CN1331878C - Chemical synthesis method of franosterol saponin and its derivative - Google Patents

Chemical synthesis method of franosterol saponin and its derivative Download PDF

Info

Publication number
CN1331878C
CN1331878C CNB200510029869XA CN200510029869A CN1331878C CN 1331878 C CN1331878 C CN 1331878C CN B200510029869X A CNB200510029869X A CN B200510029869XA CN 200510029869 A CN200510029869 A CN 200510029869A CN 1331878 C CN1331878 C CN 1331878C
Authority
CN
China
Prior art keywords
acid
saponin
alpha
base
group
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related
Application number
CNB200510029869XA
Other languages
Chinese (zh)
Other versions
CN1763076A (en
Inventor
李明
俞飚
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shanghai Institute of Organic Chemistry of CAS
Original Assignee
Shanghai Institute of Organic Chemistry of CAS
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shanghai Institute of Organic Chemistry of CAS filed Critical Shanghai Institute of Organic Chemistry of CAS
Priority to CNB200510029869XA priority Critical patent/CN1331878C/en
Publication of CN1763076A publication Critical patent/CN1763076A/en
Application granted granted Critical
Publication of CN1331878C publication Critical patent/CN1331878C/en
Expired - Fee Related legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Landscapes

  • Steroid Compounds (AREA)
  • Saccharide Compounds (AREA)

Abstract

The present invention relates to a new chemical method taking C27 spirostane type saponins and different saccharides as raw materials to synthesize furostanol saponins and derivatives thereof. The method comprises the following steps: (1) the synthesis of full protection type C27 spirostane saponins: the C27 spirostane saponins react with acyls, alkyls and glycosyl donors protected by silicone bases, under the conditions that dehydrating agents exist, and Lewis acid or protonic acid is taken as the accelerant, and full protection type oligosaccharide chains R1 are introduced into 3-bit OH; (2) oxidation: oxidation products are obtained by the reaction of the full protection type spirostane saponins with oxidants; (3) the generation replacement of saponin derivatives by 26-bit halogen: 26-bit halogenated products are obtained by the reaction of the oxidized spirostane saponins with oxytropic reagents; (4) the introduction of XR3 substituents: XR3 substituents are introduced by the nucleophilic substitution reaction of nucleophilic reagents and the halogenated products under alkaline conditions; (5) 16-bit carbonyls in saponins are selectively reduced by reducing agents; (6) furostanol saponins or analogs thereof are obtained after all protective groups are disengaged under the conditions of alkalinity or hydrogenization, etc.

Description

The chemical synthesis process of furostanol saponin and derivative thereof
Technical field
The present invention relates to spirostane type sapogenin and various carbohydrate is the chemical process of synthetic furostanol saponin of raw material and derivative thereof.
Technical background
22-OH/OMe furans gonane type saponin(e is a class formation saponin(e as shown below.Its constructional feature is: (1)
Aglycon is C 27Furans gonane type aglycon; (2) 5 can with 6 form two key (Δs 5), also can be the saturated carbon of α/β-H; (3) 12 bit substituent Y=O or H, H or OH, H; (4) 22 OH/OMe/OEt can transform mutually: reflux in MeOH/EtOH, OH can be converted into OMe/OEt; Reflux in acetone, OMe/OEt can be converted into OH[Hu, K.; Dong, A.; Yao, X.; Kobayashi, H.; Iwasaki, S.Planta Med.1997,63,161-165]; (5) R 1Can be H, but D-glucose (D-Glc), D one semi-lactosi (D-Gal), L-rhamnosyl (L-Rha), D (L)-pectinose (D monose such as (L)-Ara) and be the oligosaccharyl of 2~8 chains or ramiform by the polymerization degree that they are formed normally; (6) in present this saponins of finding, R 3Be generally β-D-Glcp, only a few is α-L-Rhap, [Sarmbi Reddy, K.; Shekhani, M.S.; Berry, D.E.; Lynn, D.G.; Hecht, S.M.J. Chem.Soc.Perkin Trans.I 1984,987-992].
Owing to 22-OH/OMe furans gonane type saponin(e can (furostanol glycoside26-O-β-glucosidase) enzymolysis specifically loses 26 glucosyl group and is converted into spirostane type saponin(e [(a) Wang, L. by the intravital F26G enzyme of plant; You, S.; Jiang, Y. H.; Yao, X.S.Chin.J. Med.Chem.2001,11,326-328; (b) Jin, J.-M.; Liu, X.-K.; Teng, R.-W.Yang, C.-R.Acta Botanica Sinica, 2002,44,1243-1249; (c) Inoue, K.; Shimomura, K.; Kobayashi, S.; Sankawa, U.; Ebizuka, Y.Phytochemistry, 1996,41,725-727; (d) Li, X.C.; Wang, D.Z.; Yang, C.R.hytochemistry, 1990,33,3559-3563; (e) Kawasaki, T.; Komori, T.; Miyahara, K.; Nohara, T.; Hosokawa, I.; Mihashi, K.Chem.Pharm.Bull.1974,22 (9), 2164-2175], this saponins is also degraded under extraction conditions such as acid, high temperature easily simultaneously, does not therefore find them in the research work in early days.Up to nineteen forty-seven, people such as Marker think that just furostanol saponin is parent compound [Marker, the R.E. of spirostanol saponin when Trillium erectum is carried out extraction separation; Wagner, R.B.; Ulshafer, P. R.; Wittbecker, E.L.; Goldsmith, D.P. J.J. Am.Chem.Soc.1947,68,2167-].Yet the discovery of-individual furans gonane type saponin(e jurubine but is thing [Schreiber, K. in 1966; Ripperger, H.Tetrahedron Lett.1966,5997].After this, along with to the intensification of furostanol saponin character and the progress of separate analytical technique, a large amount of furostanol saponins are found.From the various plants of sections such as Asparagaceae, garlic section, Liliaceae, Dioscoreaceae and Solanaceae, separated so far and obtained more than 200 furostanol saponin.
Along with the development of glycobiology, the biological activity of furans gonane type saponin(e is familiar with gradually and is used.For example: the cardiac vascular activity of (1) furostanol saponin: DIAOXINXUE KANG JIAONANG is widely used in treating cardiovascular disordeies such as coronary heart disease, stenocardia clinically.In fact, DIAOXINXUE KANG JIAONANG is to be made by the total steroid saponin that extracts from the Dioscorea panthaica Prain et Burkill root, wherein contains furostanol saponin protogracillin 1, protodioscin 2 and uncle deltoside Li 3[are firm, Zhou Zhengzhi new drug and clinical, 1994,13,75-76] (reaction formula 1).
Figure C20051002986900071
Reaction formula 1: the structure of furostanol saponin in the Diaoxinxuekang
Recently, studies show that again of fresh Dioscorea panthaica Prain et Burkill contained a large amount of furostanol saponins in the Dioscorea panthaica Prain et Burkill, and saponin(es such as spiral shell steroid may be enzymolysis product or the artifact in the extraction separation process [Geng, Y. after medicinal material is put for a long time; Tan, N.-H.; Zhou, J.; Kong, L.-Y. Chin.J. Nat.Med. 2004,2,25-27].(2) cytotoxicity of furostanol saponin: recently, Yao new life etc. has tested the activity of protodioscin 2 (Scheme 1) to 60 human cancer cell strains, and the result shows the GI of this compound to leukemia cell MOLT-4, colon cancer cell HCT-116 and SW-620, CNS cancer cells SNB-75 and kidney cancer cell 786-0 etc. 50≤ 2.0 μ M[Hu, K.; Yao, X.S.Planta Med.2002,68,297-301]; Protodioscin is to the GI of lung carcinoma cell PC-6, breast cancer cell MCF-7, stomach cancer cell NUGC-3, mouse leukemia cell P388 in addition 50<1.9 μ M[Nakamura, T.; Komori, C.; Lee, Y.-y.; Hashimoto, F.; Yahara, S.; Nohara, T.; Ejima, A.Biol.Pharm.Bull.1996,19,564-566]; Yet, when the two keys in the protodioscin 2 are the GI of furostanol saponin 4 pair cell PC-6 and NCT-116 by the saponin(e after saturated 50Value is all greater than 5 μ M[Ikeda, T.; Tsumagari, H.; Okawa, M.; Nohara, T.Chem.Pharm.Bull.2004,52,142-145].
Figure C20051002986900081
Reaction formula 2: the structure of furostanol saponin 4
In addition, furostanol saponin also has antifertility isoreactivity [Yoshi, J.; Dev, S.Indian J.Chem.1988,27B, 12-16].Zhang Jianbo etc. summarize the research work of relevant furans gonane type saponin(e in the recent period, this to understand this compounds in plant distribution and study its chemical structure helpful [Zhang Jianbo, Yu Biao, Hui Yongzheng organic chemistry, 2000,20,663].
It not merely is energy and structural material that the continuous development of glycobiology has disclosed sugar, but also [the Varki that in processes such as the unusual increment of the keeping of the permanent attitude of neural system, immunity system, inflammation, autoimmune disease, aging, cancer cells and transfer, pathogenic infection, plays an important role, A.Glycobioiogy, 1993,3,97].Yet for 2-OH/OMe furans gonane type saponin(e, since it usually with the association of other saponin(e with and the unstable of structure make their separation and purification and part degraded difficult, thereby limit the research of this compounds structure activity relationship, also restricted its further development and utilization.Utilize chemosynthesis not only can obtain having the furans saponin(e of different sugar chains, but also may suppress F26G enzyme enzymolysis at the heteroatoms beyond 26 introducing oxygen, these all provide basic substance for the structure activity study of furostanol saponin, thereby help the development and utilization of this compounds.So far, two pieces of reports about the furostanol saponin chemosynthesis [(a) Yu, B. are only arranged; Liao, J.C.; Zhang, J.B.; Hui, Y. Z.Tetrahedron Lett, 2001,42,77-79; (b) Cheng, M.S.; Wang, Q.L.; Tian, Q.; Song, H.Y.; Liu, Y. X.; Li, Q.; Xu, X.; Miao, H.D.; Yao, X.S.; Yang, Z.J. Org.Chem.2003,68,3658-60].
Summary of the invention
It is a kind of with C that this patent provides 27Spirostane type sapogenin and various carbohydrate are the new chemical method of raw material synthetic furostanol saponin and derivative thereof, and synthetic furostanol saponin structure is as follows:
Figure C20051002986900091
This method comprises the steps: (1) C 27Synthesizing of spirostanol saponin: in the presence of dewatering agent, be promotor with Lewis acid or protonic acid, C 27The glycosyl donor reaction of spiral shell sterioside unit and acyl group, alkyl and silica-based protection is at the oligonucleotide chain R of its 3 OH introducing full guard 1, obtain the spirostanol saponin of full guard: the oxidation of the spirostanol saponin of (2) full guard: full guard spirostanol saponin and oxidant reaction obtain oxidation products; (3) 26 halogens replace the generation of saponin derivative: in the presence of dewatering agent, the spirostanol saponin of oxidation and oxyphie reagent react obtain 26 halogenated product; (4) XR 3Substituent introducing: under alkaline condition, with nucleophilic reagent and halogenated product generation nucleophilic substitution reaction and introduce required XR 3Substituting group; (5) reductive agent optionally reduces 16 carbonyls in the kryptogenin saponin(e; (6) preparation of furostanol saponin or its analogue: under conditions such as alkalescence or hydrogenation, slough all protecting groups and get furostanol saponin or its analogue.
Concrete each step is as follows:
(1), C 27Synthesizing of spirostanol saponin:
In organic solvent, in the presence of dewatering agent and promotor, C27 spirostanol saponin unit obtained the monose spirostanol saponin with glycosyl donor in 3~10 hours in-40~40 ℃ of reactions; When R1 is that the polymerization degree is 2~8 oligonucleotide chain, structural formula is as follows:
Figure C20051002986900092
The time, be intermediate then, through C with corresponding monose spirostanol saponin 1~C 8Fatty acyl group, C 7~C 10Aromaticacyl radical, C 7~C 10Aromatic series halohydrocarbon (as benzyl, to methoxy-benzyl), C 1~C 6The operation of trialkyl silyl protecting group, adopting progressively, the strategy of glucosidesization obtains; The mol ratio of described glycosyl acceptor, promotor and glycosyl donor is 1: 0.05~0.5: 1.0~5.0; Described C 27The weight ratio of spiral shell sterioside unit and dewatering agent is 1.0: 3.0~10.0;
Described organic solvent is C 1~C 6Halohydrocarbon, 1,4-dioxane, ether (Et 2O), acetonitrile (CH 3CN), 2,2,2-trimethylacetonitrile (t-BuCN), tetrahydrofuran (THF) (THF), N, dinethylformamide (DMF), N,N-dimethylacetamide (DMA), hexamethylphosphoramide (HMPA), toluene (PhCH 3) or benzene (PhH) or their mixture:
Described dewatering agent is 3 , 4 , 5  molecular sieves (AW-3 , AW-4 , AW-5 ), the Na of 3 , 4 , 5  molecular sieves, pickling 2SO 4, CaSO 4, CuSO 4Or MgSO 4
Described promotor is C 1~C 6Trialkyl silyl triflate, boron trifluoride diethyl etherate (BF 3Ey 2O), silver trifluoromethanesulfonate (AgOTf), trifluoromethanesulfonic acid lithium (LiOTf), copper trifluoromethanesulfcomposite [Cu (OTf) 2], trifluoromethanesulfonic acid zinc [Zn (OTf) 2], trifluoromethanesulfonic acid scandium [Sc (OTf) 3], trifluoromethanesulfonic acid lanthanum [La (OTf) 3], Ytterbiumtriflate [Yb (OTf) 3], trifluoromethanesulfonic acid indium [In (OTf) 3], trityl group perchlorate (Ph 3CClO 4), trityl group a tetrafluoro borate (Ph 3CBF 4), trifluoromethanesulfonic acid (TfOH), perchloric acid (HClO 4), Tetrafluoroboric acid (HBF 4), four (phenyl-pentafluoride base) boric acid [HB (C 6F 5) 4] or two (fluoroform sulphonyl) imines (HNTf 2);
Described C 27Spirostanol saponin unit is that [△ 5, Y=H, H for diosgenin (diosgenin), 25 (R)], [△ 5, Y=H, H for refined nurse sapogenin (yamogenin), 25 (S)], for accusing sapogenin (tigogenin) [5 α-H, Y=H, H, 25 (R)], NSC 232021 (neotigogenin) [5 α-H, Y=H, H, 25 (S)], hecogenin (hecogenin) [5 α-H, Y=O, 25 (R)], Chinaroot Greenbier Rhizome sapogenin (smilagenin) [5 β H, Y=H, H, 25 (R)], timosaponin unit (sarsasapogenin) [5 β H, Y=H, H, 25 (S)], [△ 5 for botogenin (botogenin), Y=O, 25 (R)], [△ 5, Y=O for Neobotogenin (neobotogenin), 25 (S)] or different chiapagenin (isochiapagenin) [△ 5, Y=β-OH, α-H, 25 (S)]
Described glycosyl donor is C 1~C 8Fatty acyl group, C 7~C 10Aromaticacyl radical, C 7~C 10Aromatic series halohydrocarbon (as Benzyl Chloride, to the methoxyl group benzyl chloride), C 1~C 6D-glucose (D-Glc), D-semi-lactosi (D-Gal), D-seminose (D-Man), D-wood sugar (D-Xyl), 2-glucosamine, L-rhamnosyl (L-Rha), D (L)-pectinose (sulphur glycosides, tribromo-acetyl imines ester or trifluoroacetyl imines ester or the fluorine glycosides of the monose of D (L)-Ara), D-glucuronic acid (D-GlcA), D-galacturonic, (D-GalA) of trialkyl silyl protection.
(2) oxidation of the spirostanol saponin of full guard:
With organic solvent, water or their mixture as medium, full guard spirostanol saponin and oxone (oxone), peroxy propanone, 1,1,1-trifluoromethyl peroxy propanone, metachloroperbenzoic acid, Peracetic Acid, CrO 3, KMnO 4, NaIO 4, (Bu) 4N +IO 4 -Or the mol ratio of their mixture is to obtain oxidation products in 10~60 hours in-20~40 ℃ of reactions at 1: 3.0~25.0 o'clock.
(3) 26 halogens replace the generation of saponin derivative:
In the presence of organic solvent neutralization-40-30 ℃ and dewatering agent, above-mentioned oxidation products and oxyphie reagent react obtained 26 halogens in 0.1~2 hour and replace saponin derivatives; The mol ratio of the spirostanol saponin of oxidation and oxyphie reagent is 1: 3.0~25.0; The spirostanol saponin of oxidation and the weight ratio of dewatering agent are 1.0: 3.0~10.0; Described oxyphie reagent is MgCl 2, MgBr 2, MgI 2, AlCl 3, AlBr 3, AlI 3, NaI, LiI, LiCl, LiBr, ZrCl 4, (Cp) 2ZrCl 2, C 1~C 6Trialkyl silyl halogenide or their mixture.
(4) XR 3Substituent introducing:
Under-78~60 ℃ of the organic solvent neutralizations, alkali or reductive agent in the presence of, with nucleophilic reagent and halogenated product reaction 3~10 hours, generation nucleophilic substitution reaction and introduce required XR 3Substituting group; The mol ratio of described halogenated product, alkali or reductive agent and nucleophilic reagent is 1: 1.5~4.0: 1.5~4.0;
Described alkali can be mineral alkali, as NaOH, KOH, LiOH, CsOH, Na 2CO 3, K 2CO 3, Cs 2CO 3, NaH, KH or LiH; Also can be organic bases, as two-sec.-propyl Lithamide (LDA), potassium tert.-butoxide (t-BuOK), n-Butyl Lithium (n-BuLi), isobutyl-lithium (s-BuLi), tert-butyl lithium (t-BuLi), NaOMe, NaOEt, Mg (OMe) 2, 1,8-two-azabicyclic [5.4.0] undecane-7-alkene (DBU), 1,5-two-azabicyclic [4.3.0] nine alkane-7-alkene (DBN), 1,4-two-azabicyclic [2.2.2] heptane (DABCO), Et 2NH, Et 3N, pyridine (pyridine), imidazoles (imidazole) or 4-N, N-lutidine (DMAP);
Described reductive agent is sodium borohydride (NaBH 4), lithium borohydride (LiBH 4) or POTASSIUM BOROHYDRIDE (KBH 4);
Described nucleophilic reagent is C 1~C 7Alkyl magnesium, lithium, copper, zinc, boron, tin metal reagent, C 6~C 12Aryl magnesium, lithium, copper, zinc, boron, tin metal reagent, C 1~C 7Alkyl sulfhydryl, selenol or above-mentioned mercaptan or the corresponding acetylate of selenol, C 6~C 12Aryl mercaptan, selenol or above-mentioned mercaptan or the corresponding acetylate of selenol, different head position be OH, SH, SeH, NH2, SAc, SeAc ,-S-S-,-Se-Se-, C 1~C 7Alkylsulfonamido, C 6~C 12Aryl-sulfonyl amino by C 1~C 8Fatty acyl group, C 7~C 10Aromaticacyl radical, C 7~C 10Aromatic series halohydrocarbon (as the halogenation benzyl, to methoxyl group halogenation benzyl), C 1~C 6β-D-the glucose of trialkyl silyl protection (β-D-Glc); alpha-D-glucose (α-D-Glc); β-D-semi-lactosi (β-D-Gal); α-D-semi-lactosi (α-D-Gal); β-D-seminose (β-D-Man); α-D-seminose (α-D-Man); β-D-wood sugar (β-D-Xyl); alpha-D-xylose (α-D-Xyl); β-D-2-glucosamine; α-D-2-glucosamine; α-L-rhamnosyl (α-L-Rha); β-L-rhamnosyl (β-L-Rha); α-D-pectinose (α-D-Ara); β-D-pectinose (β-D-Ara); α-L-arabinose (α-L-Ara); β-L-arabinose (β-L-Ara); α-L-Fucose (α-L-Fuc); β-L-Fucose (β-L-Fuc); alpha-D-glucose aldehydic acid (α-D-GlcA); β-D-glucuronic acid (β-D-GlcA); α-D-galacturonic acid (α-D-GalA); β-D-galacturonic acid (β-D-GalA) or be the oligosaccharide compound of 2~8 chains or ramiform by the polymerization degree that these monose are formed; tetrabutyl fluoride amine (TBAF); trimethyl azide base silane (TMSN 3), trimethylammonium cyano group silane (TMSCN), sodiumazide (NaN 3), potassium cyanide (KCN) or sodium cyanoborohydride (NaCNBH 3).
(5) reductive agent optionally reduces 16 carbonyls:
In organic solvent, under 1.0~10.0 normal atmosphere and-40~40 ℃, the product of (4) and reductive agent reaction obtained the hemiacetal product in 15~50 hours; (4) the product and the mol ratio of reductive agent are 1: 0.2~20.0.
Described reductive agent is NaBH 4, LiBH 4, KBH 4, tetrahydrochysene lithium aluminium (LiAlH 4), three (triphenylphosphine) POTASSIUM BOROHYDRIDE [K (Ph 3P) 3BH], three (isopropoxy) POTASSIUM BOROHYDRIDE [K (i-PrO) 3BH], three (tert.-butoxy) Li-Al hydrogen [Li (t-BuO) 3AlH] or Raney nickel;
When reductive agent is NaBH 4, LiBH 4, KBH 4, LiAlH 4, K (Ph 3P) 3BH, K (i-PrO) 3BH or Li (t-BuO) 3During AlH the mol ratio of the product of (4) and reductive agent be recommended as 1: 5.0~20.0;
When reductive agent is Raney nickel, the product of (4) and the mol ratio of reductive agent be recommended as 1: 0.2~and 1.0.
(6) preparation of furostanol saponin or its analogue:
In organic solvent, water or their mixture; in the presence of inorganic or organic bases or hydrogenation catalyst; above-mentioned hemiacetal reacted 20~60 hours under-20-60 ℃ and 1.0~10.0 normal atmosphere, and all protecting groups of sloughing hemiacetal obtain furostanol saponin or its analogue.Above-mentioned hemiacetal, mol ratio inorganic or organic bases are 1: 1.0~10.0; The mol ratio of above-mentioned hemiacetal and hydrogenation catalyst is 1: 0.2~1.0
Described alkali can be mineral alkali, as NaOH, KOH, LiOH, CsOH, Na 2CO 3, K 2CO 3, Cs 2CO 3, NaH, KH or LiH; Also can be organic bases, as LDA, t-BuOK, BuLi, s-BuLi, t-BuLi, NaOMe, Mg (OMe) 2, DBU, DBN, DABCO, Et 2NH, Et 3N, pyridine, imidazoles or 4-N, the N-lutidine.
Described hydrogenation catalyst is 10% palladium charcoal, palladium hydroxide [Pd (OH) 2] or lithium+liquefied ammonia;
Method of the present invention is recommended under the rare gas element carries out, as protections such as argon gas, nitrogen down.
Method of the present invention is a kind of easy, effective chemical process of synthetic Furost saponine and its analogue.This method not only can obtain having the furans saponin(e of different sugar chains, but also may suppress F26G enzyme enzymolysis at the heteroatoms beyond 26 introducing oxygen, these all provide basic substance for the structure activity study of furostanol saponin, thereby help the development and utilization of this compounds.
Embodiment
Following embodiment will help to understand the present invention, but be not limited to content of the present invention.
Synthetic embodiment
26 sulfo-furostanol saponin VIII's is synthetic:
Figure C20051002986900131
Reaction formula 1:26 position sulfo-furostanol saponin VIII's is synthetic
Reagent and condition: a) 4  MS, trimethylsilyl trifluoromethanesulfonate (TMSOTf), CH 2Cl 2, rt; B) oxone, acetone, H 2O, C 1-C 6Halohydrocarbon, rt; C) AlI 3, CH 3CN ,-40-80 ℃; D) MeONa, MeOH, CH 2Cl 2,-78-40 ℃; E) NaBH 4, i-PrOH, CH 2Cl 2, rt; F) NaOH, CH 3OH, H 2O, 60 ℃
(1) under the glucosides condition, diosgenin and 2,2, the glucose three chlorimide esters of 2-pivaloyl (Piv) protection obtain full guard spiral steroidal saponin II for body I reaction; (2) II obtains the product III of two keys and 16 oxidations with the oxone oxidation; (3) III and AlI 3Reaction obtains epoxy and removes the product IV of 26 iodos simultaneously; 4) under alkaline condition, V and IV reaction generate the disaccharide chain saponin(e VI of 26 sulfo-s; (5) NaBH 416 carbonyls that optionally reduce get the hemiacetal product VII; (6) under the alkaline condition, remove the furostanol saponin VIII (Scheme 1) that Piv, Bz protecting group obtain 26 sulfo-s.
Experimentation and data:
Compound I I's is synthetic:
Under the argon shield, (1.50g 3.40mmol) gives body I[Zimmermann, P. with the glucose of Piv protection with diosgenin; Bommer, R.; Bar, T.; Schmidt, R.R.J.Carbohydr.Chem.1988,7,435-452] (3.45g, 5.22mmol 1.54equiv.) are dissolved in the anhydrous CH of 30ml 2Cl 2In, to wherein adding exsiccant 4  molecular sieves (10.0g), stirring at room is after half an hour, add the silica-based triflate of trifluoromethyl base (TMSOTf) (0.06ml, 0.34mmol, 0.1equiv.), continue to stir 2 hours, the TLC demonstration reacts completely, triethylamine cancellation reaction is filtered, and concentrated, rapid column chromatography gets white solid (2.80g, 3.07mmol, 90%).
[α] D 25=-50.84(c?0.90,CHCl 3);
1H?NMR(300MHz,CDCl 3)δ5.34-5.28(m,2H),5.06(t,1H,J=9.6Hz),4.98(dd,1H,J=7.5,9.9Hz),4.61(d,1H,J=7.8Hz),4.40(m,1H),4.20(d,1H,J=12.0Hz),4.01(dd,1H,J=6.6,12.0Hz),3.75-3.69(m,1H),3.54-3.36(m,3H),2.22(m,2H),1.20,1.16,1.14,1.10(s,each?9H); 13C?NMR(75MHz,CDCl 3)δ177.99,177.17,176.50,176.33,140.39,121.73,109.24,99.63,80.78,79.55,72.40,72.16,71.37,68.41,66.83,62.31,62.71,56.49,50.12,41.62,40.27,39.76,38.88,38.82,38.74,38.66,37.16,36.86,32.06,31.85,31.44,31.41,30.29,29.59,28.81,27.16,27.13,27.08,27.04,20.83,19.30,17.10,16.23,14.47;MS(ESI)m/z?91?3.7[M+H +],936.0[M+Na +];
Ultimate analysis: calculated value: C 53H 84O 121/2 H 2O, C69.02%, H9.29%, measured value: C69.13%, H, 8.94%
Synthesizing of compound III:
Under the room temperature condition, (1.70g 1.86mmol) is dissolved in by CH with compound III 2Cl 2: acetone: the mixed solvent (21ml) that water=1: 0.2: 3 is formed under the argon shield, adds NaHCO 3(2.34g, 27.90mmol, 15.0equiv.) and oxone (12equiv.), TLC detection reaction raw material disappears after 48 hours, separates organic phase, water CH for 13.72g, 22.32mmol 2Cl 2Extraction merges all organic phases, and through Na 2SO 4After the drying, filter, concentrate, rapid column chromatography gets full guard and gets Compound I I (1.32g, 1.40mmol, 75%).
α epoxide isomer (α eposide isomers)
1H?NMR(300MHz,CDCl 3)δ?5.30(t,1H,J=9.3Hz),5.07(t,1H,J=9.9Hz),4.98(dd,1H,J=7.g,9.3Hz),4.60(d,1H,J=g.4Hz),4.21(t,1H,J=9.9Hz),4.02(dd,1H,J=6.3,12.0Hz),3.80-3.55(m,4H),3.00(d,1H,J=2.1Hz);
β epoxide isomer (β eposide isomers)
1H?NMR(300MHz,CDCl3)δ?5.28(t,1H,J=9.6Hz),5.07(t,1H,J=9.3Hz),4.98(dd,1H,J=7.8,9.3Hz),4.60(d,1H,J=8.4Hz),4.21(t,1H,J=9.9Hz),4.02(dd,1H,J=6.3,12.0Hz),3.80-3.55(m,4H),2.86(d,1H,J=4.2Hz);
HRMS (MALDI) calculated value: C 53H 84O 14Na967.5759, measured value: C 53H 86O 12Na +967.5753 compound IV is synthetic:
Under the argon shield; the glucose bromine glycosides (3.48g of benzoyl (Bz) protection; 5.28mmol), (950mg 8.32mmol) is dissolved among the anhydrous DMSO of 30ml KSAc; 30 ℃ of stirring reactions 10 hours; the TLC demonstration reacts completely, and decompression is spin-dried for, and rapid column chromatography obtains white solid (2.12g; 3.23mmol, 61%).
[α] D 24=86.50(c?0.93,CHCl 3);
1H?NMR(300MHz,CDCl 3)δ?8.06~7.79(m,8H),7.58~7.24(m,12H),6.02~5.96(m,1H),5.73(t,1H,J=9.89Hz),5.67~5.61(m,2H),4.60(dd,1H,J=3.02,12.36Hz),4.46(dd,1H,J=4.65,12.36Hz),4.34~4.28(m,1H),2.31(s,3H);
13C?NMR(75MHz,CDCl 3)δ?192.13,166.10,165.65,165.13,133.52,133.26,133.08,129.92,129.82,129.71,128.69,128.42,128.23,80.63,76.82,74.16,69.84,69.11,62.89,30.82;
Ultimate analysis: calculated value: C 36H 30O 10S C66.05%, H4.62%; Measured value: C66.09%, H4.44%.
Compound V's is synthetic:
Under the argon shield, to compound III (640mg, 0.6gmol) and AlI 3(832mg, 2.04mmol add CH in mixture 3.0eqiuv.) 3CN: CH 2Cl 2=2: 3 mixed solvent 5ml stirred under-20~0 ℃ of condition after 2 hours, and the TLC demonstration reacts completely.The Na that the reaction solution impouring is saturated 2S 2O 3In the aqueous solution, CH 2Cl 2Extraction, organic phase is through Na 2SO 4After the drying, filter, concentrate, rapid column chromatography gets compound V (595mg, 0.57mmol, 85%).
[α] D 25=-82.16(c?1.19,CHCl 3);
1H?NMR(300MHz,CDCl 3)δ5.35-5.28(m,2H),5.08(t,1H,J=9.6Hz),4.99(dd,1H,J=8.1,9.3Hz),4.62(d,1H,J=8.1Hz),4.21(d,1H,J=11.7Hz),4.02(dd,1H,J=6.3,12.3Hz),3.75-3.70(m,1H),3.48-3.45(m,1H),3.27(d,2H,J=4.8Hz),2.73-2.57(m,4H),2.24-2.18(m,3H),1.21,1.15,1.12,1.10(s,each?9H),0.79(s,3H);
13C?NMR(75MHz,CDCl 3)δ217.87,212.95,177.97,177.17,176.50,176.33,140.56,121.50,99.74,79.50,72.36,72.17,71.35,68.36,66.22,62.28,51.24,49.76,43.22,41.65,39.45?38.84,38.72,38.66,38.60,37.15,36.84,36.77,33.64,31.71,30.94,29.89,29.51,27.14,27.11,27.08,27.02,20.63,20.50,19.27,17.60,15.37,12.94;
HRMS (MALDI) calculated value: C 53H 83IO 12Na1061.4827, measured value: C 53H 83IO 12Na +1061.4821.
Synthesizing of compound VI:
Under the argon shield, with compound IV (618mg, 0.94mmol, 1.68equiv.) and V (586mg 0.56mmol) is dissolved in 10ml MeOH: CH 2Cl 2In=1: 4 mixed solvents of forming, add then freshly prepd MeONa (52.42mg, 1.01mmol, 1.80equiv.),-78 ℃ were stirred 2 hours, then be warming up to 40 ℃ and stir after 1 hour, the TLC demonstration reacts completely, AcOH cancellation reaction, concentrate, column chromatography obtains thio-compounds VI (825mg, 0.54mmol, 96%).
[α] D 24=-53.45(c?0.94,CHCl 3);
1H?NMR(300MHz,CDCl 3)δ8.02-7.81(m,8H),7.52-7.27(m,12H),5.93(t,1H,J=9.6Hz),5.64(t,1H,J=9.9Hz),5.55(t,1H,J=9.6Hz),5.36-5.30(m,2H),5.08(t,1H,J=9.6Hz),5.02(d,1H,J=9.9Hz),5.01(t,1H,J=9.9Hz),4.68-4.63(m,2H),4.47(dd,1H,J=4.8,12.0Hz),4.31-4.20(m,2H),4.03(dd,1H,J=6.6,12.0Hz),3.78-3.72(m,1H),3.50-3.47(m,2H),2.91(dd,1H,J=3.9,12.9Hz),2.66-2.63(m,2H),2.53-2.39(m,3H),2.32-2.26(m,3H),1.22,1.18,1.16,1.12(s,each?9H),1.00(s,3H),0.99(d,3H,J=7.5Hz),0.89(d,3H,J=6.3Hz),0.77(s,3H);
13C?NMR(75MHz,CDCl 3)δ218.16,213.00,177.99,177.17,176.50,176.33,166.00,165.75,165.19,140.55,133.32,133.11,129.85,129.68,129.21,128.89,128.31,121.15,99.71,83.84,79.47,75.92,74.13,72.31,72.16,71.31,70.79,70.02,68.33,66.24,63.52,62.26,51.21,49.73,43.22,41.62,39.27,38.82,38.66,37.13,37.01,36.77,32.70,31.68,30.91,29.50,29.36,27.08,27.02,20.48,19.28,15.40,12.91;
HRMS (MALDI) calculated value: C 87H 110O 21SNa1545.7158, measured value; C 87H 110O 21SNa +1545.7153;
Ultimate analysis; Calculated value: C 87H 110O 21H 2O, C67.77%, H7.32%, measured value: C67.50%, H, 6.96%.
Compound VI I's is synthetic:
With compound V (600mg, 0.39mol) molten sub-10ml i-PrOH: CH 2Cl 2In=1: 5 mixed solvents, argon shield adds NaBH down 4(147.54mg, 3.90mmol 10.0equiv.), stir under-20-40 ℃ of the condition after 40 hours, in the reaction solution impouring aqueous solution, and CH 2Cl 2Extraction, organic phase is through Na 2SO 4After the drying, filter, concentrate, rapid column chromatography gets full guard and gets compound V (300mg, 0.20mmol, 50%).
[α] D 25=-8.89(c?0.82,EtOAc?containing?1%Et 3N);
1H?NMR(300MHz,pyridine-d 5)δ?8.25-7.99(m,8H),7.52-7.11(m,12H),6.73(brs,1H),6.55(t,1H,J=9.3Hz),6.24(t,1H,J=9.6Hz),6.16(t,1H,J=9.6Hz),5.74(t,1H,J=9.6Hz),5.52-5.46(m,4H),4.72-4.66(m,1H),4.55(brd,1H,J=11.1Hz),4.41(dd,1H,J=5.7,12.3Hz),4.10-4.06(m,1H),3.83-3.76(m,1H),3.10(dd,1H,J=5.7,12.6Hz),2.87(dd,1H,J=7.5,12.6Hz),2.61-2.57(m,1H),2.47-2.43(m,1H),1.00(s,3H),0.96(d,3H,J=6.6Hz),0.90(s,3H);
13C NMR (75MHz, pyridine-d 5) δ 177.84,177.24,176.68,166.22,165,75,140.57,133.72,133.47,130.37,130.08,129.90,129.52,128.88,128.78,122.25,110.55,99.68,84.52,81.14,79.54,76.52,75.25,73.15,72.49,72.02,71.77,70.50,68.78,63.82,62.54,56.66,50.42,40.84,40.68,39.98,39.39,38.88,37.67,37.44,37.11,34.13,32.49,31.69,30.89,29.99,27.32,27.27,27.12,21.19,19.40,16.54,16.42;
MS(MALDI)m/z?1529.7[M-H 2O+Na +],1547.7[M+Na +],1562.8[M+K +];
HRMS (MALDI) calculated value: C 57H 112O 21SNa1547.7315, measured value:
C 57H 112O 21SNa +1547.7309。
Synthesizing of compound VIII:
(267mg 0.17mol) is dissolved in by MeOH: CH with compound VI I 2Cl 2: water==1: the mixed solvent of forming at 4: 0.5 (10ml); under the argon shield; add NaOH (34.00mg; 0.85mmol; 5.0equiv.), in 30 ℃ of reactions 48 hours, acidic resins neutralization then, concentrate, rapid column chromatography gets full guard and gets Smilax saponin B (106mg; 0.13mmol, 79%).
[α] D 26=-67.58(c0.62,MeOH∶CH 2Cl 2=1∶1?containing?1%EtN 3);
1H NMR (400MHz, pyridine-d 5) δ 5.40 (d, 1H, J=3.68Hz), 5.12 (d, 1H, J=7.67Hz), 5.08 (d, 1H, J=9.46Hz), 4.65-4.60 (m, 2H), and 4.54-4.46 (m, 2H), 4.43 (dd, 1H, J=5.44,11.91Hz), 4.39-4.31 (m, 4H), 4.14-4.11 (m, 2H), and 4.08-4.02 (m, 3H), 3.11 (dd, 1H, J=5.62,12.81Hz), 2.87 (dd, 1H, J=7.68,12.82Hz), 2.56-2.51 (m, 1H), 2,32-2.30 (m, 1H), 2.22 (m, 1H), 1.26 (d, 3H, J=6.80Hz), 1.15 (d, 3H, J=6.58Hz), 1.00 (s, 3H), 0.91 (s, 3H);
13C NMR (100MHz, pyridine-d 5) δ 141.10,121.82,112.76,102.76,87.63,82.76,81.48,80.35,78.75,78.59,78.28,75.48,74.92,71.89,71.73,64.24,63.03,56.73,50.43,43.45,40.95,40.59,39.90,39.50,37.68,37.59,37.19,34.44,32.40,32.29,3 1.75,30.97,30.88,30.39,21.19,19.64,19.54,16.45,16.38;
MS(MALDI)m/z?777.4[M-H 2O+Na +],795.4[M-CH 2+Na +],809.4[M+Na +];
HRMS (MALDI) calculated value: C 40H 666O 13SNa 809.4122, measured value; C 40H 666O 13SNa +809.4116.

Claims (12)

1. the chemical process of a synthetic furostanol saponin or derivatives thereof, the general structure of described furostanol saponin or derivatives thereof is as follows:
Figure C2005100298690002C1
, wherein,
R 1It is β-D-glucosyl group, the alpha-D-glucose base, β-D-galactosyl, α-D-galactosyl, β-D-mannose group, α-D-mannose group, β-D-xylosyl, the alpha-D-xylose base, β-D-2-glucosamine base, α-D-2-glucosamine base, α-L-rhamanopyranosyl, β-L-rhamanopyranosyl, the Arabic glycosyl of α-D-, the Arabic glycosyl of β-D-, α-L-arabinose base, β-L-arabinose base, α-L-fucosido, β-L-fucosido, alpha-D-glucose aldehydic acid base, β-D-glucal acidic group, α-D-galacturonic acidic group, β-D-galacturonic acidic group or be the oligosaccharyl of 2~8 chains or ramiform by the polymerization degree that these monose are formed;
R 2=H or Me or Et;
XR 3Be F, N 3, guanidine radicals, CN or H; Perhaps XR 3In, X=C, O, S, N or Se; R 3It is β-D-glucosyl group, the alpha-D-glucose base, β-D-galactosyl, α-D-galactosyl, β-D-mannose group, α-D-mannose group, β-D-xylosyl, the alpha-D-xylose base, β-D-2-glucosamine base, α-D-2-glucosamine base, α-L-rhamanopyranosyl, β-L-rhamanopyranosyl, the Arabic glycosyl of α-D-, the Arabic glycosyl of β-D-, α-L-arabinose base, β-L-arabinose base, α-L-fucosido, β-L-fucosido, alpha-D-glucose aldehydic acid base, β-D-glucal acidic group, α-D-galacturonic acidic group, β-D-galacturonic acidic group or be the oligosaccharyl of 2~8 chains or ramiform by the polymerization degree that these monose are formed;
It is characterized in that this method comprises the steps:
(1) C 27Synthesizing of spirostanol saponin:
In organic solvent and dewatering agent in the presence of, at-40~40 ℃, be promotor with Lewis acid or protonic acid, C 27The glycosyl donor reaction of spiral shell sterioside unit and acyl group, alkyl and silica-based protection 3-10 hour is at the oligonucleotide chain R of its 3 OH introducing full guard 1, obtain the spirostanol saponin of full guard; Wherein, C 27Spiral shell sterioside unit is 1: 1.0~5.0: 0.05~0.5 with the mol ratio of glycosyl acceptor and promotor; C 27Spiral shell sterioside unit is 1.0: 3.0~10.0 with the weight ratio of dewatering agent;
(2) oxidation of the spirostanol saponin of full guard:
In organic solvent or water or their mixture, full guard spirostanol saponin and oxidant reaction obtained oxidation products in 10~60 hours; The mol ratio of full guard spirostanol saponin and oxygenant is 1: 3.0~25.0, and described oxygenant is oxone, peroxy propanone, 1,1,1-trifluoromethyl peroxy propanone, metachloroperbenzoic acid, Peracetic Acid, CrO 3, KMnO 4, NaIO 4Or (Bu) 4N +IO 4 -Or their mixture;
The introducing of (3) 26 halogenic substituents:
In the presence of organic solvent neutralization-40-30 ℃ and dewatering agent, the spirostanol saponin of above-mentioned oxidation and oxyphie reagent react obtained 26 halogenated product in 0.1~2 hour, and the mol ratio of the spirostanol saponin of oxidation and oxyphie reagent is 1: 3.0~25.0; Described oxyphie reagent is MgCl 2, MgBr 2, MgI 2, AlCl 3, AlBr 3, AlI 3, NaI, LiI, LiCl, LiBr, ZrCl 4, (Cp) 2ZrCl 2, C 1~C 6Trialkyl silyl halogenide or their mixture, wherein Cp is a cyclopentadienyl;
(4) XR 3Substituent introducing:
In organic solvent, under the existence of alkali or reductive agent, with nucleophilic reagent and halogenated product reaction 3~10 hours, generation nucleophilic substitution reaction and introduce required XR 3Substituting group; The mol ratio of described halogenated product, alkali or reductive agent and nucleophilic reagent is 1: 1.5~4.0: 1.5~4.0;
(5) reductive agent optionally reduces 16 carbonyls of saponin(e;
In organic solvent, under 1.0~10.0 normal atmosphere and-40~40 ℃, the product of (4) and reductive agent reaction obtained the hemiacetal product in 15~50 hours; (4) the product and the mol ratio of reductive agent are 1: 0.2~20.0;
Described reductive agent is NaBH 4, LiBH 4, KBH 4, LiAlH 4, K (Ph 3P) 3BH, K (i-PrO) 3BH, Li (t-BuO) 3AlH or Raney nickel;
(6) preparation of furostanol saponin or its analogue:
In organic solvent or water or their mixture ,-20~60 ℃ and 1.0~10.0 normal atmosphere, the hemiacetal product reacts and sloughed all protecting groups in 20~60 hours and get furostanol saponin or its analogue in the presence of alkali or hydrogenation catalyst; Described alkali is inorganic or organic bases; The mol ratio of described hemiacetal product and alkali or hydrogenation catalyst is 1: 0.2~20;
Described hydrogenation catalyst is 10% palladium charcoal, palladium hydroxide or lithium+liquefied ammonia;
Described dewatering agent is 3 , 4 , 5  molecular sieves, the Na of 3 , 4 , 5  molecular sieves, pickling 2SO 4, CaSO 4, CuSO 4Or MgSO 4
2. synthetic method as claimed in claim 1 is characterized in that described by β-D-glucose, alpha-D-glucose, β-D-semi-lactosi, α-D-semi-lactosi, β-D-seminose, α-D-seminose, β-D-wood sugar, alpha-D-xylose, β-D-2-glucosamine, α-D-2-glucosamine, α-L-rhamnosyl, β-L-rhamnosyl, α-D-pectinose, β-D-pectinose, α-L-arabinose, β-L-arabinose, α-L-Fucose, β-L-Fucose, alpha-D-glucose aldehydic acid, β-D-glucuronic acid, the polymerization degree that α-D-galacturonic acid or β-D-galacturonic acid is formed is that the oligosaccharyl structural formula of 2~8 chains or ramiform is as follows:
3. synthetic method as claimed in claim 1, the steric configuration that it is characterized in that 22 asymmetric carbons of described furostanol saponin or derivatives thereof are S or R configuration.
4. synthetic method as claimed in claim 1 is characterized in that described C 27Spiral shell sterioside unit is a diosgenin; Refined nurse sapogenin; For accusing sapogenin; NSC 232021; Hecogenin; The Chinaroot Greenbier Rhizome sapogenin; Timosaponin unit; Botogenin; Neobotogenin or different chiapagenin.
5. synthetic method as claimed in claim 1 is characterized in that described lewis acid is C 1~C 6Trialkyl silyl triflate, boron trifluoride diethyl etherate, silver trifluoromethanesulfonate, trifluoromethanesulfonic acid lithium, copper trifluoromethanesulfcomposite, trifluoromethanesulfonic acid zinc, trifluoromethanesulfonic acid scandium, trifluoromethanesulfonic acid lanthanum, Ytterbiumtriflate, trifluoromethanesulfonic acid indium, trityl group perchlorate or trityl group a tetrafluoro borate; Protonic acid is trifluoromethanesulfonic acid, perchloric acid, Tetrafluoroboric acid, four (phenyl-pentafluoride base) boric acid or two (fluoroform sulphonyl) imines.
6. synthetic method as claimed in claim 1 is characterized in that described nucleophilic reagent is C 1~C 7Alkyl magnesium, lithium, copper, zinc, boron, tin metal reagent, C 6~C 12Aryl magnesium, lithium, copper, zinc, boron, tin metal reagent, C 1~C 7Corresponding acetylate, the C of alkyl sulfhydryl, selenol or above-mentioned mercaptan or selenol 6~C 12The corresponding acetylate of aryl mercaptan, selenol or above-mentioned mercaptan or selenol, different head position are OH, SH, SeH, NH 2SAc; SeAc;-S-S-;-Se-Se-by acyl group; the following sugar of alkyl and silica-based full guard: β-D-glucose; alpha-D-glucose; β-D-semi-lactosi; α-D-semi-lactosi; β-D-seminose; α-D-seminose; β-D-wood sugar; alpha-D-xylose; β-D-2-glucosamine; α-D-2-glucosamine; α-L-rhamnosyl; β-L-rhamnosyl; α-D-pectinose; β-D-pectinose; α-L-arabinose; β-L-arabinose; α-L-Fucose; β-L-Fucose; alpha-D-glucose aldehydic acid; β-D-glucuronic acid; α-D-galacturonic acid; β-D-galacturonic acid or be the oligosaccharide compound of 2~8 chains or ramiform by the polymerization degree that these monose are formed; tetrabutyl fluoride amine; the trimethyl azide base silane; trimethylammonium cyano group silane; sodiumazide; potassium cyanide or sodium cyanoborohydride.
7. synthetic method as claimed in claim 1 is characterized in that described mineral alkali refers to oxyhydroxide, hydride or the carbonate of monovalence metal; Organic bases is two-sec.-propyl Lithamide, potassium tert.-butoxide, n-Butyl Lithium, isobutyl-lithium, tert-butyl lithium, NaOMe, NaOEt, Mg (OMe) 2, 1,8-two-azabicyclic [5.4.0] undecane-7-alkene, 1,5-two-azabicyclic [4.3.0] nine alkane-7-alkene, 1,4-two-azabicyclic [2.2.2] heptane, Et 2NH, Et 3N, pyridine, imidazoles, 4-N, the N-lutidine.
8. as claim 1 or 6 described synthetic methods, it is characterized in that described acyl group is C 1~C 8Fatty acyl group or C 7~C 10Aromaticacyl radical, silica-based be C 1~C 6Trialkyl silyl.
9. synthetic method as claimed in claim 1 is characterized in that described acyl group; the glycosyl donor of alkyl and silica-based protection is β-D-glucose; alpha-D-glucose; β-D-semi-lactosi; α-D-semi-lactosi; β-D-seminose; α-D-seminose; β-D-wood sugar; alpha-D-xylose; β-D-2-glucosamine; α-D-2-glucosamine; α-L-rhamnosyl; β-L-rhamnosyl; α-D-pectinose; β-D-Arab; α-L-arabinose; β-L-arabinose; α-L-Fucose; β-L-Fucose; alpha-D-glucose aldehydic acid; β-D-glucuronic acid; α-D-galacturonic acid; the sulphur glycosides of β-D-galacturonic acid monosaccharide groups; tribromo-acetyl imines ester; trifluoroacetyl imines ester or fluorine glycosides.
10. synthetic method as claimed in claim 1 is characterized in that the reductive agent described in the described step (5) is NaBH 4, LiBH 4, KBH 4, LiAlH 4, K (Ph 3P) 3BH, K (i-PrO) 3BH or Li (t-BuO) 3During AlH, the product of (4) and the mol ratio of reductive agent are 1: 5.0~20.0; Or described reductive agent is when being Raney nickel, and the product of (4) and the mol ratio of reductive agent are 1: 0.2~1.0.
11. synthetic method as claimed in claim 1, the mol ratio that it is characterized in that hemiacetal product described in the described step (6) and alkali is 1: 1.0~20; The mol ratio of described hemiacetal product and hydrogenation catalyst is 1: 0.2~1.0.
12. synthetic method as claimed in claim 1 is characterized in that described method is to carry out under protection of inert gas.
CNB200510029869XA 2005-09-22 2005-09-22 Chemical synthesis method of franosterol saponin and its derivative Expired - Fee Related CN1331878C (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CNB200510029869XA CN1331878C (en) 2005-09-22 2005-09-22 Chemical synthesis method of franosterol saponin and its derivative

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CNB200510029869XA CN1331878C (en) 2005-09-22 2005-09-22 Chemical synthesis method of franosterol saponin and its derivative

Publications (2)

Publication Number Publication Date
CN1763076A CN1763076A (en) 2006-04-26
CN1331878C true CN1331878C (en) 2007-08-15

Family

ID=36747415

Family Applications (1)

Application Number Title Priority Date Filing Date
CNB200510029869XA Expired - Fee Related CN1331878C (en) 2005-09-22 2005-09-22 Chemical synthesis method of franosterol saponin and its derivative

Country Status (1)

Country Link
CN (1) CN1331878C (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110054156A1 (en) * 2008-04-30 2011-03-03 Institute Of Radiation Medicine, Academy Of Military Sciences, Pla Synthesis of timosaponin bii
CN103275169A (en) * 2013-05-30 2013-09-04 中国海洋大学 Furostanol saponins and synthesis method thereof
CN105037480B (en) * 2015-06-24 2017-04-12 中国海洋大学 Furostanol saponin and application of furostanol saponin as alpha-glycosidase inhibitor to antidiabetic medicine

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Total Synthesis of Methyl Protodioscin: ...... Cheng,Mao S.,Wang,Qian L.,et al,Journal of Organic Chemistry,vol 68,no 9,page 3658.3662 2003 *

Also Published As

Publication number Publication date
CN1763076A (en) 2006-04-26

Similar Documents

Publication Publication Date Title
Gorin et al. Configuration of glycosidic linkages in oligosaccharides: IX. Synthesis of α-and β-D-mannopyranosyl disaccharides
CN104311571B (en) Intermediate and method for synthesis of halichondrin b analogs
Toshima et al. Total synthesis of elaiophylin (azalomycin B).
NZ251177A (en) Intermediates for preparing halichondrin derivatives
Morzycki et al. Synthesis of a cholestane glycoside OSW-1 with potent cytostatic activity
JP2016188220A (en) Phenyl c-glucoside derivative having deoxyglucose structure, and methods of preparation and use thereof
Carpani et al. Saponins from Albizzia anthelmintica
Gauthier et al. Synthesis of two natural betulinic acid saponins containing α-l-rhamnopyranosyl-(1→ 2)-α-l-arabinopyranose and their analogues
CN108675976A (en) A kind of halogenated glucose carbon glycosides of 6- and its preparation method and application
CN1331878C (en) Chemical synthesis method of franosterol saponin and its derivative
Cheng et al. Synthesis of β-hederin and Hederacolchiside A1: Triterpenoid saponins bearing a unique cytotoxicity-inducing disaccharide moiety
Ma et al. Synthesis of OSW-1 analogues and a dimer and their antitumor activities
Hanessian et al. Synthesis of a chiral 1, 7-dioxaspiro [5, 5] undecene. A model for the spiroacetal subunit of avermectin B1a
Lichtenthaler et al. manno-versus gluco-Selectivity in reductions of 2-keto-β-d-arabino-hexopyranosides
NO170687B (en) ANALOGY PROCEDURE FOR PREPARING FLUORO-SUBSTITUTED 4'-DEMETHYLPIPODOPHYLLOTOXIN GLUCOSIDES
Liu et al. Concise synthesis of two natural triterpenoid saponins, oleanolic acid derivatives isolated from the roots of Pulsatilla chinensis
Allanson et al. The synthesis of novel mimics of the sialyl Lewis x determinant
AU2013204005B2 (en) Improved synthesis
Jackowski et al. Formation of septanoses from hexopyranosides via 5, 6-exo-glycals
Chiba et al. A new synthesis of α-L-fucose
US4001402A (en) Cardenolide glycosides and method of making the same, and therapeutic composition
Omar et al. Nucleophilic substitution at the anomeric position of furanose carbohydrates. The case of the C-allylations
Sivets Regioselective and stereocontrolled syntheses of protected L-glycosides from L-arabinofuranosides
Li et al. Synthesis of two natural oleanolic acid saponins
CN114644678B (en) Glucuronide compound, preparation method and application thereof

Legal Events

Date Code Title Description
C06 Publication
PB01 Publication
C10 Entry into substantive examination
SE01 Entry into force of request for substantive examination
C14 Grant of patent or utility model
GR01 Patent grant
CF01 Termination of patent right due to non-payment of annual fee

Granted publication date: 20070815

Termination date: 20140922

EXPY Termination of patent right or utility model