WO2000001683A1 - Methods for synthesis of substituted tetrahydrofuran compound - Google Patents

Methods for synthesis of substituted tetrahydrofuran compound Download PDF

Info

Publication number
WO2000001683A1
WO2000001683A1 PCT/US1999/015107 US9915107W WO0001683A1 WO 2000001683 A1 WO2000001683 A1 WO 2000001683A1 US 9915107 W US9915107 W US 9915107W WO 0001683 A1 WO0001683 A1 WO 0001683A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
tetrahydrofuran
substituted
mmol
solution
Prior art date
Application number
PCT/US1999/015107
Other languages
French (fr)
Other versions
WO2000001683A8 (en
Inventor
Mukund Shankar Chorghade
Mukund Keshao Gurjar
Radha Palakodety Krishna
Sista Venkata Sai Lalitha
Kashinath Sadalapure
Shusanta Sekhar Adhikari
Andappan Murugaiah Subbaiah Murugaiah
Sunil Vyankatesh Mhaskar
Gangavaram Vasantha Madhava Sharma
Tangallapally Rajendra Prasad
Punna Sreenivas
Vavilala Goverdhan Reddy
Aminul Islam
Chittineni Hariprasad
Alla Venkata Rana Rao
Original Assignee
Leukosite, Inc.
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 Leukosite, Inc. filed Critical Leukosite, Inc.
Priority to KR1020017000008A priority Critical patent/KR20010083083A/en
Priority to MXPA01000234A priority patent/MXPA01000234A/en
Priority to AU47311/99A priority patent/AU750592B2/en
Priority to CA002342156A priority patent/CA2342156A1/en
Priority to JP2000558088A priority patent/JP2002519415A/en
Priority to EP99930868A priority patent/EP1102759A1/en
Publication of WO2000001683A1 publication Critical patent/WO2000001683A1/en
Publication of WO2000001683A8 publication Critical patent/WO2000001683A8/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/04Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • C07D307/10Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having no double bonds between ring members or between ring members and non-ring members with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D307/14Radicals substituted by nitrogen atoms not forming part of a nitro radical
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • A61K31/341Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide not condensed with another ring, e.g. ranitidine, furosemide, bufetolol, muscarine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/04Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • C07D307/10Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having no double bonds between ring members or between ring members and non-ring members with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D307/12Radicals substituted by oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/04Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • C07D307/10Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having no double bonds between ring members or between ring members and non-ring members with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D307/16Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/26Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having one double bond between ring members or between a ring member and a non-ring member
    • C07D307/30Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having one double bond between ring members or between a ring member and a non-ring member with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D307/32Oxygen atoms

Definitions

  • the present invention provides new methods for preparation of 2-(4- fluorophenoxymethyl)-5-(4-N-hydroxyureidyl- 1 -butynyl)-tetrahydrofuran ("compound 1”) and synthetic precursors thereof.
  • Leukotrienes are recognized potent local mediators, playing a significant role in inflammatory and allegeric responses, including arthritis, asthma, psoriasis and thrombotic disease.
  • Leukotrienes are produced by the oxidation of arachidonic acid by lipoxygenase. More particularly, arachidonic acid is oxidized by 5-lipooxygenase to the hydroperoxide 5-hydroperoxy-eicosatetraenoic acid (5-HPETE), that is converted to leukotriene A 4 , that in turn can be converted to leukotriene B 4 , C 4 , or D 4 .
  • the slow- reacting substance of anaphylaxis is now known to be a mixture of leukotrienes C 4 , D 4 and E 4 , all of which are potent bronchoconstrictors.
  • European Patent Application Nos. 901171171.0 and 901170171.0 report indole, benzofuran, and benzothiophene lipoxygenase inhibiting compounds.
  • Various 2,5-disubstituted tetrahydrofurans have exhibited significant biological activity, including as lipoxygenase inhibitors. See U.S. Patent Nos. 5,703,093; 5,681,966; 5,648,486; 5,434,151; and 5,358,938.
  • compound 1 and precursors thereof are provided by reacting 4-fluorophenol with an epoxide having a reactive C3 carbon, e.g. a glycidyl compound substituted at the C3 position with an electron-withdrawing group such as halo (e.g.
  • the reacted epoxide can be optically active if desired.
  • the formed epoxyphenylether is then reacted with an active methylene compound to form a lactone, preferably a ⁇ -lactone, with 5 ring members.
  • the active methylene compound can be a variety of agents.
  • Di ethyl and dimethyl malonate are generally preferred, which provide an ethyl or methyl ester as a lactone ring substituent, i.e. 2-carboalkoxy-(4-fluoro-phenoxy-methyl)- ⁇ -butyrolactone, where the alkoxy group suitably has from 1 to about 12 carbon atoms, more preferably 1 to about 6 carbon atoms, still more preferably 1 to about 3 carbons with methoxy and ethoxy particularly preferred. That ester group is then removed (e.g. via hydrolysis and decarboxylation), and the lactone suitably reduced to a hydroxy-substituted tetrahydrofuran, specifically 4-fluorophenoxymethyl-hydroxytetrahydrofuran.
  • a lactone ring substituent i.e. 2-carboalkoxy-(4-fluoro-phenoxy-methyl)- ⁇ -butyrolactone
  • the alkoxy group suitably has from 1 to about 12 carbon atom
  • the hydroxy tetrahydrofuran is further functionalized by activating the hydroxyl substituent ofthe hydroxytetrahydrofuran-phenyl ether followed by substitution ofthe corresponding position ofthe tetrahydrofuran ring with by a 1-alkyne reagent. Also, rather than directly activating the hydroxyl moiety, that group can be replaced with a halide, and the halide-substituted tetrahydrofuran reacted with a benzylsulfonic acid reagent.
  • methods ofthe invention enable such substitution of the tetrahydrofuran to proceed with extremely high stereoselectivity, e.g. at least greater than about 60 mole percent of one stereoisomer than the other, more typically greater than about 70 or 75 mole percent of one stereoisomer than the other isomer. Recrystallization has provided very high optical purities, e.g. about 95 mole %, 97 mole % or even 99 mole % or more ofthe single stereoisomer.
  • methods involve cleavage of a bis-compound to provide high yields of compound 1.
  • These methods preferably involve condensation of mannitol with an alkanoyl particularly an aldehyde such as formaldehyde to form a trialkylene mannitol such as a tri(C ⁇ - ⁇ oalkylene) mannitol e.g. trimethylene mannitol when using formaldehyde, which is then cleaved to form 2,5,-O-methylene-mannitol, which has two primary hydroxyl groups and two secondary hydroxyl groups.
  • the primary hydroxyl groups are protected (e.g. as esters) and the secondary hydroxyl groups then are suitably cyclized, e.g.
  • preparative methods include multiple reactions that surprisingly proceed as a single step without isolation of intermediates to provide compound 1.
  • the one step procedure is enantioselective.
  • the starting reagent a 2,3-epoxide
  • the resulting compound 1 also will be optically active.
  • a compound is reacted that has at least a six-carbon alkyl or alklyene chain that is activated at the 1- and 6-carbon positions such as by substitution by suitable leaving groups, and 2- and 3 -carbon positions ofthe chain form an epoxide ring.
  • the compound suitably has about 6-12 carbons in the chain.
  • the leaving groups ofthe 1- and 6-positions may be e.g.
  • halo such as chloro or bromo
  • an ester such as an alkyl or aryl sulfonic ester, e.g. mesylate or other C ⁇ -10 alkyl sulfonic ester, or a phenyl sulfonic ester such as tosylate and the like, or an arylalkyl ester such as benzylsulfonic ester.
  • the 1 -position is halo- substituted, particularly bromo-, iodo- or chloro-substituted, and the 6-position is substituted by an ester such as by a benzylsulfonyl group. That compound is reacted with a molar excess of a strong base such as an alkyllithium reagent that affords compound 1 in a single step.
  • a chiral synthon is preferably employed such as glyceraldehyde, mannitol, ascorbic acid, and the like, to provide a stereoselective route to desired stereoisomers of compound 1.
  • This route includes formation of a substituted dioxolane, typically a 1,3-dioxolane (particularly (2,2-dimethyl)- 1,3-dioxolane), which preferably is optically active.
  • a side chain ofthe dioxolane, preferably at the 4-position, is suitably extended e.g.
  • Wittig reactions typically one or two Wittig reactions that provide , ⁇ -unsaturated moieties such as an ⁇ , ⁇ -unsaturated alkyl ester.
  • asymmetric oxidation ofthe conjugated alkene to provide an optically active epoxide which then participates in an elimination reaction to yield a propargyl alcohol as the dioxolane ring substituent.
  • the dioxolane ring then can be opened, typically in the presence of acid and the acyclic intermediate cyclized to provide an optically active tetrahydrofuran that is l-alkyne-4-hydroxyalkyl- substituted, preferably l-ethynyl-4- hydroxymethyl-substituted. See generally Schemes VIII through X and the discussion related thereto below.
  • the substituted tetrahydrofuran can be further functionahzed as outlined above to provide compound 1.
  • the primary hydroxy ofthe alkylhydroxy substituent can be esterified (e.g., sulfonate such as a tosylate) and the activated carbon reacted to provide an aryl substituent, particularly para-fluorophenyl.
  • the alkynyl substituent can be extended to provided the hydroxy urea as discussed above.
  • a substituted dioxolane reagent is employed, again typically a 1,3-dioxolane (particularly (2,2-dimethyl)- 1,3-dioxolane), which preferably is optically active.
  • the alkyne reagent is preferably a butynyl compound with terminal ether group, preferably a terminal aryl or alkaryl ether such as optionally substituted 1 -(4- phenylmethylether)-butynyl.
  • the resulting substituted dioxolane can be opened suitably in the presence of acid to an acyclic intermediate, followed by cyclization under basic conditions to provide a substituted tetrahydrofuran which can be further functionahzed as discussed above with respect to Schemes VIII through X to provide compound 1. See generally Scheme XI and the discussion related thereto below.
  • a substituted dioxolane reagent is employed, again typically a 1,3-dioxolane (particularly (2,2-dimethyl)- 1,3-dioxolane), which preferably is optically active.
  • the terminal alkene is then epoxidized, e.g. by ozonolysis or other suitable oxidant.
  • the epoxidized keto alkyne then can be reduced and internally cyclized, e.g. in the presence of a suitable reducing agent such as diborone methyl sulfide, and then functionahzed to compound 1 as generally discussed above.
  • compound 1 will be useful for therapeutic applications, and may be employed to treat disorders or diseases mediated by 5 -lipoxygenase such as immune, allegeric and cardiovascular disorders and diseases, e.g. general inflammation, hypertension, skeletal-muscular disorders, osteoarthritis, gout, asthma, lung edema, adult respiratory distress syndrome, pain, aggregation of platelets, shock, rheumatoid arthritis, psoriatic arthritis, psoriasis, autoimmune uveitis, allergic encephalomyehtis, systemic lupus erythematosis, acute necrotizing hemmorrhagic encephalopathy, idiopathic thrombocytopenia, polychondritis, chronic active hepatitis, idiopathic sprue, Crohn's disease, Graves ophthalmopathy, primary biliary cirrhosis, uveitis posterior, interstitial lung fibrosis, allergic asthma and inappropriate
  • Compound 1 produced by the methods ofthe invention will be useful as a synthetic intermediate to prepare other compounds that will be useful for therapeutic applications.
  • Other aspects ofthe invention are disclosed infra.
  • Scheme I exemplifies a preferred preparative method ofthe invention wherein arylhydroxide 2 is reacted with epoxide 3 having a reactive C3 carbon.
  • Preferred epoxides are those that are enantiomerically enriched, such as the glycidyl tosylate 3 shown above that is condensed with phenol 2 for a time and temperature sufficient for reaction completion to provide epoxyaryl ether 4. See Example 1, Part 1 below for exemplary reaction conditions.
  • the reagents 2 and 3 are typically reacted in a suitable solvent, e.g. dimethyl formamide, N-methyl pyrrolidinone and the like.
  • Enantiomerically enriched epoxides suitable for condensation with an arylhydroxide are commercially available or can be readily prepared by known procedures. See, for instance, U.S. Patents Nos. 4,946,974 and 5,332,843 to Sharpless et al. for preparation of optically active derivatives of glycidol.
  • the epoxyaryl ether 4 then is reacted with an active methylene group, such a di ethyl or dimethyl malonate to provide butyrolactone 5.
  • the exocyclic ester of 5 is then suitably cleaved, e.g. with reaction with magnesium chloride hexahydrate, to provide the aryllactone ether 6. See Example 1, Part 3 which follows for an exemplary reaction conditions. That lactone 6 is then reduced to the hydroxy-tetrahydrofuran 7.
  • Suitable reducing agents include e.g. DIBAL-H and the like. See Example 1, Part 4, which follows.
  • Schemes II and III exemplify further preferred methods ofthe invention for synthesis of compound 1 and precursors thereof. More specifically, the hydroxy substituent of tetrahydrofuran 7 is preferably protected, e.g. as an ether or ester. Thus, as depicted in Schemes II and III, the hydroxy moiety of 7 can be reacted with a suitable silyl reagent, e.g. to form the t-butyldimethylsilyl ether 8, or with reagent for esterification, e.g. an anhydride such as acetic anhydride to acetyl ester 1_1. See Example 1, Part 5 and Example 2, Part 1 for suitable reaction conditions for exemplary conditions.
  • a suitable silyl reagent e.g. to form the t-butyldimethylsilyl ether 8
  • reagent for esterification e.g. an anhydride such as acetic anhydride to acetyl ester 1_1. See Example 1, Part
  • the protected phenyltetrahydrofuran ether 8 or 11 then can reacted to provide the alkynyl-substituted tetrahydrofuran 9 by treatment with a 1-alkyne in the presence of a strong base such an alkyllithium.
  • the alkyne reagent contains a protected hydroxy moiety such as a silyl ether, e.g. a tetrahydropyranyl ether as depicted in the above Schemes.
  • the hydroxy group can be readily deprotected after coupling ofthe alkynyl reagent to the tetrahydrofuran ring, e.g. by treatment with dilute acid.
  • the alkyne reagent will contain a primary or secondary hydroxy moiety.
  • Schemes IV and V above exemplify further convenient routes that can provide compound 1 and precursors thereof.
  • halo-substituted compound 12 can be reacted with an alkyne reagent as generally described above with respect to Schemes II and III to provide 9, which can be readily deprotected to provide the primary alcohol of compound 10. See generally Example 3 which follows for exemplary reaction conditions.
  • hydroxytetrahydrofuran 7 is condensed with a sulfmic acid reagent to provide the phenylsulfinic acid ester 8 which can be reacted with an alkyne reagent as generally described above to provide 9.
  • Compound K) is readily provided by treatment ofthe protected alcohol 9 with treatment with dilute acid. See Example 4 below.
  • Methylpheriylsulfmic acid including p-methylphenylsulfinic acid is particularly preferred, although phenyl ring substituents will be suitable in addition to methyl, including both electron-withdrawing and electron-donating phenyl ring substituents such as one or more C ⁇ - ⁇ alkyl groups more typically one or more C 1-8 alkyl or C ⁇ -6 alkyl groups , C 1-12 alkoxy or more typically C 1-6 alkoxy, cyano, nitro and the like. Para substitution ofthe phenyl reagent is generally preferred, although other phenyl ring positions also may be suitably substituted.
  • Scheme VI below exemplifies a further preferred method ofthe invention that can provide compound 1 and precursors thereof in high yields and involves cleavage of a bis- compound.
  • trimethylene mannitol 16 is suitably prepared by condensation of mannitol 15 with formaldehyde in the presence of acid.
  • the labile rings are cleaved and the resulting esters of 17 reduced to the primary and secondary alcohols of 18.
  • the primary alcohols are protected, e.g. as an allyl or aryl sulfonic ester, to provide intermediate 19.
  • the secondary hydroxyl groups of 19 then are functionahzed by reaction with a trialkylorthoformate, e.g. a such as triethylortho formate, to provide 20.
  • a trialkylorthoformate e.g. a such as triethylortho formate
  • the protected primary alcohols of 20 are then converted to 4-fluorophenyl ethers, preferably under basic conditions by reaction with 4-fluorophenol to provide di-(4-fluorophenyl)ether 21 . . That phenyl ether is then reacted in the presence of acid to cleave the methylene ethers to provide secondary hydroxyl groups of compound 22.
  • Compound 22 then undergoes oxidative cleavage by treatment with a suitable reagent such as Pb(OAc) , and the resulting dialdehyde is functionahzed to the acyclic bis-( ⁇ , ⁇ -unsaturated) compound, preferably bis- ⁇ , ⁇ -unsaturated ester 23 such as by reaction with carboethoxymethylenetriphenyl phosphorane.
  • a suitable reagent such as Pb(OAc)
  • the resulting dialdehyde is functionahzed to the acyclic bis-( ⁇ , ⁇ -unsaturated) compound, preferably bis- ⁇ , ⁇ -unsaturated ester 23 such as by reaction with carboethoxymethylenetriphenyl phosphorane.
  • Other ⁇ , ⁇ -unsaturated groups will be suitable for the alicyclic compound, e.g. ⁇ , ⁇ -unsaturated esters having 1 to about 12 carbon atoms, ⁇ , ⁇ -unsaturated acids, and
  • the carbon-carbon double bonds of 23 then are saturated, preferably by hydrogenation, and the resulting compound is cleaved and cyclized in the presence of acid to form the aryl ether 6.
  • the saturated compound is refluxed in a suitable solvent such as an alcohol, ethanol, for a time sufficient to provide 6. See Example 5 which follows for exemplary reagents and reaction conditions.
  • Compound 6 then can be further functionahzed, e.g. as discussed above with respect to Schemes II and III.
  • Scheme VII above exemplifies a further preferred method ofthe invention that provides compound 1 and precursors thereof and features multiple reactions that proceed as a single step without isolation of intermediates.
  • compound 2 is reacted with epoxide 24 that has a reactive C3 carbon to provide the fluorophenylepoxy ether 25.
  • the fluorophenylepoxy ether 25 may be resolved if desired such as by procedures generally depicted in Scheme VI above to provide optically active epoxide ethers 27 and 4. See Example 6, Parts 2-4 below for exemplary reagents and reaction conditions.
  • That procedure generally entails formation of optically active fluorophenyldiol ether and fluorophenylepoxide ether 26 and 27 from the racemic fluorophenylepoxide 25 with an optically active reagent, preferably an optically active catalyst such as Jacobsen's catalyst.
  • an optically active reagent preferably an optically active catalyst such as Jacobsen's catalyst.
  • the optically active diol 26 can be readily cyclized to the epoxide 4, for example by esterification (e.g. a sulfonic ester as shown exemplified by 28 above) ofthe primary hydroxyl group ofthe diol followed by epoxide formation under basic conditions (e.g. NaH).
  • An allyl halide is suitably reacted with the phenylepoxide ether, suitably in the presence of Mg, catalytic amount of iodine and cuprous cyanide to provide aryl/alkene ether 29.
  • the secondary hydroxy is suitably protected, e.g. as an ester, preferably as a sulfonic ester, to provide 30.
  • An ester group is then suitably grafted to terminal carbon- carbon double bond to the ⁇ , ⁇ -unsaturated ester 31, and the ester reduced to the alcohol, typically by treatment with strong base such as DIBAL-H.
  • the alkene is then suitably oxidized to provide epoxy group of 33.
  • the oxidation may be conducted to provide optically active epoxy carbons as generally shown in Scheme VI (compound 33) and conducted using suitable optically active reagent(s) such as an optically active catalyst or other reagent. See Example 6, Part 9 for an exemplary procedure.
  • suitable optically active reagent(s) such as an optically active catalyst or other reagent. See Example 6, Part 9 for an exemplary procedure.
  • the racemic epoxides also may be resolved, e.g. by chromatography using an optically active packing material.
  • the glycidyl compound 33 is then converted to the epihalohydrin 34.
  • the epihalohydrin 34 in a single step, is converted to the alkynyltetrahydrofuran ether 35 upon treatment with a molar excess, preferably at least about a three molar excess of a strong base such as an alkyllithium reagent or sodium amide.
  • BuLi is generally preferred, particularly n-BuLi.
  • the alkynyl group of compound 35 can be further functionahzed as desired, e.g. by reaction with ethylene oxide in the presence of base to afford the single enantiomer 10.
  • Compound K) is further functionahzed to produce compound 1 suitably by reacting compound 10 with N,O-bisphenoxycarbonyl hydroxylamine and triphenylphosphine and diisopropylazo-dicarboxylate, followed by treatment of resulting intermediate with NH 3 .
  • compound 1 is generated from 10 via a protected hydroxyurea
  • Scheme VIII shows another preferred preparative method ofthe invention that employs a polyol reagent.
  • the entire reaction is stereoselective (i.e. no separate resolution step or procedure required), begirming with the optically active mannitol 1, which is commercially available.
  • Other glyceraldehyde steroisomers can be employed in the same manner as depicted in Scheme VIII to provide the corresponding distinct stereoisomer as the reaction scheme product.
  • the chiral synthon (mannitol) is cyclized in the presence of base to the bis-dioxolane compound 2 which is then oxidized to the keto dioxolane 3 and reacted with an appropriate Wittig reagent to provide the ⁇ , ⁇ - unsaturated ester 4.
  • the term "Wittig reaction” or "Wittig-type reaction” designates any ofthe broad classes of alkene- formation reactions, typically involving ylide intermediates such as may be provided by phosphonate and phosphorane reagents.
  • the carbon-carbon double bond produced by the Wittig reaction then can be saturated, e.g. hydrogenated in the presence of a suitable catalyst such as PtO 2 , and the ester reduced and then oxidized to provide aldehyde 7.
  • Wittig reaction ofthe aldehyde moiety provides the ⁇ , ⁇ -unsaturated compound 9 which can be reduced to alcohol 9, and converted to the propargyl compound, e.g. via an epoxidized intermediate.
  • unsaturated alcohol 9 can be epoxidized to compound 10, suitably with an optically active oxidant and then elimination ofthe epihalohydrin derivative H in the presence of a suitable base e.g. LDA or other suitable agent to provide the propargyl compound 12.
  • a suitable base e.g. LDA or other suitable agent to provide the propargyl compound 12.
  • Acidic opening ofthe dioxolane ring provides diol 14 and esterification
  • Scheme IX depicts a related approach to provide another stereoisomer ofthe substituted alkyne terahydrofuran compound.
  • L-ascorbic acid can be employed as a starting reagent to provide hydroxy dioxolane compound 19, which is oxidized; subjected to multiple Witting reactions; epoxidized; and an epihalohydrin intermediate reacted in the presence of base to form a propargyl alcohol intermediate, which is converted to the optically active aryl-substituted alkyne tetrahydrofuran compounds 33 and 34.
  • the resulting tetrahydrofuran can be modified to compound 1, specifically functionalization ofthe alkynyl group as discussed above, particularly 2R,5R-(4-
  • the unsubstituted alkyne produced through the routes of Schemes VIII and IX above is a versatile intermediate that can be further reacted to provide a wide range of moieties, including groups that can be detected, either upon in vitro or in vivo applications.
  • the unsubstituted alkyne can be reacted with a group to provide radiolabeled and stable isotopic moieties e.g. 1 5 1, 3 H, 32 P, 99 Tc, 14 C, 13 C, 15 N or the like, which may be useful inter alia for mechanistic studies.
  • Scheme X depicts another route related to Schemes VIII and IX above and employs multiple Wittig reactions to provide a further stereoisomer of compound 1, specifically 2R,5 S-2-(4-fluorophenoxymethyl)-5-(4-N-hydroxyureidyl- 1 -butynyl)- tetrahydrofuran. See Example 9 which follows for exemplary reaction conditions.
  • Scheme XII depicts another convenient route to compound 1 that entails a single Wittig reaction. More particularly, the dioxolane aldehyde reagent is reacted with a Wittig reagent, the resulting alkene saturated such as by hydrogenation and the alkyl ester converted to acid 57. The acid is then reacted with a chloroformate reagent in the presence of base, and the resulting intermediate reacted with l-(4-methoxy phenylmethylether)-butynyl in the presence of a strong base, such as an alkyl lithium e.g. butyllithium.
  • a strong base such as an alkyl lithium e.g. butyllithium.
  • keto alkyne dioxolane compound 58 is then reduced asymmetrically in the presence of a suitable chiral catalyst, e.g. an optically active pinene and BBN.
  • a suitable chiral catalyst e.g. an optically active pinene and BBN.
  • the resulting optically active propargyl alcohol can be reacted as outlined above to provide
  • hydroxy tetrahydrofuran can be functionahzed as desired, e.g. esterification ofthe hydroxy moiety followed by aryl substitution and functionahzation ofthe alkynyl group provides 2-(4-fluorophenoxymethyl)-5-(4-N-hydroxyureidyl-l-butynyl)-tetrahydrofuran. See Example 12 which follows for exemplary preferred reaction conditions.
  • a lactone particularly butyrolactone such as ⁇ -butyrolactone ring-substituted (suitably at an position) by an activated ester (e.g., sulfonate ester such as tosylate, mesylate, etc.)
  • an activated ester e.g., sulfonate ester such as tosylate, mesylate, etc.
  • aryl nucleophiles include aryl compounds having one or more hydroxy ring substituents (i.e. an aryl hydroxy compound), particularly carbocyclic aryl compounds such as phenol, particularly 4-fluorophenol. See Example 13 which follows for exemplary preferred reaction conditions. It has been found that this substitution reaction proceeds without opening or other undesired attack ofthe lactone ring.
  • the substituent R is compound 60 is suitably hydrogen, alkyl e.g. C ⁇ - 8 alkyl and the like; and the substituent R' is suitably aryl as specified herein, particularly phenyl, more preferably 4-fluorophenyl.
  • Compound 60 can be readily functionahzed to provide compound 1, particularly by the procedures discussed above with respect to Schemes I and II.
  • compound 1 will be useful for numerous therapeutic applications.
  • the compounds can be administered to a subject, particularly a mammal such as a human, in need of treatment, by a variety of routes.
  • the compound can be administered orally, parenterally, intravenously, intradermally, subcutaneously, or topically.
  • the active compound may be administered to a subject as a pharmaceutically active salt, e.g. salts formed by addition of an inorganic acid such as hydrochloric acid, hydrobromic acid, phosphoric acid, etc., or an organic acid such as acetic acid, oxalic acid, tartaric acid, succinic acid, etc.
  • Base addition salts also can be formulated if an appropriate acidic group is present on the compound.
  • suitable base addition salts include those formed by addition of metal cations such as zinc, calcium, etc., or salts formed by addition of ammonium, tetraethylammonium, etc.
  • suitable dosages for a given therapy can be readily determined by the medical practitioner such as by standard dosing protocols. See also U.S. Patent 5,703,093.
  • an optically active or enantiometrically enriched mixture of compound 1 for a given therapeutic application.
  • enantiometrically enriched typically refers to a compound mixture that is at least approximately 70 mole%, 80 mole%, 85 mole% or 90 mole% of a single stereoisomer, and preferably a compound mixture that contains approximately at least about 92 mole%, 95 mole%, 97 mole%, 98 mole%, 99 mole% or 100% of a single enantiomer ofthe compound.
  • halo, halogen or the like refers to fluoro, chloro, bromo or iodo.
  • alkyl typically refers an alkyl group having 1 to about 20 carbon atoms, more typically 1 to about 12 carbon atoms, still more typically 1 to about 6 or 8 carbon atoms.
  • arylalkyl refers to a carbocyclic aryl such as phenyl that is substituted on an alkyl, particularly alkyl having 1 to about 6 to 8 carbons.
  • Example 1 Preparation of (2S)(5R)-2-(4-fluorophenoxymethyl)-5-(4-hydroxybutyn- 1 - yl)-tetrahydrofuran (Scheme II; 10)
  • (2S) (5RS)-5-bromo-2-(4-fluorophenoxymethyl)tetrahydrofuran (Scheme IV; 12) (2S) (5RS)-5-bromo-2-(4-fluorophenoxymethyl)tetrahydrofuran was prepared from (2S) (5RS)-5-O-acetyl-2-(4-fluorophenoxymethyl)tefrahydrofuran ll (1.06 g, 0.00417 mol) and TMS-Br (0.65 mL, 0.0049 mol).
  • the reaction mixture was quenched with saturated ammonium chloride solution and THF was removed on rotavapor.
  • the residue was partitioned between CH 2 C1 2 (20 mL) and water, and the organic layer was separated, washed with water, brine dried over Na SO 4 and concentrated on rotavapor to provide a crude yield of 1.73 g.
  • reaction mixture was cooled to 0°C and (2S) (5RS)-2-(4-fluorophenoxymethyl)-5-hydroxy-tetrahydrofuran (4.6g, 0.0216 mol) in dry CH 2 CI 2 (20 mL) was added.
  • the reaction mixture was filtered through celite and washed with CH 2 C1 (3 times). The combined organic layer was washed with saturated aqueous Na CO 3 , water brine and dried over Na 2 SO 4.
  • the isopropyl magnesium bromide was cannulated in a 1000 mL flame dried two-necked round bottom flask with spin-bar, nitrogen inlet and septum. 4-Tetrahydropyranoyl-l-butyne (11.0 g, 0.0714 mol) in THF (140 mL) was added. The reaction mixture was stirred for 30 min. and cooled at 0°C. Freshly prepared ZnBr 2 solution (1M, 43 mL, 0.0428 mol) in THF was introduced. After 45 min.
  • the reaction mixture was cooled in ice-water and solid was filtered washed with ethyl acetate (100 mL), and the combined filtrate was concentrated under reduced pressure.
  • the resulting residue was dissolved in ethyl acetate (800 mL) and the organic layer was washed with 2M NaOH (4x100 mL), water and brine dried over Na 2 SO 4 .
  • the reaction mixture was basified to pH 8 by saturated NaHCO 3 solution, and the solid was filtered and the filtrate concentrated to dryness to provide 125 kg (99%) of product 22, m.p. 126-127°C, [ ⁇ ] D -34.49° (c 1.148, MeCOMe).
  • reaction mixture was cooled to -22°C (CCl 4 /dry ice bath), and (2R)-l,2-epoxy-3-(4- fluoro)phenoxypropane 4 (2.05 g, 12.2 mmol) in 25 ml of ethyl ether was added.
  • the reaction was completed within 30 minutes, as determined by TLC (benzene). Saturated aqueous ammonium chloride (4 ml) then was added and the mixture stirred for 30 minutes. Inorganic material was filtered and washed with ethyl ether (25 ml).
  • reaction mixture then was filtered through a celite pad the filtrate was dried over anhydrous Na 2 SO 4 and concentrated, the residue was filtered through a short silica gel pad and concentrated to obtain (6R,2E)-ethyl-6-benzenesulfonyloxy-7-(4-fluoro)-phenoxy-hept-2- en-l-ol 32 as a solid (2.2 g, 82% yield).
  • Powdered molecular sieves 4A (3 g) were activated under N atmosphere in a 25 ml 2 necked round bottom flask. CH C1 2 (15 ml) was added followed by titanium tetraisopropoxide (1.62 ml, 5.47 mmol), (+)-diisopropyltartrate (1.07 ml, 6.56 mmol) and the mixture was cooled to -20°C with stirring. After 5 minutes cumene-hydroperoxide (2.1 ml, 10.94 mmol, 80% solution in cumene) was added dropwise.
  • reaction mixture was treated with Et 3 N (3 mL), to quench PTSA, and water (500 mL) and extracted with hexanes (3 x 200 mL) to remove the tri acetonide (5-7 g).
  • the reaction mixture was then extracted with EtO Ac (4 x 500 mL) and washed with NaCl solution. Evaporation ofthe solvent gave pure 1,2:5,6- di-O-isopropylidene-(D)- mannitol 2 (95 g) in 66% yield as a solid, m.p. 118-120 ° C (lit. m.p. 120-122 ° C (ref. Organic Synthesis 1995, 72, 6)).
  • Method B A stirred solution of (2R)-l,2-isopropylidenedioxy-5-pentanol 22 (0.800 g, 5.0 mmol) in DMSO (5 mL) was cooled to 0°C, treated with IBX (1.47 g, 5.26 mmol) in portions while maintaining the temperature below 0°C and stirred at room temperature for 4 h. The reaction mixture was treated with saturated NaHCO 3 solution, filtered through celite and washed with EtOAc (3 x 30 mL). Two layers were separated and organic layer was washed with water, brine and dried (Na 2 SO 4 ). Evaporation of solvent gave (4R)-4,5-isopropylidenedioxy-l-pentanal 23 (16.2 g) in 78% yield as a yellow liquid. [ ⁇ ] D +0.3(c 2.0, CHC1 3 ).
  • reaction mixture was cooled to room temperature and a solution of l,2-isopropylidenedioxy-9-p-methoxy phenyl methyl— 5- oxo-non-6-yn-9-ol 59 (0.25 g, 0.712 mmol) in THF (5 mL) was added and allowed to stir for 82 h.
  • the reaction mixture was treated withpropanaldehyde (0.5 mL), stirred for 15 min. and solvent was evaporated at room temperature.
  • the residue was dissolved in dry ether (10 mL), cooled to 0°C and treated with ethanolamine (0.097 g, 1.5 mmol) and stirred for 15 min. It was filtered and washed with ether (5 mL).
  • reaction mixture was quenched with aq. NH 4 C1 solution (10 mL) and extracted with EtOAc (2 x 25 mL). Organic layer was washed with brine (25 mL), dried (Na 2 SO ), evaporated and purified the residue by column chromatography (Si-gel, 8:1 Hexane-EtOAc) to afford non-8-ene- 1-p-methoxy phenyl methyl— 5-oxo-3-yn-l-ol (54; 0.35 g) in 27% yield as a colorless syrup.
  • Sodium hydride (60%, 192g) is taken in a 3 -neck round-bottom flask fitted with a mechanical stirrer and addition funnel. Dry n-hexane (1.0 L) is added and the mixture stirred for 15 minutes. The stirring is then stopped and the sodium hydride allowed to settle. After 15 minutes, n-hexane is decanted out. Dimethyl formamide (DMF) (2.0L) is charged to the flask. 4-fluorophenol (500 g) dissolved in DMF (2.0L) is added to the mixture over 0.5 hours, maintaining the temperature between 10-15°C. Tetrabutylammonium iodide (20 g) is added to the mixture maintaining temperature at 10-15°C.
  • the ethyl acetate layer is separated and washed with brine solution (2x2.0L) and then dried over sodium sulfate (250 g) and concentrated under reduced pressure.
  • the resulting crude product can be chromatographed over silica gel using 10% ethyl acetate:n-hexane as eluent.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Epoxy Compounds (AREA)
  • Furan Compounds (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Oxygen Or Sulfur (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention includes inter alia new methods for preparation of the pharmaceutically active compound 2-(4-fluorophenoxymethyl)-5-(4-N-hydroxyureidyl-1-butynyl)-tetrahydrofuran and precursors thereof.

Description

METHODS FOR SYNTHESIS OF SUBSTITUTED TETRAHYDROFURAN
COMPOUND
The present application claims the benefit of U.S. provisional application number 60/091,709, filed July 3, 1998, which is incorporated herein by reference in its entirety.
BACKGROUND
1. Field of the Invention
The present invention provides new methods for preparation of 2-(4- fluorophenoxymethyl)-5-(4-N-hydroxyureidyl- 1 -butynyl)-tetrahydrofuran ("compound 1") and synthetic precursors thereof.
2. Background
Leukotrienes are recognized potent local mediators, playing a significant role in inflammatory and allegeric responses, including arthritis, asthma, psoriasis and thrombotic disease. Leukotrienes are produced by the oxidation of arachidonic acid by lipoxygenase. More particularly, arachidonic acid is oxidized by 5-lipooxygenase to the hydroperoxide 5-hydroperoxy-eicosatetraenoic acid (5-HPETE), that is converted to leukotriene A4, that in turn can be converted to leukotriene B4, C4, or D4. The slow- reacting substance of anaphylaxis is now known to be a mixture of leukotrienes C4, D4 and E4, all of which are potent bronchoconstrictors.
Efforts have been made to identify receptor antagonists or inhibitors of leukotriene biosynthesis, to prevent or minimize pathogenic inflammatory responses mediated by leukotrienes.
For example, European Patent Application Nos. 901171171.0 and 901170171.0 report indole, benzofuran, and benzothiophene lipoxygenase inhibiting compounds. Various 2,5-disubstituted tetrahydrofurans have exhibited significant biological activity, including as lipoxygenase inhibitors. See U.S. Patent Nos. 5,703,093; 5,681,966; 5,648,486; 5,434,151; and 5,358,938.
While such compounds are highly useful therapeutic agents, current methods for synthesis of least some ofthe compounds require lengthy routes, and reagents and protocols that are less preferred in larger scale operations, such as to produce kilogram quantities.
SUMMARY OF THE INVENTION
We have now found new methods for preparation of 2-(4-fluorophenoxymethyl)- 5-(4-N-hydroxyureidyl-l-butynyl)-tetrahydrofuran and precursor compounds thereof. 2- (4-Fluorophenoxymethyl)-5-(4-N-hydroxyureidyl- 1 -butynyl)-tetrahydrofuran is sometimes referred to herein as "compound I". Preferred methods ofthe invention provide compound 1 in optically active form, particularly as an enantiomerically enriched mixture ofthe following stereoisomer (i.e. 2S,5S-trans-2-(4-fluorophenoxymethyl)-5-(4- N-hydroxyureidyl- 1 -butynyl)-tetrahydrofuran) :
Figure imgf000004_0001
The methods ofthe invention utilize reagents and synthetic protocols that facilitate large scale manufacture, and provide increased yields relative to prior approaches. In a first aspect ofthe invention, compound 1 and precursors thereof are provided by reacting 4-fluorophenol with an epoxide having a reactive C3 carbon, e.g. a glycidyl compound substituted at the C3 position with an electron-withdrawing group such as halo (e.g. epichlorohydrin, epibromohydrin), mesyl or tosyl (glycidyl mesylate and glycidyl tosylate), etc., to form an epoxyphenylether ((glycidyl-4-fluorophenyl ether) in the presence of base and preferably at or above about 0°C. The reacted epoxide can be optically active if desired. The formed epoxyphenylether is then reacted with an active methylene compound to form a lactone, preferably a γ-lactone, with 5 ring members. The active methylene compound can be a variety of agents. Di ethyl and dimethyl malonate are generally preferred, which provide an ethyl or methyl ester as a lactone ring substituent, i.e. 2-carboalkoxy-(4-fluoro-phenoxy-methyl)-γ-butyrolactone, where the alkoxy group suitably has from 1 to about 12 carbon atoms, more preferably 1 to about 6 carbon atoms, still more preferably 1 to about 3 carbons with methoxy and ethoxy particularly preferred. That ester group is then removed (e.g. via hydrolysis and decarboxylation), and the lactone suitably reduced to a hydroxy-substituted tetrahydrofuran, specifically 4-fluorophenoxymethyl-hydroxytetrahydrofuran.
The hydroxy tetrahydrofuran is further functionalized by activating the hydroxyl substituent ofthe hydroxytetrahydrofuran-phenyl ether followed by substitution ofthe corresponding position ofthe tetrahydrofuran ring with by a 1-alkyne reagent. Also, rather than directly activating the hydroxyl moiety, that group can be replaced with a halide, and the halide-substituted tetrahydrofuran reacted with a benzylsulfonic acid reagent.
It also has been found that methods ofthe invention enable such substitution of the tetrahydrofuran to proceed with extremely high stereoselectivity, e.g. at least greater than about 60 mole percent of one stereoisomer than the other, more typically greater than about 70 or 75 mole percent of one stereoisomer than the other isomer. Recrystallization has provided very high optical purities, e.g. about 95 mole %, 97 mole % or even 99 mole % or more ofthe single stereoisomer.
In another aspect, methods are provided that involve cleavage of a bis-compound to provide high yields of compound 1. These methods preferably involve condensation of mannitol with an alkanoyl particularly an aldehyde such as formaldehyde to form a trialkylene mannitol such as a tri(Cι-ιoalkylene) mannitol e.g. trimethylene mannitol when using formaldehyde, which is then cleaved to form 2,5,-O-methylene-mannitol, which has two primary hydroxyl groups and two secondary hydroxyl groups. The primary hydroxyl groups are protected (e.g. as esters) and the secondary hydroxyl groups then are suitably cyclized, e.g. with a trialkylorthoformate reagent, to provide a cyclic ether. The protected primary alcohols are then converted to aryl ethers, followed by cleavage ofthe cyclic ether to provide again the secondary hydroxyl groups. The mannitol compound then undergoes oxidative cleavage to provide the corresponding ali cyclic dialdehyde, which aldehyde groups are functionahzed to bis-α,β-unsaturated esters. The carbon-carbon double bonds of that compound are suitably saturated, and the bis-compound cleaved and the cleavage products cyclized to provide two molar equivalents of 4-fluorophenoxy-methyl-γ-butyrolactone which can be further functionahzed as described above.
In yet another aspect ofthe invention, preparative methods are provided that include multiple reactions that surprisingly proceed as a single step without isolation of intermediates to provide compound 1.
Moreover, it has been surprisingly found that the one step procedure is enantioselective. Hence, if the starting reagent (a 2,3-epoxide) is optically active, the resulting compound 1 also will be optically active. More particularly, in this aspect ofthe invention, a compound is reacted that has at least a six-carbon alkyl or alklyene chain that is activated at the 1- and 6-carbon positions such as by substitution by suitable leaving groups, and 2- and 3 -carbon positions ofthe chain form an epoxide ring. The compound suitably has about 6-12 carbons in the chain. The leaving groups ofthe 1- and 6-positions may be e.g. halo, such as chloro or bromo, or an ester, such as an alkyl or aryl sulfonic ester, e.g. mesylate or other Cι-10 alkyl sulfonic ester, or a phenyl sulfonic ester such as tosylate and the like, or an arylalkyl ester such as benzylsulfonic ester. Preferably, the 1 -position is halo- substituted, particularly bromo-, iodo- or chloro-substituted, and the 6-position is substituted by an ester such as by a benzylsulfonyl group. That compound is reacted with a molar excess of a strong base such as an alkyllithium reagent that affords compound 1 in a single step.
In another aspect ofthe invention, a chiral synthon is preferably employed such as glyceraldehyde, mannitol, ascorbic acid, and the like, to provide a stereoselective route to desired stereoisomers of compound 1. This route includes formation of a substituted dioxolane, typically a 1,3-dioxolane (particularly (2,2-dimethyl)- 1,3-dioxolane), which preferably is optically active. A side chain ofthe dioxolane, preferably at the 4-position, is suitably extended e.g. by one or more Wittig reactions, typically one or two Wittig reactions that provide ,β-unsaturated moieties such as an α,β-unsaturated alkyl ester. Such an α,β-unsaturated moiety provided then can be epoxidized, preferably by asymmetric oxidation ofthe conjugated alkene to provide an optically active epoxide, which then participates in an elimination reaction to yield a propargyl alcohol as the dioxolane ring substituent. The dioxolane ring then can be opened, typically in the presence of acid and the acyclic intermediate cyclized to provide an optically active tetrahydrofuran that is l-alkyne-4-hydroxyalkyl- substituted, preferably l-ethynyl-4- hydroxymethyl-substituted. See generally Schemes VIII through X and the discussion related thereto below. The substituted tetrahydrofuran can be further functionahzed as outlined above to provide compound 1. For instance, the primary hydroxy ofthe alkylhydroxy substituent can be esterified (e.g., sulfonate such as a tosylate) and the activated carbon reacted to provide an aryl substituent, particularly para-fluorophenyl. The alkynyl substituent can be extended to provided the hydroxy urea as discussed above.
In an alternative method ofthe invention, a substituted dioxolane reagent is employed, again typically a 1,3-dioxolane (particularly (2,2-dimethyl)- 1,3-dioxolane), which preferably is optically active. The dioxolane has an alkanoyl side chain, more particularly a propionaldehyde (-CH CH2C(=O)H) substituent that is reacted suitably in the presence of base (e.g. an alkyllithium) with a 1-alkyne to provide a proparyl alcohol. The alkyne reagent is preferably a butynyl compound with terminal ether group, preferably a terminal aryl or alkaryl ether such as optionally substituted 1 -(4- phenylmethylether)-butynyl. The resulting substituted dioxolane can be opened suitably in the presence of acid to an acyclic intermediate, followed by cyclization under basic conditions to provide a substituted tetrahydrofuran which can be further functionahzed as discussed above with respect to Schemes VIII through X to provide compound 1. See generally Scheme XI and the discussion related thereto below.
In a further synthetic route ofthe invention, a substituted dioxolane reagent is employed, again typically a 1,3-dioxolane (particularly (2,2-dimethyl)- 1,3-dioxolane), which preferably is optically active. The dioloxane has a keto alkyne side chain, preferably -CH CH2C(=O)C≡CR where R is optionally substituted alkyl , pasrticularly Cl-6 alkyl, or an alkylether or alkaryl ether such as a C]-6 ether, preferably an ethyl aryl or other (C)alkylaryl ether such as -CH CH OCH2(phenyl or substituted phenyl). The keto group is then reduced, preferably asymmetrically such as by use of a chiral catalyst, to provide a propargyl alcohol that can be further functionahzed to compound I as generally discussed above. See Scheme XII and the related discussion below. In yet a further aspect ofthe invention, an alkyne-substituted tetrahydrofuran is prepared directly (e.g., without a dioxolane intermediate) from an acyclic keto alkyne compound. More specifically, a keto alkynyl reagent with terminal alkenyl group is suitably employed, e.g. -CH2=CHCH CH2C(=O)C≡CR where R is the same as defined immediately above. The terminal alkene is then epoxidized, e.g. by ozonolysis or other suitable oxidant. The epoxidized keto alkyne then can be reduced and internally cyclized, e.g. in the presence of a suitable reducing agent such as diborone methyl sulfide, and then functionahzed to compound 1 as generally discussed above.
As mentioned above, compound 1 will be useful for therapeutic applications, and may be employed to treat disorders or diseases mediated by 5 -lipoxygenase such as immune, allegeric and cardiovascular disorders and diseases, e.g. general inflammation, hypertension, skeletal-muscular disorders, osteoarthritis, gout, asthma, lung edema, adult respiratory distress syndrome, pain, aggregation of platelets, shock, rheumatoid arthritis, psoriatic arthritis, psoriasis, autoimmune uveitis, allergic encephalomyehtis, systemic lupus erythematosis, acute necrotizing hemmorrhagic encephalopathy, idiopathic thrombocytopenia, polychondritis, chronic active hepatitis, idiopathic sprue, Crohn's disease, Graves ophthalmopathy, primary biliary cirrhosis, uveitis posterior, interstitial lung fibrosis, allergic asthma and inappropriate allergic responses to environmental stimuli.
Compound 1 produced by the methods ofthe invention will be useful as a synthetic intermediate to prepare other compounds that will be useful for therapeutic applications. Other aspects ofthe invention are disclosed infra.
DETAILED DESCRIPTION OF THE INVENTION
Particularly preferred preparative methods ofthe invention are exemplified in the following Schemes I through XIV. For purposes of exemplification only, particularly preferred compounds are depicted in the Schemes, and it will be understood that a variety of other compounds can be employed in similar manner as described below with respect to the exemplified compounds.
Scheme I
Figure imgf000010_0001
Figure imgf000010_0002
Figure imgf000010_0003
Scheme I exemplifies a preferred preparative method ofthe invention wherein arylhydroxide 2 is reacted with epoxide 3 having a reactive C3 carbon. Preferred epoxides are those that are enantiomerically enriched, such as the glycidyl tosylate 3 shown above that is condensed with phenol 2 for a time and temperature sufficient for reaction completion to provide epoxyaryl ether 4. See Example 1, Part 1 below for exemplary reaction conditions. The reagents 2 and 3 are typically reacted in a suitable solvent, e.g. dimethyl formamide, N-methyl pyrrolidinone and the like. Enantiomerically enriched epoxides suitable for condensation with an arylhydroxide are commercially available or can be readily prepared by known procedures. See, for instance, U.S. Patents Nos. 4,946,974 and 5,332,843 to Sharpless et al. for preparation of optically active derivatives of glycidol. The epoxyaryl ether 4 then is reacted with an active methylene group, such a di ethyl or dimethyl malonate to provide butyrolactone 5. The exocyclic ester of 5 is then suitably cleaved, e.g. with reaction with magnesium chloride hexahydrate, to provide the aryllactone ether 6. See Example 1, Part 3 which follows for an exemplary reaction conditions. That lactone 6 is then reduced to the hydroxy-tetrahydrofuran 7. Suitable reducing agents include e.g. DIBAL-H and the like. See Example 1, Part 4, which follows.
SCHEME II
Figure imgf000011_0001
SCHEME III
Figure imgf000012_0001
Schemes II and III exemplify further preferred methods ofthe invention for synthesis of compound 1 and precursors thereof. More specifically, the hydroxy substituent of tetrahydrofuran 7 is preferably protected, e.g. as an ether or ester. Thus, as depicted in Schemes II and III, the hydroxy moiety of 7 can be reacted with a suitable silyl reagent, e.g. to form the t-butyldimethylsilyl ether 8, or with reagent for esterification, e.g. an anhydride such as acetic anhydride to acetyl ester 1_1. See Example 1, Part 5 and Example 2, Part 1 for suitable reaction conditions for exemplary conditions.
The protected phenyltetrahydrofuran ether 8 or 11 then can reacted to provide the alkynyl-substituted tetrahydrofuran 9 by treatment with a 1-alkyne in the presence of a strong base such an alkyllithium. Preferably the alkyne reagent contains a protected hydroxy moiety such as a silyl ether, e.g. a tetrahydropyranyl ether as depicted in the above Schemes. The hydroxy group can be readily deprotected after coupling ofthe alkynyl reagent to the tetrahydrofuran ring, e.g. by treatment with dilute acid. Typically, the alkyne reagent will contain a primary or secondary hydroxy moiety. SCHEME IV
Figure imgf000013_0001
SCHEME V
Figure imgf000013_0002
Schemes IV and V above exemplify further convenient routes that can provide compound 1 and precursors thereof. Thus, in Scheme IV, halo-substituted compound 12 can be reacted with an alkyne reagent as generally described above with respect to Schemes II and III to provide 9, which can be readily deprotected to provide the primary alcohol of compound 10. See generally Example 3 which follows for exemplary reaction conditions.
In Scheme V, hydroxytetrahydrofuran 7 is condensed with a sulfmic acid reagent to provide the phenylsulfinic acid ester 8 which can be reacted with an alkyne reagent as generally described above to provide 9. Compound K) is readily provided by treatment ofthe protected alcohol 9 with treatment with dilute acid. See Example 4 below.
It also has been found that enhanced yields can be obtained by use of a phenylsulfinic acid reagent that is substituted at one or more positions on the phenyl ring, rather than the unsubstituted phenylsulfinic reagent. Methylpheriylsulfmic acid, including p-methylphenylsulfinic acid is particularly preferred, although phenyl ring substituents will be suitable in addition to methyl, including both electron-withdrawing and electron-donating phenyl ring substituents such as one or more Cι-ι alkyl groups more typically one or more C1-8alkyl or Cι-6alkyl groups , C1-12 alkoxy or more typically C1-6alkoxy, cyano, nitro and the like. Para substitution ofthe phenyl reagent is generally preferred, although other phenyl ring positions also may be suitably substituted.
Scheme VI below exemplifies a further preferred method ofthe invention that can provide compound 1 and precursors thereof in high yields and involves cleavage of a bis- compound.
SCHEME VI
D-M
Figure imgf000015_0001
Figure imgf000015_0002
More specifically, as depicted above, trimethylene mannitol 16 is suitably prepared by condensation of mannitol 15 with formaldehyde in the presence of acid. The labile rings are cleaved and the resulting esters of 17 reduced to the primary and secondary alcohols of 18. The primary alcohols are protected, e.g. as an allyl or aryl sulfonic ester, to provide intermediate 19. The secondary hydroxyl groups of 19 then are functionahzed by reaction with a trialkylorthoformate, e.g. a
Figure imgf000016_0001
such as triethylortho formate, to provide 20. The protected primary alcohols of 20 are then converted to 4-fluorophenyl ethers, preferably under basic conditions by reaction with 4-fluorophenol to provide di-(4-fluorophenyl)ether 21.. That phenyl ether is then reacted in the presence of acid to cleave the methylene ethers to provide secondary hydroxyl groups of compound 22.
Compound 22 then undergoes oxidative cleavage by treatment with a suitable reagent such as Pb(OAc) , and the resulting dialdehyde is functionahzed to the acyclic bis-(α,β-unsaturated) compound, preferably bis-α,β-unsaturated ester 23 such as by reaction with carboethoxymethylenetriphenyl phosphorane. Other α,β-unsaturated groups will be suitable for the alicyclic compound, e.g. α,β-unsaturated esters having 1 to about 12 carbon atoms, α,β-unsaturated acids, and other Michael-type acceptors. The carbon-carbon double bonds of 23 then are saturated, preferably by hydrogenation, and the resulting compound is cleaved and cyclized in the presence of acid to form the aryl ether 6. In one system, the saturated compound is refluxed in a suitable solvent such as an alcohol, ethanol, for a time sufficient to provide 6. See Example 5 which follows for exemplary reagents and reaction conditions. Compound 6 then can be further functionahzed, e.g. as discussed above with respect to Schemes II and III. SCHEME VII
Figure imgf000017_0001
Figure imgf000017_0002
Figure imgf000017_0003
SCHEME VII (Continued)
Figure imgf000018_0001
Figure imgf000018_0002
Figure imgf000018_0003
Figure imgf000018_0004
Scheme VII above exemplifies a further preferred method ofthe invention that provides compound 1 and precursors thereof and features multiple reactions that proceed as a single step without isolation of intermediates.
More specifically, as shown above compound 2 is reacted with epoxide 24 that has a reactive C3 carbon to provide the fluorophenylepoxy ether 25. If the epoxide 24 is not enantiomerically enriched such as 3, the fluorophenylepoxy ether 25 may be resolved if desired such as by procedures generally depicted in Scheme VI above to provide optically active epoxide ethers 27 and 4. See Example 6, Parts 2-4 below for exemplary reagents and reaction conditions. That procedure generally entails formation of optically active fluorophenyldiol ether and fluorophenylepoxide ether 26 and 27 from the racemic fluorophenylepoxide 25 with an optically active reagent, preferably an optically active catalyst such as Jacobsen's catalyst. See E. Jacobsen, Science, 277:936-938 (1997). The optically active diol 26 can be readily cyclized to the epoxide 4, for example by esterification (e.g. a sulfonic ester as shown exemplified by 28 above) ofthe primary hydroxyl group ofthe diol followed by epoxide formation under basic conditions (e.g. NaH).
An allyl halide is suitably reacted with the phenylepoxide ether, suitably in the presence of Mg, catalytic amount of iodine and cuprous cyanide to provide aryl/alkene ether 29. The secondary hydroxy is suitably protected, e.g. as an ester, preferably as a sulfonic ester, to provide 30. An ester group is then suitably grafted to terminal carbon- carbon double bond to the α,β-unsaturated ester 31, and the ester reduced to the alcohol, typically by treatment with strong base such as DIBAL-H.
The alkene is then suitably oxidized to provide epoxy group of 33. The oxidation may be conducted to provide optically active epoxy carbons as generally shown in Scheme VI (compound 33) and conducted using suitable optically active reagent(s) such as an optically active catalyst or other reagent. See Example 6, Part 9 for an exemplary procedure. The racemic epoxides also may be resolved, e.g. by chromatography using an optically active packing material. The glycidyl compound 33 is then converted to the epihalohydrin 34.
The epihalohydrin 34, in a single step, is converted to the alkynyltetrahydrofuran ether 35 upon treatment with a molar excess, preferably at least about a three molar excess of a strong base such as an alkyllithium reagent or sodium amide. BuLi is generally preferred, particularly n-BuLi.
While not being bound by theory, it is believed the single step reaction proceeds through the mechanism shown immediately below, where Ar is 4-fluorophenyl and Ms is mesyl (-S(O)2CH3):
Figure imgf000020_0001
Figure imgf000020_0002
The alkynyl group of compound 35 can be further functionahzed as desired, e.g. by reaction with ethylene oxide in the presence of base to afford the single enantiomer 10. Compound K) is further functionahzed to produce compound 1 suitably by reacting compound 10 with N,O-bisphenoxycarbonyl hydroxylamine and triphenylphosphine and diisopropylazo-dicarboxylate, followed by treatment of resulting intermediate with NH3.
More preferably, compound 1 is generated from 10 via a protected hydroxyurea
(e.g., a compound ofthe formula NH2C(O)NHOR, where R is a hydroxy protecting group such as oαrα-methoxybenzyl-) is reacted with a substituted alcohol compound such as 10 of Scheme II, preferably in the presence of suitable dehydrating agent(s) such as triphenyl phosphine and diethylazodicarboxylate (DEAD), to provide an amino ester, i.e. a moiety ofthe formula -NRC(O)OR!R where R is as defined immediately above and R1 is a non-hydrogen group such as aryl, particularly phenyl, alkyl, e.g. CM O alkyl, etc. That amino ester is then treated with ammonia and a Lewis acid such as boron trifluoride etherate and the like to provide compound .
Scheme VIII shows another preferred preparative method ofthe invention that employs a polyol reagent. As depicted in the below Scheme, the entire reaction is stereoselective (i.e. no separate resolution step or procedure required), begirming with the optically active mannitol 1, which is commercially available. Other glyceraldehyde steroisomers can be employed in the same manner as depicted in Scheme VIII to provide the corresponding distinct stereoisomer as the reaction scheme product.
In the following Schemes VIII through XIV, the compound numerals in the discussions of those Schemes are made in reference to the compound depicted in the particular Scheme, with the exception of compound 1, i.e. 2-(4-fluorophenoxymethyl)-5- (4-N-hydroxyureidyl- 1 -butynyl)-tetrahydrofuran.
As generally exemplified in Scheme VIII below, the chiral synthon (mannitol) is cyclized in the presence of base to the bis-dioxolane compound 2 which is then oxidized to the keto dioxolane 3 and reacted with an appropriate Wittig reagent to provide the α,β- unsaturated ester 4. As referred to herein, unless specified otherwise, the term "Wittig reaction" or "Wittig-type reaction" designates any ofthe broad classes of alkene- formation reactions, typically involving ylide intermediates such as may be provided by phosphonate and phosphorane reagents. Additionally, as referred to herein, unless otherwise specified, to "keto", "carbonyl", or "carboxy" or like term designate any functional group that includes a carbon-oxygen double bond (C=O). The carbon-carbon double bond produced by the Wittig reaction then can be saturated, e.g. hydrogenated in the presence of a suitable catalyst such as PtO2, and the ester reduced and then oxidized to provide aldehyde 7. Wittig reaction ofthe aldehyde moiety provides the α,β-unsaturated compound 9 which can be reduced to alcohol 9, and converted to the propargyl compound, e.g. via an epoxidized intermediate. More specifically, unsaturated alcohol 9 can be epoxidized to compound 10, suitably with an optically active oxidant and then elimination ofthe epihalohydrin derivative H in the presence of a suitable base e.g. LDA or other suitable agent to provide the propargyl compound 12. Acidic opening ofthe dioxolane ring provides diol 14 and esterification
10 (e.g. sulfonate ester such as a tosylate) provides the substituted tetrahydrofuran 16. The resulting hydroxy tetrahydrofuran can be functionahzed as desired, e.g. esterification of the hydroxy followed by aryl substitution and functionalization of the alkynyl group provides compound 1, particularly 2S,5S-trans-(4-fluorophenoxymethyl)-5-(4-N- hydroxyureidyl-l-butynyl)-tetrahydrofuran. See, generally, Example 7 which follows for
15 exemplary preferred reaction procedures.
Scheme VIII
Figure imgf000022_0001
5 K2C&., MeOH
v J ) -^_ 16 TsCI Et3N O^ ^ • /~~~ =^- u 17 ArOH aH, DMF Compound 1 16 R = H
17 R = Ts
+ 18 R = p-F-C3H4
Tosylate (17 R = τs) Scheme IX depicts a related approach to provide another stereoisomer ofthe substituted alkyne terahydrofuran compound. As shown in Scheme IX, L-ascorbic acid can be employed as a starting reagent to provide hydroxy dioxolane compound 19, which is oxidized; subjected to multiple Witting reactions; epoxidized; and an epihalohydrin intermediate reacted in the presence of base to form a propargyl alcohol intermediate, which is converted to the optically active aryl-substituted alkyne tetrahydrofuran compounds 33 and 34. See Example 8 which follows, for exemplary preferred reaction conditions. The resulting tetrahydrofuran can be modified to compound 1, specifically functionalization ofthe alkynyl group as discussed above, particularly 2R,5R-(4-
10 fluorophenoxymethyl)-5-(4-N-hydroxyureidyl- 1 -butynyl)-tetrahydrofuran.
Scheme IX
NaD«
Figure imgf000023_0001
Figure imgf000023_0002
7. (-) DIPT
8. P 3P / CCI *
Figure imgf000023_0003
+ Ditosylate (31a)
Compound 1
Figure imgf000023_0004
It should be appreciated that the unsubstituted alkyne produced through the routes of Schemes VIII and IX above is a versatile intermediate that can be further reacted to provide a wide range of moieties, including groups that can be detected, either upon in vitro or in vivo applications. For instance, the unsubstituted alkyne can be reacted with a group to provide radiolabeled and stable isotopic moieties e.g. 1 51, 3H, 32P, 99Tc, 14C, 13C, 15N or the like, which may be useful inter alia for mechanistic studies.
Scheme X below depicts another route related to Schemes VIII and IX above and employs multiple Wittig reactions to provide a further stereoisomer of compound 1, specifically 2R,5 S-2-(4-fluorophenoxymethyl)-5-(4-N-hydroxyureidyl- 1 -butynyl)- tetrahydrofuran. See Example 9 which follows for exemplary reaction conditions.
Scheme X
Figure imgf000024_0001
Compound _!__
Figure imgf000024_0002
Scheme XI below exemplifies further methods ofthe invention that utilize dioxolane intermediate 7. Rather than employing multiple Wittig reactions, a 1-alkyne is reacted with aldehyde reagent 7, the resulting dioxolane 35 esterified and the dioxolane ring opened in the presence of acid, followed by cyclization in the presence of base, suitable a relatively weak base such as K2CO3. The cyclization of 38 to optically active substituted tetrahydrofuran 39 can proceed with high stereoselectivity. The resulting tetrahydrofuran then can be further functionahzed to provide 2-(4-fiuorophenoxyrnethyl)- 5-(4-N-hydroxyureidyl-l-butynyl)-tetrahydrofuran. See Example 10 which follows, for exemplary preferred reaction conditions. 0 Scheme XI
Figure imgf000025_0001
35 R = H 36 R = Ac
Figure imgf000025_0002
Compouud 1
Scheme XII below depicts another convenient route to compound 1 that entails a single Wittig reaction. More particularly, the dioxolane aldehyde reagent is reacted with a Wittig reagent, the resulting alkene saturated such as by hydrogenation and the alkyl ester converted to acid 57. The acid is then reacted with a chloroformate reagent in the presence of base, and the resulting intermediate reacted with l-(4-methoxy phenylmethylether)-butynyl in the presence of a strong base, such as an alkyl lithium e.g. butyllithium. The keto alkyne dioxolane compound 58 is then reduced asymmetrically in the presence of a suitable chiral catalyst, e.g. an optically active pinene and BBN. The resulting optically active propargyl alcohol can be reacted as outlined above to provide
10 compound L particularly 2S,5S-trans-2-(4-fluorophenoxymethyl)-5-(4-N- hydroxyureidyl-l-butynyl)-tetrahydrofuran. See Example 11 which follows for exemplary reaction conditions.
Scheme XII
Figure imgf000026_0001
Figure imgf000026_0002
Scheme XIII below depicts a highly efficient route to 2-(4-fluorophenoxymethyl)- 5-(4-N-hydroxyureidyl-l-bu1ynyl)-tetrahydrofuran. As shown in the Scheme, butynyl reagent 52 is treated with base, preferably a strong base such as an alkyl lithium e.g. butyl lithium, and then reacted with an unsarurated anhydride 53 to provide the keto alkynyl compound 54 with terminal alkene group. The alkene group is oxidized, e.g. via ozonolysis, and the keto-epoxide compound 55 reduced and cyclized typcially in the presence of a suitable reducing agent, e.g. diborane methyl sulfide. The resulting hydroxy tetrahydrofuran can be functionahzed as desired, e.g. esterification ofthe hydroxy moiety followed by aryl substitution and functionahzation ofthe alkynyl group provides 2-(4-fluorophenoxymethyl)-5-(4-N-hydroxyureidyl-l-butynyl)-tetrahydrofuran. See Example 12 which follows for exemplary preferred reaction conditions.
Scheme XIII
Figure imgf000027_0001
5. TsCl, Py
6. p-FCeHtOH ,K2C03
7. DDQ
Compound 1
Figure imgf000027_0002
R = MP R = H In another aspect ofthe invention, it has been found that that a lactone, particularly butyrolactone such as γ -butyrolactone ring-substituted (suitably at an position) by an activated ester (e.g., sulfonate ester such as tosylate, mesylate, etc.) can be reacted with an aryl nucleophile to provide in good yields a methylene aryloxy group. More specifically, with reference to Scheme XIV below, compound 60 will undergo a displacement reaction with an aryl nucleophile in the presence of a hydride reagent (base), such as potassium hydride or more preferably sodium hydride to yield the tetrahydrofuran arylether 6L Preferred aryl nucleophiles include aryl compounds having one or more hydroxy ring substituents (i.e. an aryl hydroxy compound), particularly carbocyclic aryl compounds such as phenol, particularly 4-fluorophenol. See Example 13 which follows for exemplary preferred reaction conditions. It has been found that this substitution reaction proceeds without opening or other undesired attack ofthe lactone ring. In Scheme XIV, the substituent R is compound 60 is suitably hydrogen, alkyl e.g. Cι-8 alkyl and the like; and the substituent R' is suitably aryl as specified herein, particularly phenyl, more preferably 4-fluorophenyl. Compound 60 can be readily functionahzed to provide compound 1, particularly by the procedures discussed above with respect to Schemes I and II.
Scheme XIV
Figure imgf000028_0001
60 61
As discussed above, compound 1 will be useful for numerous therapeutic applications. The compounds can be administered to a subject, particularly a mammal such as a human, in need of treatment, by a variety of routes. For example, the compound can be administered orally, parenterally, intravenously, intradermally, subcutaneously, or topically. The active compound may be administered to a subject as a pharmaceutically active salt, e.g. salts formed by addition of an inorganic acid such as hydrochloric acid, hydrobromic acid, phosphoric acid, etc., or an organic acid such as acetic acid, oxalic acid, tartaric acid, succinic acid, etc. Base addition salts also can be formulated if an appropriate acidic group is present on the compound. For example, suitable base addition salts include those formed by addition of metal cations such as zinc, calcium, etc., or salts formed by addition of ammonium, tetraethylammonium, etc. Suitable dosages for a given therapy can be readily determined by the medical practitioner such as by standard dosing protocols. See also U.S. Patent 5,703,093.
Often, it will be preferable to use an optically active or enantiometrically enriched mixture of compound 1 for a given therapeutic application. As used herein, the term "enantiometrically enriched" typically refers to a compound mixture that is at least approximately 70 mole%, 80 mole%, 85 mole% or 90 mole% of a single stereoisomer, and preferably a compound mixture that contains approximately at least about 92 mole%, 95 mole%, 97 mole%, 98 mole%, 99 mole% or 100% of a single enantiomer ofthe compound.
As used herein, the term halo, halogen or the like refers to fluoro, chloro, bromo or iodo. The term alkyl typically refers an alkyl group having 1 to about 20 carbon atoms, more typically 1 to about 12 carbon atoms, still more typically 1 to about 6 or 8 carbon atoms. The term arylalkyl refers to a carbocyclic aryl such as phenyl that is substituted on an alkyl, particularly alkyl having 1 to about 6 to 8 carbons.
All documents mentioned herein are incorporated herein by reference.
The following non-limiting examples are illustrative ofthe invention. Example 1 : Preparation of (2S)(5R)-2-(4-fluorophenoxymethyl)-5-(4-hydroxybutyn- 1 - yl)-tetrahydrofuran (Scheme II; 10)
Part 1 : (S)-Glycidyl-4-fluorophenyl ether (Scheme I; 4) In a 100 ml two-necked round bottom flask equipped with magnetic stir bar, nitrogen inlet and a septum, was taken sodium hydride (60% dispersion in oil, 0.742g, 0.0185 mol) and lOmL of dry dimethyl formamide (DMF). The reaction mixture was cooled to 0°C and 4-fluorophenol 2 (1.9 g, 0.017 mol) in dry DMF (20 mL) was introduced. The reaction mixture was stirred at room temperature for 1 hour and cooled to 0°C. (S)-Glycidyl tosylate 3 (3.52 g, 0.015 mol) in DMF (10 mL) was added, and the reaction mixture was stirred at room temperature and monitored by TLC (EtO Ac-light petroleum ether 1 :4, Rf=0.5). After 4 hours, the reaction mixture was quenched by addition of ice-water (lmL) and extracted with (2x25 mL) ethyl ether. The ether layer was washed with water, brine, dried over Na2SO4 and concentrated under reduced pressure to afford (S)-glycidyl-4-fluorophenyl ether 4, crude yield 3.6 g. The crude compound was purified by distillation at 160°-170°C/9mm, to yield 1.98 g (76%) of purified product 4, [α]D + 4.96° (c 2.335, CHC13). 1H NMR (200 MHz, CDC13) : δ 2.68 (dd, J = 4.5, 2.2 Hz, 1 H), 2.85 (t, J = 4.5 Hz, 1 H), 3.27 (m, 1 H), 3.89 (dd, J = 15.7, 6.7 Hz, 1 H), 4.11 (dd, J = 15.7, 4.5 Hz, 1 H), 6.74 - 7.02 (m, 4 H).
Part 2: (4S)-2-carboethoxy-(4-fluoro-phenoxy-methyl)-γ-butyrolactone (Scheme i; 5)
In a 50 ml two-necked round bottom flask equipped with magnetic stir bar, nitrogen inlet septum, sodium salt of diethyl malonate (prepared from 1.8 mL/0.0118 mol of diethyl malonate and 0.245 g/0.0106 mol of sodium) in dry THF (10 mL) was taken. The reaction mixture was cooled to 0°C and (S)-glycidyl-4-fluorophenyl ether 4_(1.788 g, 0.0106 mol) in tetrahydrofuran (THF) (10 mL) was added. The reaction mixture was stirred at room temperature and monitored by TLC, (EtO Ac-light petroleum 1:3, Rf=0.30). After 12 hours, THF was removed on rotavapor. The residue was dissolved in ethyl acetate (25 mL) and washed with water, brine, dried over Na SO4 and concentrated on rotavapor to afford (4S)-2-carboethoxy-(4-fluoro-phenoxy-methyl)-γ-butyrolactone 5, with a crude yield of 2.816 g. That crude product was purified on silica gel column chromatography using EtOAc-light petroleum ether (1:8) to provide 2.10 g (70%) of purified product 5, m.p.69-71°C, [α]D + 16.95° (c 1.51, CHC13). 1H NMR (200 MHz, CDC13): δ 1.3 (m, 3 H), 2.37 - 2.9 (m, 2 H), 3.52 - 3.8 (m, 1 H), 3.95 4.32 (m, 4 H), 4.68 - 4.82 (m, 1/3 H), 4.82 - 4.98 (m, 2/3 H), 6.72 - 7.01 (m, 4 H). It is also noted that the crude product can be suitably employed directly in the decarboxylative elimination of Part 3 below.
Part 3: (4S)-4-fluorophenoxy-methyl)-γ-butyrolactone (Scheme I; 6)
(4S)-2-carboethoxy-(4-fluoro-phenoxy-methyl)-γ-butyrolactone 5 (2.1 g, 0.0074 mol) and N,N-dimethylacetamide (10 mL) were taken in a 25 mL round bottom flask equipped with a stir bar and reflux condenser. MgCl 6H O (1.51 g, 0.0074 mol) was added, and the reaction mixture was heated under reflux for 4 hours and monitored by TLC (EtO Ac-light petroleum 1 :2, Rf=0.2). The reaction mixture then was partitioned between ethyl ether and water (50 mL each). The ether layer was separated, washed twice with water, brine, dried over Na SO4 and concentrated on rotavapor to afford (4S)- 4-fluorophenoxy-methyl)-γ-butyrolactone 6, yield 1.40 g (90%), m.p. 58-59°C, [α]D + 23° (c 1.99, CHC13), e.e. 92%. 1H NMR (200 MHz, CDC13) : δ 2.13 - 2.80 (m, 4 H), 4.02 (dd, 1 H, J = 4.5, 9.0 Hz), 4.11 (dd, 1 H, J = 4.5, 9.0 Hz), 4.80 (m, 1 H), 6.75 = 7.02 (m, 4 H).
Part 4: (2S)-(4-Fluorophenoxymethyl)-5-hydroxytetrahydrofuran (Scheme I; 7) A flame dried 100 mL two neck round bottom flask equipped with a magnetic stir bar and nitrogen inlet was charged with a solution of 3.5 g (0.0167 mol) of (4S)-4- fluorophenoxy-methyl)-γ-butyrolactone 6 in 30 mL of CH2C12. That solution was cooled to -78°C and 7.34 mL (0.018 mol) diisobutylaluminum hydride (DIBAL-H; 2.5M solution in hexane) was added dropwise. The reaction mixture was stirred at -78°C for 3 hours. The reaction mixture was quenched with methanol (5 mL) and saturated aqueous solution of potassium sodium tartrate. The organic layer was separated, dried over Na2SO4 and concentrated on rotavapor to provide (2S)-(4-fluorophenoxymethyl)-5- hydroxytetrahydrofuran 7 as a solid (3.47 g). This crude lactol was used in the next reaction (Part 5) without further purification.
Part 5: (2S) (4-fluorophenoxymethyl)-5-(tert-butyldimethylsiloxy)- tetrahydrofuran) (Scheme II; 8)
A solution of 3.47 g of (2S)-(4-fluorophenoxymethyl)-5-hydroxytetrahydrofuran 7 in 30 mL of CH2C12 was taken in anlOO mL round bottom flask equipped with a magnetic stir bar and nitrogen inlet. That solution was cooled in an ice-water bath and 2.18 g (0.032 mol) of imidazole was added, followed by a solution of 3.6 g (0.024 mol) of tert-butyldimethylsilylchloride (TBDMSC1) in 30 mL of CH2C12. The reaction mixture then was stirred at room temperature for 3 hours, and the reaction then quenched with ice water, the organic layer separated, dried over Na2SO and concentrated under reduced pressure. The residue was purified by column chromatography using light petroleum ether: ethyl acetate (9:1) to yield (2S) (4-fluorophenoxymethyl)-5-(tert- butyldimethylsiloxy)-tetrahydrofuran) 8 as an oil (5.1 g, 95%). 1H NMR (200 MHz, CDC13) : δ 0.09 (s, 6 H), 0.88 (s, 9 H), 1.72 - 2.34 (m, 4 H), 3.76 = 4.08 (m, 2 H), 4.28 - 4.54 (m, 1 H), 5.47 (s, 1/3 H), 5.54 (d, J=4.5 Hz, 2/3 H), 6.75 - 7.0 (m, 4H).
Part 6: (2S) (5SR) (4-fluorophenoxymethyl)-5-(l-butynyl-4-tert- butyldimethylsiloxy)-tetrahydrofuran (Scheme II; 9)
To a flame dried 100 mL two neck round bottom flack equipped with a magnetic stir bar and nitrogen inlet and septum was added a solution of 5 g (0.0154 mol) of (2S) (4-fluorophenoxymethyl)-5-(tert-butyldimethylsiloxy)-tetrahydrofuran) 8 in 25 mL of CH2C12. That solution was cooled to -78°C and 2.82 mL (0.0184 mol) of trimethylsilylbromide (TMSBr) was added dropwise. The reaction mixture was then stirred at -78°C for 3 hours. In a separate flame dried 50 mL two neck round bottom flask equipped with a magnetic stir bar, nitrogen inlet and septum was added a solution of 3.4 g (0.0184 mol) of 4-tert-butyl-dimethylsiloxy-l-butyne in 30 mL of THF. That solution was cooled to - 78°C and 15.4 mL (1.5M solution in hexane; 0.023 mol) of n-BuLi was added dropwise. That reaction mixture was stirred at -78°C for 1 hour, and then transferred via syringe to the TMSBr solution. The combined solutions were stirred at -78°C for 2 hours, and then the reaction quenched with saturated ammonium chloride solution (20 mL) and the organic layer separated. The aqueous layer was extracted with CH2C1 and the combined organic layers were dried over Na2SO4 and then concentrated under reduced pressure to afford (2S) (5 SR) (4-fluorophenoxymethyl)-5 -( 1 -butynyl-4-tert-butyldimethylsiloxy)- tetrahydrofuran 9 as a thick syrup (6.0 g; 97%).
Part 7: (2S) (5RS)-2-(4-Fluorophenoxymethyl)-5-(4-hydroxybutyn-l-yl)- tetrahydrofuran (Scheme II; 10) Without further purification, (2S) (5SR) (4-fluorophenoxymethyl)-5-(l-butynyl-4- tert-butyldimethylsiloxy)-tetrahydrofuran 9 as prepared in Part 6 above was dissolved in 25 mL of methanol in a 50 mL single neck round bottom flask. That methanol solution was cooled in an ice-water bath and 3 mL of 1% HC1 solution in methanol was added. The reaction mixture was then stirred at room temperature for 3 hours, followed by neutralization with saturated aqueous sodium bicarbonate solution. After removal of methanol under reduced pressure, the resulting residue was dissolved in 100 mL of ethyl acetate. The organic layer was washed with water and brine, dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography using light petroleum etheπethyl acetate (1:1) to provide (2S) (5RS)-2- (4-fluorophenoxymethyl)-5-(4-hydroxybutyn-l-yl)-tetrahydrofuran 10 as a thick syrup (4.0 g, 96%). 1H NMR (200 MHz, CDC13) : δ 1.76 - 2.32 (m, 4 H), 2.46 (dt, 2 H, J - 2.2, 6.7 Hz), 3.69 (t, 2 H, J = 6.7 Hz), 3.89 (d, 2 H, J = 4.5 Hz), 4.41 (m, 1 H), 4.70 (m, 1 H), 6.73 - 6.98 (m, 4 H). Example 2: Alternate preparation of (2S) (5RS)-2-(4-fluorophenoxymethyl)-5-(4- hydroxybutyn-l-yl)-tetrahydrofuran (Scheme III; 10)
Part 1: (2S) (5RS)-5-0-acetyl-2-(4-fluoro-phenoxymethyl) tetrahydrofuran (Scheme III; U).
To a 25 ml round bottom flask with magnetic stir bar, (2S) (5RS)-2-(4- fluorophenoxymethyl)-5 -hydroxy tetrahydrofuran 7 (1.0 g, 0.0047 mol) in CH CI2 (5 mL) was added. The solution was cooled in an ice-bath, pyridine (0.8 mL), acetic anhydride (0.9 mL) and DMAP (catalytic amount) were added in succession. The reaction was monitored by TLC (EtOAc-light petroleum ether 1 :3, Rf=0.5). The reaction mixture was diluted with CH2C1 (10 mL) washed with 5% HCI, brine and dried over Na2SO4. The solvent was removed on rotavapor to give (2S) (5RS)-5-O-acetyl-2-(4- fluoro-phenoxymethyl) tetrahydrofuran U (1.05 g, 88%). 1H NMR (200 MHz, CDC13): δ 1.98, 2.05 (2s, 3 H), 1.89 - 2.3 (m, 4 H), 3.85 - 4.09 (m, 2 H), 4.36 - 4.61 (m, 1 H), 6.26 (s, Vi H), 6.33 (d, J = 4.5 Hz, lΛ H), 6.75 - 7.01 (m, 4 H).
Part 2: (2S) (5SR) (4-fluorophenoxymethyl)-5-(l-butynyl-4-tert- butyldimethylsiloxy)-tetrahydrofuran (Scheme III; 9).
To a flame dried 25 ml two-necked round bottom flask equipped with magnetic stir bar, nitrogen inlet and a septum, was added a solution of (2S) (5RS)-2-(4- fluoroρhenoxy-methyl)-5-O-acetyl tetrahydrofuran 11 (1.05 g, 0.004 mol) in CH2C12 (12 mL). The solution was cooled to 78°C and TMS-Br (0.65 ml, 0.0049 mol) was added dropwise. The reaction mixture was stirred at -78°C for 3hours (monitored by TLC, EtOAc-light petroleum 1 :4, Rf==0.4). In a separate flame dried 50 mL two-necked round bottom flask equipped with magnetic stir bar, nitrogen inlet and a septum, a solution of 4- tert-butyldimethylsiloxy-1-butyne (0.913 g, 0.0049 mol) in THF (15 mL) was taken. The solution was cooled to -78°C and n-BuLi in hexane (1.5M, 4.13 mL, 0.0062 mol) was added dropwise. The reaction mixture was stirred at -78°C for 1 hour. This solution was transferred via cannula to the reaction mixture of step 3 at -78°C. The reaction was monitored by TLC (EtOAc-light petroleum 1 :4, Rf=0.7) and completed in 2 hours. The reaction mixture was quenched with saturated ammonium chloride solution (10 mL). THF was removed under reduced pressure and extracted with CH2C12 (2 x 10 mL) dried over Na2SO4 and concentrated, to provide a crude yield of 1.7 g of (2S) (5SR) (4- fluorophenoxymethyl)-5-( 1 -butynyl-4-tert-butyldimethylsiloxy)-tetrahydrofuran 9. Part 3: (2S) (5RS)-2-(4-Fluorophenoxymethyl)-5-(4-hydroxybutyn-l-yl)- tetrahydrofuran (Scheme III; 10)
The crude product 9 (1.7 g) as prepared in Part 2 above was dissolved in methanol (10 mL), and 1% HCI solution in methanol (5 mL) was added. After 3 h the reaction mixture was neutralized with saturated aqueous sodium bicarbonate. After removal of methanol on rotavapor, the residue was dissolved in ethyl acetate (15 mL). The EtO Ac fraction was washed with water, brine, dried over Na2SO4 and concentrated on rotavapor. The residue afforded (2S) (5Rs)-2-(4-fluorophenoxymethyl)-5-(4-hydroxybutyn-l-yl)- tetrahydrofuran 10 as a thick syrup (0.957 g, 88%).
Example 3: Further alternate preparation of (2S) (5RS)-2-(4-fluorophenoxymethyl)-5- (4-hydroxybutyn-l-yl)-tetrahydrofuran (Scheme TV; 10)
Part 1 : (2S) (5RS)-5-bromo-2-(4-fluorophenoxymethyl)tetrahydrofuran (Scheme IV; 12) (2S) (5RS)-5-bromo-2-(4-fluorophenoxymethyl)tetrahydrofuran was prepared from (2S) (5RS)-5-O-acetyl-2-(4-fluorophenoxymethyl)tefrahydrofuran ll (1.06 g, 0.00417 mol) and TMS-Br (0.65 mL, 0.0049 mol).
Part 2: (2S) (5RS)-2-(4-fluorophenoxymethyl)-5-(4-tetrahydropyranoyloxybutyn- 1 -yl)-tetrahydrofuran (Scheme IV; 13)
In a flame dried 50 mL two-necked RB flask equipped with a magnetic stir bar, nitrogen inlet and a septum 4-tetrahydropyranoyl-l-butyne (0.774 g, 0.005 mol) in THF (10 mL) was taken and cooled to -78°C. A solution of n-BuLi in hexane (1.5 M, 4.2 mL, 0.0063 mol) was added dropwise, and the reaction mixture was stirred at -78°C for 1 hour. This solution was transferred via cannula to the reaction mixture of Part 1 of this example at -78°C. That reaction mixture was stirred at -78°C for 2h and monitored by TLC (EtOAc-light petroleum 1 :4, Rf=0.7). The reaction mixture was quenched with saturated ammonium chloride solution and THF was removed on rotavapor. The residue was partitioned between CH2C12 (20 mL) and water, and the organic layer was separated, washed with water, brine dried over Na SO4 and concentrated on rotavapor to provide a crude yield of 1.73 g.
Part 3: (2S) (5RS)-2-(4-fluoroρhenoxymethyl)-5-(4-hydroxybutyn-l-yl)- tetrahydrofuran (Scheme IV; 10) That crude product 13 ( 1.73 g) was dissolved in MeOH ( 10 mL) and 1 % HCI in methanol (5 mL) was added. After 2.5 h, the reaction mixture was quenched by saturated aqueous NaHCO3, and concentrated under reduced pressure. The residue was dissolved in EtOAc (20 mL), washed with water, brine, dried over Na2SO4 and concentrated to give (2S) (5RS)-2-(4-fluorophenoxymethyl)-5-(4-hydroxybutyn- 1 -yl)-tetrahydrofuran 10 (1.03 g, 93%). HPLC analysis: Column ODS; flowrate: 1.0 mL/min.; UV: 225nm. Mobile phase 60% methanol in water. Transxis ratio (65:35).
Example 4: Further alternate preparation of (2S)(5RS)-2-(4-fluorophenoxymethyl)-5- (4-hydroxybutyn-l-yl)-tetrahydrofuran (Scheme V: 10)
Part 1: (2S) (5RS)-5 -benzenesulfonyl-2-(4-fluorophenoxymethyl)tetrahydrofuran (Scheme V; 14)
To benzenesulfmic acid sodium salt (lO.Og, 0.061 mol), 25% HCI was added dropwise with stirring until the solid dissolved. The reaction mixture was extracted (100 mL each, 3 times) with EtO Ac, dried over Na2SO4 and concentrated to give benzenesulfonic acid (7.8 g, 90%). To a 100 mL round bottom with a magnetic stir bar, benzenesulfonic acid (4.61 g, 0.0324 mol), CaCl2(3.6 g, 0.0324 mol) and dry dichloromethane (30 mL) were added. The reaction mixture was cooled to 0°C and (2S) (5RS)-2-(4-fluorophenoxymethyl)-5-hydroxy-tetrahydrofuran (4.6g, 0.0216 mol) in dry CH2CI2 (20 mL) was added. The reaction mixture was stirred for 3 h and monitored by TLC (EtOAc-light petroleum ether 1 :4, Rf=0.25). The reaction mixture was filtered through celite and washed with CH2C1 (3 times). The combined organic layer was washed with saturated aqueous Na CO3, water brine and dried over Na2SO4. Solvent was removed under reduced pressure to afford the crude (2S) (5RS)-5-benzenesulfonyl-2-(4- fluorophenoxymethyl)tetrahydrofuran 14 which was crystallized from chloroform-hexane to give pure white solid, yield 6.8 g (93%), m.p. 102°C - 104°C. Η NMR (200 MHz, CDC13) : δ 1.90 - 3.0 (m, 4 H), 3.85 - 5.0 (m, 4 H), 6.70 - 7.05 (m, 4 H), 7.45 - 7.72 (m, 3 H), 7.77 - 8.0 (m, 2 H).
Part 2: (2S) (5RS)-2-(4-fluorophenoxymethyl)-5-(4-tetrahydropyranoyl-l- butyne)-tetrahydrofuran (Scheme V; 9)
To a 250 ml two-necked RB flask equipped with magnetic stir bar, nitrogen inlet and a septum, Grignard grade magnesium (2.0 g, 0.0833 mol) was taken and the flask flame dried along with magnesium. The flask was cooled to room temperature and dry THF (5 mL) was added followed by 1,2-dibromoethane (catalytic amount) to activate the magnesium. Isopropylbromide (8.78 g, 0.0714 mol) in THF (140 mL) as added dropwise over 15 min. The reaction mixture was stirred for 1 hour. The isopropyl magnesium bromide was cannulated in a 1000 mL flame dried two-necked round bottom flask with spin-bar, nitrogen inlet and septum. 4-Tetrahydropyranoyl-l-butyne (11.0 g, 0.0714 mol) in THF (140 mL) was added. The reaction mixture was stirred for 30 min. and cooled at 0°C. Freshly prepared ZnBr2 solution (1M, 43 mL, 0.0428 mol) in THF was introduced. After 45 min. at room temperature (2S) (5RS)-5-benzenesulfonyl-2-(4-fluorophenoxy- methyl)tetrahydrofuran (12.0 g, 0.0357 mol) in THF (70 mL) was added at room temperature and stirred for 3h. (TLC, EtOAc-light petroleum 1 :4, Rf=0.7). Saturated aqueous NH4C1 solution was added at 0°C to quench the reaction. THF was removed on rotavapor and the reaction mixture was partitioned between water and EtO Ac. The EtO Ac layer was washed with water, brine, dried over Na2SO4 and concentrated to provide (2S) (5RS)-2-(4-fluorophenoxymethyl)-5-(4-tetrahydropyranoyl- 1 -butyne)- tetrahydrofuran 9, crude yield 18.9 g. Part 3: (2S) (5RS)-2-(4-fluorophenoxy-methyl)-5-(4-hydroxybutyn-l- yl)tetrahydrofuran (Scheme V; 10)
That crude product 9 (18.9 g) was dissolved in methanol (60 mL) in 100 mL round bottom flask fitted with magnetic stirring arrangement. 1% HCI in methanol (25 mL) was introduced, and the reaction mixture was stirred at room temperature for 2 hours (TLC, EtOAc-light petroleum ether 1 :1, Rf=0.4). The reaction mixture was neutralized by saturated aqueous Na2CO3 solution and then concentrated under reduced pressure. The residue was extracted with ethyl acetate, washed with water, brine, dried over Na SO4 and concentrated on rotavapor. The residue was dried under vacuum on hot water bath to give (2S) (5RS)-2-(4-fluorophenoxy-methyl)-5-(4-hydroxybutyn-l- vDtetrahydrofuran 10, yield 10.9g. HPLC analysis: Column ODS; flowrate: 1.0 mL/min.; UV: 225nm. Mobile phase 60% methanol in water. Transxis ratio (69:31). That crude product of 10 was crystallized (2 times) from ether-light petroleum ether by seeding to yield the pure product (3.3 g, 35%), m.p. 76°C. [α]D -34.26° (c 1.36, CHC13). HPLC purity above 95%.
Example 5 : Further preparation of (4S)-4-fluorophenoxy-methyl)-γ-butyrolactone (Scheme VI; 6) Part 1: Tiimethylene D-mannitol (Scheme VI; 16)
D-mannitol (2.0 kg, 10.98 mol) (Scheme VII; 15) formaldehyde solution (35 % by weight, 4.4 lit, 51.2 rnmol) and cone. HCI (4.0 lit.) were taken in a 10 lit. flask with mechanical stirring arrangement. The reaction mixture was kept at room temperature for 72 hours. The solid was filtered, washed with water and dried to provide 2.2 kg (91.9%) of tiimethylene D-mannitol 16, m.p. 228°-230°, [α]D -108° (c 2.0, CHC13), TLC (silica gel), 1 :2, ethyl acetate: hexane, Rf = 0.4. 1H NMR (CDC13): δ 3.4 - 3.75 (m, 6H), 4.18 (dd, J-4.0, 8.0 Hz), 4.59 (d, 2H, J-4.0 Hz), 4.76 (s, 2H), 5.05 (d, 2H, J=4.0 Hz). Part 2: l,3,4,6-Tetra-O-acetyl-2,5-O-methylene-D-mannitol (Scheme VI; 17) Ice cold acetylating mixture (10.1 lit.) prepared from 7.0 liters of acetic anhydride, 3.0 liters of acetic acid and 0.1 liters of concentrated H2SO4 was taken in 20 lit. round bottom flask with mechanical stirring arrangement. Tiimethylene D-mannitol 16 (2.2 kg, 10.1 mol) was slowly added in portions (45 min. - 1 hour). After 3 h the reaction mixture was poured over ice-water with vigorous stirring (50-60 lit.). The solid was filtered, washed with water and dried to provide 2.8 kg (78%) of 17 , m.p. 126°-128°, [α]D + 57.8° (c 3.6, CHC13); TLC (silica gel), 2:1, ethyl acetate: hexane, Rf=0.5.
Part 3: 2,5-O-methylene-D-mannitol (Scheme VI; 18) l,3,4,6-Tetra-O-acetyl-2,5-O-methylene-D-mannitol 17 (2.8 kg, 7.73 mol) was added to chloroform (14 lit.) in 25 lit. round bottom flask with mechanical stirring. The reaction mixture was cooled to 0°C, and 0.5% NaOMe solution (6.5 lit.) was added slowly. The reaction mixture was stirred for 3 hours. The solid was filtered and dried to provide 1.0 kg (67%) of 2,5-O-methylene-D-mannitol 18, m.p. 172°-173°C, [α]D -52° (c 1.18, CHC13), TLC (silica gel), 1 :4, methanol: chloroform, Rf = 0.8. 1H NMR (D20): δ 3.42 (m, 2H), 3.72 (m, 4 H), 3.97 (m, 2 H), 4.91 (s, 2 H).
Part 4: l,6-Di-O-tosyl-2,5-O-methylene-D-mannitol (Scheme VI; 19) 2,5-O-methylene-D-mannitol 18 (200 g, 1.03 mol) was dissolved in pyridine (1.2 lit.) in 3 liter two neck R B flask fitted with an addition funnel and mechanical stirring arrangement. The reaction mixture was cooled to 0°C, tosyl chloride (430.9 g, 2.26 mol) dissolved in pyridine (0.8 lit.) was added slowly, and the reaction mixture was stirred at room temperature for 12 h. Pyridine then was removed on rotavapour under vacuo. The thick slurry was poured over ice-water (10 lit.) with mechanical stirring. After 2 hours the solid was filtered, washed with water, dried (yield, 400 g crude) and crystallized from methanol to provide 260 g of product 19, m.p. 142°C, [α]D - 23.39° (c 1.7, MeCOMe), TLC (silica gel), 4:1, ethyl acetate: hexane, Rf- 0.4. ]H NMR (CD3COCD3): δ 2.45 (s, 6H), 2.85 (s, 2 H), 3.27 (m, 2 H), 3.65 (m, 2 H), 4.12 (dd, 2 H, J- 6.2, 10.0 Hz), 4.45 (m, 2 H), 4.46 (s, 2 H), 7.38, 7.63 (Abq, 8 H, J=8.0 Hz).
Part 5: 3,4-O-Ethoxymetyhlene-2,5-O-methylene-l-6-di-O-tosyl-D-mannitol (Scheme VI, 20)
2,5-O-methylene-l,6-di-O-tosyl-D-mannitol 19, (185 g, 0.368 mol) triethylorthoformate (613 mL) and PTSA (100 mg) were stirred in a 1 lit. round bottom flask fitted with mechanical stirring arrangement at room temperature. After 3 hours of stirring potassium carbonate was added to neutralize PTSA. Solid was filtered and filtrate concentrated under reduced pressure and dried under vacuo to provide 206 g
(100%) of product 20, m.p. 87-88°C, [α]D + 46.02° (c 0.93, CHC13), TLC (silica gel) 7:3 hexane: EtOAc, Rf=0.4. 1H NMR (CDC13): δ 1.21 (t, 3 H, J=7.6 Hz), 2.45 (s, 6 H), 3.55 (q, 2 H, J= 7.6 Hz), 3.7 - 3.85 (m, 2 H), 3.97 (t, 1 H, J=8.5 Hz), 4.08 - 4.31 (m, 5 H). 4.74 (s, 2 H), 5. 76 (s, 1 H), 7.34, 7.77 (ABq, 8 H, J=8.5 Hz).
Part 6: 3,4-O-ethoxymethylene-l,6-di-O-p-fluorophenyl-2,5-O-methylene-D- mannitol (Scheme VI: 21)
4-Fluorophenol 2 (124 g, 1.107 mol) was dissolved in CH3CN (250 mL) and then KOH solution (62 g, in 45 mL, H20, 1.107 mol) was added. The reaction mixture was stirred for 15 minutes. 3,4-O-Ethoxymethylene-2,5-O-methylene-l-6-di-O-tosyl-D- mannitol 20 (206 g, 0.369 mol) (used as prepared in Part 5 above without further purification) in CH3CN (400 mL) was separately taken in 1 liter two neck round bottom flask fitted with reflux condenser, guard tube and mechanical stirring arrangement. To this solution the potassium salt of 4-fluorophenol was added at room temperature. The reaction mixture was heated under reflux for 6 hours and monitored by TLC (silica gel, 3:7, ethyl acetate: hexane, Rf=0.7). The reaction mixture was cooled in ice-water and solid was filtered washed with ethyl acetate (100 mL), and the combined filtrate was concentrated under reduced pressure. The resulting residue was dissolved in ethyl acetate (800 mL) and the organic layer was washed with 2M NaOH (4x100 mL), water and brine dried over Na2SO4. Concentration under reduced pressure afforded 3,4-0- ethoxymethylene-l,6-di-O-p-fluorophenyl-2,5-O-methylene-D-mannitol 21 (147 g, 90.9%). 1H NMR (CDC13): δ 1.3 (t, 3 H, J= 6.25 Hz), 3.70 (q, 2 H, J = 6.25 Hz), 4.0 - 4.45 (m, 7 H), 4.56 (t, 1 H, J=9.6 Hz), 5.19 (s, 2 H), 5.97 (s, 1 H), 6.89 - 7.10 (m, 8 H).
Part 7: l,6-Di-O-p-fluorophenyl-2,5-O-methylene-D-mannitol (Scheme VI: 22) 3,4-O-Ethoxymethylene-l,6-di-O-p-fluorophenyl-2,5-O-methylene-D-mannitol 21 (145 g 0.331 mol), tetrahydrofuran (350mL) and 0.1% aqueous HCI (40 mL) were mixed in a 1 lit two neck round bottom flask fitted with mechanical stirring arrangement at 0°C. The reaction mixture was allowed to attain room temperature and further stirred for 6 hours and monitored by TLC (silica gel, 1:1, ethyl acetate: hexane, Rf=0.3). The reaction mixture was basified to pH 8 by saturated NaHCO3 solution, and the solid was filtered and the filtrate concentrated to dryness to provide 125 kg (99%) of product 22, m.p. 126-127°C, [α]D -34.49° (c 1.148, MeCOMe). 1H NMR (CDC13): δ 2.7 (s, 2 H), 3.72 (m, 2 H), 3.90 (m, 2 H), 4.12 (m, 4 H), 4.87 (s, 2 H), 6.77 - 7.0 (m, 8 H).
Part 8: 4,4'-methylenedioxy-bis[(R)ethyl, (E)-2-ene-5-p-fluorophenoxy- pentanoate] (Scheme VI: 23)
In a 250 ml two neck round bottom flask equipped with magnetic stirring arrangement and fitted with a guard tube was taken a solution of 1 ,6-di-O-p- fluorophenyl-2,5-O-methylene-D-mannitol 22 (10.0 g, 0.026 mol) in CH2C12 (100 ml). The solution was cooled to 0°C and Pb(OAc)4 (12.8 g, 0.0288 mol) was added in portions. After 3 hours, ethylene glycol (1 ml) was added to quench excess Pb(OAc)4. The reaction mixture was filtered over celite, and the filtrate was washed successively with water and brine. The organic layer was dried over Na SO and concentrated under reduced pressure to afford the di-aldehyde as a thick syrup. That crude dialdehyde was taken in CH2C12 (100 ml) in 250 ml two necked round bottom flask with magnetic stirring arrangement and fitted with a nitrogen inlet. Carboethoxymethylenetriphenyl phosphorane (27.3 g, 0.0785 mol) was added in portions. The reaction mixture then was stirred for 3 hours, concentrated and purified on silica gel chromatography with 85:15 hexane: ethyl acetate as the eluent. The isolated fractions on concentration under reduced pressure yielded 4,4'-methylenedioxy-bis[ethyl, (E)-2-ene-5-p-fluorophenoxypentanoate] 23 (10.0 g, 74%) as an oil. 1H NMR (CDC13): δ 1.24 - 1.40 (m, 6 H), 3.86 - 4.30 (m, 8 H), 4.70 (m, 1 H), 4.84 (s, 2 H), 5.70 (brs, 1 H), 5.9 - 6.32 (m, 4 H), 6.76 - 7.02 (m, 8 H). Part 9: 4S-(4-Fluorophenoxymethyl)-γ-butyrolactone (Scheme VI: 6) A solution of 4,4' - methylenedioxy-bis[(R)ethyl, (E)-2-ene-5-p- fluorophenoxypentanoate] 23 (10.0 g, 0.0192 mol) in methanol (10 ml) was taken in a 200 ml parr hydrogenation flask. Pd C (500 mg) was added to that solution and the mixture shaken in a parr apparatus at 40-50 psi for 6 hour and monitored by TLC. The reaction mixture was filtered over celite and the filtrate concentrated to afford 4,4'- methylenedioxy-bis[(R) ethyl, 5-p-fluorophenoxypentanoate] as an oil (10.0 g, 100%).
A 250 ml round bottom flask equipped with magnetic stirring arrangement and fitted with a reflux condenser was then charged with 4,4' - methylenedioxy-bis [® ethyl, 5-p- fluorophenoxypentanoate] (10.0 g, 0.019 mol) in ethanol (60 ml). To that solution 10% aqueous solution H2SO4 (15 ml) was added. The mixture was heated under reflux for 10-12 hours and monitored by TLC, silica gel, 1 :1, ethyl acetate: hexane, Rf=0.25. The reaction was cooled to 0°C and neutralized with saturated sodium bicarbonate solution. The reaction mixture was concentrated on a rotavapour to dryness and redissolved in ethyl acetate (100 ml). The organic layer was washed with water and brine dried over Na2SO4 and concentrated. The residue was purified by column chromatography to afford off white crystalline solid of 4S-(4-fluorophenoxymethyl)-γ- butyrolactone 6 (7.0 g, 87%), .m.p 60-61°C, [ ]D +25° (c 2.18, CHC13). 1H NMR (CDC13): δ 2.13 - 2.80 (m, 4 H), 4.02 (dd, 1 H, J=4.5, 9.0 Hz), 4.11 (dd, 1 H, J=4.5, 9.0 Hz), 4.80 (m, 1 H), 6.75 - 7.02 (m, 4 H). Example 6: Further alternate preparation of (2S) (5RS)-2-(4-Fluorophenoxymethyl)- 5-(4-hydroxybutyn-l-yl)-tetrahydrofuran (Scheme VII; 10)
Part 1: (+)-l,2-Epoxy-(4-fluoro)phenoxy propane (Scheme VII; 25) p-Fluorophenol 2 (5 g, 44.6 mmol) and epichlorohydrin 24 (16.5 g, 178.4 mmol
13) were admixed in anhydrous acetone (100 ml). Anhydrous K CO3 (24.0 g, 178.4 mmol) was added in 10 minutes and the reaction mixture was heated at reflux for 18 hours until the complete consumption of p-fluorophenol as monitored by TLC (4:1 hexane: ether). The reaction mixture then was filtered off, the filtrate was concentrated under vacuo to afford a light yellow oil, excess epichlorohydrin was distilled off, the residue was subjected to column chromatography on silica gel (2:8, ethyl acetate-hexane) to afford (+)-l,2-epoxy-(4-fluoro)phenoxy propane 25 in quantitative yield (8.5 g).
Part 2: (2R)-3-(4-fluoro)phenoxy-propane-l,2-diol (Scheme VII; 26) (2R)-3 -(4-fluoro)phenoxy-propane- 1 ,2-diol 26 was prepared using Jacobsen' s catalyst as generally described in E. Jacobsen, Science, 277:936-938 (1997). More specifically (+)-l,2-epoxy-3-(4-fluoro)phenoxy propane 25 (10 g, 59.5 mmol) and (R,R)- Jacobsen's catalyst (215 mg, 0.29 mmol) were taken in a 50 ml round bottom flask and cooled to 0°C. Water (0.6 ml, 32.7 mmol) was then added dropwise for 1 hour and stirred for 5 hours at room temperature, monitored by TLC (1:1 ethyl acetate:hexane).
Ethyl acetate (50 ml) was added, followed by anhydrous Na2SO4 (200 mg), stirred for 10 minutes filtered, concentrated to afford dark colored residue of a mixture of 26 and 27, which on column chromatography gave isolated epoxide 27 (4.36 g, 43%, 1 :9 ethyl acetate-hexane) and (2R)-3-(4-fluoro)phenoxy-propane-l,2-diol 26 (5.06 g, 46%, 1:1 ethyl acetate-hexane).
Part 3: (2S)-3-(4-fluoro)phenoxy-l-tosyloxy-propan-2-ol (Scheme VII; 28) A mixture of (2R)-3-(4-fluoro)phenoxy-propan-l,2-diol 26 (5.0 g, 26.8 mmol) and pyridine (4.5 ml) in CH2C1 (60 ml) were cooled to 0°C, and then p-toluenesulphonyl chloride (5.0 g, 26.8 mmol) was added portionwise to the cooled mixture. The mixture was stirred at room temperature overnight (TLC 2:3, ethyl acetate-hexane). The solvent was then removed by codistillation with toluene, and the resulting residue purified by silica gel column chromatography (2:3, ethyl acetate-hexane) to afford the product 28 (7.7 g, 85%).
Part 4: (2R)-l,2-epoxy-3-(4-fluoro)phenoxypropane (Scheme VII; 4) (2R)-(4-Fluoro)phenoxy-l-tosyloxy-propan-2-ol 28 (5.0 g, 14.7 mmol) in a solvent mixture of THF and DMF (100 ml, 4:1) was cooled to 0°C and NaH (0.75 g, 19.2 mmol) was added portionwise, followed by stirring ofthe reaction mixture for 1 hour at room temperature with monitoring ofthe reaction by TLC (20% ethyl acetate in hexane). The THF was removed and the residue was taken in ethyl ether (50 ml). That ether solution was washed successively with water (3 x 50 ml), brine (1 x 50 ml) dried (Na2SO4) and concentrated to afford (2R)-l,2-epoxy-3-(4-fluoro)phenoxypropane 4 as a colorless oil (2.53 g, 95%).
Part 5: (2R)-l-(4-fluoro)phenoxyhex-5-en-2-ol (Scheme VII; 29) Magnesium (0.89 g, 36.6 mmol) and iodine (catalytic amount) were taken in a 50 ml 2-neck round bottom flask provided with a reflux condenser and a septum, under N2 atmosphere. A solution of allyl bromide (3.0 g, 24.4 mmol) in 10 ml of ethyl ether was slowly added and stirred for 30 minutes at room temperature. Cuprous cyanide (22 mg) then was added, and the color ofthe reaction mixture became dark brown. The reaction mixture was cooled to -22°C (CCl4/dry ice bath), and (2R)-l,2-epoxy-3-(4- fluoro)phenoxypropane 4 (2.05 g, 12.2 mmol) in 25 ml of ethyl ether was added. The reaction was completed within 30 minutes, as determined by TLC (benzene). Saturated aqueous ammonium chloride (4 ml) then was added and the mixture stirred for 30 minutes. Inorganic material was filtered and washed with ethyl ether (25 ml). The ether layer was dried (sodium sulphate) concentrated to give a colorless oil of (2R)-l-(4- fluoro)phenoxyhex-5-en-2-ol 29 (2.3 g, 90%). Part 6: (2R)-2-benzenesulfonyloxy-l-(4-fluoro)-phenoxy-5-hexane (Scheme VII; 30)
(2R)-(4-Fluoro)phenoxyhex-5-en-2-ol, 29 (7.4 g, 35.2 mmol), triethylamine (10 ml) and 4-N,N'-dimethylammopyridine (DMAP, 0.43 g, catalytic) were dissolved in 50 ml of dry CH2C12 and cooled in ice bath while stirring. Benzenesulfonyl chloride (5 ml, 38.7 mmol) in CH2C12 (10 ml) was then added dropwise to the mixture. The reaction mixture was stirred at room temperature for 6 hours and monitored by TLC (benzene)]. Solvent then was removed and the residue was poured onto a short silica gel column and eluted with 1 :4 ethyl acetate-hexane to afford (2R)-2-benzenesulfonyloxy-l-(4-fluoro)- phenoxy-5-hexane 30 as a colorless oil (11.3 g, 92%).
Part 7: (6R,2E)-ethyl-6-benzenesulfonyloxy-7-(4-fluoro)-phenoxy-hept-2-en- 1 - oate (Scheme VII; 3l)
(2R)-2-Benzenesulfonyloxy-l-(4-fluoro)-phenoxy-5-hexane 30 (11.3 g, 32.5 mmol 19) in 30 ml of dry CH2C12 was cooled to -78°C. O3 then was bubbled through the mixture until the blue color persisted (30 minutes). A stream of N2 then was purged for 5 minutes through the mixture to remove excess of ozone. Dimethylsulfide (13.9 ml, 325 mmol) was added, and the mixture stirred for 2 hours. The reaction mixture was washed with water (2 x 25 ml), brine (1 x 30 ml) and concentrated to afford the crude product (10.8 g, 95%). (2R)-Benzenesulfonyloxy-l-(4-fluoro)-phenoxy-5-pentanal (10.5 g, 30 mmol) was added and heated at reflux for 5 hours. Ethoxycarbonylmethylene triphenylphosphorane (11.5 g, 33 mmol) was added and heated at reflux for 5 hours. Completion ofthe reaction was checked by TLC (1:10, EtO Ac-benzene) and the solvent was removed, the residue was purified by column chromatography on silica gel (1 :3, ethyl acetate-hexane) to afford (6R,2E)-ethyl-6-benzenesulfonyloxy-7-(4-fluoro)- phenoxy-hept-2-en-l-oate 31 (8.8 g, 70%) as a colorless oil. Part 8: (6R,2E)-ethyl-6-benzenesulfonyloxy-7-(4-fluoro)-phenoxy-hept-2-en- 1 -ol (Scheme VII; 32)
(6R,2E)-Ethyl-6-benzenesulfonyloxy-7-(4-fluoro)-phenoxy-hept-2-en- 1 -oate (3 g, 7.1 mmol) 31 was dissolved in 30 ml of CH2C12 under N2 atmosphere and cooled to - 78°C. DIBAL-H (14.2 ml, 14.2 mmol, IM solution in toluene) was added dropwise over 5 minutes and the solution was stirred at -78°C for 45 minutes. At reaction completion as monitored by TLC (2:5, ethyl acetate-hexane), saturated aqueous ammonium chloride solution (3 ml) was added and the mixture stirred for another 30 minutes. The reaction mixture then was filtered through a celite pad the filtrate was dried over anhydrous Na2SO4 and concentrated, the residue was filtered through a short silica gel pad and concentrated to obtain (6R,2E)-ethyl-6-benzenesulfonyloxy-7-(4-fluoro)-phenoxy-hept-2- en-l-ol 32 as a solid (2.2 g, 82% yield).
Part 9: (2S,3S,6R)-6-benzenesulfonyloxy-2,3-epoxy-7-(4-fluoro)-phenoxy-7- heptan- 1 -ol (Scheme VII; 33)
Powdered molecular sieves 4A (3 g) were activated under N atmosphere in a 25 ml 2 necked round bottom flask. CH C12 (15 ml) was added followed by titanium tetraisopropoxide (1.62 ml, 5.47 mmol), (+)-diisopropyltartrate (1.07 ml, 6.56 mmol) and the mixture was cooled to -20°C with stirring. After 5 minutes cumene-hydroperoxide (2.1 ml, 10.94 mmol, 80% solution in cumene) was added dropwise. The mixture was stirred for 15 minutes at -20°C (6R,2E)-benzenesulfonyloxy-7-(4-fluoro)-phenoxy-hept- 2-en-l-ol 32 (2.0 g, 5.47 mmol) in 10 ml of CH2C12 was then added and the reaction mixture was stirred for 2.5 hours at -20°C. The reaction mixture was checked for the completion by TLC (1:1, ethyl acetate-hexane), 1 ml of 10% aqueous tartaric acid solution was added at -20°C and the reaction mixture was warmed to room temperature in 30 minutes. The reaction mixture was filtered through a celite pad dried over Na2SO4, concentrated and the residue was subjected to column chromatography on silica gel (1:1, ethyl acetate-hexane) to afford (2S,3S,6R)-6-benzenesulfonyloxy-2,3-epoxy-7-(4-fluoro)- phenoxy-7-heptan-l-ol 33 (2.4 g, 98% yield) as a solid. Part 10: (2S,3S,6R)-6-benzenesulfonyloxy-l-chloro-2,3-epoxy-7-(4-fluoro)- phenoxy-heptane (Scheme VII; 34)
(2S,3S,6R)-6-Benzenesulfonyloxy-2,3-epoxy-7-(4-fluoro)-phenoxy-7-heptan-l-ol (2.25 g, 5.7 mmol) 33 and triphenylphosphine (1.5 g, 5.7 mmol) were dissolved in solvent mixture of CHC13 and CC14 (40 ml, 1 : 1) and NaHCO3 (0.3 g) was added. The reaction mixture was refluxed for 3 hours and monitored by TLC (2:5, ethyl acetate- hexane). Solvent was removed, the residue was purified by column chromatography on silica gel (1:4, ethyl acetate-hexane) to afford (2S,3S,6R)-6-benzenesulfonyloxy-l- chloro-2,3-epoxy-7-(4-fluoro)-phenoxy-heptane 34 (1.5 g, 64% yield) as a solid.
Part 11 : (2S,5S)-5-ethynyl-2-(4-fluoro)-phenoxymethyl-tetrahydrofuran (Scheme VII; 35) n-BuLi (7.2 ml, 7.2 mmol) was added to a solution of freshly distilled diisopropylamine (1.12 ml, 8.6 mmol) in 6 ml of dry THF at -40°C and stirred for 15 minutes. A solution of (2S,3S,6R)-6-benzenesulfonyloxy-l-chloro-2,3-epoxy-7-(4- fluoro)-phenoxy-heptane 34 (1.0 g, 2.42 mmol) was added in 8 ml of dry THF. The reaction mixture was stirred at -40°C for 1 hour and then at room temperature for 1 hour. When TLC showed complete consumption of starting material the reaction was quenched at 40°C with aqueous ammonium chloride (1 ml), THF was removed under vacuo, the residue was taken in ethyl acetate, filtered, dried over Na2SO and concentrated. Crude product was subjected to column chromatography on silica gel (1 :9, ethyl acetate-hexane) to afford (2S,5S)-5-ethynyl-2-(4-fluoro)-phenoxymethyl-tetrahydrofuran 35 (0.32 g, 60% yield).
Part 12: Preparation of (2S,5S)-5-(2'-hydroxyethyl)-ethynyl-2-(4-fluoro)- phenoxymethyltetrahydrofuran (Scheme VII; 10)
To a solution of (2S,5S)-5-ethynyl-2-(4-fluoro)-phenoxymethyl-tetrahydrofuran 35 (0.8 g, 3.6 mmol) in 15 ml of dry THF at -78°C, n-BuLi (5 ml, IM solution in hexane), stirred for 15 minutes. Freshly distilled BF3Et O (1.4 ml, 11 mmol) was added followed by ethyleneoxide (excess, THF solution). The reaction mixture was allowed to stir at -78°C until completion (30 minutes). Saturated aqueous ammonium chloride solution (1 ml) was added at -78°C stirred for 5 minutes, warmed to room temperature, THF was removed, residue was extracted with ether (2 x 20 ml), combined organic layer was dried over Na2SO4, concentrated to afford a residue. That residue was purified by column chromatography on silica gel (2:5, ethyl acetate-hexane) to afford (2S,5S)-5-(2'- hydroxyethyl)-ethynyl-2-(4-fluoro)-phenoxymethyltetrahydrofuran 10 (0.87 g, 90% yield) as a white solid. That product K) was found to be identical (NMR, optical rotation, TLC) with samples prepared by Example 1 above.
Example 7: Further alternate preparation of (2S) (5RS)-2-(4-
Fluorophenoxymethyl)-5-(4-hydroxybutyn- 1 -yl)-tetrahydrofuran (Scheme VIII; 10)
References in this Example 7 to compound numerals (generally underlined) designate the compounds depicted structurally in Scheme VIII above.
Part i: 1,2:5,6- Di-O-isoproρylidene-(D)-mannitol (Scheme VIII; 2): A solution of D-mannitol 1 (100 g, 0.549 mol) in a mixture of DMSO (160 mL, distilled and stored over molecular sieves) and dimethoxy propane (149 mL, 1.209 mol), at room temperature, was treated with p-toluene sulfonic acid (PTSA; 0.5 g) and stirred for 18h. The reaction mixture was treated with Et3N (3 mL), to quench PTSA, and water (500 mL) and extracted with hexanes (3 x 200 mL) to remove the tri acetonide (5-7 g). The reaction mixture was then extracted with EtO Ac (4 x 500 mL) and washed with NaCl solution. Evaporation ofthe solvent gave pure 1,2:5,6- di-O-isopropylidene-(D)- mannitol 2 (95 g) in 66% yield as a solid, m.p. 118-120 ° C (lit. m.p. 120-122 ° C (ref. Organic Synthesis 1995, 72, 6)). Part 2: Ethyl (2E,4S)-4,5-isopropylidenedioxy-2-pentenoate (Scheme VIII; 4): A solution of 1,2:5,6- di-O-isopropylidene-(D)-mannitol 2 (100 g, 0.38 mol) in
CH2C12 (1000 mL) containing saturated NaHCO3 solution (40 mL) was cooled to 0°C, treated with NaIO4 (123 g, 0.57 mol) and allowed to stir at 0°C to 20° C. After 2 to 3 h (TLC analysis), solid Na2SO4 (35 g) was added and the reaction mixture was stirred further for 15 min. The reaction mixture was filtered and concentrated (below 25°C bath temperature) to half the volume.
The above solution of (R)-glyceraldehyde 3 in CH C12 (500 mL) was cooled to below 10°C and treated with (carbethoxymethylene) triphenyl phosphorane (132 g, 0.38 mol) in portions. After stirring at room temperature for 3-4 h, the solvent was evaporated, the residue treated with hexane (500 mL) and the solution decanted through silica gel. The residue was further treated with 10% EtO Ac in hexane (4 x 500 mL) and decanted through silica gel. Evaporation of solvent afforded ethyl (2E,4S)-4,5- isopropylidenedioxy-2-pentenoate 4 (105 g) in 73 % yield as a colorless liquid. 'HNMR (CDC13, 200 MHz): δ 1.2 (t, 3H, J 7.1 Hz, CH3), 1.3, 1.35 (2s, 6H, CH3), 3.48 (dd, 1H, J 6.1 Hz, H-5), 4.05 (q, 2H, J 7.1 Hz, OCH2), 4.25 (dd, 1H, J 8.0 Hz, H-5a), 5.3-5.40 (m, 1H, H-4), 5.72 (dd, 1H, J 2.3, 11.0 Hz, H-2), 6.72 (dd, 1H, J 6.1, 12.2 Hz, H-3).
Part 3: Ethyl (4S)-4,5-isopropylidenedioxy-l-pentanoate (Scheme VIII; 5):
A solution of ethyl (2E,4S)-4,5-isopropylidenedioxy-2-pentenoate 4 (90 g, 0.45 mol) in EtO Ac (100 mL) was treated with PtO2 (0.225 g) and subjected to hydrogenation till there was no further consumption of hydrogen (3-4 h). At the end of reaction, the reaction mixture was filtered and the solvent evaporated to afford ethyl (4S)-4,5- isopropylidenedioxy-1-pentanoate 5 (90 g) in 99% yield as a colorless liquid. [α]π +4.99 (c 2.2, CHC13); HNMR (CDC13, 200 MHz): δ 1.28 (t, 3H, J 7.1 Hz, CH3), 1.30, 1.45 (2s, 6H), 1.8-1.95 (m, 2H, H-3), 2.38-2.5 (m, 2H, H-2), 3.52 (dd, 1H, J 7.1 Hz, H-5), 4.0-4.2 (m, 4H, H-4,5a,-OCH2). Part 4: (2S)-l,2-Isopropylidenedioxy-5-pentanol (Scheme VIII; 6): A suspension of LAH (16.9 g, 0.44 mol) in THF (300 mL) was cooled to 0°C and treated dropwise with a solution of ethyl (4S)-4,5-isopropylidenedioxy-l-pentanoate 5 (90 g, 0.44 mol) in THF (200 mL). The reaction mixture was then allowed to stir at room temperature for 10-12 h and treated with a saturated solution of Na2SO (38 mL) at 0°C for an additional 30 min. period, it was filtered through celite and washed with EtOAc (3 x 200 mL). The combined organic layers were washed with saturated NaCl solution and evaporated in vacuo to provide (2S)-l,2-isopropylidenedioxy-5-pentanol 6 (65.5 g) in 92% yield as a colorless liquid. [α]D +14.18 (c 2.2 CHC13). 1HNMR (CDC13, 200 MHz): δ 1.3, 1.4 (2s, 6H), 1.6-1.8 (m, 5H, H-3,4,OH), 3.5 (t,lH, J 8.3 Hz ,H-1), 3.6-3.7 (m, 2H, H-5), 4.0-4.18 (m, 2H, H-la,2); 13CNMR (CDC13, 50 MHz): δ 25.5, 26.7, 28.9, 30.0, 62.2, 69.3, 76.3, 108.8; MS: 145 (M+-15); HRMS: Calculated for C73O3 (M+-15): 145.086469: Observed: 145.086081.
Part 5: (4S)-4,5-Isopropylidenedioxy-l-pentanal (Scheme VIII; 7):
A stirred solution of (2S)-l,2-isopropylidenedioxy-5-pentanol 6 (20 g, 0.125 mol) in DMSO (60 mL) was cooled to 0°C, treated with IBX (38.5 g, 0.137 mol) in portions while maintaining the temperature below 0°C and allowed to stir at room temperature for 4h. The reaction mixture was treated with saturated NaHCO3 solution (25 mL), filtered through celite and washed with EtO Ac (3 x 150 mL). The layers were separated and the organic layer was washed with water, brine and dried (Na2SO4). Evaporation of solvent furnished (4S)-4,5-isopropylidenedioxy-l-pentanal 7 (16.1 g) in 81% yield as a dark yellow liquid. [α]D +3.8 (c 1.0, CHC13); ]HNMR (CDC13, 200 MHz): δ 1.3, 1.35 (2s, 6H, CH3), 1.6-2.1 (m, 4H, H-2,3), 2.6 (t, 2H, J2;36.2 Hz, H-2), 3.5 (t, 2H, J4>5 6.2 Hz, H-5), 3.98-4.2 (m, 2H, H-4,5a), 9.8 (s, 1H, CHO).
Part 6: Ethyl (2E,6S)-6,7-isopropylidenedioxy hept-2-enoate (Scheme VIII; 8): A solution of (4S)-4,5-isopropylidenedioxy-l-pentanal 7 (24g, 0.15 mol) in benzene (250 mL) was treated with (carbethoxymethylene) triphenyl phosphorane (57.8 g, 0.166 mmol) and heated at reflux for 6h. Solvent was evaporated, the residue was treated with hexane (500 mL) and decanted through silica gel. Further, the residue was treated with 10% EtO Ac in hexane (3 x 500 mL) and the solution filtered through silica gel. Evaporation ofthe combined solvents afforded ethyl (2E,6S)-6,7- isopropylidenedioxy hept-2-enoate 8 (26 g) in 76% yield as a pale yellow liquid. [α]o +6.9 (c 1.0, CHC13); 1HNMR (CDC13, 200 MHz): δ 1.28 (t, 3H, J 6.75 Hz, CH3), 1.32, 1.38 (2s, 6H), 1.6-1.8 (m, 2H, H-5), 2.2-2.45 (m, 2H, H-4), 3.5 (t, 1H, J 6.75 Hz, H-7), 3.95 -4.25 (m, 4H, H-6,7a,OCH2), 5.8 (d, 1H, J 15.75 Hz, H-2), 6.82-7.02 (dt, 1H, J 7.8, 15.75 Hz, H-3); MS: 213 (M+-15).
Part 7: (2E,6S)-6,7-Isopropylidenedioxy hept-2-ene-l-ol (Scheme VIII; 9): A stirred solution of ethyl (2E,6S)-6,7-isopropylidenedioxy hept-2-enoate 8 (15 g, 65.7 mmol) in dry CH2C12 (80 mL) was cooled to -20°C (CC14 + dry ice bath) and treated drop wise with a solution of DIBAL-H (66 mL, 134 mmol; 2N solution in hexane). After stirring for 2h, the reaction mixture was warmed to 0°C, treated with a saturated solution of Na-K tartarate (20 mL) drop wise and stirred for an additional 45 min. The organic layer was separated and the aqueous layer extracted with CH2C12 (100 mL). The combined organic layers were washed with brine, dried (Na2SO ) and evaporated in vacuo to provide (2E,6S)-6,7-isopropylidenedioxy hept-2-ene-l-ol 9 (10.8 g) in 88% yield a colorless liquid. [α]D +12.83 (c 2.17, CHC13); 'HNMR (CDC13, 200 MHz): δ 1.35, 1.40 (2s, 6H), 1.4-1.8 (m, 2H, H-5), 2.05-2.3 ( m, 2H, H-4), 3.5 (t, 1H, J 7.95 Hz, H-7), 3.95-4.2 (m, 4H, H-l,6,7a), 5.65-5.72 (m, 2H, H-2,3); 13CNMR (CDC13, 50 MHz): δ 25.60, 26.81, 28.26, 32.96, 63.20, 69.19, 75.38, 108.63, 129.68, 131.47; MS: 175 (M+ - 15); HRMS: Calculated for C9H15O3 (M+-15): 171.102120; Observed: 171.102318.
Part 8: (2S,3S,6S)-2,3-Epoxy-6,7-isopropylidenedioxy heptan-1-ol (Scheme VIII; 10):
Method A (Stoichiometric) - To a stirred and cooled (-20°C) suspension of molecular sieves (4A, 4g), in CH2C1 (15 mL) under N2 atmosphere, (+) Diisopropyl L- tartarate (18.1 g, 77.4 mmol) in CH2C12 (20 mL) , Titanium(IV) isopropoxide (19.1 mL, 64.54 mmol) and cumene hydroperoxide (19.6 mL, 103mmol; 80% solution in cumene) were added sequentially. After 20 min. the resulting mixture was treated with a solution of (2E,6S)-6,7-isopropylidenedioxy hept-2-ene-l-ol 9 (12 g, 64.5 mmol) in CH2C12 (30 mL) drop wise and stirred further for 3h at the same temperature. The reaction mixture was quenched with 10% NaOH solution saturated with NaCl (20 mL) and filtered through celite. Evaporation of solvent and purification of residue by column chromatography (Si-gel, 1:1.5 EtOAc-hexane) furnished (2S,3S,6S)-2,3-epoxy-6,7- isopropylidenedioxy heptan-1-ol 10 (7.5 g) in 57% yield as a colorless liquid. [α]o - 15.06(c 3.0, CHC13); 1HNMR (CDC13, 200 MHz): δ 1.35, 1.4 (2s, 6H, CH3), 1.45-1.85 (m, 5H, H-4,5,-OH), 2.85-3.05 (m, 2H, H-2,3), 3.5 (t, 1H, J 6.5 Hz, H-7), 3.65 (dd, 1H, J 4.08, 12.2 Hz, H-l), 3.9 (dd, 1H, J 3.06, 12.2 Hz, H-la), 3.98-4.2 (m,2H, H-6,7a); 13CNMR (CDC13, 50 MHz): δ 25.33, 26.31, 27.33, 29.87, 55.34, 58.41, 61.83, 69.12, 75.13, 100.84. Method B (Catalytic - 10 mol%) - A stirred mixture of (+) diisopropyl L-tartarate
(1.5 g, 6.45 mmol) and molecular sieves (4A, 4 g) in CH2C1 (30 mL) was cooled to - 20°C under N2 atmosphere and treated with titanium(IV) isopropoxide (1.59 mL, 5.37 mmol). After 20 min. cumene hydroperoxide (16.3 mL, 86.0 mmol) was added followed by addition of a solution of (2E,6S)-6,7-isopropylidenedioxy hept-2-ene-l-ol 9 (10.0 g, 53.7 mmol) in CH2C12 (25 mL) drop wise. The reaction was conducted out for 12 h at - 20°C and worked up as already described above to give (2S,3S,6S)-2,3-epoxy-6,7- isopropylidenedioxy heptan-1-ol 10 (6.2 g) in 58% yield as a liquid, whose spectral data was comparable with that ofthe above compound.
Part 8: (2S,3S,6S)-l-Chloro-2,3-epoxy-6,7-isopropylidenedioxy heptane
(Scheme VIII; 11):
A stirred mixture of (2S,3S,6S)-2,3-epoxy-6,7-isopropylidenedioxy heptan-1-ol 10 (6.0 g, 29.7 mmol), Ph3P (9.69 g, 35.6mmol) and NaHCO3 (0.8 g) in CC14 (30 mL) was heated at reflux for 3 h. The solvent was evaporated and the residue obtained on chromatographic purification (Si- gel, 15% EtOAc-hexane) gave (2S,3S,6S)-l-chloro- 2,3-epoxy-6,7-isopropylidenedioxy heptane 11 (6.2 g) in 95% yield as a colorless liquid. [α]D -33.19(c 3.0, CHC13); 'HNMR (CDC13, 200 MHz): δ 1.35, 1.4 (2s, 6H, CH3), 1.6- 1.82 (m, 4H, H-4,5), 2.85-3.05 (m, 2H, H-2,3), 3.35-3.7 (m, 3H, H-1,7), 3.2-3.98 (m, 2H, H-6,7a); 13CNMR (CDC13, 50 MHz): δ 25.5, 26.6, 26.8, 27.5, 29.5, 44.4, 57.1, 58.3, 69.1, 75.0, 108.8.
Part 9: (3S,6S)-3-Hydroxy-6,7-isopropylidenedioxy-hept-l-yne (Scheme VIII;
12): To freshly prepared LDA [prepared from diisopropylamine (14.9 mL, 114.5 mmol) and n-BuLi (70 mL, 109 mmol; 1.5 N hexane solution)] in THF (100 mL), (2S,3S,6S)-l-chloro-2,3-epoxy-6,7-isopropylidenedioxy heptane H (6.0 g, 27.2 mmol) in THF (20 mL) was added at -40°C (CH3CN + dry ice bath). After 3h, the reaction was quenched with aq. NH4C1 solution. The aqueous layer separated and extracted with EtO Ac. Combined organic layers were dried (Na2SO4), evaporated and the residue purified by column chromatography (Si-gel, 30% EtOAc-hexane) to furnish (3S,6S)-3- hydroxy-6,7-isopropylidenedioxy-hept-l-yne 12 ( 3.9 g) in 94% yield as a yellow liquid. [α]D +2.59 (c 1.54, CHC13); ]HNMR (CDCI3, 200 MHz): δ 1.35, 1.4 (2s, 6H) 1.6-198 (m, 4H, H-4,5), 2.4 (d, IH, J 1.86 Hz, H-l), 3.5 (t, IH, J 6.9 Hz, H-7), 3.98-4.2 (m, 2H, H-6,7a), 4.4 (m, IH, H-3); 13CNMR (CDC13, 50 MHz): δ 25.62, 26.84, 29.09, 33.85, 61.83, 69.24, 72.92, 75.55, 84.66, 109.01; MS: 169 (M+-15); HRMS: Calculated for C9H13O3 (M+-15): 169.086469: Observed: 169.086063.
Part 10: (3S,6S)-3-Acetoxy-6,7-isopropylidenedioxy-hept-l-yne (Scheme I; 13): A solution of (3S,6S)-3-hydroxy-6,7-isopropylidenedioxy-hept-l-yne 12 (3.9 g,
25.65 mmol) and Et3N (10 mL, 76.9 mmol) in CH2C12 (40 mL) containing DMAP (catalytic) at 0°C was treated with Ac2O (3.65 mL, 38.48 mmol) and stirred at room temperature for 30 min. The reaction mixture was treated with saturated aqueous NaHCO3 solution (20 mL) for 30 min. and the organic layer was separated. It was washed with water (3 x 20 mL), brine (25 mL) and dried (Na SO4). Evaporation of solvent and purification of residue by column chromatography (Si-gel, 20% EtOAc- hexane) gave (3S,6S)-3-acetoxy-6,7-isopropylidenedioxy-hept-l-yne 13 (4.15 g) in 83% yield as a pale yellow liquid. [α]D -2.55 (c 1.96, CHC13); 'HNMR (CDC13, 200 MHz): δ 1.25,1.35 (2s, 6H), 1.5-1.9 ( m, 4H, H-4,5), 2.02 (s, 3H, -OCH3), 2.35 (d, IH, J 2.3 Hz, H-l), 3.45 (t, IH, J 6.5 Hz, H-7), 3.9-4.1 (m, 2H, H-6,7a), 5.25-5.4 (m, IH, H-3); 13CNMR (CDC13, 50 MHz): δ 20.54, 25.58, 26.89, 29.71, 30.88, 63.88, 69.12, 73.81, 75.99, 80.80, 108.93, 169.87.
Part 11: (3S,6S)-3-Acetoxy-6,7-dihydroxy-hept-l-yne (Scheme VIII; ]4):
A mixture of (3S,6S)-3-acetoxy-6,7-isopropylidenedioxy-hept-l-yne 13 (4.1 g) in 60% aqueous . AcOH (20 mL) was stirred at room temperature for 12 h. The reaction mixture was neutralised with saturated NaHCO3 solution and extracted with EtO Ac (3 x 50 mL). The combined organic layers were evaporated and the residue purified by filtration through a small pad of silica gel with 1:1 EtOAc-hexane to afford (3S,6S)-3- acetoxy-6,7-dihydroxy-hept-l-yne 14 (3.1 g) in 92% yield as a pale yellow liquid. [α]o - 49.6 (c 0.5, CHC13); 'HNMR (CDC13, 200 MHz): δ 1.5-1.65 (m, 2H, H-4), 1.75-2.10 (m, 2H, H-5), 2.10 (s, 3H, -OAc), 2.45 (d, IH, J 2.7 Hz, H-l), 3.38-3.5 (m, IH, H-7), 3.55- 3.78 ( m, 2H, H-6,7a), 5.38 (dt, IH, J 2.7, 5.0 Hz, H-3); 13CNMR (CDC13, 50 MHz): δ 20.87, 28.17, 30.73, 63.57, 66.55, 71.52, 73.91, 80.86, 170.04.
Part 12: (3S,6S)-3-Acetoxy-6-hydroxy-7-p-toluene sulfonyloxy-hept-1-yne (Scheme VIII; 15):
A solution of (3S,6S)-3-acetoxy-6,7-dihydroxy-hept-l-yne 14 (4.3 g, 23.1 mmol) in CH C12 (50 mL) containing pyridine (3.7 mL, 46.2 mmol) was cooled to 0°C and treated with p-toluene sulfonyl chloride (4.4 g, 23.1 mmol) and stirred at room temperature for 5h. The reaction mixture was diluted with CH2C1 (25 mL) and washed with aqueous CuSO4 solution (2x25 mL) followed by water (2x25 mL). Organic layer was dried (Na2SO4), evaporated and purified by chromatography (Si-gel, 15% EtO Ac- hexane); first eluted was (3S,6S)-3-acetoxy-6,7-di-p-toluene sulfonyloxy-hept-1-yne l_5a (1.14 g) in 10% yield as a yellow syrup. 'HNMR (CDC13, 200 MHz): δ 1.48-1.8 (m, 4H, H-3,4), 2.0 (s, 3H, OAc), 2.31 (d, IH, J 2.5 Hz, H-7) 2.4 (s, 3H, Ar-CH3), 3.94 (d, 2H, J 4.5 Hz, H-l), 4.5-4.64 (m, IH, H-2), 5.06-5.18 (m, IH, H-5), 7.20-7.3, 7.56-7.7 (m, 4H each, Ar-H).
The second eluted was (3S,6S)-3-acetoxy-6-hydroxy-7-p-toluene sulfonyloxy- hept-1-yne 15 (3 .77 g) as a yellow syrup. [α]D -28.10 (c 2.0, CHCI3); 'HNMR (CDCI3, 200 MHz): δ 1.5-1.65 (m, 2H, H-4), 1.7-2.08 (m, 2H, H-5), 2.10 (s, 3H, OAc), 2.43 (d, IH, J 1.86 Hz, H-l) 2.55 (s, 3H, Ar-CH3), 2.60 (br.s, IH, OH), 3.8-4.05 (m, 3H, H- 6,7,7a), 5.35 (dt, IH, J 1.3, 6.9 Hz, H-3), 7.35, 7.8 (2d, 4H, J 7.9 Hz, Ar-H); EIMS m/z (relative intensity): 281 (M+l, 16), 155 (60), 136 (95), 109 (55), 91 (100).
Part 13: (2S,5S)-5-Ethynyl-2-(hydroxymethyl)-tetrahydrofuran (Scheme VIII; 16): A solution of (3S,6S)-3-acetoxy-6-hydroxy-7-p-toluene sulfonyloxy-hept-1-yne
15 (3.67 g, 10.79 mmol) in MeOH (50 mL) at room temperature, was treated with K2CO3 (3.2 g, 23.7 mmol) and stirred for 3h. It was further treated with NH4C1 solution, evaporated MeOH and residue extracted with EtO Ac (3x 50 mL). Organic layer was washed with water (2 x 25 mL), brine (25 mL), dried (Na2SO4), and evaporated. The residue obtained was purified by column chromatography (Si-gel, 20% EtOAc-hexane) to afford (2S,5S)-5-ethynyl-2-(hydroxymethyl)-tetrahydrofuran 16 (1.35 g) in 90% yield as a colorless liquid. [α]D -15.07 (c 1.26, CHC13); !HNMR (CDCI3, 200 MHz): δ 1.9-2.38 (m, 4H, H-3,4), 2.45 (d, IH, J 2.1 Hz, H-7), 3.55 (dd, IH, J 6.3,12.75 Hz, H-l), 3.72 (dd, IH, J 3.8, 12.75 Hz, H-la), 4.0-4.18 (m, IH, H-2), 4.55-4.65 (m, IH, H-5); !3CNMR (CDCI3, 50 MHz): δ 26.6, 29.64, 64.62, 68.33, 73.11, 80.7, 83.92. Part 14: (2S,5S)-5-Ethynyl-2-(p-toluene sulfonyloxymethyl)- tetrahydrofuran (Scheme VIII; 17):
Procedure A: A solution of (2S,5S)-5-ethynyl-2-(hydroxymethyl)-tetrahydrofuran 16 (1.3 g, 10.2 mmol), Et3N (6.7 mL, 51.5 mmol) in CH2C12 (25 mL) containing DMAP (10 mg) was treated with p-TsCl (2.39 g, 12.2 mmol) and stirred at room temperature for 4h. Evaporation of solvent and purification of residue by column chromatography (Si-gel, 20% EtOAc-hexane) gave (2S,5S)-5-ethynyl-2-(p-toluene sulfonyloxymethyl)- tetrahydrofuran 17 (2.23 g) in 82 % yield as a yellow syrup. [α]D -30.86(c 1.38, CHC13); 'HNMR (CDCI3, 200 MHz): δ 1.8-2.25 (m, 4H, H-3,4), 2.35 (d, IH, J 2.2 Hz, H-7), 2.45 (s, 3H, Ar-CH3), 3.95-4.20 (m, 3H, H-2,l,la), 4.55-4.60 (m, IH, H-5), 7.32, 7.8 (2d, 4H, J 9.0 Hz, Ar-H).
Procedure B: A solution of (3S,6S)-3-acetoxy-6,7-dihydroxy-hept-l-yne 14 (3.5 g, 18.81 mmol) in CH2C12 (50 mL) containing Et3N (5.8 mL, 41.39 mmol) was cooled to 0°C and treated with p-toluene sulfonyl chloride (4.3 g, 22.5 mmol) and stirred at room temperature for δhours. Evaporated the solvent and the residue was purified by column chromatography (Si-gel, 15% EtOAc-hexane) to give a mixture of compounds 15 and 17
(6.7g).
The above mixture was treated with K2CO3 (6.4 g, 46.7 mmol) and stirred for 4h. The reaction mixture was further treated with NH4C1 solution, evaporated MeOH and residue extracted with EtOAc (3x 50 mL). Organic layer was washed with water (2 x 25 mL), brine (25 mL), dried (Na2SO4), and evaporated. The residue obtained was purified by column chromatography (Si-gel, 15% EtOAc-hexane), first to afford (2S,5S)-5- ethynyl-2-(p-toluene sulfonyloxymethyl)-tetrahydrofuran 17 (1.5 g) as a yellow syrup. The second eluted was (2S,5S)-5-ethynyl-2-(hydroxymethyl)-tetrahydrofuran 16 (1.61 g) as a liquid. Compound 16 (1.61 g, 12.77 mmol), was further tosylated with p- TsCl (2.65 g, 14mmol) and Et3N (4.1 mL, 31.9 mmol), as described in procedure A of this example to afford 17 (3.34 g) in 88% yield as a yellow syrup. Part 15: (2S,5S)-5-Ethynyl-2-(4-fluorophenoxymethyl)-tetrahydrofuran (Scheme VIII; 18):
To a stirred suspension of NaH (0.57 g, 23.86 mmol) in DMF (5 mL), a solution of (2S,5S)-5-ethynyl-2-(p-toluene sulfonyloxymethyl)-tetrahydrofuran l7 (3.34 g, 11.92 mmol) in DMF (5 mL) was added, followed by the addition of 4-fluoro phenol (1.74 g, 15.5 mmol) in DMF (5 mL) and heating at 80°C for 2hours. The reaction mixture was cooled to room temperature and treated with NH4C1 solution (20 mL). It was extracted with ether (3 x 40 mL) and organic layer was washed with water (2 x 25 mL), brine (25 mL) and dried (Na2SO4). Evaporation of solvent and purification of residue by column chromatography (Si-gel, 2% EtOAc-hexane) afforded (2S,5S)-5-ethynyl-2-(4- fluorophenoxymethyl)-tetrahydrofuran 18 (1.55 g) in 59.6% yield as a colorless liquid, whose spectral data was comparable with the reported reference values. [α]D -15.07(c 1.26, CHC13); 1HNMR (CDC13, 200 MHz): δ 2.0-2.35 (m, 4H, H-3,4'), 2.45 (d, IH, J 2.2 Hz, H-7), 3.95 (dd, IH, J 4.5, 9.0 Hz, H-l), 4.12 (dd, IH, J 4.54, 9.0 Hz, H-l a), 4.35 (quin, IH, H-2), 4.65 (m, IH, H-5), 6.85-7.05 (m, 4H, Ar-H); 13CNMR (CDC13, 50 MHz): δ 28.24, 33.1, 68.45, 72.9, 79.9, 83.8, 71.28, 115.43, 115.52, 115.68, 115.89, 154.91, 159.66; MS: m/z 220 (M+); HRMS: Calculated for Cι3H13O2 (M+): 220.089958; Observed; 220.089905.
Example 8: Synthesis of (2R,5R)-5-Ethynyl-2-(hydroxymethyl)-tetrahydrofuran from L-Glyceraldehyde
References in this Example 8 to compound numerals (generally underlined) designate the compounds depicted structurally in Scheme IX above. Part 1 : Ethyl (2E,4R)-4,5-isopropylidenedioxy-2-pentenoate (Scheme IX; 20):
A solution of (2S,3R)-l,2-O-isopropylidene-butane-l,2,3,4-tetrol 19 (11.0 g, 68.1 mmol) in CH2C12 (120 mL) containing saturated NaHCO3 solution (4.5 mL) was cooled to 0°C, treated with NaIO4 (29.1 g, 136.3 mmol) and allowed to stir at 0°C to 20° C. After 2 to 3 h (TLC analysis), solid Na2SO4 (6 g) was added and the reaction mixture was stirred further for 15 min. The reaction mixture was filtered and solvent evaporated (below 25°C bath temperature) to give (S)-glyceraldehyde 19a (8.7 g) in 98% yield as a colorless liquid. Compound 19 was prepared by procedures described in J. Am. Chem. Soc, 102, 6304 (1980); and J Org. Chem., 53, 2598 (1988). A solution of (S)-glyceraldehyde 19a ( 15 g, 115.4 mmol) in MeOH (200 mL) was cooled to 0°-10°C (ice-salt bath) and treated with (carbethoxymethylene) triphenyl phosphorane (48.1 g, 138.4 mmol) in portions. After stirring at room temperature for 9 h, the solvent was evaporated, the residue obtained on purification by column chromatography (Si-gel, 10% EtOAc- Hexane) gave ethyl (2E,4R)-4,5- isopropylidenedioxy-2-pentenoate 20 (23 g) in 99% yield as a pale yellow liquid. [α]o - 116.3(c 0.71, CHC13); 'HNMR (CDC13, 200 MHz): δ 1.2 (t, 3H, j 6.8 Hz, CH3), 1.3, 1.35 (2s, 6H, CH3), 3.5 (dd, IH, j 5.9 Hz, H-5), 4.07 (q, 2H, j 6.8 Hz, -OCH2), 4.27 (dd, IH, j 5.9 Hz, H-5a), 5.32-5.43 (m, IH, H-4), 5.72 (dd, IH, j 2.2, 11.3 Hz, H-2), 6.27 (dd, IH, j 5.4, 11.3 Hz, H-3); 13CNMR (CDC13, 50 MHz): δ 13.0, 25.2, 26.3, 60.1, 69.21, 73.3, 109.4, 120.5, 149.1, 165.3; EIMS m/z (relative intensity): 185 (M+-15, 15), 173 (6), 149 (23), 125 (20), 97 (45), 43 (100); HRMS: Calculated for C9H13O4 (M+-15): 145.086469; Observed: 145.087162.
Part 2: Ethyl (4R)-4,5-isopropylidenedioxy-l-pentanoate (Scheme IX; 21): A solution of ethyl (2E,4R)-4,5-isopropylidenedioxy-2-pentenoate_20 (23 g, 115 mmol) in EtOAc (50 mL) was treated with PtO2 (0.100 g, mmol) and hydrogenated till there was no additional consumption of hydrogen (3-4 h). At the end of reaction, the reaction mixture was filtered and concentrated to afford ethyl (4R)-4,5- isopropylidenedioxy-1-pentanoate 21 (23 g) in 99% yield as a colorless liquid. [α]o - 4.0(c 2.0, CHC13); 'HNMR (CDCI3, 200 MHz): δ 1.25 (t, 3H, J6.8 Hz, CH3), 1.29, 1.32 (2s, 6H, CH3), 1.75-1.89 (m, 2H, H-3), 2.3-2.45 (m, 2H, H-2), 3.5 (t, IH, J 6.5 Hz, H-5), 3.92-4.15 (m, 4H, H-4,5a, -OCH2); 13CNMR (CDC13, 50 MHz): δ 14.0, 25.4, 26.8, 28.6, 30.2, 60.1, 68.8, 74.7, 108.7, 172.6. EIMS m/z (relative intensity): 203 (M++l, 23), 173 (16.4), 143 (13.4), 101 (100), 43 (97); HRMS: Calculated for C83O4 (M+-29): 173.081384; Observed: 1173.081619.
Part 3: (2R)-l,2-Isopropylidenedioxy-5-pentanol (Scheme IX; 22): A suspension of LAH (4.93 g, 130.4 mmol) in THF (50 mL) was cooled to 0°C and treated drop wise with a solution of ethyl (4R)-4,5-isopropylidenedioxy-l-pentanoate 21 (22 g, 108.9 mmol) in THF (75 mL). The reaction mixture was warmed to room temperature, then allowed to stir for 3 h and treated with a saturated solution of Na SO4 (15 mL). After stirring for additional 30 min., it was filtered through celite and washed with EtOAc (3 x 75 mL). The combined organic layers were washed with NaCl solution and evaporated to provide the (2R)-l,2-isopropylidenedioxy-5-pentanol 22 (17 g) in 97% yield as a colorless liquid. [α]D -10.3(c 0.75, CHC13); 1HNMR (CDC13, 200 MHz): δ 1.35, 1.4 (2s, 6H), 1.6-1.75 (m, 4H, H-3,4), 1.92 (br.s, IH, OH), 3.5 (t, IH, J 6.1 Hz, H- 1), 3.6-3.72 (m, 2H, H-5), 3.98-4.16 (m, 2H, H-la,2); 13CNMR (CDC13, 50 MHz): δ 25.6, 26.8, 29.0, 30.1, 62.4, 69.4, 75.9, 108.8; EIMS m/z (relative intensity): 145 (M+ -15, 13.4), 85 (32), 72 (18), 57 (13.4), 43 (100); HRMS: Calculated for C7H13O3 (M+-15): 145.086468; Observed: 145.087162.
Part 4: (4R)-4,5-Isopropylidenedioxy-l-pentanal (Scheme IX; 23): Method A: A stirred solution of (2R)-l,2-isopropylidenedioxy-5-pentanol 22 (17 g, 106.3 mmol) in CH2C12 (200 mL) was treated with PDC (59.9 g, 159.3 mmol) in portions and allowed stir at 40°C for 5 h. The reaction mixture was diluted with ether (4x300 mL) and decanted through a small pad of silica gel. Evaporation of solvent afforded (4R)-4,5-isopropylidenedioxy-l-pentanal 23 (15 g) in 89% yield as a pale yellow liquid.
Method B: A stirred solution of (2R)-l,2-isopropylidenedioxy-5-pentanol 22 (0.800 g, 5.0 mmol) in DMSO (5 mL) was cooled to 0°C, treated with IBX (1.47 g, 5.26 mmol) in portions while maintaining the temperature below 0°C and stirred at room temperature for 4 h. The reaction mixture was treated with saturated NaHCO3 solution, filtered through celite and washed with EtOAc (3 x 30 mL). Two layers were separated and organic layer was washed with water, brine and dried (Na2SO4). Evaporation of solvent gave (4R)-4,5-isopropylidenedioxy-l-pentanal 23 (16.2 g) in 78% yield as a yellow liquid. [α]D +0.3(c 2.0, CHC13).
Part 5: Ethyl (2E,6R)-6,7-isopropylidenedioxy hept-2-enoate (Scheme IX; 24): A solution of (4R)-4,5-isopropylidenedioxy-l-pentanal 23 (15 g, 94.9 mmol) in benzene ( 200 mL) was treated with (carbethoxymethylene) triphenyl phosphorane (39.6 g, 113.8 mmol) and heated at reflux for 6h. Solvent was evaporated and the residue purified by column chromatography (Si-gel, 10% EtOAc-hexane) to afford ethyl
(2E,6R)-6,7-isopropylidenedioxy hept-2-enoate 24 (14 g) in 65% yield as a pale yellow liquid. [α]D -5.4(c 1.2, CHCI3); 1HNMR (CDCI3, 200 MHz): δ 1.3 (t, 3H, J 6.8 Hz,CH3), 1.34, 1.4 (2s, 6H), 1.61-1.7 (m, 2H, H-6), 2.2-2.42 (m, 2H, H-4), 3.5 (t, IH, J 6.8Hz, H- 7a), 3.99-4.26 (m, 4H, H-6,7,-OCH2), 5.82 (td,lH, J 2.25, 15.75 Hz, H-2), 6.94 (dt, IH, J 6.8, 15.75 Hz, H-3); 13CNMR (CDC13, 50 MHz): δ 14.0, 25.4, 26.7, 28.2, 31.9, 60.0, 69.0, 74.9, 108.7, 121.7, 147.7, 166.3; EIMS m/z(relative intensity): 213 (M+-15, 9), 95 (40.2), 67 (25.3), 55 (53.7), 41 (100); HRMS: Calculated for C,,Hι7O4 (M+-15): 213.112684; observed: 213.112732.
Part 6: (2E,6R)-6,7-Isoρropylidenedioxy hept-2-ene-l-ol (Scheme IX; 25):
A stirred solution of ethyl (2E,6R)-6,7-isopropylidenedioxy hept-2-enoate 24 (13.87 g, 60.8 mmol) in dry CH2C12 (60 mL) was cooled to -20°C (CC14 + dry ice bath) and treated with a solution of DIBAL-H (17.27g, 121.6 g, mmol; 2.5M solution in hexane) drop wise. After stirring for 2h, the reaction mixture was warmed to 0°C, treated drop wise with MeOH (10 mL) to obtain a gelatin cake. The mixture was diluted with CH2C12 (100 mL) and stirred for 15 min. A solution of Na-K tartarate (90 mL) was added drop wise and stirred for an additional 45 min. Reaction mixture was filtered through celite and washed with CH C1 (2 x 50 mL). The organic layer was washed with water (2 x 100 mL), brine (50 mL), dried (Na2SO ) and evaporated to give (2E,6R)-6,7- isopropylidenedioxy hept-2-ene-l-ol 25 (11 g) in 98.2% yield as a colorless liquid. [α]π - 13.2 (c 2.5, CHC13); 'HNMR (CDCI3, 200 MHz): δ 1.16,1.2 (2s, 6H, CH3), 1.46-1.74 (m, 2H, H-5), 1.79-198 (m, IH, -OH), 2.02-219 (m, 2H, H-4), 3.36-3.78(m, 3H, H-6,7), 4.02-412 (m, 2H, H-l), 5.61-5.71 (m, 2H, H-2,3); 13CNMR (CDC13, 50 MHz): δ 25.3, 26.5, 28.0, 32.7, 62.8, 68.9, 75.1, 108.3, 129.8 (2C); EIMS m/z (relative intensity): 171 (M+-15, 35.8), 93 (22.3), 67 (37.3), 55 (26.8), 43 (100); HRMS: Calculated for C9H15O3 (M+-15): 171102120; observed: 171102195.
Part 7: (2R,3R,6R)-2,3-Epoxy-6,7-isopropylidenedioxy heptan-1-ol (Scheme IX; 26):
To a stirred and cooled (-20°C) suspension of molecular sieves (4 A, 1.25 g) in CH2C12 (10 mL) under N atmosphere, (-)-diisopropyl D-tartarate (7.6 g, 32.4 mmol), titanium(rV) isopropoxide (7.68 g, 27.02 mmol) and cumene hydroperoxide (8.22 g, 54 mmol) were added sequentially. After 20 min., the resulting mixture was treated drop wise addition of a solution of (2E,6R)-6,7-isopropylidenedioxy hept-2-ene-l-ol 25 (5 g, 26.88 mmol) in CH2C12 (15 mL) and stirred for additional 3h. The reaction mixture was quenched with 10% NaOH solution saturated with NaCl (15 mL) and filtered through celite. Evaporation of solvent and purification of residue by column chromatography (Si- gel, 1:1 EtOAc-hexane) gave (2R,3R,6R)-2,3-epoxy-6,7-isopropylidenedioxy heptan-1-ol 26 (4.15 g) in 76.4% yield as a colorless liquid. [α]D +24.3(c 0.3, CHC13); 1HNMR
(CDCI3, 200 MHz): δ 1.32, 1.38 (2s, 6H, CH3), 1.58-1.78 (m, 4H, H-4,5), 2.84-3.01 (m, 2H, H-2,3), 3.5 (t, IH, J 6.1 Hz, H-7), 3.6 (dd, IH, J 4.7, 11.75 Hz, H-l), 3.85 (dd, IH, J 3.29, 11.75, H-la), 3.98-4.2 (m, 2H, H-6,7'); 13CNMR (CDC13, 50 MHz): δ 25.5, 26.8, 27.6, 29.6, 55.3, 58.3, 61.6, 691, 75.1, 108.8; EIMS M/Z (relative intensity): 188 (M+ - 15, 14.9), 144 (85), 101 (47.7), 83 (95), 43 (100); HRMS: Calculated for C95O4 (M- 15): 187.097034; Observed: 187.096634. Part 8: (2R,3R,6R)-l-Chloro-2,3-epoxy-6,7-isopropylidenedioxy heptane (Scheme IX; 27):
A stirred mixture of (2R,3R,6R)-2,3-epoxy-6,7-isopropylidenedioxy heptan-1-ol 26 (3.8 g, 18.8 mmol), Ph3P (7.4 g, 28.3 mmol) and NaHCO3 (0.6 g) in CC14 (50 mL) was heated at reflux for 3 h. The solvent was evaporated and residue obtained purified by column chromatography (Si- gel, 20% EtOAc-hexane) to give (2R,3R,6R)-l-chloro-2,3- epoxy-6,7-isopropylidenedioxy heptane 27 (2.8 g) in 67.8% yield as a colorless liquid. [CC]D +816(C 0.7, CHC13); 'HNMR (CDCI3, 200 MHz): δ 1.31, 1.36 (2s, 6H, CH3), 1.63- 1.72 (m, 4H, H-4,5), 2.8-2.9 (m, IH, H-2), 2.91-3.02 (m, IH, H-3), 3.32-3.68 (m, 3H, H- 1,7), 3,95-419 (m, 2H, H-6,7a); 13CNMR (CDCI3, 50 MHz): δ 25.6, 26.9, 27.6, 29.6,
44.5, 57.0, 58.3, 69.2, 75.1, 108.9; EIMS m/z (relative intensity): 205 (M+-15, 35.8), 145 (23), 83 (61), 72 (98), 43 (100); HRMS: Calculated for C94ClO3 (M+-15): 205.063147; Observed: 205.062719.
Part 9: (3R,6R)-3-Hydroxy-6,7-isopropylidenedioxy-hept-l-yne (Scheme IX;
To freshly prepared LDA [prepared from diisopropyl amine (4.6 g, 45.45 mmol) and n-BuLi (2.91 g, 45.54 mmol; 1.4N hexane solution)] in THF (10 mL),a solution of (2R,3R,6R)-l-chloro-2,3-epoxy-6,7-isopropylidenedioxy heptane 27 (2.5 g, 11.36 mmol) in THF (20 mL) was added at -40°C (CH3CN + dry ice bath). After 3h, the reaction was quenched with aq. NH4C1 solution and diluted with CH C1 (50 mL). The organic layer was separated, washed with water (3 x 20 mL), brine (200 mL) and dried (Na2SO4), evaporated and residue purified by column chromatography (Si-gel, 15% EtOAc-hexane) to furnish (3R,6R)-3-hydroxy-6,7-isopropylidenedioxy-hept-l-yne 28 ( 2.0 g) in 95.2% yield as a pale yellow liquid. [α]D -3.02(c 2.2, CHC13); 'HNMR (CDCI3, 200 MHz): δ
1.32, 1.39 (2s, 6H, CH3), 1.64-1.94 (m, 4H, H-4,5), 219-2.21 (br.s, IH, OH), 2.39 (d, IH, J 2.3Hz, H-l), 3.5 (t, IH, J 5.7 Hz, H-7), 3.96-416 (m, 2H, H-6,7a), 4.34-4.45 (m, IH, H- 3); 13CNMR (CDCI3, 50 MHz): δ 25.4, 26.6, 28.8, 33.5, 61.3, 69.0, 72.7, 75.3, 84.7, 108.7; EIMS m/z (relative intensity): 169 (M+-15, 22.3), 109 (20.8), 81 (37.3), 55 (35.8), 43 (100); HRMS: Calculated for C9H13O3 (M-15): 169.086469; Observed: 169.086140.
Part 10: (3R,6R)-3-Acetoxy-6,7-isopropylidenedioxy-hept-l-yne (Scheme IX; 29):
A solution of hydroxy-6,7-isopropylidenedioxy-hept-l-yne 28 (1.8 g, 9.8 mmol) and pyridine (3.1 g, 39.2 mmol) in CH2C12 (15 mL) containing DMAP (catalytic) at 0°C was treated with Ac2O (1.2 g, 11.7 mmol) and stirred at room temperature for 30 min. After completion, the reaction was diluted with CH2C1 (50 mL), sequentially washed with CuSO4 solution (3 x 20 mL), saturated aq. NaHCO3 solution (20 mL), water ( 20 mL), brine (20 mL) and dried. Evaporation of solvent and purification of residue by column chromatography (Si-gel, 10% EtOAc-hexane) gave (3R,6R)-3-acetoxy-6,7- isopropylidenedioxy-hept-1-yne 29 (2.15 g) in 97.2% yield as a yellow liquid. [α]o +37.5(c 2.1, CHC13); 'HNMR (CDC13, 200 MHz): δ 1.3, 1.39 (2s, 6H, CH3), 1.64-2.0 (m, 2H, H-4,5), 2.06 (s, 3H, CH3), 2.4 (d, IH, J 2.0 Hz, H-l), 3.5 (t, IH, J 5.7 Hz, H-7), 3.95- 4.13 (m, 2H, H-6,7a), 5.31-5.41 (m, IH, H-3); 13CNMR (CDCI3, 50 MHz): δ 20.8, 25.5, 26.8, 28.8, 30.7, 63.3,691, 73.7, 75.1, 80.7, 108.9, 169.6; EIMS m/z (relative intensity): 211 (M+ -15, 29.8), 169 (11.9), 91 (22.3), 72 (23), 43 (100); HRMS: Calculated for d ιH15O4 (M+-l 5): 211.097034; Observed; 211.095947.
Part 11 : (3R,6R)-3-Acetoxy-6,7-dihydroxy-hept-l-yne (Scheme IX; 30): A solution of (3R,6R)-3-acetoxy-6,7-isopropylidenedioxy-hept-l-yne 29 (2 g, 8.8 mmol) in MeOH (150 mL) containing catalytic amount of PTSA was stirred at 0°C for 8 h. The reaction mixture was neutralised with saturated sat. NaHCO3 solution, evaporated to remove MeOH and extracted with EtOAc (3 x 50 mL). Organic layer were evaporated and the residue filtered through a small pad of silica gel with 1 :1 EtOAc- hexane to afford (3R,6R)-3-acetoxy-6,7-dihydroxy-hept-l-yne 30 (1.2 g) in 72.9% yield as a colorless syrup. [α]D +83.2 (c 1.2, CHC13); 'HNMR (CDC13, 200 MHz): δ 1.5-1.7 (m, 2H, H-4), 1.75-2.05 (m, 2H, H-5), 2.14 (s, 3H, -OAc), 2.45 (d, 2H, H-l), 2.57 (br.s, IH, OH), 3.35-3.5 (m, H, H-7), 3.57-3.8 (m, 2H, H-6,7a), 5.32-5.47 (m, IH, H-3); CIMS m/z (relative intensity): 187 (M+l, 74.6), 127 (59.7), 109 (35.8), 81 (56.7), 43 (100); HRMS Calculated for C9H15O4 ( M+l): 187.097034; Observed: 187.096547.
Part 12 (3R,6R)-3-Acetoxy-6-hydroxy-7-p-toluene sulfonyloxy-hept-1-yne
(Scheme IX; 31):
A solution of (3R,6R)-3-acetoxy-6,7-dihydroxy-hept-l-yne 30 (1.1 g, 5.9 mmol) in CH2C12 (20 mL) containing pyridine (0.934 g, 11.82 mmol) was cooled to 0°C, treated with p-TsCl (1.12 g, 5.91 mmol) and stirred at room temperature for 8 h. The reaction mixture was diluted with CH2C1 and washed sequentially with water (20 mL), CuSO4 solution (3 x 20 mL) and water (20 mL). Organic layer was dried (Na2SO4), evaporated and residue obtained was purified by column chromatography (Si-gel, 10% EtOAc- Hexane); first eluted was (3R,6R)-3-acetoxy-6,7-di-p-toluene sulfonyloxy-hept-1-yne 31_a (0.23 g) in 8% yield as a yellow syrup. 1HNMR (CDC13, 200 MHz): δ 1.5-1.85 (m, 4H, H-3,4), 2.05 (s, 3H, OAc), 2.41-2.52 (m, 7H, H-7, Ar-CH3), 4.0 (d, 2H, J4.8 Hz, H-l), 4.58-4.62 (m, IH, H-2), 512-5.26 (m,lH, H-5), 7.28-7.44, 7.64-7.81 (m, 4H each, Aril).
Second eluted was (3R,6R)-3-acetoxy-6-hydroxy-7-p-toluene sulfonyloxy-hept-1- yne^l (11 g) in 55% yield as a yellow syrup. [α]D +28.1 (c 1.0, CHC13); 'HNMR (CDC13, 200 MHz): δ 1.35-1.68 (m, 3H, H-4,-OH), 1.68-2.0 (m, 2H, H-5), 2.08 (s, 3H, CH3), 2.4 (d, IH, J 2.4 Hz, H-l), 2.46 (s, 3H, Ar-CH3), 3.79-4.06 (m, 3H, H-6,7), 5.35 (td,lH, J 4.8, 7.2 Hz, H-3), 7.36 (d, 2H, J 7.2 Hz, Ar-H), 7.8 (d, 2H, J 7.2 Hz, Ar-H). FABMS m/z (relative intensity): 341(M+1, 13.8), 281(50), 155(541), 133(52.7), 109(100). HRMS: Calculated for Cι6H2ιO6S (M+l):341105885; Observed:341104916.
Part 13: (2R,5R)-5-Ethynyl-2-(hydroxymethyl)-tetrahydrofuran (Scheme IX; 32): To a solution of (3R,6R)-3-acetoxy-6-hydroxy-7-p-toluene sulfonyloxy-hept-1- yne 31 (0.6 g, 1.76 mmol) in MeOH (10 mL) at room temperature, K2CO3 (0.536 g, 3.88 mmol) was added and the mixture was stirred for 2h. It was treated with NH C1 solution, evaporated MeOH and the residue extracted with EtOAc (3 x 20 mL). Organic layer was washed with water (10 mL), brine (10 mL), dried (Na SO4) evaporated. The residue obtained was purified by column chromatography (Si-gel, 20% EtOAc-hexane) to furnish (2R,5R)-5-ethynyl-2-(hydroxymethyl)-tetrahydrofuran 32 (0.22 g) in 99% yield as a colorless liquid. [α]D +20.0 (c 1.0, CHC13); 'HNMR (CDC13, 200 MHz): δ 1.89-2.38 (m, 4H, H-3,4), 2.4 (br.s, IH, OH), 2.46 (d, IH, J 2.2 Hz, H-7), 3.55 (dd, IH, J 4.5, 11.25 Hz, H-l), 3.72 (dd, IH, J 4.0, 11.25 Hz, H-la), 4.0-415 (m, IH, H-2), 4.52-4.66 (m,lH, H-5); 13CNMR (CDC13, 50 MHz): δ 26.6, 29.6, 33.6, 64.6, 68.3, 73.0, 80.7; EIMS m/z (relative intensity): 125 (MM, 8), 95 (74.6), 67 (100), 53 (40), 41 (80); HRMS: Calculated for C7H9O2 (M-l): 125.060255; Observed: 125.060322.
Part 14: (2R,5R)-5-Ethynyl-2-(p-toluene sulfonyloxymethyl)-tetrahydrofuran (Scheme IX; 33):
A solution of alcohol (2R,5R)-5-ethynyl-2-(hydroxymethyl)-tetrahydrofuran 32 (0.22 g, 1.75 mmol) in pyridine (0.6 mL) was treated with p-TsCl (0.354 g, 1.86 mmol) and the mixture stirred at room temperature for 3 h. The reaction mixture was diluted with CH C12 (20 mL) and washed sequentially with water (10 mL), CuSO4 solution (2 x 10 mL), brine (10 mL) and dried (Na SO4). Evaporation of solvent and purification of residue by column chromatography (Si-gel, 15% EtOAc-hexane) gave (2R,5R)-5- ethynyl-2-(p-toluene sulfonyloxymethyl)-tetrahydrofuran 33 (0.33 g) in 63.9% yield as a yellow syrup. [α]D +10.0 (c 0.54, CHC13); 'HNMR (CDC13, 200 MHz): δ 1.84-211 (m, 4H, H-3,4) 2.32 (d, IH, J 2.1 Hz, H-7), 2.45 (s, 3H, CH3), 3.92-4.2 (m, 3H, H-2,l,la), 4.48-4.58 (m, IH, H-5), 7.34 (d, 2H, J 7.6 Hz, Ar-H), 7.8 (d, 2H, J 7.6 Hz, Ar-H); CIMS m/z (relative intensity): 281(M+1, 100), 109(49.2), 117(31.3), 81(7.0), 43(100); HRMS: Calculated for C14H17O4S (M+l):281.084756; Observed: 281.083610. Part 15: (2R,5R)-5-Ethynyl-2-(4-fluoro phenoxymethyl)-tetrahydrofuran (Scheme IX; 34):
To a stirred suspension of NaH (0.032 g, 1.33 mmol) in DMF (3 mL), a solution of (2R,5R)-5-ethynyl-2-(p-toluene sulfonyloxy methyl)-tetrahydrofuran 33 (0.33 g, 1.1 mmol) in DMF (3 mL) was added and heated at 80°C for 5h. The reaction mixture was cooled to room temperature and treated with NHjCl solution. It was extracted with ether (2 x 10 mL) and the organic layer was washed with water (2 x 10 mL), brine (10 mL) and dried (Na SO4). Evaporation of solvent and purification of residue by column chromatography (Si-gel, 7% EtOAc-hexane) afforded (2R,5R)-5-ethynyl-2-(4-fluoro phenoxy methyl)-tetrahydrofuran 34 (0.21 g) in 85.7% yield as a colorless liquid, whose spectral data is accordance with the reported reference values. [α]o +16.0 (c 1.0, CHCI3); 'HNMR (CDCI3, 200 MHz): δ 1.88-2.32 (m, 4H, H-3,4), 2.41 (d, IH, J 2.3 Hz, H-7), 3.9 (dd, IH, J 4.6, 9.1 Hz, H-l), 4.06 (dd, IH, J 5.9, 9.1 Hz, H-la), 4.22-4.36 (m, IH, H-2), 4.58-4.69 (m, IH, H-5), 6.75-7.02 (m, 4H, Ar-H); 13CNMR (CDCI3, 50 MHz): δ 28.2, 33.1, 68.5, 71.2, 72.9, 76.3, 83.7, 115.4, 115.6, 115.8, 115.9, 154.9, 159.6; EIMS m/z (relative intensity): 220 (M+, 10.4), 125 (14.9), 95 (94), 67 (100), 41 (59.7); HRMS: Calculated for C133O2F (M+): 220.089958; Observed: 220.089497.
Example 9: Preparation of (2R,5S)-5-Ethynyl-2-(hydroxymethyl)-tetrahydrofuran)
References in this Example 9 to compound numerals (generally underlined) designate the compounds depicted structurally in Scheme X above.
Part 1: (2S,3S,6R)-2,3-Epoxy-6,7-isopropylidenedioxy heptan-1-ol (Scheme X; 43): To a stirred and cooled (-20°C) suspension of molecular sieves (4 A, 0.55 g) in
CH2C12 (10 mL) under N2 atmosphere, (+)-diisopropyl D-tartarate (4.0 g, 17.41 mmol), titanium(IV) isopropoxide (4.12 g, 14.51 mmol) and cumene hydroperoxide (4.4 g, 29.0 mmol) were added sequentially. After 20 min., the resulting mixture was treated drop wise addition of a solution of (2E,6R)-6,7-isopropylidenedioxy hept-2-ene-l-ol 25 (2.7 g, 14.51 mmol) in CH2C12 (10 mL) and stirred for additional 3h. The reaction mixture was quenched with 10% NaOH solution saturated with NaCl (15 mL) and filtered through celite. Evaporation of solvent and purification of residue by column chromatography (Si- gel, 1:1 EtOAc-hexane) gave (2R,3R,6R)-2,3-epoxy-6,7-isopropylidenedioxy heptan-1-ol 43 (2.1 g) in 72.4% yield as a colorless liquid. [ ]D -26.9(c 1.1, CHC13); 1HNMR (CDCI3, 200 MHz): δ 1.32, 1.38 (2s, 6H, CH3), 1.58-1.79 (m, 4H, H-4,5), 2.3-2.52 (br.s, IH, OH), 2.84-3.0 (m, 2H, H-2,3), 3.5 (t, IH, J 6.1 Hz, H-7), 3.6 (dd, IH, J 4.5, 12.0 Hz, H-l), 3.85 (dd, IH, J 3.3, 11.2 Hz, H-la), 3.98-4.2 (m, 2H, H-6,7').
Part 2: (2S,3S,6R)-l-Chloro-2,3-epoxy-6,7-isopropylidenedioxy heptane (Scheme
X; 44):
A stirred mixture of (2S,3S,6R)-2,3-epoxy-6,7-isopropylidenedioxy heptan-1-ol 43 (1.9 g, 9.4 mmol), Ph3P (3.7 g, 14.1 mmol) and NaHCO3 (0.3 g) in CC14 (25 mL) was heated at reflux for 3 h. The solvent was evaporated and residue obtained purified by column chromatography (Si- gel, 20% EtOAc-hexane) to give (2S,3S,6R)-l-chloro-2,3- epoxy-6,7-isopropylidenedioxy heptane 44 (1.5 g) in 75% yield as a colorless liquid. [CC]D +12.9 (c 1.2, CHCI3); 'HNMR (CDC13, 200 MHz): δ 1.32, 1.39 (2s, 6H, CH3), 1.58-1.72 (m, 4H, H-4,5), 2.8-2.9 (m, IH, H-2), 2.91-3.02 (m, IH, H-3), 3.32-3.68 (m, 3H, H-1,7), 3.94-416 (m, 2H, H-6,7a).
Part 3: (3S,6R)-3-Hydroxy-6,7-isopropylidenedioxy-hept-l-yne (Scheme X; 45): To freshly prepared LDA [prepared from diisopropyl amine (2.3 g, 23.6 mmol) and n-BuLi (15.7 mL, 23.6 mmol; 1.5N hexane solution)] in THF (6 mL), a solution of (2S,3S,6R)-l-chloro-2,3-epoxy-6,7-isopropylidenedioxy heptane 44 (1.5 g, 5.9 mmol) in THF (10 mL) was added at -40°C (CH3CN + dry ice bath). After 3h, the reaction was quenched with aq. NH C1 solution and diluted with CH2C12 (25 mL). The organic layer was separated, washed with water (3 x 15 mL), brine (100 mL) and dried (Na2SO4), evaporated and residue purified by column chromatography (Si-gel, 15% EtOAc-hexane) to furnish (3S,6R)-3-hydroxy-6,7-isopropylidenedioxy-hept-l-yne 45 ( 1.0 g) in 83% yield as a pale yellow liquid. [α]D -21.0 (c 1.0, CHC13); 'HNMR (CDC13, 200 MHz): δ 1.34, 1.4 (2s, 6H, CH3), 1.64-1.9 (m, 4H, H-4,5), 2.49 (d, IH, J 2.3Hz, H-l), 2.55-2.7 (br.s, IH, OH), 3.52 (t, IH, J 5.8 Hz, H-7), 3.98-416 (m, 2H, H-6,7a), 4.35-4.49 (m, IH, H-3).
Part 4: (3S,6R)-3-Acetoxy-6,7-isopropylidenedioxy-hept-l-yne (Scheme X; 46): A solution of (3S,6R)-3-hydroxy-6,7-isopropylidenedioxy-hept-l-yne 45 (1.0 g, 5.43 mmol) and Et3N (2.74 g, 27.15 mmol) in CH2C12 (10 mL) containing DMAP (catalytic) at 0°C was treated with Ac2O (0.665 g, 6.52 mmol) and stirred at room temperature for 30 min. After completion, the reaction was diluted with CH C1 (25 mL), washed with water (2 x 20 mL), brine (20 mL) and dried. Evaporation of solvent and purification of residue by column chromatography (Si-gel, 10% EtOAc-hexane) gave (3S,6R)-3-acetoxy-6,7-isopropylidenedioxy-hept-l-yne 46 (1.1 g) in 91% yield as a yellow liquid. [α]D -56.5 (c 1.0, CHC13); 1HNMR (CDCI3, 200 MHz): δ 1.32, 1.39 (2s, 6H, CH3), 1.46-2.01 (m, 2H, H-4,5), 2.08 (s, 3H, CH3), 2.41 (d, IH, J 2.3 Hz, H-l), 3.5 (t, IH, J 6.9 Hz, H-7), 3.98-414 (m, 2H, H-6,7a), 5.31-5.42 (m, IH, H-3).
Part 5: (3S,6R)-3-Acetoxy-6,7-dihydroxy-hept-l-yne (Scheme X; 47):
A solution of (3S,6R)-3-acetoxy-6,7-isopropylidenedioxy-hept-l-yne 46 (1.1 g, 4.86 mmol) in 60% aqueous AcOH (10 mL) was stirred at room temperature for 12 h. The reaction mixture was neutralised with saturated sat. NaHCO3 solution and extracted with EtOAc (3 x 50 mL). Organic layer was dried (Na SO4), evaporated and the residue filtered through a small pad of silica gel with 1:1 EtOAc-hexane to afford (3S,6R)-3- acetoxy-6,7-dihydroxy-hept-l-yne 47 (0.6 g) in 66.6% yield as a colorless syrup. [CC]D - 46.3 (c 1.0, CHCI3); 'HNMR (CDC13, 200 MHz): δ 1.45-1.7 (m, 2H, H-4), 1.75-2.05 (m, 2H, H-5), 2.09 (s, 3H, -OAc), 2.4 (br.s, 2H, H-l, OH), 3.32-3.5 (m, H, H-7), 3.52-3.8 (m, 2H, H-6,7a), 5.3-5.44 (m, IH, H-3). Part 6: (3S,6R)-3-Acetoxy-6-hydroxy-7-p-toluene sulfonyloxy-hept-1-yne (Scheme X; 48):
A solution of (3S,6R)-3-acetoxy-6,7-dihydroxy-hept-l-yne 47 (0.6 g, 3.22 mmol) and Et3N (0.975 g, 9.66 mmol) in CH2C12 (10 mL) was cooled to 0°C, treated with p- TsCl (0.737 g, 3.87 mmol) and stirred at room temperature for 10 hours. The reaction mixture was diluted with CH2C1 (20 mL) and washed with water (2 x 15 mL) and brine (20 mL). Organic layer was dried (Na2SO4), evaporated and residue obtained was purified by column chromatography (Si-gel, 10% EtOAc-Hexane) to afford (3S,6R)-3- acetoxy-6-hydroxy-7-p-toluene sulfonyloxy-hept-1-yne 48 (0.94 g) in 89% yield as a colorless syrup. [α]D -37.2 (c 1.3, CHC13); 'HNMR (CDC13, 200 MHz): δ 1.51-2.0 (m, 5H, H-4, 5, -OH), 2.08 (s, 3H, CH3), 2.4 (d, IH, J 2.3 Hz, H-l), 2.48 (s, 3H, Ar-CH3), 3.8-4.06 (m, 3H, H-6,7), 5.35 (td,lH, J 4.8, 7.2 Hz, H-3), 7.36 (d, 2H, J 7.9 Hz, Ar-H), 7.8 (d, 2H, J 7.9 Hz, Ar-H).
Part 7: (2R,5S)-5-Ethynyl-2-(hydroxymethyl)-tetrahydrofuran (Scheme X; 49):
To a solution of (3S,6R)-3-acetoxy-6-hydroxy-7-p-toluene sulfonyloxy-hept-1- yne 48 (0.9 g, 2.64 mmol) in MeOH (15 mL) at room temperature, K2CO3 (0.805 g, 5.83 mmol) was added and the mixture was stirred for 2h. It was treated with NH4C1 solution, evaporated MeOH and the residue extracted with EtOAc (3 x 20 mL). Organic layer was washed with water (10 mL), brine (10 mL), dried (Na2SO4) evaporated. The residue obtained was purified by column chromatography (Si-gel, 20% EtOAc-hexane) to furnish (2R,5S)-5-ethynyl-2-(hydroxymethyl)-tetrahydrofuran 49 (0.4 g) in 95% yield as a colorless liquid. [α]D -10.5 (c 1.0, CHC13); 'HNMR (CDC13, 200 MHz): δ 1.50-2.3 (m, 4H, H-3,4), 2.4 (br.s, IH, OH), 2.42 (d, IH, J 2.2 Hz, H-7), 3.5 (dd, IH, J 4.5, 11.3 Hz, H-l), 3.75 (dd, IH, J 3.3, 11.35 Hz, H-l a), 416-4.34 (m, IH, H-2), 4.6-4.74 (m,lH, H- 5). Part 8: (2R,5S)-5-Ethynyl-2-(p-toluene sulfonyloxymethyl)-tetrahydrofuran (Scheme X; 50):
A solution of alcohol(2R,5S)-5-ethynyl-2-(hydroxymethyl)-tetrahydrofuran 49 (0.33 g, 2.6 mmol) and Et3N (0.263 g, 7.8 mmol) in CH2C12 (10 mL) containing DMAP (catalytic) was treated with p-TsCl (0.499 g, 2.6 mmol) at 0°C and the mixture stirred at room temperature for 3 hours. The reaction mixture was diluted with CH2C12 (20 mL) and washed sequentially with water (2 x10 mL), brine (10 mL) and dried (Na SO4). Evaporation of solvent and purification of residue by column chromatography (Si-gel, 15%) EtOAc-hexane) gave (2R,5S)-5-ethynyl-2-(p-toluene sulfonyloxy methyl)- tetrahydrofuran 50 (0.4 g) in 55% yield as a colorless syrup. [α]D -7.0(c 1.0, CHCI3);
'HNMR (CDCI3, 200 MHz): δ 1.64-2.0 (m, 4H, H-3,4) 2.22 (d, IH, J 2.1 Hz, H-7), 2.35 (s, 3H, CH3), 3.72-4.05 (m, 3H, H-2,l,la), 4.28-4.48 (m, IH, H-5), 7.34 (d, 2H, J 7.6 Hz, Ar-H), 7.8 (d, 2H, J 7.6 Hz, Ar-H).
Part 9: (2R,5S)-5-Ethynyl-2-(4-fluoro phenoxymethyl)-tetrahydrofuran (Scheme
X; 51):
To a stirred suspension of NaH (0.041 g, 1.71 mmol) in DMF (3 mL), a solution of (2R,5S)-5-ethynyl-2-(p-toluene sulfonyloxy methyl)-tetrahydrofuran 50 (0.32 g, 1.1 mmol) in DMF (3 mL) was added and heated at 80°C for 3h. The reaction mixture was cooled to room temperature and treated with NH C1 solution. It was extracted with hexane (2 x 10 mL) and the organic layer was washed with water (2 x 10 mL), brine (10 mL) and dried (Na SO4). Evaporation of solvent and purification of residue by column chromatography (Si-gel, 7% EtOAc-hexane) afforded (2R,5S)-5-ethynyl-2-(4-fluoro phenoxy methyl)-tetrahydrofuran 5_1 (0.19 g) in 76% yield as a colorless liquid, [α]π - 21.5 (c 1.1, CHCI3); 'HNMR (CDC13, 200 MHz): δ 1.8-2.34 (m, 4H, H-3,4), 2.38 (d, IH, J 2.3 Hz, H-7), 3.81-4.0 (m, 2H, H-l, la), 4.36-4.51 (m, IH, H-2), 4.64-4.78 (m, 2H, H- 5), 6.75-7.02 (m, 4H, Ar-H). Example 10: Preparation of (2S,5R)-5-Ethynyl-2-(hydroxymethyl)-tetrahydrofuran from homopropargyl alcohol
References in this Example 10 to compound numerals (generally underlined) designate the compounds depicted structurally in Scheme XI above.
Part 1 : (2S,5RS)-5-Hydroxy-l,2-isopropylidenedioxy-9-p-methoxyphenyl methyl-non-6-yn-9-ol (Scheme XI; 35):
A stirred solution of 1-p-methoxy phenyl methyl but-3-yn-l-ol (1.0 g, 5.3 mmol) in THF (5 mL) at -78°C was treated with n-BuLi (3.8 mL, 3.8 mmol; 1.5 N hexane solution). After 30 min, a solution of (4S)-4,5-isopropylidenedioxy-l-pentanal 7 (0.6 g, 3.8 mmol) in THF (5 mL) was added dropwise and stirred for lhour at the same temperature. The reaction mixture was warmed to room temperature, quenched with aqueous NH4C1 solution. Aqueous layer was separated and back extracted with EtOAc. Combined organic layers were dried (Na2SO4), evaporated and residue purified by column chromatography (Si-gel, EtOAc-hexane) to furnish (2S,5RS)-5-hydroxy-l,2- isopropylidenedioxy-9-p-methoxyphenyl methyl-non-6-yn-9-ol 35 (0.75 g) in 57% yield as a pale yellow syrup. [α]D +11.0 (c 1.0, CHC13); 'HNMR (CDC13, 200 MHz): δ 1.35, 1.4 (2s, 6H) 1.64-1.85 (m, 5H, H-3,4,-OH), 2.5 (dt, IH, J 2.1, 8.6 Hz, H-8), 3.45-3.58 (m, 3H, H-l, 9), 3.8 (s, 3H, -OCH3), 3.96-415 (m, 2H, H-la,2), 4..32-4.42 (m, IH, H-5), 4.46 (S,2H, H-10), 6.85, 7.22 (2d, 2H each, J 7.5 Hz, Ar-H); FABMS m/z (relative intensity): 348(12.3), 347(39.7), 289(261), 189(100); HRMS: Calculated for C20H27O5 (M+ -l): 347185849; Observed: 347185567.
Part 2: (2S,5RS)-5-Acetoxy-l,2-isopropylidenedioxy-9-p-methoxy phenyl methyl-non-6-yn-9-ol (Scheme XI; 36):
A solution of (2S,5RS)-5-hydroxy-l ,2-isopropylidenedioxy-9-p-methoxyphenyl methyl-non-6-yn-9-ol 35 (0.7 g, 2.01 mmol) and pyridine (0.7 mL, 8.04 mmol) in CH2C12 (10 mL) containing DMAP (catalytic) at 0°C was treated with Ac2O (0.23 mL, 2.4 mmol) and stirred at ambient temperature for 30 min. The reaction mixture was diluted with CH2C12 (50 mL), washed sequentially with CuSO4 solution (3 x 20 mL), NaHCO3 solution (20 mL), water (20 mL), brine (20 mL) and dried (Na2SO4). Evaporation of solvent and purification of residue by column chromatography (Si-gel, EtOAc-hexane) gave (2S,5RS)-5-acetoxy-l,2-isopropylidenedioxy-9-p-methoxy phenyl methyl-non-6- yn-9-ol 36 (0.684 g) in 87% yield as a pale yellow sOOOOOyrup. [α]D +9.5 (c 1.0, CHC13); 'HNMR (CDC13, 200 MHz): δ 1.36,1.42 (2s, 6H), 1.6-2.0 ( m, 4H, H-3,4), 2.1 (s, 3H, OAc), 2.45-2.56 (dt, IH, J 2.17, 8.2 Hz, H-8), 3.45-3.60 (m, 3H, H-l, 9), 3.82 (s, 3H, - OCH3), 3.96-414 (m, 2H, H-la,2), 4.48 (s, 2H, -OCH2), 5.32-5.45 ( m, IH, H-5), 6.86, 7.25 (2d, 2H each, J 7.6 Hz, Ar-H); FABMS m/z (relative intensity): 413(M+23, 4), 391(6), 337(19), 253(10), 143(100).
Part 3: (2S,5RS)-5-Acetoxy-l,2-dihydroxy-9-p-methoxy phenyl methyl-non-6-yn- 9-ol (Scheme XI; 37):
A mixture of (2S,5RS)-5-acetoxy-l,2-isopropylidenedioxy-9-p-methoxy phenyl methyl-non-6-yn-9-ol 36 (0.8 g) in 60% aqueous AcOH (8 mL) was stirred at room temperature for 12 h. The reaction mixture was neutralised with saturated NaHCO3 solution and extracted with EtOAc (3 x 50 mL). Organic layer was evaporated and residue was purified by filtration through a small pad of silica gel with 1 :1 EtOAc-hexane to afford (2S,5RS)-5-acetoxy-l,2-dihydroxy-9-p-methoxy phenyl methyl-non-6-yn-9-ol 37 (0.6 g) in 83.5% yield as a syrup. [α]D +6.5 (c 1.0, CHCI3); 'HNMR (CDC13, 200 MHz): δ 1.46-1.65 (m, 2H, H-4), 1.69-2.0 (m, 2H, H-3), 2.09 (s, 3H, OAc), 2.02-2.26 (br.s, 2H, -OH), 2.5 (t, 2H, H-8), 3.3-3.46 (m, IH, H-l), 3.46-3.78 ( m, 4H, H-1,2,9), 3.8 (s, IH, -OCH3), 4.45 (s, 2H, -OCH2), 5.3-5.42 (m, IH, H-5), 6.82, 7.2 (d, 4H, J 7.2 Hz, Ar-H); FABMS m/z (relative intensity): 335(M-15, 12.5), 215(5.5), 183(27.7), 154(64.3), 107(100). HRMS: Calculated for Cι8H23O6 (M+ -15): 335149464; Observed: 335149249. Part 4: (2S,5RS)-5-Acetoxy-2-hydroxy-9-p-methoxy phenyl methyl-2-p-toluene sulfonyloxy-non-6-yn-9-ol (Scheme XI; 38):
A solution of (2S,5RS)-5-acetoxy-l,2-dihydroxy-9-p-methoxy phenyl methyl- non-6-yn-9-ol 37 (0.6 g, 1.71 mmol) in CH2C12 (20 mL) containing pyridine (0.27 g, 3.42 mmol) was cooled to 0°C and treated with p-TsCl (0.327 g, 1.71 mmol) and stirred at room temperature for 4h. The reaction mixture was diluted with CH2C1 (15 mL), washed with water (2 x 20 mL), NaHCO3 solution (2 x 20 mL), water (20 mL), brine (20 mL), dried (Na2SO4) and evaporated. Purification ofthe residue obtained by column chromatography (Si-gel, 10% EtO AC -Hexane) gave (2S,5RS)-5-acetoxy-2-hydroxy-9-p- methoxy phenyl methyl-2-p-toluene sulfonyloxy-non-6-yn-9-ol 38 (0.8 g) in 92.5% yield as a yellow syrup. 'HNMR (CDC13, 200 MHz): δ 1.52-1.98 (m, 4H, H-3,4), 1.98 (s, 3H, OAc), 2.41-2.56 (m, 5H, H-8, Ar-CH3), 3.34-3.72 m, 2H, H-9), 3.8 (s, 3H, -OCH3), 3.94- 4.2 (m, 2H, H-l, 2), 4.45 (s, 2H, -OCH2), 4.56-4.72 (m, IH, H-5), 5.26-5.42 (m, IH, H- la), 6.82, 7.21 (2d, 2H each, J 7.9 Hz, Ar-H), 7.26-7.3 (m, 2H, Ar-H), 7.62-7.82 (m, 2H, Ar-H).
Part 5: (2S,5RS)-2-(Hydroxymethyl)-5-(l-p-methoxyphenylmethyleneoxy-but-3- yn-4-yl)-tetrahydrofuran (Scheme XI; 39):
To a solution of (2S,5RS)-5-acetoxy-2-hydroxy-9-p-methoxy phenyl methyl-2-p- toluene sulfonyloxy-non-6-yn-9-ol 38 (0.8 g, 1.58 mmol) in MeOH (15 mL) at room temperature, K2CO3 (0.482g, 3.49 mmol) was added and stirred for 4 h. It was treated with NH4C1 solution, evaporated MeOH and residue extracted with EtOAc (3x 30 mL). Organic layer were washed with water (2 x 30 mL), brine (20 mL), dried (Na2SO4) and evaporated. The residue obtained was purified by column chromatography (Si-gel, 15% EtOAc-hexane) to afford (2S,5RS)-2-(hydroxymethyl)-5-(l-p-methoxyphenylmethylen oxy-but-3-yn-4-yl)-tetrahydrofuran 39 (0.45 g) in 97.8% yield as a yellow syrup. 'HNMR (CDC13, 200 MHz): δ 1.85-212 (m, 4H, H-3,4), 2.4-2.54 (m, 2H, H-8), 3.39- 3.59 (m, 2H, H-9), 3.68 (dd, 2H, J 2.38, 11.9Hz, H-l), 3.8 (s, 3H, -OCH3), 41-4.26 (m, 1H, H-2), 4.44 (s, 2H, -OCH2), 4.58-4.70 (m, IH, H-5), 6.81, 7.2 (2d, 2H each, J 7.6Hz, Ar-H).
Part 6: (2S,5RS)-5-(l-p-Methoxyphenylmethylenoxy-but-3-yn-4-yl)-2-(p- toluenesulfonyloxymethyl)-tetrahydrofuran (Scheme XI; 40):
A solution of (2S,5RS)-2-(hydroxymethyl)-5-(l-p-methoxyphenylmethylenoxy- but-3-yn-4-yl)-tetrahydrofuran 39 (0.45 g, 1.55 mmol), pyridine (0.25 mL, 3.1 mmol) in CH2C1 (10 mL) was treated with p-TsCl (0.325 g, 1.70 mmol) and stirred at room temperature for 4h. Reaction mixture was diluted with CH2C12 (20 mL) and washed sequentially with CuSO4 solution (3 x 20 mL), NaHCO3 solution (20 mL), water (20 mL), brine (20 mL) and dried (Na2SO4). Evaporation of solvent and purification of residue by column chromatography (Si-gel, 10% EtOAc-hexane) gave (2S,5RS)-5-(l-p- methoxyphenylmethylenoxy-but-3-yn-4-yl)-2-(p-toluenesulfonyloxymethyl)-tetrahydro furan 40 (0.45 g) in 65.3% yield as a yellow syrup. 'HNMR (CDC13, 200 MHz): δ 1.74- 2.21 (m, 4H, H-3,4), 2.39-2.49 (m, 2H, H-8), 2.5 (s, 3H, Ar-CH3), 3.5 (t, 2H, J 8.1Hz, H- 9), 4.44 (s, 2H, -OCH2), 4.5-4.6 (m, IH, H-2), 6.82 (d, 2H, J 7.9 Hz, Ar-H), 716-7.38 (m, 4H, Ar-H), 7.79 (d, 2H, J 7.9Hz, Ar-H).
Part 7: (2S,5RS)-2-(p-Fluorophenoxymethyl)-5-(l-ρ- methoxyphenylmethylenoxy-but-3-yn-4-yl)-tetrahydrofuran (Scheme XI; 41):
To a stirred suspension of NaH (25.9 g, 1.08 mmol) in DMF (3 mL), a solution of (2S,5RS)-5-(l-p-methoxyphenylmethylenoxy-but-3-yn-4-yl)-2-(p- toluenesulfonyloxymethyl)-tetrahydrofuran 40 (0.4 g, 0.9 mmol) in DMF (3 mL) was added, followed by the addition of 4-fluoro phenol (0.121 g, 1.08 mmol) in DMF (2 mL) and heated at 80°C for 2h. The reaction mixture was cooled to room temperature and treated with NH4C1 solution (5 mL). It was extracted with ether (3 x 15 mL) and organic layer was washed with water (3 x 15 mL), brine (15 mL) and dried (Na SO4). Evaporation of solvent and purification of residue by column chromatography (Si-gel, 6% EtOAc-hexane) afforded (2S,5RS)-2-(p-fluorophenoxymethyl)-5-(l-p- methoxyphenylmethylenoxy-but-3-yn-4-yl)-tetrahydrofuran 41 (0.325 g) in 93.9% yield as a colorless syrup. 1HNMR (CDC13, 200 MHz): δ 1.82-2.29 (m, 4H, H-3,4), 2.42-2.58 (m, 2H, H-8), 3.45-3.59 (m, 2H, H-9), 3.81 (s, 3H, -OCH3), 3.85-3.96 (m, 2H, H-l), 4.0- 414 (m, IH, H-2), 4.66-4.76 (m, IH, H-5), 6.76-7.0 (m, 6H, Ar-H), 718-7.3 (m, 2H, Ar- H); FABMS m/z (relative intensity): 384(18), 383(69), 369(10), 313(20), 121(100); HRMS: Calculated for C23H24O4F (M+ -l): 383165863; Observed: 383164866.
Part 8: (2S,5RS)-2-(ρ-Fluorophenoxymethyl)-5-(l-hydroxy-but-3-yn-4-yl)- tetrahydrofuran (Scheme XI; 42): A mixture of (2S,5RS)-2-(p-fluorophenoxy methyl)-5-(l-p- methoxyphenylmethylenoxy-but-3-yn-4-yl)-tetrahydrofuran 41 (0.3 g, 0.78 mmol) and DDQ (0.212 g, 0.937 mmol) in aqueous CH2C12 (20 mL; 1:19) was stirred at room temperature for 5h. The reaction mixture was treated with saturated NaHCO3 solution (10 mL) and diluted with CH2C1 (40 mL). Organic layer was separated and washed with water (3 x 30 mL), brine (20 mL) ansd dried (Na2SO4). Evaporation of solvent afforded (2S,5RS)-2-(p-fluorophenoxymethyl)-5-(l-hydroxy-but-3-yn-4-yl)-tetrahydrofuran 42 (0.140 g) in 67.9% yield as a liquid, whose spectral data was comparable with the reported (Org Process & Development, 3, 73-76, 1999) values. Η NMR (CDCI3, 200 MHz): δ 1.8-2.34 (m, 5H, H-6.7, -OH), 2.4-2.52 (m, 2H, H-2), 3.6-3.75 (m, 2H, H-l), 3.84-4.0 (m, 2H, H-9), 416-4.5 (m, IH, H-5), 4.5-4.75 (m, IH, H-8), 6.72-7.0(m, 4H, Aril).
Example 11 : Asymmetric reduction
References in this Example 11 to compound numerals (generally underlined) designate the compounds depicted structurally in Scheme XII above.
Part 1: (4S)-4,5-Isopropylidenedioxy-l-pentanoic acid (Scheme XII; 57): A solution of ethyl (4S)-4,5-isopropylidenedioxy-l-pentanoate (5; 5 g, 24.7 mmol) in DME (30 mL) was cooled to 0°C and treated with a solution of LiOH (60 mL; 1N solution in water) and allowed to stir at room temperature for 3 hours. The reaction mixture was quenched with AcOH (till neutral to pH) and extracted with EtOAc (3 x 75 mL). Organic layer was dried over (Na2SO4) and evaporation of solvent gave (4S)-4,5- isopropylidenedioxy-1-pentanoic acid 57 (4.1 g) in 95% yield as a colorless liquid. 'HNMR (CDC13, 200 MHz): δ 1.3, 1.35 (2s, 6H, CH3), 1.8-1.95 (m, 2H, H-3), 2.4-2.6 (m, 2H, H-2), 3.55 (t, IH, J 6.1 Hz, H-5), 3.8- 4.2 (m, 2H, H-4,5a).
Part 2: 1 ,2-Isopropylidenedioxy-9-p-methoxy phenyl methyl~5-oxo-non-6-yn-9- ol (Scheme XII; 59): A. Preparation of mixed anhydride (Scheme XII; 58): A stirred and cooled (-
10°C to 0°C) solution of (4S)-4,5-isopropylidenedioxy-l-pentanoic acid 57 (lg, 5.74 mmol) and freshly distilled Et3N (0.58 g, 5.7 mmol) in dry ether (5 mL), was treated with ethyl chloro formate (0.62 g, 5.7 mmol). The reaction mixture was allowed to reach room temperature and stirred for 3 h. The reaction mixture was filtered and washed with ether. Organic layer was washed with saturated NaHCO solution (25 mL), water (25 mL), brine (20 mL) and dried (Na2SO4). Evaporation of solvent under vacuum at room temperature afforded mixed anhydride 58 (lg) in 71% yield as a colorless syrup.
B. l,2-Isopropylidenedioxy-9-p-methoxy phenyl methyl~5-oxo-non-6-yn-9-ol (Scheme XII; 59): A stirred solution of 1-p-methoxy phenyl methyl-but-3-yne-l-ol (0.5g, 2.03 mmol) in dry THF (5 mL) was cooled to -78°C and treated with n-BuLi (1.35 mL, 2.03 mmol; 1.5 N hexane solution) dropwise. After 30 min., a solution of anhydride 58 (0.5 g, 2.03 mmol) in THF (5 mL) was added and stirred at the same temperature for 2hours. The reaction mixture was quenched with aqueous NH4C1 solution (10 mL) and extracted with EtOAc (2 x 25 mL). Organic layer was washed with brine (25 mL), dried (Na2SO4), evaporated and purified the residue by column chromatography (Si-gel, 8:1 Hexane-EtOAc) to afford l,2-isopropylidenedioxy-9-p-methoxy phenyl methyl— 5-oxo- non-6-yn-9-ol 59 (0.095 g) in 13.5% yield as a colorless syrup. 1HNMR (CDCI3, 200 MHz): δ 1.32, 1.4 (2s, 6H, CH3), 1.75-2.2 (m, 2H, H-3), 2.6-2.8 (m, 4H, H-4,8), 3.45-3.7 (m, 3H, H-l, 9), 3.8 (s, 3H, -OMe), 3.8-415 (m, 2H, H-la,2), 4.5 (s, 2H, -OCH2), 6.85, 7.25 (2d, 2H each, J 9.3 Hz, Ar-H).
Part 3: (2S,5RS)-5-Hydroxy- 1 ,2-isopropylidenedioxy-9-p-methoxyphenyl methyl-non-6-yn-9-ol (Scheme XII; 60): A solution of 9-BBN (0.087 g, 0.72 mmol) in dry THF (5 mL) under N atmosphere was treated with -pinene (0.118 g, 0.86 mmol; technical grade) and heated at reflux for 2.5 h. The reaction mixture was cooled to room temperature and a solution of l,2-isopropylidenedioxy-9-p-methoxy phenyl methyl— 5- oxo-non-6-yn-9-ol 59 (0.25 g, 0.712 mmol) in THF (5 mL) was added and allowed to stir for 82 h. The reaction mixture was treated withpropanaldehyde (0.5 mL), stirred for 15 min. and solvent was evaporated at room temperature. The residue was dissolved in dry ether (10 mL), cooled to 0°C and treated with ethanolamine (0.097 g, 1.5 mmol) and stirred for 15 min. It was filtered and washed with ether (5 mL). The ethereal layer was washed with brine (20 mL), dried (Na SO ) and evaporated. The residue obtained was purified by column chromatography (Si-gel, 1:4 EtOAc-hexane) to afford (2S,5RS)-5- hydroxy-l,2-isopropylidenedioxy-9-p-methoxyphenyl methyl-non-6-yn-9-ol 60 (0.098 g) in 40% yield as a colorless syrup. Compound 60 prepared by the present approach is identical to compound 35 prepared in Scheme IX by TLC analysis as well as 'HNMR data.
Example 12: Keto-epoxide Cyclisation
References in this Example 12 to compound numerals (generally underlined) designate the compounds depicted structurally in Scheme XIII above. Part 1: Non-8-ene-l-p-methoxy phenyl methyl— 5-oxo-3-yn-l-ol (Scheme XIII;
54):
A. Mixed anhydride (Scheme XIII; 53): A stirred and cooled (-10°C to 0°C) solution of pent-4-enoic acid (0.5 g, 5 mmol) and freshly distilled Et3N (0.505 g, 5 mmol) in dry ether (5 mL), was treated with ethyl chloro formate (0.542 g, 5 mmol). The reaction mixture was allowed to reach room temperature and stirred for 3 h. The reaction mixture was filtered and washed with ether. Organic layer was washed with saturated NaHCO3 solution (25 mL), water (25 mL), brine (20 mL) and dried (Na2SO4). Evaporation of solvent under vacuum at room temperature afforded mixed anhydride 53 (0.79 g) in 91.8% yield as a colorless syrup.
B. Non-8-ene-l-p-methoxy phenyl methyl-5-oxo-3-yn-l-ol (Scheme XIII; 54): A stirred solution of 1-p-methoxy phenyl methyl-but-3-yn-l-ol (52: 1.12 g, 5.91 mmol) in dry THF (5 mL) was cooled to -78°C and treated with n-BuLi (4 mL, 5.91 mmol; 1.5 N hexane solution) dropwise. After 30 min., a solution of anhydride 53 (0.78 g, 4.54 mmol) in THF (5 mL) was added and stirred at the same temperature for 2hours. The reaction mixture was quenched with aq. NH4C1 solution (10 mL) and extracted with EtOAc (2 x 25 mL). Organic layer was washed with brine (25 mL), dried (Na2SO ), evaporated and purified the residue by column chromatography (Si-gel, 8:1 Hexane-EtOAc) to afford non-8-ene- 1-p-methoxy phenyl methyl— 5-oxo-3-yn-l-ol (54; 0.35 g) in 27% yield as a colorless syrup. 1HNMR (CDC13, 200 MHz): δ 2.32-2.46 (m, 2H, H-7), 2.56-2.69 (m, 4H, H-6,2), 3.59 (t, 2H, J 8.37 Hz, H-l), 3.8 (s, 3H, -OMe), 4.47 (s, 2H, -OCH2), 4.95-511 (m, 2H, H-9), 5.67-5.9 (m, IH, H-8), 6.84, 7.22 (2d, 2H each, J 9.3 Hz, Ar-H).
Part 2: l,2-Epoxy-9-p-methoxy phenyl methyl-5-oxo-non-6-yn-9-ol (Scheme XIII; 55): A solution of non-8-ene- 1-p-methoxy phenyl methyl— 5-oxo-3-yn-l-ol 54 (0.2 g,
0.73 mmol) in acetone (5 mL) was sequentially treated with solid NaHCO3 (0.306 g, 3.65 mmol), water (5 mL) followed by a solution of oxone (0.448 g, 073 mmol) in aqueous. 4 x 10"4 M EDTA disodium solution (10 mL) dropwise at 0°C and stirred at room temperature for 4h. The reaction mixture was filtered and washed with EtOAc (10 mL). The aqueous layer was extracted with EtOAc (2 x 10 mL) and combined organic layers were washed with brine (20 mL) and dried (Na2SO4). Evaporation of solvent and purification of residue by column chromatography (Si-gel, 15% EtOAc in hexane) gave l,2-epoxy-9-p-methoxy phenyl methyl— 5-oxo-non-6-yn-9-ol 55 (0.1 g) in 48% yield as a colorless syrup. 'HNMR (CDC13, 200 MHz): δ 1.62-1.82 (m, IH, H-3), 1.9-2.1 (m, IH, H-3'),2.41-2.57 (m, IH, H-l), 2.57-2.74 (m, 5H, H-l',4,8), 2.85-2.96 (m, IH, H-2), 3.58 (t, 2H, J 8.13 Hz, H-9), 3.8 (s, 3H, -OMe), 4.45 (s, 2H, -OCH2), 6.84, 7.22 (2d, 2H each, J 9.3 Hz, Ar-H).
Part 3: (2S,5RS)-2-(Hydroxymethyl)-5-(l -p-methoxyphenylmethylenoxy-but-3- yn-4-yl)-tetrahydrofuran (Scheme XIII; 56):
To a stirred and cooled -78°C solution of l,2-epoxy-9-p-methoxy phenyl methyl— 5-oxo-non-6-yn-9-ol 55 (0.075 g. 0.26 mmol) in CH2C12 (52 mL; 0.005M solution), a solution of BH3-DMS (0.25 mL, 0.26 mmol; 1 M solution in CH2C12) was added dropwise. After 3 hours, the reaction mixture was quenched with aq. NH4CI solution (lOmL) at 0°C and extracted with EtOAc (2 xlO mL). Organic layer was washed with water (2 x 10 mL), brine (10 mL) and dried (Na2SO4). Evaporation of solvent and purification of residue by column chromatography (Si-gel, 25% EtOAc in hexane) gave racemic 2-(Hydroxymethyl)-5-(l-p-methoxyphenylmethylenoxy-but-3-yn-4-yl)- tetrahydrofuran 56 (0.025 g) in 34% yield as a colorless syrup. The compound 56 thus obtained by this approach is comparable to compound 39 (Scheme IX) by TLC analysis as well as 'HNMR data.
Example 13 : 5-methyl substitution of γ-butyrolactone
Sodium hydride (60%, 192g) is taken in a 3 -neck round-bottom flask fitted with a mechanical stirrer and addition funnel. Dry n-hexane (1.0 L) is added and the mixture stirred for 15 minutes. The stirring is then stopped and the sodium hydride allowed to settle. After 15 minutes, n-hexane is decanted out. Dimethyl formamide (DMF) (2.0L) is charged to the flask. 4-fluorophenol (500 g) dissolved in DMF (2.0L) is added to the mixture over 0.5 hours, maintaining the temperature between 10-15°C. Tetrabutylammonium iodide (20 g) is added to the mixture maintaining temperature at 10-15°C. 5-hydroxymethyl- γ-butyrolactone (compound 60 in Scheme XIV where R is hydrogen) dissolved in DMF (3.0L) is added slowly over 1.5 hours while maintaining the temperature between 10-15°C. The reaction mixture is heated at 60-65°C and temperature maintained for 1.0 hours. Thin layer chromatography can be checked to ensure disappearance of starting material. The reaction mixture is quenched with saturated NH4C1 solution (500 ml), followed by acidification with 18% HCI (500 ml) and then stirred for 0.5 hours at room temperature. Water (12.0L) is added to the reaction mixture and the product (compound 6_1 in Scheme XIV above where R' is para- fluorophenyl) extracted into ethyl acetate (5.0L). The ethyl acetate layer is separated and washed with brine solution (2x2.0L) and then dried over sodium sulfate (250 g) and concentrated under reduced pressure. The resulting crude product can be chromatographed over silica gel using 10% ethyl acetate:n-hexane as eluent.
The invention has been described in detail including preferred embodiments thereof. However, it will be understood that those skilled in the art, upon consideration of this disclosure, may make modifications and improvements thereon without departing from the spirit and scope ofthe invention as set forth in the following claims.

Claims

What is claimed is:
1. A method for preparing a 4-fluorophenoxymethyl-(hydroxy)- tetrahydrofuran, comprising: a) reacting 4-fluorophenol and an epoxy compound to form an epoxy-phenyl ether; b) reacting the epoxy-phenyl ether with an active methylene compound to form a lactone having five ring members; and c) reducing the lactone to provide the 4-fluorophenoxymethyl-(hydroxy)- tetrahydrofuran.
2. The method of claim 1 wherein the epoxy compound is a glycidyl compound substituted with an electron- withdrawing group.
3. The method of claim 1 wherein the epoxy compound is an epihalohydrin or a glycidyl sulfonyl ester compound.
4. The method of claim 1 wherein the epoxy compound is optically active.
5. The method of claim 1 wherein the epoxy compound is racemic.
6. The method of claim 1 or 5 wherein the 4-fluorophenol and the epoxide are reacted in the presence of an optically active compound.
7. The method of claim 1 wherein the epoxide is racemic and 4-fluorophenol and epoxide are reacted in the presence of an optically active compound to form an optically active epoxy-phenyl ether.
8. The method of claim 1 wherein the active methylene compound is a diester or a half-ester thereof.
9. The method of claim 1 wherein the active methylene compound is a dialkyl malonate.
10. The method of claim 1 further comprising activating the hydroxy group of the hydroxy tetrahydrofuran and substituting the activated tetrahydrofuran position.
11. The method of claim 10 wherein the tetrahydrofuran position is substituted with a 1 -alkynyl compound.
12. The method of claim 10 wherein the substitution provides a 4- fluorophenoxymethyl-( 1 -butynyl)tetrahydrofuran.
13. The method of claim 10 wherein a 2-(4-fluorophenoxymethyl)-5-(4-N- hydroxyureidyl-l-butynyl)-tetrahydrofuran is provided.
14. The method of claim 10 wherein 2S,5S-trans-2-(4-fluorophenoxymethyl)- 5-(4-N-hydroxyureidyl-l-butynyl)-tetrahydrofuran is provided.
15. The method of claim 10 or 11 wherein the substitution produces an enantiomeric excess of a stereoisomer.
16. The method of claim 15 wherein the substitution produces a stereoisomer that is present in at least about 60 percent relative to the other stereoisomer.
17. The method of claim 15 wherein the substitution produces a stereoisomer that is present in at least about 70 percent relative to the other stereoisomer.
18. The method of claim 15 wherein the substitution produces a trans stereoisomer that is present in at least about 60 percent relative to the cis stereoisomer.
19. The method of claim 15 wherein the substitution produces a trans stereoisomer that is present in at least about 70 percent relative to the cis stereoisomer.
20. The method of claim 15 wherein the substitution produces a cis steoroisomer that is present in at least about 60 percent relative to the trans steroisomer.
21. The method of claim 15 wherein the substitution produces a cis steoroisomer that is present in at least about 70 percent relative to the trans steroisomer.
22. A method for preparing a substituted ╬│-butyrolactone, comprising: a) reacting mannitol with an aldehyde compound to form a trialkylene mannitol; b) hydrolyzing the trialkylene mannitol to provide a 2,5-O-alkylene- mannitol; and c) functionalizing secondary hydroxy groups ofthe 2,5-O-alkylene-mannitol to provide a fused ring cyclic ether comprising a first cyclic ether fused to a second cyclic ether; d) reacting the fused ring cyclic ether with 4-fluorophenol to form a bis-4- fluorophenylether; and e) cleaving the bis-4-fluorophenylether to form a substituted ╬│- butyrolactone.
23. The method of claim 22 wherein primary hydroxy-substituted carbons of the fused ring cyclic ether are activated prior to reaction with 4-fluorophenol.
24. The method of claim 22 wherein the fused ring cyclic ether is cleaved to an acyclic ether prior to forming the substituted ╬│-butyrolactone.
25. The method of claim 24 wherein the acyclic ether is ofthe following formula:
Figure imgf000084_0001
wherein each X is the same or different ╬▒, ╬▓-unsaturated electron-withdrawing group.
26. The method of claim 22 wherein an enantiomeric excess of a stereoisomer ofthe ╬│-butyrolactone moiety is formed.
27. The method of claim 22 wherein cleavage ofthe bis-4-fluorophenylether produces two molar equivalents ofthe substituted ╬│-butyrolactone.
28. A method for preparing 4-fluorophenoxymethyl-(alkynyl)- tetrahydrofuran, comprising: treating with base a compound comprising a substituted alkyl group to form a 4- fluorophenoxymethyl-(alkynyl)-tetrahydrofuran, wherein the substituted alkyl group has 6 or more carbon atoms, the 2,3-positions of alkyl group forming an epoxide ring, the 1 -position ofthe alkyl group substituted with a first leaving group, and the 6-position ofthe alkyl group substituted with a second leaving group.
29. The method of claim 28 wherein the substituted alkyl compound is treated with a molar excess of base.
30. The method of claim 28 wherein the substituted alkyl compound is treated with about a three molar excess of base.
31. The method of claim 28 wherein the base is an alkyllithium reagent, an amide salt or a hydride.
32. The method of claim 28 wherein the first and second leaving groups are each independently a halogen, a sulfonic alkyl ester, a sulfonic aryl ester or a sulfonic arylalkyl ester.
33. The method of claim 28 wherein one or both ofthe epoxide carbons are optically active.
34. The method of claim 28 wherein the formed 4-fluorophenoxymethyl- (alkynyl)-tetrahydrofuran is optically active.
35. The method of claim 28 wherein both ofthe epoxide carbons are optically active.
36. The method of claim 28 wherein the two carbons adjacent to the ring oxygen ofthe formed 4-fluorophenoxymethyl-(alkynyl)-tetrahydrofuran are each optically active.
37. The method of claim 28 wherein the 4-fluorophenoxymethyl-(alkynyl)- tetrahydrofuran is formed from the substituted alkyl compound without isolation of intermediate compounds.
38. The method of claim 28 wherein the 4-fluorophenoxymethyl-(alkynyl)- tetrahydrofuran is formed from the substituted alkyl compound in a single reaction step.
39. The method of claim 28 wherein the tetrahydrofuran is ofthe following structure:
Figure imgf000086_0001
40. A method of preparing an alkynyl-tetrahydrofuran, comprising: subjecting a keto-substituted dioxolane compound to at least one Wittig-type reaction; forming an epoxide moiety from a carbon-carbon double bond produced by the Wittig-type reaction; ring-opening the dioxolane group to form an acylic compound and cyclizing the acyclic compound to provide an alkynyl-substituted tetrahydrofuran.
41. The method of claim 40 wherein the tetrahydrofuran is optically active.
42. The method of claim 40 wherein the epoxide undergoes an elimination reaction to form a propargyl alcohol substituent ofthe dioxalone group.
43. A method of preparing an alkynyl-tetrahydrofuran, comprising: a) subjecting a keto-substituted dioxolane compound to a Wittig-type reaction; b) reacting a carbonyl group of the reaction product of step a) with a compound having a terminal alkynyl group; and c) reducing the carbonyl group ofthe reaction product of step b).
44. The method of claim 43 wherein the reduction provides a propargyl alcohol.
45. The method of claim 43 wherein the carbonyl group is reduced asymmetrically.
46. The method of claim 43 wherein after step c), the dioxolane group is opened to an acyclic compound, and the acyclic compound is cyclized to an alkynyl- substituted tetrahydrofuran.
47. A method for preparing an alkynyl-tetrahydrofuran, comprising: reacting a compound having a terminal alkynyl moiety with an unsaturated anhydride compound to form a keto alkynyl compound having a terminal alkene group; epoxidizing the alkene group ofthe compound and then cyclizing the compound to provide an alkynyl-substituted tetrahydrofuran.
48. The method of claim 47 wherein the compound is cyclized in the presence of a reducing agent.
49. The method of claim 48 wherein the reducing agent is a borane reagent.
50. A method for preparing an aryl-substituted ╬│-butyrolactone, comprising: reacting a ╬│-butyrolactone compound having a substituted 5-methyl moiety with an aryl nucleophile in the presence of a hydride reagent to yield a ╬│-butyrolactone having a 5-aryloxymethylene moiety.
51. The method of claim 50 wherein the aryl nucleophile is an arylhydroxy compound.
52. The method of claim 50 wherein the aryl nucleophile is a phenol.
53. The method of claim 50 wherein the aryl nucleophile is a 4-fluorophenol.
PCT/US1999/015107 1998-07-03 1999-07-02 Methods for synthesis of substituted tetrahydrofuran compound WO2000001683A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
KR1020017000008A KR20010083083A (en) 1998-07-03 1999-07-02 Methods for synthesis of substituted tetrahydrofuran compound
MXPA01000234A MXPA01000234A (en) 1998-07-03 1999-07-02 Methods for synthesis of substituted tetrahydrofuran compound.
AU47311/99A AU750592B2 (en) 1998-07-03 1999-07-02 Methods for synthesis of substituted tetrahydrofuran compound
CA002342156A CA2342156A1 (en) 1998-07-03 1999-07-02 Methods for synthesis of substituted tetrahydrofuran compound
JP2000558088A JP2002519415A (en) 1998-07-03 1999-07-02 Method for synthesizing substituted tetrahydrofuran compounds
EP99930868A EP1102759A1 (en) 1998-07-03 1999-07-02 Methods for synthesis of substituted tetrahydrofuran compound

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US9170998P 1998-07-03 1998-07-03
US60/091,709 1998-07-03

Publications (2)

Publication Number Publication Date
WO2000001683A1 true WO2000001683A1 (en) 2000-01-13
WO2000001683A8 WO2000001683A8 (en) 2001-02-22

Family

ID=22229262

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1999/015107 WO2000001683A1 (en) 1998-07-03 1999-07-02 Methods for synthesis of substituted tetrahydrofuran compound

Country Status (9)

Country Link
US (2) US6310221B1 (en)
EP (1) EP1102759A1 (en)
JP (1) JP2002519415A (en)
KR (1) KR20010083083A (en)
CN (1) CN1313857A (en)
AU (1) AU750592B2 (en)
CA (1) CA2342156A1 (en)
MX (1) MXPA01000234A (en)
WO (1) WO2000001683A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050107838A1 (en) * 2003-09-18 2005-05-19 Lovett Eric G. Subcutaneous cardiac rhythm management with disordered breathing detection and treatment
JP5712510B2 (en) * 2010-06-18 2015-05-07 Jnc株式会社 Compound having 5-membered ring, liquid crystal composition, and liquid crystal display device

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5750565A (en) * 1995-05-25 1998-05-12 Cytomed, Inc. Compounds and methods for the treatment of cardiovascular, inflammatory and immune disorders

Family Cites Families (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5543531A (en) 1987-06-16 1996-08-06 Sandoz Ltd. Thiophen compounds and their preparation
CA1334975C (en) 1987-11-13 1995-03-28 James H. Holms Furan and pyrrole containing lipoxygenase inhibiting compounds
US5175183A (en) 1989-02-01 1992-12-29 Abbott Laboratories Lipoxygenase inhibiting compounds
US5037853A (en) 1989-12-28 1991-08-06 Abbott Laboratories Cyclopropyl derivative lipoxygenase inhibitors
ATE205084T1 (en) 1991-03-04 2001-09-15 Ct For Innovative Technology 2,4-DIARYL-1,3-DITHIOLANES AS A PLATELET ACTIVATING FACTOR RECEPTOR BLOCKER AND 5-LIPOXYGENASE INHIBITOR
US5183818A (en) 1991-08-27 1993-02-02 Abbott Laboratories Arylalkylether and arylalkylthioether inhibitors of lipoxygenase enzyme activity
US5169854A (en) 1992-02-26 1992-12-08 Abbott Laboratories N-substituted-furylalkenyl hydroxamic acid and N-hydroxyurea compounds having lipoxygenase inhibitory activity
US5530141A (en) 1992-03-04 1996-06-25 Center For Innovative Technology 2,4-diaryl-1,3-dithiolanes; 2,4-diaryl-1,3-dioxolanes; 2,4-diaryl-1,3-oxathiolanes; and 2,5-diaryl-1,3-oxathiolanes for the treatment of disorders mediated by platelet activating factor or products of 5-lipoxygenase
US5639782A (en) 1992-03-04 1997-06-17 Center For Innovative Technology Neolignan derivatives as platelet activating factor receptor antagonists and 5-lipoxygenase inhibitors
US5187192A (en) 1992-03-13 1993-02-16 Abbott Laboratories Cyclobutyl derivatives having lipoxygenase inhibitory activity
US5326787A (en) 1992-05-12 1994-07-05 Abbott Laboratories Cycloalkyl N-hydroxy derivatives having lipoxygenase inhibitory activity
JPH08502243A (en) 1992-07-13 1996-03-12 サイトメッド,インコーポレイテッド 2,5-Diaryltetrahydro-thiophenes, -furans and analogues for the treatment of inflammatory and immune disorders
US5358938A (en) 1992-07-13 1994-10-25 Cytomed, Inc. Compounds and methods for the treatment of disorders mediated by platelet activating factor or products of 5-lipoxygenase
US5463083A (en) 1992-07-13 1995-10-31 Cytomed, Inc. Compounds and methods for the treatment of cardiovascular, inflammatory and immune disorders
US5434151A (en) 1992-08-24 1995-07-18 Cytomed, Inc. Compounds and methods for the treatment of disorders mediated by platelet activating factor or products of 5-lipoxygenase
US5288751A (en) 1992-11-06 1994-02-22 Abbott Laboratories [(Substituted) phenyalkyl]furylalkynyl-and [substituted) phenyalkyl] thienylalkynyl-N-hydroxyurea inhibitors or leukotriene biosynthesis
US5703093A (en) 1995-05-31 1997-12-30 Cytomed, Inc. Compounds and methods for the treatment of cardiovascular, inflammatory and immune disorders
US5792776A (en) 1994-06-27 1998-08-11 Cytomed, Inc., Compounds and methods for the treatment of cardiovascular, inflammatory and immune disorders
PL317873A1 (en) 1994-06-27 1997-04-28 Cytomed Inc Compounds for and method of treating cardiovascular, inflammatory and immunological disorders
JP3777408B2 (en) 1994-12-28 2006-05-24 株式会社カネカ Method for producing carboxylic acid derivative

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5750565A (en) * 1995-05-25 1998-05-12 Cytomed, Inc. Compounds and methods for the treatment of cardiovascular, inflammatory and immune disorders

Also Published As

Publication number Publication date
CA2342156A1 (en) 2000-01-13
CN1313857A (en) 2001-09-19
AU4731199A (en) 2000-01-24
MXPA01000234A (en) 2003-02-10
AU750592B2 (en) 2002-07-25
KR20010083083A (en) 2001-08-31
US20020035278A1 (en) 2002-03-21
US6541647B2 (en) 2003-04-01
EP1102759A1 (en) 2001-05-30
WO2000001683A8 (en) 2001-02-22
JP2002519415A (en) 2002-07-02
US6310221B1 (en) 2001-10-30

Similar Documents

Publication Publication Date Title
Tietze et al. Stereodivergent Hetero‐Diels‐Alder Reactions of Chiral 1‐Oxa‐1, 3‐butadienes through a Conformational Switch Induced by Lewis Acids
Reitz et al. Stereoselectivity of electrophile-promoted cyclizations of. gamma.-hydroxyalkenes. An investigation of carbohydrate-derived and model substrates
JP5735277B2 (en) Intermediates and methods for the synthesis of halichondrin B analogues
KR100894673B1 (en) Method for the preparation of hexahydro-furo[2,3-B]furan-2-ol
EP2089371B1 (en) Methods for the preparation of hexahydrofuro[2,3-b]furan-3-ol
Hashimoto et al. Total synthesis of the meso-triterpene polyether teurilene
Paolucci et al. Dihydro-and Tetrahydrofuran Building Blocks from 1, 4: 3, 6-Dianhydrohexitols. 2. Synthesis of Acetal, Alcohol, Diol, Epoxide, Hydrocarbon, and Lactone Pheromones
Srinivas et al. Stereoselective total synthesis of (+)-varitriol
US6310221B1 (en) Methods for synthesis of substituted tetrahydrofuran compound
US6433197B1 (en) Substituted oxygen alicyclic compounds, including methods for synthesis thereof
US20040063968A1 (en) Process for use in 11-oxa prostaglandin synthesis
Miyaoka et al. Total synthesis of constanolactone E, a marine eicosanoid from the alga constantinea simplex; Absolute structure of constanolactone E
EP0159528A1 (en) Tetrahydrofuranyl substituted prostaglandin analogs
CA2045863A1 (en) Process of making 2,5-diaryl tetrahydrofurans and analogs thereof useful as paf antagonists
DK147070B (en) ANALOGY PROCEDURE FOR THE PREPARATION OF 13,14-DEHYDRO-11-DEOXY PROSTAGLANDINES
Giacomini et al. Synthesis of ambergris odorant ent-ambrox
Mootoo et al. Synthesis of Carbohydrate Analogues of the THF-Acetogenin 4-Deoxyannomontacin and Their Cytotoxicity Against Human Prostate Cancer Cell Lines
US7501535B2 (en) Chemical synthons and intermediates
EP1115702A1 (en) Substituted nitrogen and sulfur alicyclic compounds, including methods for synthesis thereof
US5099033A (en) Process of making 2,5-diaryl tetrahydrofurans and analogs thereof useful as PAF antagonists
JPS6323181B2 (en)
Li Achmatowicz Rearrangement in Total Syntheses of Diarylheptanoid Natural Products
김태우 Enantioselective Total Synthesis of (-)-Galiellalactone and Biological Evaluation of Novel Galiellalactone-Based Analogues as STAT3 inhibitors
JPH0651694B2 (en) Optically active compound
Reddy et al. A novel, concise and highly efficient stereospecific synthesis of deacetylanisomycin

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 99809837.X

Country of ref document: CN

AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG UZ VN YU ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SL SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
ENP Entry into the national phase

Ref document number: 2342156

Country of ref document: CA

Ref document number: 2342156

Country of ref document: CA

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 1020017000008

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 1200100007

Country of ref document: VN

ENP Entry into the national phase

Ref document number: 2000 558088

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: PA/a/2001/000234

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 47311/99

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 1999930868

Country of ref document: EP

AK Designated states

Kind code of ref document: C1

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG UZ VN YU ZW

AL Designated countries for regional patents

Kind code of ref document: C1

Designated state(s): GH GM KE LS MW SD SL SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

CFP Corrected version of a pamphlet front page
CR1 Correction of entry in section i
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 1999930868

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1020017000008

Country of ref document: KR

WWG Wipo information: grant in national office

Ref document number: 47311/99

Country of ref document: AU

WWW Wipo information: withdrawn in national office

Ref document number: 1999930868

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1020017000008

Country of ref document: KR