 133.18, 137.70, 154.18, 174.72; MS (FAB+) m/z 814 [M+H+Na]+; HR-MS (FAB+) m/z Calcd for C40H64NO8Cl3Na: 814.3595. Found 814.3601.

Allyl 6-O-Benzyl-2-deoxy-3-O-(3-nonyldodecanoyl)-4-O-(3-oxido-1,5-dihydrobenzo[e][1,3,2]dioxaphosphepin-3-yl)-2-(2,2,2-trichloroethoxycarbonylamino)-α-D-glucopyranoside (13)

To a solution of 12 (2.10 g, 2.70 mmol) in 1,2-dichloroethane (5.4 mL) were added MS 4 Å (1.50 g), N,N-diethyl-1,5-dihydro-2,3,4-benzodioxaphosphepin-3-amine (0.8 mL, 3.70 mmol) and 1H-tetrazole (476 mg, 6.80 mmol) at room temperature. After the mixture was stirred for 10 min at 0°C, m-chloroperoxybenzoic acid (contain 55%) (467 mg, 2.70 mmol) was added to the mixture at −20°C. After the mixture was stirred for 10 min at −20°C, it was filtrated by Celite®. The filtrate was diluted with AcOEt and quenched with aqueous NaHCO3 and Na2S2O3. The combined organic layer was dried over MgSO4, and concentrated in vacuo. The residue was purified by silica gel chromatography (hexane–AcOEt=4 : 1) to give 13 as colorless syrup (2.61 g, 2.67 mmol, 99%). 13: [α]D22 +29.2 (c=1.0, CHCl3); FT-IR (neat) 3437, 3331 3064, 3029, 2927, 2855, 1958, 1864, 1804, 1747, 1647 cm−1; 1H-NMR (300 MHz in CDCl3) δ: 0.87 (3H, t, J=6.6 Hz, CH3), 0.88 (3H, t, J=6.6 Hz, CH3), 1.24 (32H, m, CH2×16), 1.82 (1H, m, β-CH), 2.34 (2H, d, J=6.6 Hz, α-CH2), 3.73 (1H, dd, J=10.8, 4.8 Hz, H-6a), 3.80 (1H, dd, J=10.8, 2.1 Hz, H-6b), 4.01 (3H, m, H-2, H-5 and OCH2CH=CH2), 4.21 (1H, dd, J=12.9, 5.4 Hz, OCH2CH=CH2), 4.55 (1H, d, J=12.3 Hz, CH2 of Troc), 4.58 (1H, d, J=12.3 Hz, CH2 of Troc), 4.66 (1H, d, J=12.3 Hz, PhCH2), 4.74 (1H, m, H-4), 4.81 (1H, d, J=12.3 Hz, PhCH2), 4.95 (1H, d, J=3.9 Hz, H-1), 5.10 (4H, m, o-C6H4(CH2O)2P), 5.30 (4H, m, H-3, NH and OCH2CH=CH2), 5.88 (1H, m, OCH2CH=CH2), 7.18–7.40 (9H, m, o-C6H4(CH2O)2P and PhCH2); 13C-NMR (75 MHz in CDCl3) δ: 14.14, 22.69, 26.47, 26.61, 29.36, 29.66, 29.73, 30.02, 31.91, 33.38, 33.62, 34.33, 38.69, 54.13, 68.28, 68.59, 68.72, 69.84, 70.75, 73.62, 74.42, 74.64, 95.29, 96.06, 118.47, 127.59, 127.85, 128.33, 128.41, 128.49, 128.95, 133.02, 134.68, 137.89, 154.03, 173.86; MS (FAB+) m/z 996 [M+Na]+; HR-MS (FAB+) m/z Calcd for C48H71NO11Cl3PNa: 996.3728. Found 996.3715.

6-O-Benzyl-2-deoxy-3-O-(3-nonyldodecanoyl)-4-O-(3-oxido-1,5-dihydrobenzo[e][1,3,2]dioxaphosphepin-3-yl)-2-(2,2,2-trichloroethoxycarbonylamino)-α-D-glucopyranose (14)

To a solution of 13 (2.50 g, 2.60 mmol) in THF (52 mL, degassed with Ar) was added [Ir(cod)(MePh2P)2]PF6 (60.0 mg, 0.20 mmol) at room temperature. The mixture was stirred for 1 h under bubbling H2 gas at room temperature. H2O (32.5 mL) and I2 (660 mg, 2.60 mmol) were added to the mixture at room temperature. After the mixture was stirred for 10 min at room temperature, it was diluted with AcOEt and quenched with saturated Na2S2O3 solution. The organic layer was separated and washed with saturated NaHCO3 solution and then brine. The combined organic layer was dried over MgSO4, and concentrated in vacuo. The residue was purified by silica gel chromatography (hexane–AcOEt=2 : 1) to give 14 as colorless syrup (2.00 g, 2.14 mmol, 80%). 14: [α]D22 +6.6 (c=1.0, CHCl3); FT-IR (neat) 3374, 3109, 3066, 3026, 2955, 2925, 2853, 2737, 1949, 1910, 1742, 1725 cm−1; 1H-NMR (500 MHz in CDCl3) δ: 0.87 (3H, t, J=7.0 Hz, CH3), 0.87 (3H, t, J=6.5 Hz, CH3), 1.25 (32H, m, CH2×16), 1.80 (1H, m, β-CH), 2.32 (2H, t, J=6.0 Hz, α-CH2), 3.69 (1H, dd, J=11.0, 6.0 Hz, H-6a), 3.78 (1H, dd, J=11.0, 2.0 Hz, H-6b), 3.95 (1H, dt, J=10.5, 3.5 Hz, H-2), 4.24 (1H, m, H-5), 4.55 (1H, d, J=12.0 Hz, CH2 of Troc), 4.58 (1H, d, J=12.0 Hz, CH2 of Troc), 4.60 (2H, m, H-4 and PhCH2), 4.75 (1H, d, J=12.0 Hz, PhCH2), 5.04 (4H, m, o-C6H4(CH2O)2P), 5.25 (1H, d, J=3.5 Hz, H-1), 5.39 (2H, m, H-3 and NH), 7.16–7.37 (9H, m, o-C6H4(CH2O)2P and PhCH2); 13C-NMR (75 MHz in CDCl3) δ: 14.14, 22.70, 26.46, 26.62, 29.37, 29.68, 29.75, 29.99, 30.04, 31.92, 33.38, 33.61, 34.28, 38.68, 54.33, 68.32, 68.72, 69.17, 70.40, 74.70, 91.45, 95.24, 127.85, 128.14, 128.43, 128.52, 129.00, 134.58, 134.63, 137.46, 154.06, 173.78; MS (FAB+) m/z 956 [M+Na]+; HR-MS (FAB+) m/z Calcd for C45H69NO11Cl3PNa: 956.3415. Found 956.3415.

Allyl 6-O-{6′-O-Benzyl-2′-deoxy-3′-O-(3-nonyldodecanoyl)-4′-O-(3-oxido-1,5-dihydrobenzo[e][1,3,2]dioxaphosphepin-3-yl)-2′-(2,2,2-trichloroethoxycarbonylamino)-β-D-glucopyranosyl}-2-deoxy-3-O-[(R)-3-(benzyloxy)tetradecanoyl]-2-O-[(R)-3-(benzyloxy)tetradecanoylamino]-α-D-glucopyranoside (15)

To a solution of 14 (1.00 g, 1.00 mmol) in CH2Cl2 (50 mL) were added Cs2CO3 (160 mg, 0.450 mmol) and trichloroacetonitrile (1.40 g, 10.0 mmol) at room temperature. After the mixture was stirred for 30 min at room temperature, it was quenched with saturated NaHCO3 solution. The organic layer was separated, and the aqueous layer was extracted with CH2Cl2 three times. The combined organic layer was dried over MgSO4, and concentrated in vacuo to afford the imidate derivative as a yellow syrup. The imidate derivative was dried over P2O5 under vacuum for 1 d, and it was dissolved in CH2Cl2 (20 mL). 1026) (866 mg, 1.00 mmol) and MS 4 Å (4.0 g) were added to the solution of imidate derivative at room temperature. Next, TMSOTf (20.0 µL, 0.100 mmol) was slowly added to the mixture at −20°C, which was stirred for 45 min at −20°C. The reaction was quenched with saturated NaHCO3 solution at 0°C. The organic layer was separated and washed with saturated NaHCO3 solution and then brine. The organic layer was dried over MgSO4 and concentrated in vacuo. The residue was purified by silica gel chromatography (hexane–AcOEt=2 : 1) to give 15 as colorless syrup (1.47 g, 0.820 mmol, 82%). 15: [α]D21 +14.3 (c=1.0, CHCl3); FT-IR (neat) 3376, 3064, 3029, 2925, 2854, 1741 cm−1; 1H-NMR (500 MHz in CDCl3) δ: 0.87 (12H, m, CH3×4), 1.26 (72H, m, CH2×36), 1.82 (1H, m, β-CH of C3′-O-acyl), 2.30 (2H, m, α-CH2 of N-acyl), 2.35 (2H, m, α-CH2 of C3′-O-acyl), 2.45 (1H, dd, J=15.5, 5.0 Hz, α-CH2 of C3-O-acyl), 2.63 (1H, dd, J=15.5, 8.5 Hz, α-CH2 of C3-O-acyl), 3.70 (10H, m, H-2, H-4, H-5, H-6a, H-2′, H-6′ab, β-CH×2 and OCH2CH=CH2), 4.02 (1H, dd, J=12.5, 5.5 Hz, H-6b), 4.08 (1H, d, J=10.0 Hz, OCH2CH=CH2), 4.27 (1H, dt, J=9.5, 3.5 Hz, H-5′), 4.50 (4H, m, PhCH2×2), 4.68 (5H, m, H-4′, PhCH2 and CH2 of Troc), 4.76 (1H, d, J=4.0 Hz, H-1), 5.09 (9H, m, H-3, H-1′, o-C6H4(CH2O)2P, OCH2CH=CH2 and TrocNH), 5.43 (1H, t, J=10.0 Hz, H-3′), 5.72 (1H, m, OCH2CH=CH2), 6.24 (1H, d, J=9.5 Hz, acylNH), 7.16–7.38 (19H, m, o-C6H4(CH2O)2P and PhCH2×3); 13C-NMR (100 MHz in CDCl3) δ: 14.12, 22.67, 25.05, 25.27, 26.50, 29.35, 29.61, 29.64, 29.67, 31.91, 33.39, 33.58, 33.98, 34.03, 34.38, 38.62, 39.81, 41.73, 51.32, 56.28, 68.27, 68.39, 68.59, 68.65, 68.85, 70.74, 71.06, 71.38, 71.49, 73.62, 74.11, 74.18, 74.54, 74.70, 74.76, 75.73, 76.29, 95.24, 96.55, 100.83, 117.93, 127.56, 127.60, 127.63, 127.66, 127.91, 127.94, 128.32, 128.35, 128.38, 128.42, 128.51, 128.97, 133.35, 134.56, 134.60, 137.79, 138.19, 138.40, 153.98, 171.01, 172.58, 173.41; MS (FAB+) m/z 1805 [M+K]+; HR-MS (FAB+) m/z Calcd for C96H146N2O19Cl3PK: 1805.8960. Found 1805.8953.

Allyl 6-O-{2′-Amino-6′-O-benzyl-2′-deoxy-3′-O-(3-nonyldodecanoyl)-4′-O-(3-oxido-1,5-dihydrobenzo[e][1,3,2]dioxaphosphepin-3-yl)-β-D-glucopyranosyl}-2-deoxy-3-O-[(R)-3-(benzyloxy)tetradecanoyl]-2-[(R)-3-(benzyloxy)tetradecanoylamino]-α-D-glucopyranoside (16)

Compound 15 (1.10 g, 0.620 mmol) was dissolved in acetic acid (16 mL). Zinc-copper couple powder (4.40 g, 400% (w/w)) was added to the mixture, which was stirred for 2 h at room temperature. After the suspension was filtrated by Celite®, the filtrate was cooled in ice bath. One molar KOH solution was added dropwise to the mixture at 0°C. The mixture was diluted with AcOEt, and the organic layer was separated and washed with saturated NaHCO3 solution and then brine. The combined organic layer was dried over Na2SO4 and concentrated in vacuo. The residue was purified by silica gel chromatography (hexane–AcOEt–NEt3=1 : 4 : 0.01) to give 16 as colorless syrup (959 mg, 0.601 mmol, 97%). 16: [α]D21 +14.1 (c=1.0, CHCl3); FT-IR (neat) 3370, 3063, 3029, 2925, 2854, 1738 cm−1; 1H-NMR (300 MHz in CDCl3) δ: 0.88 (12H, m, CH3×4), 1.25 (72H, m, CH2×36), 1.94 (1H, m, β-CH of C3′-O-acyl), 2.31 (2H, m, α-CH2 of N-acyl), 2.39 (2H, d, J=6.9 Hz, α-CH2 of C3′-O-acyl), 2.45 (1H, dd, J=15.0, 4.8 Hz, α-CH2 of C3-O-acyl), 2.63 (1H, dd, J=15.0, 7.5 Hz, α-CH2 of C3-O-acyl), 2.90 (1H, dd, J=10.2, 8.1 Hz, H-2′), 3.77 (8H, m, H-2, H-4, H-5, H6′ab, OCH2CH=CH2 and β-CH×2), 4.03 (1H, dd, J=12.6, 5.4 Hz, H-6a), 4.12 (1H, m, OCH2CH=CH2), 4.31 (2H, m, H-6b and H-5′), 4.53 (5H, m, H-4′, PhCH2×2), 4.56 (1H, d, J=12.0 Hz, PhCH2), 4.65 (1H, d, J=12.0 Hz, PhCH2), 4.78 (1H, d, J=3.6 Hz, H-1), 5.08 (9H, m, H-3, H-1′, H-3′, o-C6H4(CH2O)2P and OCH2CH=CH2), 5.72 (1H, m, OCH2CH=CH2), 6.26 (1H, d, J=9.6 Hz, acylNH), 7.15–7.39 (19H, m, o-C6H4(CH2O)2P and PhCH2×3); 13C-NMR (125 MHz in CDCl3) δ: 14.13, 22.68, 25.10, 25.29, 26.40, 26.60, 29.35, 29.62, 29.67, 30.00, 31.91, 33.60, 33.79, 33.99, 34.12, 34.59, 39.00, 39.80, 41.71, 51.26, 56.07, 68.18, 68.23, 68.33, 68.57, 68.64, 68.96, 69.07, 69.38, 70.84, 71.04, 71.40, 73.59, 74.25, 74.29, 74.95, 75.00, 75.76, 76.29, 76.77, 96.57, 104.58, 117.89, 127.59, 127.60, 127.62, 127.86, 127.91, 128.33, 128.36, 128.47, 128.91, 128.92, 133.33, 134.59, 134.63, 137.95, 138.17, 138.39, 171.06, 172.57, 173.64; MS (FAB+) m/z 1616 [M+Na]+; HR-MS (FAB+) m/z Calcd for C93H145N2O17PNa: 1616.0152. Found 1616.0352.

Allyl 6-O-{6′-O-Benzyl-2′-deoxy-3′-O-(3-nonyldodecanoyl)-2′-(3-nonyldodecanoylamino)-4′-O-(3-oxido-1,5-dihydrobenzo[e][1,3,2]dioxaphosphepin-3-yl)-β-D-glucopyranosyl}-2-deoxy-3-O-[(R)-3-(benzyloxy)tetradecanoyl]-2-[(R)-3-(benzyloxy)tetradecanoylamino]-α-D-glucopyranoside (17)

To a solution of 16 (924 mg, 0.580 mmol) in CH2Cl2 (62 mL) were added 3-nonyldodecanoic acid (400 mg, 1.20 mmol), MS 4 Å (10.0 g) and EDCI (240 mg, 1.20 mmol) at room temperature. After the reaction mixture was stirred for 11 h at room temperature, it was filtrated by Celite®. The filtrate was diluted with CH2Cl2 and quenched by H2O. The organic layer was separated and washed with brine. The combined organic layer was dried over MgSO4, and concentrated in vacuo. The residue was purified by silica gel chromatography (hexane–AcOEt=2 : 1) to give 17 as colorless syrup (550 mg, 0.290 mmol, 50%). 17: [α]D21 +10.9 (c=1.0, CHCl3); FT-IR (neat) 3514, 3311, 3063, 3031, 2954, 2922, 2852, 1736, 1651 cm−1; 1H-NMR (300 MHz in CDCl3) δ: 0.88 (18H, m, CH3×6), 1.25 (104H, m, CH2×52), 1.81 (2H, m, β-CH of N′-acyl and β-CH of C3′-O-acyl), 2.01 (2H, d, J=7.2 Hz, α-CH2 of N-acyl), 2.31 (4H, m, α-CH2 of N′-acyl and α-CH2 of C3′-O-acyl), 2.43 (1H, dd, J=15.3, 5.4 Hz, α-CH2 of C3-O-acyl), 2.63 (1H, dd, J=15.3, 7.2 Hz, α-CH2 of C3-O-acyl), 3.77 (10H, m, H-2, H-4, H-5, H-6a, H-2′, H-6′ab, OCH2CH=CH2, β-CH of N-acyl and β-CH of C3-O-acyl), 4.01 (2H, m, H-6b and OCH2CH=CH2), 4.28 (1H, dt, J=10.5, 3.3 Hz, H-5′), 4.45 (1H, d, J=11.4 Hz, PhCH2), 4.50 (2H, s, PhCH2), 4.57 (1H, d, J=11.4 Hz, PhCH2), 4.58 (3H, m, H-4′ and PhCH2), 4.76 (1H, d, J=3.6 Hz, H-1), 4.85 (1H, d, J=8.4 Hz, H-1′), 5.06 (7H, m, H-3′, o-C6H4(CH2O)2P and OCH2CH=CH2), 5.39 (1H, dd, J=10.5, 9.3 Hz, H-3), 5.46 (1H, d, J=8.4 Hz, N′H), 5.71 (1H, m, OCH2CH=CH2), 6.21 (1H, d, J=9.3 Hz, NH), 7.15–7.34 (19H, m, o-C6H4(CH2O)2P and PhCH2×3); 13C-NMR (75 MHz in CDCl3) δ: 14.14, 22.71, 25.18, 25.28, 25.62, 26.41, 26.47, 26.55, 26.59, 29.39, 29.43, 29.72, 29.78, 30.07, 30.14, 31.95, 33.26, 33.47, 33.71, 34.09, 38.17, 38.75, 39.78, 41.86, 51.62, 54.59, 67.98, 68.21, 68.43, 68.94, 70.01, 71.14, 71.43, 72.00, 73.61, 73.79, 74.08, 74.72, 75.66, 76.30, 96.52, 100.79, 117.78, 127.53, 127.62, 127.68, 127.82, 128.02, 128.27, 128.33, 128.39, 128.46, 128.53, 128.97, 133.37, 134.64, 134.69, 137.68, 138.39, 138.46, 148.87, 160.52, 171.00, 172.39, 173.15, 173.52; MS (FAB+) m/z 1940 [M+K]+; HR-MS (FAB+) m/z Calcd for C114H185N2O18PK: 1940.2997. Found 1940.3008.

6-O-{6′-O-Benzyl-2′-deoxy-3′-O-(3-nonyldodecanoyl)-2′-(3-nonyldodecanoylamino)-4′-O-(3-oxido-1,5-dihydrobenzo[e][1,3,2]dioxaphosphepin-3-yl)-β-D-glucopyranosyl}-2-deoxy-3-O-[(R)-3-(benzyloxy)tetradecanoyl]-2-[(R)-3-(benzyloxy)tetradecanoylamino]-α-D-glucopyranose (18)

To a solution of 17 (500 mg, 0.260 mmol) in THF (43 mL, degassed with Ar) was added [Ir(cod)(MePh2P)2]PF6 (20.0 mg, 0.0260 mmol) at room temperature. After the mixture was stirred for 10 min under bubbling H2 gas at room temperature, it was stirred for another hour. H2O (10 mL) and I2 (66.0 mg, 0.540 mmol) were added to the mixture, which was stirred for 30 min at room temperature. The mixture was diluted with AcOEt and quenched with saturated Na2S2O3 solution. The organic layer was separated and washed with saturated NaHCO3 solution and then brine. The combined organic layer was dried over MgSO4, and concentrated in vacuo. The residue was purified by silica gel chromatography (hexane–AcOEt=2 : 1) to give 18 (421 mg, 0.226 mmol, 87%). 18: [α]D18 −1.9 (c=1.0, CHCl3); FT-IR (neat) 3409, 3327, 3063, 3030, 2954, 2925, 2853, 1742, 1650 cm−1; 1H-NMR (300 MHz in CDCl3) δ: 0.86 (18H, m, CH3×6), 1.23 (104H, m, CH2×52), 1.79 (2H, m, β-CH of N′-acyl and β-CH of C3′-O-acyl), 2.00 (2H, d, J=6.9 Hz, α-CH2 of N-acyl), 2.31 (4H, m, α-CH2 of N′-acyl and α−CH2 of C3′-O-acyl), 2.42 (1H, dd, J=14.7, 4.8 Hz, α-CH2 of C3-O-acyl), 2.59 (1H, dd, J=14.7, 7.8 Hz, α-CH2 of C3-O-acyl), 3.30 (2H, m, β-CH of N-acyl and β−CH of C3-O-acyl), 3.74 (6H, m, H-4, H-5, H-6a, H-2′ and H-6′ab), 3.98 (3H, m, H-2, H-6b and H-5′), 4.52 (7H, m, PhCH2×3 and H-4′), 5.01 (6H, m, H-1, H-1′ and o-C6H4(CH2O)2P), 5.42 (1H, d, J=8.1 Hz, N′H), 5.60 (2H, m, H-3 and H-3′), 6.21 (1H, d, J=9.3 Hz, NH), 7.16–7.37 (19H, m, o-C6H4(CH2O)2P and PhCH2×3); 13C-NMR (100 MHz in CDCl3) δ: 14.13, 22.70, 25.03, 25.10, 25.22, 26.35, 26.42, 26.54, 26.62, 26.70, 29.36, 29.41, 29.64, 29.66, 29.69, 29.72, 29.79, 29.94, 30.04, 30.09, 30.11, 30.19, 31.92, 31.95, 33.29, 33.37, 33.48, 33.73, 33.86, 33.91, 34.07, 34.17, 34.39, 35.10, 38.72, 39.78, 41.90, 51.59, 56.30, 68.28, 68.62, 68.71, 69.70, 70.98, 71.43, 71.56, 71.65, 73.57, 73.77, 74.70, 75.05, 75.11, 75.81, 76.61, 91.56, 99.57, 127.74, 127.81, 127.86, 127.89, 127.98, 128.03, 128.12, 128.35, 128.38, 128.41, 128.46, 128.53, 128.99, 134.66, 137.62, 138.11, 138.44, 171.20, 172.70, 173.13, 174.15; MS (FAB+) m/z 1900 [M+K]+; HR-MS (FAB+) m/z Calcd for C111H181N2O18PK: 1900.2684. Found 1900.2678.

6-O-{2′-Deoxy-3′-O-(3-nonyldodecanoyl)-2′-(3-nonyldodecanoylamino)-β-D-glucopyranosyl}-2-deoxy-3-O-[(R)-3-hydroxytetradecanoyl]-2-[(R)-3-hydroxytetradecanoylamino]-α-D-glucopyranose 4,4′-Diphosphate (5)

Compound 18 (370 mg, 0.200 mmol) was dissolved in THF (20 mL), and then 1.6 M solution of n-BuLi (0.16 mL in hexane, 0.260 mmol) was added to the solution at −78°C. The mixture was stirred for 5 min at −78°C, and pyrophosphate (130 mg, 0.240 mmol) was added to the mixture at −78°C. After stirring for 30 min at room temperature, the reaction was quenched with saturated NaHCO3 solution. The organic layer was separated and dried over MgSO4, and it was concentrated to afford the crude product as a yellow syrup. Next, it was dissolved in THF (20 mL), and then Pd-Black (300 mg, 95% (w/w)) was added to the solution at room temperature. The mixture was stirred for 4 h under H2 (10.0 kg/cm2) at room temperature. Triethylamine was added to the mixture at room temperature, and Pd/C was removed by filtration. The filtrate was concentrated in vacuo and the residue was purified by reverse phase silica gel chromatography (MeOH only) to give 5 (213 mg, 0.136 mmol, 68%).

N,O-3NDDA-Dicondenced Analog 5

[α]D20 +23.2 (c=1.0, CHCl3); FT-IR (neat) 3360, 3062, 2957, 2933, 2855, 2710, 2500, 1739, 1731, 1660 cm−1; 1H-NMR (300 MHz in C5D5N) δ: 0.87 (18H, m, CH3×6), 1.23 (106H, m, CH2×52, β-CH of N′-acyl and β-CH of C3′-O-acyl), 2.23 (1H, m, α-CH2 of N′-acyl), 2.36 (1H, m, α-CH2 of C3′-O-acyl), 2.69 (6H, m, α-CH2 of N-acyl, α-CH2 of C3-O-acyl, α-CH2 of N′-acyl and α-CH2 of C3′-O-acyl), 3.68 (1H, d, J=9.3 Hz, H-5′), 4.04 (1H, t, J=9.6 Hz, H-4), 4.13 (1H, d, J=12.0 Hz, H-6′a), 4.42 (4H, m, H-6ab, β-CH of N-acyl and β-CH of C3-O-acyl), 4.57 (1H, d, J=12.0 Hz, H-6′b), 4.73 (1H, q, J=9.0 Hz, H-2′), 4.90 (2H, m, H-2 and H-5), 5.01 (1H, q, J=9.9 Hz, H-4′), 5.72 (1H, d, J=8.7 Hz, H-1′), 5.96 (2H, m, H-3 and H-3′), 6.30 (1H, dd, J=7.8, 3.0 Hz, H-1), 8.28 (1H, d, J=9.0 Hz, NH), 9.39 (1H, d, J=9.0 Hz, N′H); 13C-NMR (125 MHz in C5D5N) δ: 8.94, 14.29, 14.32, 22.94, 22.97, 23.00, 26.25, 26.79, 27.14, 27.19, 29.63, 29.70, 29.75, 29.77, 29.87, 29.94, 30.01, 30.05, 30.09, 30.11, 30.15, 30.21, 30.30, 30.40, 30.52, 30.60, 30.70, 32.14, 32.18, 32.23, 33.83, 34.02, 34.10, 34.15, 34.81, 35.63, 38.05, 38.61, 39.90, 41.91, 43.98, 45.56, 45.65, 53.13, 53.69, 54.79, 61.44, 68.24, 68.43, 69.53, 71.97, 74.86, 75.22, 75.35, 77.33, 93.67, 101.76, 172.74, 173.39, 173.46; MS (FAB) m/z 1568 [M−H]; HR-MS (FAB) m/z Calcd for C82H157N2O21P2: 1568.0754. Found 1568.0743.

Allyl 6-O-{6′-O-Benzyl-2′-deoxy-3′-O-(3-nonyldodecanoyl)-2′-[(R)-3-(dodecanoyloxy)tetradecanoylamino]-4′-O-(3-oxido-1,5-dihydrobenzo[e][1,3,2]dioxaphosphepin-3-yl)-β-D-glucopyranosyl}-2-deoxy-3-O-[(R)-3-(benzyloxy)tetradecanoyl]-2-[(R)-3-(benzyloxy)tetradecanoyl Amino]-α-D-glucopyranoside (20)

Compound 20 (750 mg, 56%) was prepared from 16 (1.30 g, 0.67 mmol) and (R)-3-(tetradecanoyloxy)tetradecanoic acid (19) (600 mg, 1.30 mmol) according to the procedure described for compound 17. 20: [α]D20 +13.0 (c=1.0, CHCl3); FT-IR (neat) 3514, 3308, 3087, 3065, 3031, 2955, 2925, 2853, 1739, 1653 cm−1; 1H-NMR (500 MHz in CDCl3) δ: 0.88 (18H, m, CH3×6), 1.25–1.58 (112H, m, CH2×56), 1.81 (1H, m, β-CH of C3′-O-acyl), 2.31 (8H, m, α-CH2 of N-acyl, α-CH2 of C3′-O-acyl and N′HCOCH2CHROCOCH2R), 2.42 (1H, m, α-CH2 of C3-O-acyl), 2.63 (1H, dd, J=15.5, 7.5 Hz, α-CH2 of C3-O-acyl), 3.77 (10H, m, H-2, H-4, H-5, H-6a, H-2′, H-6′ab, OCH2CH=CH2, β-CH of N-acyl and β-CH of C3-O-acyl), 4.02 (2H, m, OCH2CH=CH2 and H−6b), 4.28 (1H, dt, J=10.5, 3.5 Hz, H-5′), 4.45 (1H, d, J=11.5 Hz, PhCH2), 4.50 (5H, m, PhCH2×2 and H−4′), 4.54 (1H, d, J=11.5 Hz, PhCH2), 4.77 (1H, d, J=3.5 Hz, H-1), 4.86 (1H, d, J=8.0 Hz, H-1′), 5.11 (8H, m, H-3′, OCH2CH=CH2, o-C6H4(CH2O)2P and β-CH of N′-acyl), 5.39 (1H, dd, J=10.5, 9.0 Hz, H-3), 5.71 (1H, m, OCH2CH=CH2), 6.00 (1H, d, J=8.0 Hz, N′H), 6.22 (1H, d, J=9.5 Hz, NH), 7.16–7.38 (19H, m, o-C6H4(CH2O)2P and PhCH2×3); 13C-NMR (100 MHz in CDCl3) δ: 14.12, 22.69, 24.95, 25.19, 25.26, 25.29, 26.55, 29.25, 29.38, 29.54, 29.58, 29.65, 29.70, 29.78, 30.00, 30.08, 30.11, 31.92, 31.94, 33.41, 33.64, 34.09, 34.32, 34.43, 34.48, 34.57, 38.61, 39.75, 41.88, 42.03, 45.61, 51.62, 51.92, 54.96, 58.72, 67.22, 67.84, 68.26, 68.61, 70.80, 71.17, 71.21, 71.44, 71.90, 73.59, 73.90, 74.14, 74.82, 75.62, 76.29, 96.63, 100.69, 117.77, 127.50, 127.53, 127.62, 127.65, 127.79, 127.95, 128.26, 128.34, 128.38, 128.46, 128.52, 128.96, 129.00, 133.41, 134.63, 134.69, 137.79, 138.45, 170.08, 171.01, 172.35, 173.44, 174.26; MS (FAB+) m/z 2040 [M+K]+; HR-MS (FAB+) m/z Calcd for C119H193N2O20PK: 2040.3521. Found 2040.3530.

6-O-{6′-O-Benzyl-2′-deoxy-3′-O-(3-nonyldodecanoyl)-2′-[(R)-3-(dodecanoyloxy)tetradecanoylamino]-4′-O-(3-oxido-1,5-dihydrobenzo[e][1,3,2]dioxaphosphepin-3-yl)-β-D-glucopyranosyl}-2-deoxy-3-O-[(R)-3-(benzyloxy)tetradecanoyl]-2-[(R)-3-(benzyloxy)tetradecanoyl Amino]-α-D-glucopyranose (21)

Compound 21 (430 mg, 87%) was prepared from 20 (500 mg, 0.25 mmol) according to the procedure described for compound 18. 21: [α]D22 +15.4 (c=0.26, CHCl3); FT-IR (neat) 3417, 3331, 3087, 3063, 3030, 2954, 2924, 2853, 1741, 1732, 1652 cm−1; 1H-NMR (500 MHz in CDCl3) δ: 0.88 (18H, m, CH3×6), 1.25–1.58 (112H, m, CH2×56), 1.81 (1H, m, β-CH of C3′-O-acyl), 2.29 (8H, m, α-CH2 of N-acyl, α-CH2 of C3′-O-acyl and N′HCOCH2CHROCOCH2R), 2.43 (1H, m, α-CH2 of C3-O-acyl), 2.61 (1H, dd, J=15.0, 7.5 Hz, α-CH2 of C3-O-acyl), 3.38 (1H, t, J=9.0 Hz, β-CH of N-acyl), 3.55 (1H, q, J=8.5 Hz, β-CH of C3-O-acyl), 3.79 (6H, m, H-4, H-5, H-6a, H-2′ and H-6′ab), 4.01 (2H, m, H-2 and H-6b), 4.19 (1H, dt, J=10.0, 3.5 Hz, H-5′), 4.53 (7H, m, H-4′ and PhCH2×3), 5.07 (7H, m, H-1, H-1′, o-C6H4(CH2O)2P and β-CH of N′-acyl), 5.17 (1H, t, J=8.5 Hz, H-3′), 5.46 (1H, t, J=9.0 Hz, H-3), 6.11 (1H, d, J=8.0 Hz, N′H), 6.29 (1H, d, J=9.5 Hz, NH), 7.14–7.41 (19H, m, o-C6H4(CH2O)2P and PhCH2×3); 13C-NMR (125 MHz in CDCl3) δ: 14.10, 22.68, 24.94, 25.11, 25.21, 25.24, 25.59, 26.56, 29.22, 29.35, 29.49, 29.57, 29.65, 29.68, 29.71, 29.77, 30.01, 30.09, 30.12, 31.92, 33.44, 33.67, 34.13, 34.35, 34.45, 34.49, 38.62, 39.79, 41.89, 42.09, 51.65, 55.57, 67.95, 68.06, 68.28, 68.62, 68.72, 69.56, 70.98, 71.26, 71.52, 71.74, 71.81, 73.59, 74.13, 74.53, 74.98, 75.79, 76.50, 91.60, 99.86, 127.60, 127.66, 127.77, 127.81, 127.85, 127.93, 127.99, 128.33, 128.38, 128.44, 128.49, 128.93, 128.97, 134.67, 134.73, 137.81, 138.16, 138.48, 170.40, 171.18, 172.68, 173.32, 174.58; MS (FAB+) m/z 2000 [M+K]+; HR-MS (FAB+) m/z Calcd for C116H189N2O20PK: 2000.3208. Found 2000.3236.

6-O-{2′-Deoxy-3′-O-(3-nonyldodecanoyl)-2′-[(R)-3-(dodecanoyloxy)tetradecanoylamino]-β-D-glucopyranosyl}-2-deoxy-3-O-[(R)-3-hydroxytetradecanoyl]-2-[(R)-3-hydroxytetradecanoylamino]-α-D-glucopyranose 4,4′-Diphosphate (6)

Compound 6 (120 mg, 45%) was prepared from 21 (320 mg, 0.16 mmol) according to the procedure described for compound 5.

O-3NDDA-monocondenced Analog 6

[α]D20 +31.0 (c=1.0, CHCl3); FT-IR (neat) 3361, 3064, 2955, 2924, 2853, 2685, 1732, 1660 cm−1; 1H-NMR (300 MHz in C5D5N) δ: 0.87 (18H, m CH3×6), 1.26 (111H, m, CH2×55, β-CH of C3′-O-acyl), 2.22 (1H, m, α-CH2 of C3′-O-acyl), 2.75 (8H, m, α-CH2 of N-acyl, α-CH2 of C3-O-acyl, α-CH2 of C3′-O-acyl and NHCOCH2CHROCOCH2R), 3.21 (1H, dd, J=14,7, 5.1 Hz, N′HCOCH2CHROCOCH2R), 3.71 (1H, d, J=9.9 Hz, H-5′), 3.99 (1H, t, J=9.6 Hz, H-4), 4.16 (1H, d, J=12.0 Hz, H-6′a), 4.45 (5H, m, H-6ab, H-6′b, β-CH of N-acyl and β-CH of C3-O-acyl), 4.71 (1H, q, J=9.9 Hz, H-2′), 4.90 (2H, m, H-2 and H-5), 5.05 (1H, q, J=9.3 Hz, H-4′), 5.78 (1H, d, J=7.8 Hz, H-1′), 5.87 (1H, t, J=5.4 Hz, N′HCOCH2CHROCOCH2R), 5.97 (2H, m, H-3 and H-3′), 6.34 (1H, m, H-1), 8.37 (1H, d, J=7.5 Hz, NH), 9.69 (1H, d, J=8.4 Hz, N′H); 13C-NMR (125 MHz in C5D5N) δ: 8.73, 14.29, 18.67, 22.94, 22.96, 22.99, 23.68, 25.48, 25.82, 26.05, 26.24, 26.77, 27.06, 29.55, 29.63, 29.70, 29.75, 29.82, 29.92, 29.95, 29.99, 30.02, 30.12, 30.20, 30.22, 30.50, 30.60, 30.77, 32.14, 32.18, 32.21, 33.75, 34.07, 34.80, 38.04, 38.45, 39.83, 41.31, 43.92, 45.32, 45.64, 53.45, 54.87, 61.36, 68.21, 68.47, 69.44, 71.52, 72.06, 74.73, 75.23, 77.22, 93.73, 101.52, 170.49, 172.71, 173.35, 173.45; MS (FAB) m/z 1668 [M−H]+; HR-MS (FAB) m/z Calcd for C87H165N2O23P2: 1668.1278. Found 1668.1261.

Allyl 6-O-{6′-O-Benzyl-2′-deoxy-3′-O-[(R)-3-(tetradecanoyloxy)tetradecanoyl]-2′-(3-nonyldodecanoylamino)-4′-O-(3-oxido-1,5-dihydrobenzo[e][1,3,2]dioxaphosphepin-3-yl)-β-D-glucopyranosyl}-2-deoxy-3-O-[(R)-3-(benzyloxy)tetradecanoyl]-2-[(R)-3-(benzyloxy)tetradecanoylamino]-α-D-glucopyranoside (23)

Compound 23 (1.01 g, 73%) was prepared from 2226) (1.40 g, 0.740 mmol) and 3NDDA (500 mg, 1.5 mmol) according to the procedure described for compound 17. 23: [α]D23 +18.9 (c=1.0, CHCl3); FT-IR (neat) 3517, 3305, 3087, 3063, 3030, 2922, 2852, 1736, 1651 cm−1; 1H-NMR (300 MHz in CDCl3) δ: 0.88 (18H, m, CH3×6), 1.25 (116H, m, CH2×58), 1.82 (1H, m, β-CH of N′-acyl), 2.03 (2H, m, α-CH2 of N-acyl), 2.27 (4H, m, α-CH2 of N′-acyl and OCOCH2CHROCOCH2R), 2.43 (1H, dd, J=15.3, 5.4 Hz, α-CH2 of C3-O-acyl), 2.63 (3H, m, α-CH2 of C3-O-acyl and OCOCH2CHROCOCH2R), 3.74 (10H, m, H-2, H-4, H-5, H-6a, H-2′, H-6′ab, OCH2CH=CH2, β-CH of N-acyl and β-CH of C3-O-acyl), 4.03 (2H, m, H-6b and OCH2CH=CH2), 4.28 (1H, dt, J=10.8, 3.9 Hz, H-5′), 4.46 (1H, d, J=11.4 Hz, PhCH2), 4.53 (1H, d, J=11.4 Hz, PhCH2), 4.55 (5H, m, H-4′ and PhCH2×2), 4.77 (1H, d, J=3.6 Hz, H-1), 5.12 (8H, m, H-1′, H-3′, OCH2CH=CH2, o-C6H4(CH2O)2P and OCOCH2CHROCOCH2R), 5.44 (1H, dd, J=10.2, 9.0 Hz, H-3), 5.72 (1H, m, OCH2CH=CH2), 5.99 (1H, d, J=7.5 Hz, N′H), 6.23 (1H, d, J=9.3 Hz, NH), 7.17–7.38 (19H, m, o-C6H4(CH2O)2P and PhCH2×3); 13C-NMR (75 MHz in CDCl3) δ: 14.13, 22.70, 25.09, 25.19, 25.23, 25.28, 26.39, 26.44, 29.21, 29.37, 29.43, 29.59, 29.68, 29.72, 29.78, 30.04, 30.11, 31.94, 33.24, 33.47, 34.08, 34.58, 34.80, 39.76, 41.61, 41.86, 51.62, 55.51, 68.22, 68.54, 68.85, 70.26, 70.99, 71.17, 71.43, 72.82, 73.61, 73.80, 75.67, 76.31, 96.54, 100.72, 117.77, 127.53, 127.62, 127.65, 127.82, 127.97, 128.27, 128.32, 128.38, 128.49, 128.78, 129.04, 133.38, 134.72, 134.79, 137.74, 138.37, 138.45, 170.35, 171.00, 172.41, 173.60, 173.80; MS (FAB+) m/z 2068 [M+K]+; HR-MS (FAB+) m/z Calcd for C121H197N2O20PK: 2068.3835. Found 2068.3826.

6-O-{6′-O-Benzyl-2′-deoxy-3′-O-[(R)-3-(tetradecanoyloxy)tetradecanoyl]-2′-(3-nonyldodecanoylamino)-4′-O-(3-oxido-1,5-dihydrobenzo[e][1,3,2]dioxaphosphepin-3-yl)-β-D-glucopyranosyl}-2-deoxy-3-O-[(R)-3-(benzyloxy)tetradecanoyl]-2-[(R)-3-(benzyloxy)tetradecanoylamino]-α-D-glucopyranose (24)

Compound 24 (490 mg, 82%) was prepared from 23 (600 mg, 0.300 mmol) according to the procedure described for compound 18. 24: [α]D23 −10.5 (c=1.0, CHCl3); FT-IR (neat) 3412, 3320, 3087, 3064, 3031, 2954, 2924, 2852, 1738, 1725, 1649 cm−1; 1H-NMR (500 MHz in CDCl3) δ: 0.88 (18H, m, CH3×6), 1.25 (116H, m, CH2×58), 1.79 (1H, m, β-CH of N′-acyl), 2.02 (2H, t, J=6.0 Hz, α-CH2 of N-acyl), 2.29 (4H, m, α-CH2 of N′-acyl and OCOCH2CHROCOCH2R), 2.44 (1H, dd, J=15.0, 5.0 Hz, α-CH2 of C3-O-acyl), 2.58 (1H, dd, J=13.0, 7.5 Hz, OCOCH2CHROCOCH2R), 2.62 (1H, dd, J=12.0, 7.5 Hz, OCOCH2CHROCOCH2R), 2.66 (1H, dd, J=15.0, 4.0 Hz, α-CH2 of C3-O-acyl), 3.27 (1H, q, J=8.0 Hz, β-CH of N-acyl), 3.37 (1H, t, J=9.5 Hz, β-CH of C3-O-acyl), 3.77 (6H, m, H-4, H-5, H-6a, H-2′ and H-6′ab), 4.02 (2H, m, H-2, H-6b), 4.20 (1H, dt, J=9.5, 3.0 Hz, H-5′), 4.55 (7H, m, H-4′ and PhCH2×3), 5.06 (7H, m, H-1, H-1′, H-3′ and o-C6H4(CH2O)2), 5.27 (1H, m, OCOCH2CHROCOCH2R), 5.59 (1H, t, J=9.0 Hz, H-3), 5.68 (1H, d, J=8.5 Hz, N′H), 6.25 (1H, m, NH), 7.17–7.41 (19H, m, o-C6H4(CH2O)2P and PhCH2×3); 13C-NMR (125 MHz in CDCl3) δ: 14.11, 22.69, 22.71, 25.09, 25.12, 25.23, 26.34, 26.58, 29.23, 29.37, 29.42, 29.56, 29.59, 29.64, 29.66, 29.68, 29.72, 29.78, 30.06, 30.15, 31.93, 31.95, 33.40, 33.52, 34.12, 34.21, 34.62, 34.81, 34.94, 39.80, 40.26, 41.77, 41.96, 51.65, 57.03, 67.97, 68.35, 68.69, 69.74, 70.61, 70.98, 71.59, 71.98, 72.61, 73.64, 73.73, 74.87, 75.15, 75.85, 76.54, 91.61, 99.17, 127.62, 127.67, 127.72, 127.82, 127.89, 127.97, 128.03, 128.10, 128.35, 128.36, 128.41, 128.48, 128.78, 129.01, 134.75, 134.81, 137.73, 138.16, 138.49, 170.12, 171.19, 172.75, 173.72, 174.65; MS (FAB+) m/z 2028 [M+K]+; HR-MS (FAB+) m/z Calcd for C118H193N2O20PK: 2028.3521. Found 2028.3512.

6-O-{2′-Deoxy-3′-O-[(R)-3-(tetradecanoyloxy)tetradecanoyl]-2′-(3-nonyldodecanoylamino)-β-D-glucopyranosyl}-2-deoxy-3-O-[(R)-3-hydroxytetradecanoyl]-2-[(R)-3-hydroxytetradecanoylamino]-α-D-glucopyranose 4,4′-Diphosphate (7)

Compound 7 (120 mg, 35%) was prepared from 24 (390 mg, 0.200 mmol) according to the procedure described for compound 5.

N-3NDDA-monocondenced Analog 7

[α]D23 +22.3 (c=1.0, CHCl3); FT-IR (neat) 3376, 2954, 2929, 2853, 2699, 1739, 1659, 1652 cm−1; 1H-NMR (300 MHz in C5D5N) δ: 0.86 (18H, m, CH3×6), 1.25 (115H, m, CH2×57, β-CH of N′-acyl), 2.33 (1H, m, α-CH2 of N′-acyl), 2.60 (7H, m, α-CH2 of N-acyl, α-CH2 of C3-O-acyl, α-CH2 of N′-acyl and OCOCH2CHROCOCH2R), 3.04 (1H, m, OCOCH2CHROCOCH2R), 3.30 (1H, dd, J=16.5, 6.6 Hz, OCOCH2CHROCOCH2R), 3.70 (1H, m, H-5), 4.01 (1H, t, J=9.6 Hz, H-4), 4.16 (1H, d, J=12.0 Hz, H-6′a), 4.38 (4H, m, H-6a, H-6′b, β-CH of N-acyl and β-CH of C3-O-acyl), 4.56 (1H, d, J=12.3 Hz, H-6b), 4.77 (1H, dd, J=9.3 Hz, H-2′), 4.91 (2H, m, H-2 and H-5), 5.05 (1H, q, J=9.6 Hz, H-4′), 5.77 (2H, m, H-1′ and OCOCH2CHROCOCH2R), 5.96 (2H, m, H-3 and H-3′), 6.33 (1H, dd, J=7.5, 3.0 Hz, H-1), 8.34 (1H, d, J=9.3 Hz, NH), 9.44 (1H, d, J=8.7 Hz, N′H); 13C-NMR (75 MHz in C5D5N) δ: 8.72 11.57, 14.26, 14.41, 22.78, 22.93, 25.48, 25.63, 25.79, 26.21, 27.12, 27.22, 29.61, 29.74, 29.83, 29.98, 30.09, 30.25, 30.36, 30.50, 30.66, 30.84, 31.99, 32.12, 32.22, 33.83, 33.97, 34.07, 34.39, 34.57, 34.70, 34.89, 35.57, 38.03, 38.34, 38.47, 39.35, 39.83, 41.72, 41.88, 42.06, 43.42, 43.57, 43.90, 45.32, 45.54, 45.69, 51.25, 53.48, 54.50, 54.71, 61.42, 68.12, 68.23, 68.47, 68.57, 68.80, 69.47, 69.82, 70.49, 71.94, 72.97, 74.83, 75.20, 76.12, 76.40, 77.14, 93.12, 93.81, 101.25, 101.64, 170.88, 172.71, 173.28, 173.34, 173.54; MS (FAB) m/z 1696 [M−H]; HR-MS (FAB) m/z Calcd for C89H169N2O23P2: 1696.1591. Found 1696.1578.

6-O-{2′-Deoxy-3′-O-(3-pentyloctanoyl)-2′-(3-pentyloctanoylamino)-β-D-glucopyranosyl}-2-deoxy-3-O-[(R)-3-hydroxytetradecanoyl]-2-[(R)-3-hydroxytetradecanoylamino]-α-D-glucopyranose 4,4′-Diphosphate (25)

Compound 25 was synthesized by using 3-pentyloctanoic acid instead of 3NDDA in accordance with the procedure described for compound 5. 25: [α]D19 +24.0 (c=1.0, CHCl3), FT-IR (neat) 3360, 2955, 2931, 2855, 2678, 1738, 1666 cm−1; 1H-NMR (500 MHz in C5D5N) δ: 0.86 (18H, m, CH3×6), 1.25 (74H, m, CH2×36, β-CH of N′-acyl and β-CH of C3′-O-acyl), 2.15 (1H, m, α-CH2 of N′-acyl), 2.28 (1H, m, α-CH2 of C3′-O-acyl), 2.67 (6H, m, α-CH2 of N-acyl, α-CH2 of C3-O-acyl, α-CH2 of N′-acyl and α-CH2 of C3′-O-acyl), 3.64 (1H, d, J=10.0 Hz, H-5′), 4.05 (1H, t, J=9.5 Hz, H-4), 4.10 (1H, d, J=11.5 Hz, H-6′a), 4.40 (4H, m, H-6ab, β-CH of N-acyl and β-CH of C3-O-acyl), 4.54 (1H, d, J=11.5 Hz, H-6′b), 4.65 (1H, q, J=9.0 Hz, H-2′), 4.86 (2H, m, H-2 and H-5), 4.93 (1H, dd, J=10.0, 9.5 Hz, H-4′), 5.66 (1H, d, J=8.5 Hz, H-1′), 5.86 (1H, t, J=9.5 Hz, H-3′), 5.95 (1H, t, J=10.0 Hz, H-3), 6.25 (1H, dd, J=8.0, 3.0 Hz, H-1), 8.26 (1H, d, J=8.5 Hz, NH), 9.30 (1H, d, J=9.0 Hz, N′H); 13C-NMR (75 MHz in C5D5N) δ: 8.78, 14.24, 14.31, 14.34, 22.90, 22.96, 23.00, 23.06, 23.07, 26.20, 26.23, 26.28, 26.66, 26.68, 26.71, 29.58, 29.90, 29.95, 29.98, 30.06, 30.08, 32.09, 32.51, 32.53, 32.57, 32.66, 33.60, 33.82, 33.91, 33.95, 34.64, 35.46, 38.00, 38.39, 39.70, 41.75, 43.92, 45.21, 45.66, 53.40, 53.45, 54.72, 61.30, 68.13, 68.40, 69.25, 71.94, 74.82, 74.86, 75.20, 77.14, 93.84, 101.80, 172.67, 173.24, 173.30, 173.33; MS (FAB) m/z 1343 [M−H]; HR-MS (FAB) m/z Calcd for C66H125N2O21P2: 1343.8250. Found 1343.8238.

6-O-{2′-Deoxy-3′-O-(3-heptyldecanoyl)-2′-(3-heptyldecanoylamino)-β-D-glucopyranosyl}-2-deoxy-3-O-[(R)-3-hydroxytetradecanoyl]-2-[(R)-3-hydroxytetradecanoylamino]-α-D-glucopyranose 4,4′-Diphosphate (26)

Compound 26 was synthesized by using 3-heptyldecanoic acid instead of 3NDDA in accordance with the procedure described for compound 5. 26: [α]D19 +21.3 (c=1.0, CHCl3), FT-IR (neat) 3370, 2955, 2925, 2854, 2728, 1737, 1658 cm−1; 1H-NMR (500 MHz in C5D5N) δ: 0.85 (18H, m, CH3×6), 1.28 (90H, m, CH2×44, β-CH of N′-acyl and C3′-O-acyl), 2.19 (1H, m, α-CH2 of N′-acyl), 2.32 (1H, m, α-CH2 of C3′-O-acyl), 2.70 (6H, m, α-CH2 of N-acyl, α-CH2 of C3-O-acyl, α-CH2 of N′-acyl and α-CH2 of C3′-O-acyl), 3.68 (1H, d, J=9.0 Hz, H-5′), 4.03 (1H, t, J=9.5 Hz, H-4), 4.14 (1H, d, J=13.0 Hz, H-6′a), 4.38 (4H, m, H-6ab, β-CH of N-acyl and β-CH of C3-O-acyl), 4.54 (1H, d, J=11.5 Hz, H-6′b), 4.74 (1H, m, H-2′), 4.90 (2H, dd, J=9.0, 8.0 Hz, H-2 and H-5), 5.04 (1H, dd, J=10.0, 9.5 Hz, H-4′), 5.72 (1H, d, J=8.5 Hz, H-1′), 5.99 (2H, m, H-3 and H-3′), 6.34 (1H, dd, J=7.5, 3.0 Hz, H-1), 8.44 (1H, d, J=8.5 Hz, NH), 9.38 (1H, d, J=9.0 Hz, N′H); 13C-NMR (125 MHz in C5D5N) δ: 8.61, 14.25, 14.28, 14.31, 22.90, 22.96, 22.98, 26.22, 26.70, 26.86, 27.03, 27.09, 27.22, 29.60, 29.65, 29.68, 29.71, 29.74, 29.79, 29.82, 29.91, 29.97, 30.20, 30.34, 30.37, 30.42, 30.54, 32.10, 32.15, 32.19, 32.25, 33.76, 33.98, 34.05, 34.72, 35.53, 38.06, 38.16, 38.32, 39.77, 41.91, 43.92, 45.09, 45.66, 53.31, 53.37, 54.69, 61.32, 68.14, 68.21, 68.49, 68.53, 69.33, 72.37, 72.41, 74.63, 75.14, 77.01, 94.11, 94.14, 101.78, 172.64, 173.29, 173.39, 173.44; MS (FAB) m/z 1455 [M−H]; HR-MS (FAB) m/z Calcd for C74H141N2O21P2: 1455.9502. Found 1455.9531.

6-O-{2′-Deoxy-3′-O-(3-undecyltetradecanoyl)-2′-(3-undecyltetradecanoylamino)-β-D-glucopyranosyl}-2-deoxy-3-O-[(R)-3-hydroxytetradecanoyl]-2-[(R)-3-hydroxytetradecanoylamino]-α-D-glucopyranose 4,4′-Diphosphate (27)

Compound 27 was synthesized by using 3-undecyltetradecanoic acid instead of 3NDDA in accordance with the procedure described for compound 5. 27: [α]D20 +17.4 (c=1.0, CHCl3); FT-IR (neat) 3367, 2955, 2932, 2855, 2737, 2622, 2605, 2530, 2498, 1738, 1665 cm−1; 1H-NMR (500 MHz in C5D5N) δ: 0.88 (18H, m, CH3×6), 1.25 (122H, m, CH2×60, β-CH of N′-acyl and β-CH of C3′-O-acyl), 2.24 (1H, m, α-CH2 of N′-acyl), 2.36 (1H, m, α-CH2 of C3′-O-acyl), 2.64 (6H, m, α-CH2 of N-acyl, α-CH2 of C3-O-acyl, α-CH2 of N′-acyl and α-CH2 of C3′-O-acyl), 3.67 (1H, d, J=9.0 Hz, H-5′), 4.06 (1H, dd, J=10.0, 9.0 Hz, H-4), 4.13 (1H, d, J=12.0 Hz, H-6′a), 4.41 (4H, m, H-6ab, β-CH of N-acyl and β-CH of C3-O-acyl), 4.57 (1H, d, J=12.0 Hz, H-6′b), 4.74 (1H, dd, J=9.5, 8.5 Hz, H-2′), 4.90 (2H, m, H-2 and H-5), 5.01 (1H, q, J=10.0 Hz, H-4′), 5.49 (1H, d, J=8.5 Hz, H-1′), 5.97 (2H, m, H-3 and H-3′), 6.31 (1H, dd, J=8.0, 3.0 Hz, H-1), 8.31 (1H, d, J=9.0 Hz, NH), 9.41 (1H, d, J=8.5 Hz, N′H); 13C-NMR (125 MHz in C5D5N) δ: 8.61, 14.28, 14.30, 22.94, 22.97, 26.24, 26.26, 26.78, 27.13, 29.63, 29.67, 29.70, 29.95, 30.01, 30.04, 30.06, 30.08, 30.13, 30.17, 30.24, 30.32, 30.51, 30.55, 30.60, 30.71, 32.13, 32.15, 32.19, 33.80, 34.01, 39.86, 41.94, 43.97, 45.32, 45.64, 53.50, 54.74, 61.36, 68.24, 68.48, 69.40, 72.19, 74.69, 75.17, 75.27, 77.17, 79.83, 93.92, 101.85, 172.71, 173.34, 173.47; MS (FAB+) m/z 1704 [M+Na]+; HR-MS (FAB+) m/z Calcd for C87H174N2O21P2Na: 1704.1982. Found 1704.1995.

6-O-{2′-Deoxy-3′-O-(3-tridecylhexadecanoyl)-2′-(3-tridecylhexadecanoylamino)-β-D-glucopyranosyl}-2-deoxy-3-O-[(R)-3-hydroxytetradecanoyl]-2-[(R)-3-hydroxytetradecanoylamino]-α-D-glucopyranose 4,4′-Diphosphate (28)

Compound 28 was synthesized by using 3-tridecylhexadecanoic acid instead of 3NDDA in accordance with the procedure described for compound 5. 28: [α]D20 +16.9 (c=1.0, CHCl3); FT-IR (neat) 3373, 2924, 2853, 2679, 2498, 1739, 1659 cm−1; 1H-NMR (500 MHz in C5D5N) δ: 0.88 (18H, m, CH3×6), 1.25 (138H, m, CH2×68, β-CH of N′-acyl and β-CH of C3′-O-acyl), 2.24 (1H, m, α-CH2 of N′-acyl), 2.37 (1H, m, α-CH2 of C3′-O-acyl), 2.69 (6H, m, α-CH2 of N-acyl, α-CH2 of C3-O-acyl, α-CH2 of N′-acyl and α-CH2 of C3′-O-acyl), 3.68 (1H, d, J=9.0 Hz, H-5′), 4.05 (1H, dd, J=10.0, 9.5 Hz, H-4), 4.14 (1H, d, J=12.5 Hz, H-6′a), 4.42 (4H, m, H-6ab, β-CH of N-acyl and β-CH C3-O-acyl), 4.57 (1H, d, J=12.5 Hz, H-6′b), 4.75 (1H, q, J=9.0 Hz, H-2′), 4.91 (2H, m, H-2 and H-5), 5.03 (1H, q, J=10.0 Hz, H-4′), 5.74 (1H, d, J=8.0 Hz, H-1′), 5.98 (2H, m, H-3 and H-3′), 6.33 (1H, dd, J=8.0, 3.0 Hz, H-1), 8.33 (1H, d, J=8.5 Hz, NH), 9.42 (1H, d, J=9.0 Hz, N′H); 13C-NMR (125 MHz in C5D5N) δ: 8.74, 14.42, 23.07, 23.09, 26.09, 26.38, 26.90, 27.25, 29.76, 29.80, 30.08, 30.10, 30.14, 30.17, 30.22, 30.23, 30.28, 30.33, 30.36, 30.39, 30.45, 30.64, 30.67, 30.74, 30.84, 32.27, 32.29, 33.92, 34.14, 34.24, 34.89, 35.71, 38.21, 38.57, 39.99, 42.07, 44.07, 45.42, 45.80, 53.60, 54.87, 61.46, 68.36, 68.60, 69.51, 72.29, 74.83, 75.22, 75.37, 77.26, 79.46, 79.72, 79.93, 79.98, 94.06, 101.99, 172.84, 173.47, 173.61; MS (FAB+) m/z 1816 [M]+; HR-MS (FAB+) m/z Calcd for C98H190N2O21P2: 1816.3234. Found 1816.3251.

Docking Simulation Analysis

The docking study of 5 was carried out using Molecular Operating Environment software (MOE 2014.09, Chemical Computing Group, Montreal, Canada). The X-ray crystallographic structures of MD2 (PDB ID: 2E59) and TLR4/MD2 complex (PDB ID: 3FXI) were used. The minimizations of MD2 and TLR4/MD2 complex were performed using MMFF94s force-field. The active sites of MD2 and TLR4/MD2 complex were identified using MOE a Site Finder. The docking simulations with 5 and these proteins (Fig. 6 and graphic abstract) were performed using ASE Dock program (Ryoka Systems, Tokyo, Japan).

Conflict of Interest

The authors declare no conflict of interest.

Supplementary Materials

The online version of this article contains supplementary materials.

References
 
© 2016 The Pharmaceutical Society of Japan
feedback
Top