US20060270862A1 - Analogs of discodermolide and dictyostatin-1, intermediates therefor and methods of synthesis thereof - Google Patents

Analogs of discodermolide and dictyostatin-1, intermediates therefor and methods of synthesis thereof Download PDF

Info

Publication number
US20060270862A1
US20060270862A1 US11/497,746 US49774606A US2006270862A1 US 20060270862 A1 US20060270862 A1 US 20060270862A1 US 49774606 A US49774606 A US 49774606A US 2006270862 A1 US2006270862 A1 US 2006270862A1
Authority
US
United States
Prior art keywords
group
alkyl
alkyl group
chr
sir
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/497,746
Inventor
Dennis Curran
Youseung Shin
Nakyen Choy
Billy Day
Raghavan Balachandran
Charitha Madiraju
Tiffany Turner
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Pittsburgh
Original Assignee
Individual
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 Individual filed Critical Individual
Priority to US11/497,746 priority Critical patent/US20060270862A1/en
Publication of US20060270862A1 publication Critical patent/US20060270862A1/en
Assigned to PITTSBURGH, UNIVERSITY OF reassignment PITTSBURGH, UNIVERSITY OF ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BALACHANDRAN, RAGHAVAN, CHOY, NAKYEN, CURRAN, DENNIS P., DAY, BILLY W., MADIRAJU, CHARITHA, SHIN, YOUSEUNG, TURNER, TIFFANY
Priority to US12/235,720 priority patent/US7968736B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D407/00Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00
    • C07D407/02Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing two hetero rings
    • C07D407/06Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C33/00Unsaturated compounds having hydroxy or O-metal groups bound to acyclic carbon atoms
    • C07C33/02Acyclic alcohols with carbon-to-carbon double bonds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C59/00Compounds having carboxyl groups bound to acyclic carbon atoms and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups
    • C07C59/01Saturated compounds having only one carboxyl group and containing hydroxy or O-metal groups
    • C07C59/10Polyhydroxy carboxylic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C59/00Compounds having carboxyl groups bound to acyclic carbon atoms and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups
    • C07C59/40Unsaturated compounds
    • C07C59/42Unsaturated compounds containing hydroxy or O-metal groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C69/00Esters of carboxylic acids; Esters of carbonic or haloformic acids
    • C07C69/66Esters of carboxylic acids having esterified carboxylic groups bound to acyclic carbon atoms and having any of the groups OH, O—metal, —CHO, keto, ether, acyloxy, groups, groups, or in the acid moiety
    • C07C69/73Esters of carboxylic acids having esterified carboxylic groups bound to acyclic carbon atoms and having any of the groups OH, O—metal, —CHO, keto, ether, acyloxy, groups, groups, or in the acid moiety of unsaturated acids
    • C07C69/732Esters of carboxylic acids having esterified carboxylic groups bound to acyclic carbon atoms and having any of the groups OH, O—metal, —CHO, keto, ether, acyloxy, groups, groups, or in the acid moiety of unsaturated acids of unsaturated hydroxy carboxylic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C69/00Esters of carboxylic acids; Esters of carbonic or haloformic acids
    • C07C69/66Esters of carboxylic acids having esterified carboxylic groups bound to acyclic carbon atoms and having any of the groups OH, O—metal, —CHO, keto, ether, acyloxy, groups, groups, or in the acid moiety
    • C07C69/73Esters of carboxylic acids having esterified carboxylic groups bound to acyclic carbon atoms and having any of the groups OH, O—metal, —CHO, keto, ether, acyloxy, groups, groups, or in the acid moiety of unsaturated acids
    • C07C69/734Ethers
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D309/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings
    • C07D309/02Heterocyclic compounds containing six-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
    • C07D309/08Heterocyclic compounds containing six-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 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
    • C07D309/10Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D309/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings
    • C07D309/16Heterocyclic compounds containing six-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
    • C07D309/28Heterocyclic compounds containing six-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
    • C07D309/30Oxygen atoms, e.g. delta-lactones
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D313/00Heterocyclic compounds containing rings of more than six members having one oxygen atom as the only ring hetero atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D319/00Heterocyclic compounds containing six-membered rings having two oxygen atoms as the only ring hetero atoms
    • C07D319/041,3-Dioxanes; Hydrogenated 1,3-dioxanes
    • C07D319/061,3-Dioxanes; Hydrogenated 1,3-dioxanes not condensed with other rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F7/00Compounds containing elements of Groups 4 or 14 of the Periodic System
    • C07F7/02Silicon compounds
    • C07F7/08Compounds having one or more C—Si linkages
    • C07F7/18Compounds having one or more C—Si linkages as well as one or more C—O—Si linkages
    • C07F7/1804Compounds having Si-O-C linkages
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers

Definitions

  • the present invention relates to analogs of discodermolide and dictyostatin-1, intermediates for the synthesis of such analogs and methods of synthesis of such intermediates and analogs.
  • Taxol paclitaxel
  • a number of analogs of Taxol, including Taxotere (docetaxel), are also powerful anticancer agents.
  • discodermolide is not readily available in large quantities from natural sources. Accordingly, assuring a sufficient supply of discodermolide is problematic. Simplified analogs that retain high anti-cancer activity but are easier to make are in urgent need.
  • dictyostatin has also been shown to stabilize microtubules, like discodermolide and Taxol. See Wright, A. E.; Cummins, J. L.; Pomponi, S. A.; Longley, R. E.; Isbrucker, R. A. Dictyostatin compounds for stabilization of microtubules. In PCT Int. Appl.; WO 62239, 2001. Accordingly, dictyostatin 1 and its analogs show great promise as new anticancer agents. There is an urgent need for a synthetic route to make dictyostatin 1 and its analogs in order to fully assign the structure of dictyostatin 1, to produce analogs to study the structure/activity relationship and to identify and produce the best possible drugs in this family.
  • the inventors of the present invention as one aspect of the present invention, herein set forth a number of analogs of discodermolide, as well as methods and intermediates for the synthesis thereof.
  • the inventors of the present invention as another aspect of the present invention, herein set forth a family of both closed and open analogs of dictyostatin 1 with methods and intermediates for the synthesis of this family.
  • the present invention provides a compound of the following structure: wherein R 1 is H, an alkyl group, an aryl group, an alkenyl group, an alkynyl group, or a halogen atom; R 2 is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiR a R b R c , CH 2 OR d , or COR e ; R a , R b and R c are independently an alkyl group or an aryl group; R d is an alkyl group, an aryl group, an alkoxylalkyl group, —R i SiR a R b R c or a benzyl group, wherein R i is an alkylene group; R e is an alkyl group, an allyl group, a benzyl group, an aryl group, an alkoxy group, or —NR g R h
  • the compound has the following stereostructure, or its enantiomer: wherein R 1 is alkenyl; R 2 is H; R 3 is —CH 2 CH(CH 3 ) or —CH ⁇ C(CH 3 ); and R 4 is —(CHR k1 ) y1 (CHR k2 ) y2 (CHR k3 ) y3 (CHR k4 ) y4 (CHR k5 ) y5 C(R s1 ) ⁇ C(R s2 )C(R s3 )C(R s4 )— wherein y1-y4 are 1, y5 is 0, R k1 and R k3 are OH, R k2 is H, R k4 is CH 3 , R s1 , R s2 , R s3 and R s4 are H, and R 5 is OH.
  • R 1 is —CH ⁇ CH 2 and R 4 is
  • the present invention provides a compound of the following structure: wherein R 1 is H, an alkyl group, an aryl group, an alkenyl group, an alkynyl group, or a halogen atom; R 2 and R 2d are independently H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiR a R b R c , CH 2 OR d , or COR E ; R a , R b and R c are independently an alkyl group or an aryl group; R d is an alkyl group, an aryl group, an alkoxylalkyl group, —R i SiR a R b R c or a benzyl group, wherein R i is an alkylene group; R e is an alkyl group, an allyl group, a benzyl group, an aryl group, an alkoxy group, or —NR
  • the compound has the following stereostructure, or its enantiomer wherein R 1 is alkenyl; R 2 is H; R 2d is H, OC(O)CH 3 or OC(O)NR g R h wherein R g and R h are independently H, an alkyl group or an aryl group; R 3 is CH 2 CH(CH 3 ) or CH ⁇ C(CH 3 ); and R 4 is —(CHR k1 ) y1 (CHR k2 ) y2 (CHR k3 ) y3 (CHR k4 ) y4 (CHR k5 ) y5 C(R s1 ) ⁇ C(R s2 )C(R s3 ) ⁇ C(R s4 )— wherein y1-y4 are 1, y5 is 0, R k1 and R K3 are OH, R k2 is H, R k4 is CH 3 , R s1 , R s2 ,
  • R 1 is —CH ⁇ CH 2
  • R 2d is H, OC(O)CH 3 or OC(O)NH 2 .
  • the present invention provides a compound of the following structure: wherein R 1 is H, an alkyl group, an aryl group, an alkenyl group, an alkynyl group, or a halogen atom; R 2 and R 2d are independently H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiR a R b R c , CH 2 OR d , or COR e ; R a , R b and R c are independently an alkyl group or an aryl group; R d is an alkyl group, an aryl group, an alkoxylalkyl group, —R i SiR a R b R c or a benzyl group, wherein R i is an alkylene group; R e is an alkyl group, an allyl group, a benzyl group, an aryl group, an alkoxy group, or
  • the compound has the following stereostructure, or its enantiomer wherein R 1 is alkenyl; R 2d is H, OC(O)CH 3 or OC(O)NR g R h wherein R g and R h are independently H, an alkyl group or an aryl group; R 3 is CH 2 CH(CH 3 ) or CH ⁇ C(CH 3 ); R 11a and R 11b are H or together form a portion of a six-membered acetal ring containing C(H)(p-C 6 H 4 OCH 3 ) or C(CH 3 ) 2 ; R 12 is a halogen atom, CH 2 OR 2c , CHO, CO 2 R 10 , CH ⁇ CHCH 2 OR 2c , CH ⁇ CHCHO, wherein R 2c is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiR a R b R c , CH 2 OR d
  • R 1 is —CH ⁇ CH 2
  • R 2d is H, —OC(O)CH 3 or —OC(O)NH 2
  • R 12 is —CH 2 OH, —CHO or —CO 2 R 10 .
  • the present invention provides a compound having the following structure: wherein R 2 is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiR a R b R c , CH 2 OR d , or COR e ; R a , R b and R c are independently an alkyl group or an aryl group; R d is an alkyl group, an aryl group, an alkoxylalkyl group, —R i SiR a R b R c or a benzyl group, wherein R i is an alkylene group; R e is an alkyl group, an allyl group, a benzyl group, an aryl group, an alkoxy group, or —NR g R h , wherein R g and R h are independently H, an alkyl group or an aryl group; R 3 is (CH 2 ) n where n is
  • the compound has the following stereostructure, or its enantiomer
  • R 2 is H
  • R 2d is H, OC(O)CH 3 or OC(O)NR g R h wherein R g and R h are independently H, an alkyl group or an aryl group
  • R 3 is CH 2 CH(CH 3 ) or CH ⁇ C(CH 3 )
  • R 11a and R 11b are H or together form a portion of a six-membered acetal ring containing C(H)(p-C 6 H 4 OCH 3 ) or C(CH 3 ) 2
  • R 12 is a halogen atom, CH 2 OR 2c , CHO, CO 2 R 10 , CH ⁇ CHCH 2 OR 2 C, CH ⁇ CHCHO or CH ⁇ CHCO 2 R 10 , wherein R 2c is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiR a R b R c ,
  • the present invention provides a compound having the following formula wherein R 1 is H, an alkyl group, an aryl group, an alkenyl group, an alkynyl group, or a halogen atom; R 2 is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiR a R b R c , CH 2 OR d , or COR e ; R a , R b and R c are independently an alkyl group or an aryl group; R d is an alkyl group, an aryl group, an alkoxylalkyl group, —R i SiR a R b R c or a benzyl group, wherein R i is an alkylene group; R e is an alkyl group, an allyl group, a benzyl group, an aryl group, an alkoxy group, or —NR g R h
  • the compound has the following stereostructure, or its enantiomer wherein R 3 is CH 2 CH(CH 3 ) or CH ⁇ C(CH 3 ); R 11a and R 11b are H or together form a portion of a six-membered acetal ring containing C(H)(p-C 6 H 4 OCH 3 ) or C(CH 3 ) 2 ; R 12 is a halogen atom, CH 2 OR 2c , CHO, CO 2 R 10 , CH ⁇ CHCH 2 OR 2c , CH ⁇ CHCHO or CH ⁇ CHCO 2 R 10 , wherein R 2c is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiR a R b R c , CH 2 OR d , or COR e , and R 10 is H or alkyl; and R 14a and R 14b are H or together form a portion of a six-membered acetal ring containing
  • the present invention provides a compound having the following formula wherein R 2 is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiR a R b R c , CH 2 OR d , or COR e ; R a , R b and R c are independently an alkyl group or an aryl group; R d is an alkyl group, an aryl group, an alkoxylalkyl group, —R i SiR a R b R c or a benzyl group, wherein R i is an alkylene group; R e is an alkyl group, an allyl group, a benzyl group, an aryl group, an alkoxy group, or —NR g R h , wherein R g and R h are independently H, an alkyl group or an aryl group; R 11a and R 11b are independently H, an alkyl group or an
  • the compound has the following stereostructure, or its enantiomer wherein R 2 is H, an alkyl group, a benzyl group, a trityl group, —SiR a R b R c , CH 2 OR d , or COR e , R 11a and R 11b are H or together form a portion of a six-membered acetal ring containing C(H)(p-C 6 H 4 OCH 3 ) or C(CH 3 ) 2 ; and R 16 is H or alkyl.
  • the present invention provides a compound having the following formula wherein R 1 is H, an alkyl group, an aryl group, an alkenyl group, an alkynyl group, or a halogen atom; R 2 , R 2d and R 2e are independently H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiR a R b R c , CH 2 OR d , or COR e ; R a , R b and R c are independently an alkyl group or an aryl group; R d is an alkyl group, an aryl group, an alkoxylalkyl group, —R i SiR a R b R c or a benzyl group, wherein R i is an alkylene group; R e is an alkyl group, an allyl group, a benzyl group, an aryl group, an alk
  • the compound has the following stereostructure, or its enantiomer
  • R 1 is a CH ⁇ CH 2 and R 3 is (Z)-CH ⁇ CH—, or —CH 2 CH 2 —.
  • R 11a and R 11b are independently H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiR a R b R c , CH 2 OR d , COR e , or R 11a and R 11b together form a portion of six-membered acetal ring containing CR t R u ;
  • R t and R u are independently H, an alkyl group, an aryl group or an alkoxyarly group;
  • R a , R b and R c are independently an alkyl group or an aryl group;
  • R d is an alkyl group, an aryl group, an alkoxylalkyl group, —R i SiR a R b R c or a benzyl group, wherein R i is an alkylene group;
  • R e is an alkyl group, an allyl group,
  • the compound has the following stereostructure, or its enantiomer wherein R 11a and R 11b are independently H, an alkyl group, and aryl group, a benzyl group, a trityl group, —SiR a R b R c , CH 2 OR d , COR e , or R 11a and R b together form a portion of six-membered acetal ring incorporating CR t R u ; R t and R u are independently H, an alkyl, an aryl group or an alkoxyarly group; R a , R b and R c are independently an alkyl group or an aryl group; R d is an alkyl group, an aryl group, an alkoxylalkyl group, —R i SiR a R b R c or a benzyl group, wherein R i is an alkylene group; R e is an alkyl group, an ally
  • R 11a and R 11b are H or together form a portion of a six-membered acetal ring containing C(H)(p-C 6 H 4 OCH 3 ) or C(CH 3 ) 2 ;
  • R 21 is a halogen atom, CH 2 OR 2c , CHO, CO 2 R 10 , CH ⁇ CHCH 2 OR 2c , CH ⁇ CHCHO, wherein R 2c is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiR a R b R c , CH 2 OR d , or COR e , and R 10 is H or alkyl.
  • R 1 is CH ⁇ CH 2
  • R 21 is CH 2 OH, CHO or CO 2 R 10 .
  • the present invention provides a compound having the following formula wherein R 13 is H or an alkyl group, R 14 is H, an aryl group, an alkoxyaryl group or an alkyl group, and R 22 is a halogen atom or —P(Ar) 3 X, wherein X is a counteranion selected from the groups halide, tetrafluoroborate, hexafluorophosphate and sulfonate, provided that when R 13 and R 14 are methyl groups, X is not I. In one embodiment, when R 13 and R 14 are alkyl groups, X is not halogen.
  • the compound has the following stereostructure, or its enantiomer wherein R 13 is H or an alkyl group, and R 14 is H, an aryl group, an alkoxyaryl group, or an alkyl group, an aryl group or an alkoxyarly group, R 22 is a halogen atom or —P(Ar) 3 X, wherein X is a counteranion selected from the groups halide, tetrafluoroborate, hexafluorophosphate and sulfonate, provided that when R 13 and R 14 are methyl groups, X is not 1.
  • R 13 is H
  • R 14 is aryl
  • R 22 is P(C 6 H 5 ) 3 X.
  • R 14 is C 6 H 4 -p-OCH 3 .
  • the present invention provides a process for conversion of a first compound with the formula wherein R 1 is H, an alkyl group, an aryl group, an alkenyl group, an alkynyl group, or a halogen atom; R 2 is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiR a R b R c , CH 2 OR d , or COR e ; R 2d is H R a , R b and R c are independently an alkyl group or an aryl group; R d is an alkyl group, an aryl group, an alkoxylalkyl group, —R i SiR a R b R c or a benzyl group, wherein R i is an alkylene group; R e is an alkyl group, an allyl group, a benzyl group, an aryl group, an alkylene group; R
  • the process is for conversion of a compound with the following stereostructure or its enantiomer
  • R 1 is H, an alkyl group, an alkenyl group, an alkynyl group, or a halogen atom
  • R 2 is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiR a R b R c , CH 2 OR d , or COR e
  • R 2d is H R a , R b and R c are independently an alkyl group or an aryl group
  • R d is an alkyl group, an aryl group, an alkoxylalkyl group, —R i SiR a R b R c or a benzyl group, wherein R i is an alkylene group
  • R e is an alkyl group, an allyl group, a benzyl group, an aryl group, an alkoxy group, or
  • R 1 is alkenyl;
  • R 3 is CH 2 CH(CH 3 ) or CH ⁇ C(CH 3 ); and
  • R 4 is —(CHR k1 ) y1 (CHR k2 ) y2 (CHR k3 ) y3 (CHR k4 ) y4 (CHR k5 ) y5 C(R s1 ) ⁇ C(R s2 )C(R s3 ) ⁇ C(R s4 )— wherein y1-y4 are 1, y5 is 0, R k1 and R k3 are R 2a , R k2 is H, R k4 is CH 3 , R s1 -R s4 are H, and R 5 is OR 2b .
  • R 1 is CH ⁇ CH 2 and R 4 is
  • the first compound is converted by reacting the first compound with 2,4,6-trichlorobenzoylchloride.
  • the above general structures for the compounds of the present invention include all stereoisomers thereof (other than the natural compound dictyostatin 1). Moreover, the structures of the compounds of the present invention include the compounds in racemic form, enantiomerically enriched form or enantiomerically pure form.
  • alkyl refer generally to both unsubstituted and substituted groups unless specified to the contrary.
  • the groups set forth above can be substituted with a wide variety of substituents to synthesize analogs retaining biological activity.
  • alkyl groups are hydrocarbon groups and are preferably C 1 -C 15 (that is, having 1 to 15 carbon atoms) alkyl groups, and more preferably C 1 -C 10 alkyl groups, and can be branched or unbranched, acyclic or cyclic.
  • alkyl group refers to a substituent on another group (for example, an alkyl group as a substituent of an alkylamino group or a dialkylamino group).
  • aryl refers to phenyl or naphthyl.
  • halogen or halo refer to fluoro, chloro, bromo and iodo.
  • alkoxy refers to —OR, wherein R is an alkyl group.
  • alkenyl refers to a straight or branched chain hydrocarbon group with at least one double, bond, preferably with 2-15 carbon atoms, and more preferably with 2-10 carbon atoms (for example, —CH ⁇ CHR or —CH 2 CH ⁇ CHR; wherein R can be a group including, but not limited to, an alkyl group, an alkoxyalkyl group, an amino alkyl group, an aryl group, or a benzyl group).
  • alkynyl refers to a straight or branched chain hydrocarbon group with at least one triple bond, preferably with 2-15 carbon atoms, and more preferably with 2-10 carbon atoms (for example, —C ⁇ CR or —CH 2 —C ⁇ CR; wherein R can be a group including, but not limited to, an alkyl group, an alkoxyalkyl group, an amino alkyl group, an aryl group, or a benzyl group).
  • alkylene alkenylene” and alkynylene” refer to bivalent forms of alkyl, alkenyl and alkynyl groups, respectively.
  • trityl refers to a triphenyl methyl group or —C(Ph) 3 .
  • Certain groups such as amino and hydroxy groups may include protective groups as known in the art.
  • Preferred protective groups for amino groups include tert-butyloxycarbonyl, formyl, acetyl, benzyl, p-methoxybenzyloxycarbonyl, trityl.
  • Other suitable protecting groups as known to those skilled in the art are disclosed in Greene, T., Wuts, P. G. M., Protective Groups in Organic Synthesis , Wiley (1991), the disclosure of which is incorporated herein by reference.
  • the present invention provides a method of treating a patient for cancer, including the step of administering a pharmaceutically effective amount of a biologically active compound of the present invention or a pharmaceutically acceptable salt thereof.
  • FIG. 1 illustrates one embodiment of the syntheses of a representative isomeric aldehyde for incorporation of the left part of the configuration of (+)-discodermolide.
  • FIG. 2 illustrates one embodiment of the syntheses of another representative isomeric aldehyde for incorporation of the left part of the configuration of (+)-discodermolide.
  • FIG. 2 illustrates one embodiment of the syntheses of an intermediate for the center part of the configuration of (+)-discodermolide.
  • FIG. 4 illustrates one embodiment of the construction of the right fragment or part of the molecule.
  • FIG. 5 illustrates an embodiment of the synthesis of several discodermolide analogs of the present invention.
  • FIGS. 6A through D illustrate the tubulin polymerization-inducing properties of discodermolide ( Figure A), as well as discodermolide analog compounds 40 ( Figure B), 41 ( Figure C) and 42 ( Figure D) of the present invention in comparison to 10 ⁇ M paclitaxel (PTX).
  • FIG. 7 illustrates a retrosynthetic analysis of a dictyostatin analog of the present invention.
  • FIG. 8 illustrates an embodiment of the coupling of three fragments of a dictyostatin analog of the present invention.
  • FIG. 9 illustrates the structures of several acyclic compounds of the present invention that were tested for biological activity.
  • FIG. 10 illustrates an embodiment of the synthesis of a lower fragment of a dictyostatin analog of the present invention.
  • FIG. 11 illustrates an embodiment of the synthesis of a macrolactone of the present invention.
  • FIG. 12 illustrates a second representative general scheme for synthesis of stereoisomers and analogs of dictyostatin.
  • FIG. 13 illustrates a summary of an embodiment of the synthesis of the bottom fragment of dictyostatin analogs of the present invention.
  • FIG. 14 illustrates an embodiment of the coupling of the bottom fragment of FIG. 13 with the center fragment of the dictyostatin analogs of the present invention.
  • FIG. 15 illustrates an embodiment of the introduction of the C16 stereocenter and introduction of the top fragment of the dictyostatin analog of the present invention.
  • FIG. 16 illustrates an embodiment of the completion of the synthesis of the dictyostatin analog of the present invention.
  • FIG. 17 illustrates embodiments of representative methods to make analogs of the terminal diene fragment of the dictyostatin analog of the present invention.
  • FIG. 18 illustrates a summary of an embodiment of the synthesis of the bottom fragment of the dictyostatin analog of the present invention.
  • FIG. 19 illustrates an embodiment of the synthesis of two fragments with anti/anti configurations as assigned to dictyostatin 1 at C13-C15.
  • FIGS. 1 and 2 The syntheses of two representative isomeric aldehydes 9a and 9b for, incorporation of the left part of these molecules are shown in FIGS. 1 and 2 .
  • he synthesis of the left display bearing the configuration of (+)-discodermolide started with the commercially available methacrolein 3 ( FIG. 1 ).
  • Reaction of 3 with the boron enolate of Evans oxazolidinone 4 gave the corresponding alcohol, which was silylated to give compound 5 in 90% yield. Lactonization followed by the introduction of allyl group was performed by the previously reported method to give 6 in good yield. See Day, B. W.; Kangani, C. O.; Avor, K. S.
  • Oxazolidinone 18 was prepared from (S)-3-hydroxy-2-methylpropionic methyl ester 16 by the known procedure for the preparation of ent-18. See Clark, D. L.; Heathcock, C. H. Studies on the alkylation of chiral enolates: application toward the total synthesis of discodermolide. J. Org. Chem. 1993, 58 5878-5879. Reduction of 18 with lithium borohydride gave the diol, which was protected by anisaldehyde dimethyl acetal 19 to give the acetal 20.
  • the construction of the right fragment 34 featured aldol reactions.
  • Syn-Aldol reaction of aldehyde 24 with 4 provided 25, which was reduced to a diol and protected with anisaldehyde dimethyl acetal 19 to give 26.
  • Selective opening of the benzylidine ring of 26 with DIBAL gave a primary alcohol, which was oxidized to aldehyde 27.
  • the subsequent anti-aldol condensation using Heathcock's aldol reaction with dimethylphenyl propionate 28 furnished compound 29 as the major product in 73% yield. See Heathcock, C. H.; Pirrung, M. C.; Montgomery, S. H.; Lampe, J.
  • the first Wittig reaction of 9a with 21 provided 35 (see FIG. 5 ).
  • DIBAL reductive cleavage of the acetal followed by Dess-Martin oxidation gave aldehyde 37.
  • aldehyde 38 was made from 9b via 36.
  • the second Wittig olefination was accomplished with 38 as an example and phosphonium salt 33 ( FIG. 5 ).
  • Tetrabutylammonium fluoride deprotection followed by carbamoylation using Kocovsky's method See Kocovsky, P. Carbamates: a method of synthesis and some synthetic applications. Tetrahedron Lett. 1986, 27 5521-5524 afforded the C19 carbamate-containing compound 39.
  • the lactone was built from the methyl acetal in the left fragment by using aqueous 60% acetic acid in THF followed by Dess-Martin oxidation.
  • Deprotected analog 40 containing a free C3 hydroxy group on the lactone was obtained by the removal of MOM group with 4N HCl followed by removal of the PMB protecting groups with DDQ oxidation.
  • Two additional example compounds, 41 and 42, were prepared from the intermediate 39 by using appropriate conditions. All three of these analogs exhibited significant activity, as shown in FIGS. 6 A-D and Table 1. Surprisingly, the C3-MOM-protected analogs 41 and 42 showed better microtubule hypernucleation activities than the analog 40 with a free C3-hydroxy group. As can be seen in FIG.
  • discodermolide is superior to paclitaxel (taxol) in that it causes equivalent microtubule assembly at both lower concentrations and temperatures (the increase in absorbance caused by discodermolide occurs at a time point earlier than that caused by paclitaxel). Additionally, the polymer induced to form by discodermolide is more resistant to cold-induced disassembly than is the paclitaxel-induced polymer. Both analogs 41 ( FIG. 6C ) and 42 ( FIG. 6D ) showed these more rapid polymer-inducing and cold-resistant properties, albeit at somewhat lower potencies (for example, higher concentrations of the analogs were necessary for these effects to be detected) than discodermolide. MOM ether lactone 41 was the most potent among these analogs.
  • Table 1 shows microtubule stabilizing, antiproliferative, and paclitaxel-displacing properties of 40-42.
  • the lactone, MOM ether 41 was more potent than the lactol 42 or free hydroxy 40 relatives.
  • the cellular activity of 41 was good, showing a submicromolar 50% growth inhibitory (GI 50 ) concentration. This compound also showed considerable affinity for the paclitaxel binding site on tubulin. A 2-fold molar excess of 41 displaced [ 3 H]paclitaxel from microtubules better than paclitaxel and at almost the same potency as discodermolide.
  • Macrocycle 43 is a representative example of a dictyostatin analog with an alkyl chain bridging the lactone carbonyl group and the C10/C11 alkene and with two Z-double bonds in the macrocycle. It can also be considered as a macrocyclic analog of discodermolide. This can be synthesized convergently from three components-33, 21 and 44-via sequential Wittig couplings and a macrocyclization ( FIG. 7 ). This design allows the synthesis of substantially any stereoisomer by employing the desired isomer of the relevant precursor—21 or 33.
  • Fragment 45 was synthesized from 1,10-decanediol (not shown) by mono-TBS protection (NaH/TBSCl, 42%) followed by Dess-Martin oxidation (80%). Fragment 21 was prepared as shown in FIG. 3 . Fragment 33 was prepared as shown in FIG. 4 .
  • Acyclic compounds 47, 48 and 49 were readily made from appropriate synthetic intermediates (45 or 46) in reasonable yields ( FIG. 9 ).
  • Macrolactone 43 and non-cyclized alcohol 47 and ester 48 exhibited similar 50% growth inhibitory concentrations, in the 15-30 ⁇ M range.
  • Carboxylic acid 49 was inactive (>50 ⁇ M) possibly due to poor cell membrane penetration. The modest activity of these compounds is encouraging given the simplicity and flexibility of their lower chain.
  • FIG. 10 The synthesis of a lower fragment more closely related to dictyostatin is shown in FIG. 10 .
  • Synthesis of the needed aldehyde 51 ( FIG. 10 ) started from the intermediate 25, which was reduced to an alcohol with LiBH 4 , followed by PMB acetal protection as in FIG. 4 .
  • Selective acetal opening produced alcohol 50, which was subjected to Dess-Martin oxidation to give aldehyde 27 (see FIG. 4 ).
  • Wittig-Homer reaction, and removal of the TBS group with HF-pyridine gave a primary alcohol, which was oxidized to aldehyde 51.
  • Compound 54 proved to be quite potent in terms of antiproliferative activity against human carcinoma cells (Table 2) showing a 50% growth inhibitory concentration against breast and ovarian cancer cells of about 1 ⁇ M. Furthermore compound 54 displaced [ 3 H]paclitaxel stoichiometrically bound to microtubules at about 1 ⁇ 3 rd the potency of discodermolide.
  • FIG. 12 A second representative general strategy for the synthesis of stereoisomers and close analogs of dictyostatin is shown in FIG. 12 . Again the molecule is dissected such that every stereocenter can be controlled and modified either by starting with an appropriate precursor or through an asymmetric reaction allowing access to both possible isomers.
  • FIG. 13 summarizes the synthesis of the bottom fragment.
  • (S)-Diethyl maleate was reduced and the resulting diol was converted to acetonide 55.
  • Reduction to the aldehyde and standard Evans aldol reaction gave 56.
  • Reduction of this to the aldehyde and Wittig-Homer Emmons reaction gave 57.
  • Removal of the acetonide and silyation gave 58, which was mono-desilylated to 59 and oxidized to aldehyde 60.
  • FIG. 15 Introduction of the C16 stereocenter and introduction of the top fragment are shown in FIG. 15 .
  • Evans asymmetric alkylation to 69 followed by removal of the chiral auxiliary by reduction gave 70.
  • reagent 71 was made from the Evans oxazolindinone by displacement with LiCH 2 P(O)(OMe) 2 .
  • Dess-Martin oxidation and Horner-Emmons olefination with 71 then gave 72, which was reduced with NiCl 2 /NaBH 4 to 73.
  • Now reduction with sodium borohydride gave a 2.8/1 mixture of stereoisomers, which could be separated and converted to the final products independently.
  • Silylation to 75 followed by DIBAL reduction gave 76, which was converted to diene 77 as described above. Detriylation then gave 78.
  • FIG. 16 Completion of the synthesis is shown in FIG. 16 .
  • Dess-Martin oxidation and Still-Gennari olefination gave 79 which was deprotected with DDQ to 80. Saponification then gave the hydroxy acid 81 ready for macrocyclization.
  • Treatment of 81 under the Yamaguchi protocol gave 82, which was finally deprotected to give the target product 83, an isomer of dictyostatin 1 called dictyostatin 5.
  • FIG. 17 Representative methods to make analogs of the terminal diene fragment are shown in FIG. 17 .
  • Alkene 84 was ozonized to give the aldehyde, which was subjected to a Wittig reaction to give analogs like 85.
  • 84 can be converted to the Z-vinyl iodide 86, which can in turn be coupled with organometallic reagents like phenyl zinc iodide to give 87.
  • organometallic reagents like phenyl zinc iodide
  • FIG. 18 summarizes the synthesis of a fully elaborated bottom fragment 92.
  • Acetal 88 readily prepared from (D)-malic acid, was silylated with t-butyidiphenylsilyl chloride (TBDPSCl). Reductive cleavage of the acetal with DIBAL followed by Swem oxidation provided aldehyde 89. Reaction of 89 with the indicated Z-crotyl boronate according to Roush (See: Roush, W. R.; Hoong, L. K.; Palmer, M. A. J.; Straub, J.
  • FIG. 19 shows the synthesis of two fragments with anti/anti configurations as assigned to dictyostatin 1 at C13-C15.
  • Evan anti-aldol reaction of ent-4 and methacrolein See: Evans, D. A.; Tedrow, J. S.; Shaw, J. T.; Downey, C. W. Diastereoselective magnesium halide-catalyzed anti-aldol reactions of chiral N-acyloxazolidinones. J. Am. Chem. Soc. 2002, 124, 392-393) followed by TFA treatment gave 93 in 78% yield. A minor diastereomer of 93 (about 16/1 ratio) was separated by chromatography.
  • N,N-Diisopropylethylamine (7.5 mL) and chloromethyl methyl ether (1.13 mL, 15 mmol) were added to a solution of the alcohol in CH 2 Cl 2 (15 mL).
  • the reaction mixture was heated to reflux and stirred overnight.
  • the reaction was quenched with aqueous saturated NaHCO 3 (50 mL) followed by washing with brine.
  • the aqueous layer was extracted with CH 2 Cl 2 (2 ⁇ 50 mL).
  • the combined organic layer was dried over MgSO 4 .
  • the solvent was removed under reduced pressure and the crude product was purified by column chromatography (hexane/EtOAc 7:3) to provide the pure anomers of 8 ( ⁇ , 33%; ⁇ , 32%).
  • N,N-Diisopropylethylamine (1.9 mL, 11 mmol) was added to a solution of propionyloxaiolidinone (2.33 g, 10 mmol) in anhydrous CH 2 Cl 2 (110 mL) at 0° C., followed by dropwise addition of Bu 2 BOTf (1.0 M in CH 2 Cl 2 , 11 mL, 11 mmol). The solution was stirred for 0.5 h at 0° C. Crude 10 in anhydrous CH 2 Cl 2 (30 mL) was added at ⁇ 78° C. The mixture was stirred for 10 min at ⁇ 78° C. followed by an additional 2 h at 0° C.
  • reaction was quenched by addition of phosphate buffer, pH 7.0 (50 mL). A solution of hydrogen peroxide (30%, 10 mL) in methanol (20 mL) was added and the mixture was allowed to stir for 1 h at 0° C. After separation of organic and aqueous layers, the aqueous layer was extracted with CH 2 Cl 2 .
  • TBDMSOTf (1.7 mL, 7.5 mmol) was added to a stirred solution of the above alcohol (2.20 g, 5 mmol) and 2,6-lutidine (1.2 mL, 10 mmol) in CH 2 Cl 2 (50 mL) at ⁇ 78° C. and the mixture was stirred for 2 h at ambient temperature. The reaction was quenched by addition of aqueous HCl (0.5 N, 100 mL). The reaction mixture was extracted with CH 2 Cl 2 and dried over MgSO 4 followed by the evaporation of the solvent under reduced pressure.
  • N,N-Diisopropylethylamine (0.97 mL, 5.5 mmol) was added to a solution of propionyloxazolidinone (1.16 g, 5 mmol) in anhydrous CH 2 Cl 2 (11 mL) at 0° C., followed by dropwise addition of Bu 2 BOTf (1.0 M in CH 2 Cl 2 , 5.5 mL, 5.5 mmol). The solution was stirred for 0.5 h at 0° C. A solution of crude aldehyde 12 from above in anhydrous CH 2 Cl 2 (10 mL) was added at ⁇ 78° C. The reaction mixture was stirred for 10 min at ⁇ 78° C. then for 2 h at 0° C.
  • reaction mixture was quenched with phosphate buffer, pH 7.0 (50 mL).
  • a solution of hydrogen peroxide (30%, 10 mL) in methanol (20 mL) was slowly added and the mixture was stirred for 1 h. After the separation of organic and aqueous layers, the aqueous layer was extracted with CH 2 Cl 2 .
  • reaction was quenched with aqueous sat'd NaHCO 3 (50 mL) and washed with brine.
  • the aqueous layer was extracted with CH 2 Cl 2 (2 ⁇ 50 mL).
  • the combined organic layer was dried over MgSO 4 .
  • the reaction was quenched by the careful addition of aqueous sat'd potassium sodium tartrate (50 mL) and stirring for 3 h at room temperature. Once the organic and aqueous layers separated, the aqueous layer was extracted with CH 2 Cl 2 . The combined organic layer was washed with brine and dried over MgSO 4 followed by the evaporation of solvent under reduced pressure. The crude lactol obtained was used without further purification.
  • the reaction mixture was stirred at ambient temperature for 1 h, diluted with ethyl ether (50 mL) and washed with aqueous HCl (0.5 N, 50 mL) and brine (10 ml). The separated organic layer was dried over MgSO 4 . Filtration and concentration followed by short flash column chromatography (hexane/EtOAc 4:1) provided the crude aldehyde 37 (270 mg, 0.55 mmol) as a colorless oil which was used without further purification.
  • Dess-Martin periodinane reagent (31.8 mg, 0.075 mmol) was added to a solution of the lactol in CH 2 Cl 2 (5 mL). The resultant solution was stirred for 1 h and quenched by the simultaneous addition of saturated aqueous Na 2 S 2 O 3 (5 mL) and saturated aqueous NaHCO 3 . The aqueous layer was extracted with CH 2 Cl 2 (2 ⁇ 10 mL) and the combined extracts were dried over anhydrous MgSO 4 .
  • Carbamic acid (1S,2S,3R,6Z,8S,9S,10S,11Z)-3,9-dihydroxy-14-1(2S,3S,4S,5R)-4-methoxymethoxy-3,5-dimethyl-6-oxotetrahydropyran-2-yl]-2,8,10-trimethyl-1-[(1S,2Z)-1-methylpenta-2,4-dienyl]tetradeca-6,11-dienyl ester (41).
  • Carbamate 39 (8.49 mg, 0.01 mmol) was subjected to the lactonization procedure described above. The removal of the PMB protecting group was accomplished by treating with NaHCO 3 and DDQ.
  • the above macrolactone (12 mg, 16. ⁇ mol) was dissolved in CH 2 Cl 2 (2 ml)—H 2 O (0.2 ml) and DDQ (12 mg, 53 ⁇ mol) was added at 0° C. After 1 h of stirring at 0° C., the reaction mixture was quenched by adding sat'd NaHCO 3 (5 ml). The organic phase was washed by sat'd NaHCO 3 solution (3 ⁇ 20 ml) and brine, dried over MgSO 4 and concentrated.
  • Trimethylsilyl chloride (0.24 ml, 1.9 mmol) was added dropwise to a stirred mixture containing above acetal (0.177 g, 0.31 mmol), NaCNBH 3 (0.12 g, 1.9 mmol) and 4 ⁇ molecular sieve in acetonitrile (6 ml) at 0° C.
  • the reaction mixture was stirred for 1 h at 0° C. and filtered through Celite, poured into 1N HCl (10 ml). The aqueous phase was extracted by CH 2 Cl 2 (2 ⁇ 20 ml), dried (MgSO 4 ), filtered and concentrated.
  • reaction mixture was quenched by addition of a sat'd NH 4 Cl solution (1 ml) and diluted with diethyl ether (10 ml). The layer was separated and organic phase was washed with brine (10 ml) and dried with MgSO 4 , filtered, and concentrated.
  • the ethanolic solution was concentrated and then diluted with EtOAc (50 ml). After the solution was acidified to pH 3 with 1N HCl solution, organic phase was separated and aqueous phase was extracted with EtOAc (2 ⁇ 10 ml). The combined organic phase were dried with MgSO 4 , concentrated and used as crude in next step without further purification.
  • the carboxylic acid was treated with NEt 3 (1.5 ml) and ethyl chloroformate (0.67 ml) in dry THF (50 ml) at ⁇ 10° C. After 15 min, the mixture was warmed to 0° C. and a solution of NaBH, (1.2 g) in H 2 O (10 ml) were added.
  • reaction was quenched by addition of sat'd Rochelle salt solution and Et 2 O. The layers were separated and the organic layer was washed with H 2 O, sat'd NaHCO 3 solution and brine, dried with MgSO 4 .
  • reaction mixture was quenched by EtOAc (10 ml) and sat'd sodium potassium tartrate solution (50 mL) followed by vigorously stirring for 4 h.
  • the aqueous phase was extracted with CH 2 Cl 2 (3 ⁇ 20 mL) and the combined organic layers were washed with brine (30 mL).
  • reaction mixture was treated with MeI (0.29 ml) at ⁇ 78° C., stirred for an additional 4 h, quenched with sat'd aqueous NH 4 Cl, and extracted with ether (2 ⁇ 10 ml).
  • reaction mixture was quenched by addition of a sat'd NH 4 Cl solution (1 ml) and diluted with diethyl ether (5 ml). The layers were separated and organic phase was washed with brine (5 ml) and dried with MgSO 4 , filtered, and concentrated.
  • the reaction mixture was treated with dimethysulfide (2.0 ml) and pyridine (32 ⁇ l) and stirred for 3.0 h at ambient temperature.
  • the reaction mixture was concentrated and diluted with Et 2 O (80 ml).
  • the organic layer was washed with saturated aqueous CuSO 4 (2 ⁇ 20 ml) and brine (20 ml), dried over MgSO 4 , filtered and concentrated.
  • a suspension of propyltriphenylphosphonium bromide 0.383 g 98% purity, 0.97 mmol
  • THF 1.0 M solution in THF, 0.98 ml
  • reaction mixture was quenched with saturated aqueous NaHCO 3 (20 ml) and extracted with Et 2 O (2 ⁇ 40 ml). The combined extracts were washed with brine, (20 ml), dried over MgSO 4 , filtered and concentrated.
  • reaction mixture was treated with dimethylsulfide (1.5 ml) and pyridine (23 ⁇ l) and stirred for 3.0 h at ambient temperature.
  • the reaction mixture was concentrated and diluted with Et 2 O (80 ml).
  • the organic layer was washed with saturated aqueous CuSO 4 (2 ⁇ 20 ml) and brine (20 ml), dried over MgSO 4 , filtered and concentrated.
  • a suspension of (iodomethyl)triphenylphosphonium iodide 0.213 g, 0.40 mmol) in THF (3.0 ml) was added NaN(TMS) 2 (1.0 M solution in THF, 0.40 ml).
  • reaction mixture was quenched with saturated aqueous NaHCO 3 (20 ml) and extracted with Et 2 O (2 ⁇ 40 ml). The combined extracts were washed with brine (20 ml), dried over MgSO 4 , filtered and concentrated.
  • the aqueous layer was extracted with CH 2 Cl 2 (4 ⁇ 15 mL) and the organic layers were combined, dried over MgSO 4 , filtered and concentrated in vacuo.
  • the crude oil was purified by flash chromotaography (20% to 50% EtOAc in hexanes) providing 4.01 g (80%) of the corresponding alcohol as a clear, yellow oil.
  • a solution of (R,R)-diisopropyl tartrate (Z)-crotylboronate (15.0 mmol) was added to 4 ⁇ powdered molecular sieves (0.170 g) in toluene (8.4 mL) under argon and the mixture was stirred for 20 min at room temperature. The mixture was cooled to ⁇ 78° C.
  • [2R,4R,5R]-[2,4-bis-(4-Methoxybenzyloxy)-5-methylhept-6-enyloxy]tert-butyidiphenyl silane A mixture of NaH (1.8 g, 7.23 mmol) in THF (5 mL) was cooled to 0° C. then DMF (5 mL), the alcohol (1.25 g, 2.41 mmol) in THF (5 mL), and PMBBr (1.14 g, 6.03 mmol) were added. The reaction mixture was warmed to room temperature and stirred for 48 h. The resulting mixture was poured into a pH 7 phosphate buffer and diluted with ether (85 mL).
  • reaction mixture was poured into a vigorously stirred solution of saturated Rochelle salt in H 2 O (8 mL) and EtOAc (12 mL) and stirred overnight; The aqueous layer was separated and extracted with EtOAc (3 ⁇ 5 mL), dried over MgSO 4 , filtered and concentrated in vacuo.
  • reaction mixture was slowly quenched with saturated NaHCO 3 (5 mL) and the aqueous layer was separated and extracted with CH 2 Cl 2 (5 ⁇ 2 mL). The combined organic layers were washed with saturated CuSO 4 (2 mL), dried over MgSO 4 , filtered and concentrated in vacuo.
  • n-BuLi 100 mL, 2.5 M in hexane
  • Triisopropylborate 57.8 mL, 250 mmol
  • the reaction mixture was maintained at ⁇ 78° C. for 30 min and then rapidly poured into a 2 L separatory funnel containing 470 mL of 1 N HCl saturated with NaCl.
  • the aqueous layer was adjusted to pH 1 by using 1 N HCl (100-150 mL), and then a solution of (R,R)-diisopropyl tartrate (52.8 g, 250 mmol) in 88 mL of Et 2 O was added. The phases were separated, and the aqueous layer was extracted with additional Et 2 O (4 ⁇ 120 mL). The combined extracts were dried over MgSO 4 for 1 h then vacuum filtered through a fritted glass funnel under Ar blanket into an oven-dried round-bottom flask. The filtrate was concentrated in vacuo, and pumped to constant weight at under vacuum. Anhydrous toluene (170 mL) was added to the flask make a 1M solution.
  • n-BuLi 100 mL, 2.5 M in hexane
  • Triisopropylborate 57.8 mL, 250 mmol
  • the reaction mixture was maintained at ⁇ 78° C. for 30 min and then rapidly poured into a 2L separatory funnel containing 470 mL of 1 N HCl saturated with NaCl.
  • the aqueous layer was adjusted to pH 1 by using 1 N HCl (100-150 mL), and then a solution of (R,R)-diisopropyl tartrate (52.8 g, 250 mmol) in 88 mL of Et 2 O was added. The phases were separated, and the aqueous layer was extracted with additional Et 2 O (4 ⁇ 120 mL). The combined extracts were dried over MgSO 4 for 1 h then vacuum filtered through a fritted glass funnel under Ar blanket into an oven-dried round-bottom flask. The filtrate was concentrated in vacuo, and pumped to constant weight at under vacuum. Anhydrous toluene (170 mL) was added to the flask make a 1M solution.
  • Tubulin without microtubule-associated proteins was prepared from fresh bovine brains[32] The normoisotopic and tritiated forms of paclitaxel and normoisotopic docetaxel were provided by the Drug Synthesis and Chemistry Branch, National Cancer Institute. (+)-Discodermolide was from Novartis Pharmaceutical Corporation. Ca 2+ - and Mg 2+ -free RPMI-1640 culture medium were from GIBCO/BRL-Life Technologies. Fetal bovine serum (FBS) was from Hyclone. Cell lines were obtained from American Type Culture Collection (Manassas, Va.).
  • Tubulin Polymerization[32] Tubulin assembly was followed in a Beckman-Coulter 7400 spectrophotometer, equipped with an electronic Peltier temperature controller, reading absorbance (turbidity) at 350 nm.
  • Reaction mixtures (0.25 mL final volume) contained tubulin (final concentration 10 ⁇ M; 1 mg/mL), monosodium glutamate (0.8 M from a stock solution adjusted to pH 6.6 with HCl), DMSO (final volume 4% v/v), and differing concentrations of test agent where indicated.
  • Reaction mixtures without test agent were cooled to 0° C. and added to cuvettes held at 0.25-0.5° C. in the spectrophotometer. Test agent in DMSO was then rapidly mixed in the reaction mixture.
  • Each run contained one positive control (paclitaxel, 10 ⁇ M final concentration) and one negative control (DMSO only). Baselines were established at 0.25-2.5° C. and temperature was rapidly raised to 30° C. (in approximately 1 min) and held there for 20 min. The temperature was then rapidly lowered back to 0.25-2.5° C.
  • Cell Growth Inhibition [34] Cells were plated (500-2000 cells/well depending on the cell line) in 96-well microplates, allowed to attach and grow for 48 h, then treated with vehicle (4% DMSO, a concentration that allowed doubling times of 24 h or less) or test agent (50, 10, 2, 0.4 and 0.08 ⁇ M for the new agents; 0.001, 0.005, 0.010, 0.020 and 0.100 ⁇ M for paclitaxel and discodermolide) for the given times.
  • vehicle 4% DMSO, a concentration that allowed doubling times of 24 h or less
  • test agent 50, 10, 2, 0.4 and 0.08 ⁇ M for the new agents; 0.001, 0.005, 0.010, 0.020 and 0.100 ⁇ M for paclitaxel and discodermolide
  • One plate consisted of cells from each line used for a time zero cell number determination.
  • the other plates in a given determination contained eight wells of control cells, eight wells of medium and each agent concentration tested in quadrup
  • GI 50 growth inhibitory concentration
  • Paclitaxel binding site inhibition assay [34] A stock solution of [ 3 H]paclitaxel (26.8 ⁇ M, 16.2 Ci/mmol), obtained from the NCI, was prepared in 37% (v/v) DMSO. The test agents were prepared in 25% (v/v) DMSO-0.75 M monosodium glutamate (prepared from a 2M stock solution adjusted to pH 6.6 with HCl). The radiolabeled paclitaxel and test agents, as indicated in terms of final concentrations, were mixed in equal volumes and warmed to 37° C.
  • a reaction mixture (50 ⁇ L) containing 0.75 M monosodium glutamate, 4.0 ⁇ M tubulin, and 40 ⁇ M ddGTP (a non-hydrolyzable analog of GTP) was prepared and incubated at 37° C. for 30 min to preform microtubules.
  • An equivalent volume of drug mixture with [ 3 H]paclitaxel was added to preformed polymer and incubated for 30 min at 37° C.
  • Bound [ 3 H]paclitaxel was separated from free drug by centrifugation of the reaction mixtures at 14000 rpm for 20 min at room temperature. Non-specific binding was determined by addition of a 12-fold excess of docetaxel.
  • Radioactive counts from the supernatant were determined by scintillation spectrometry.
  • Bound [ 3 H]paclitaxel was calculated from the following: total paclitaxel added to each reaction mixture minus the paclitaxel present in the supernatant (free drug). The % bound values were normalized to the control values with no inhibitor added.

Abstract

A compound of the following structure:
Figure US20060270862A1-20061130-C00001

wherein R1 is H, an alkyl group, an aryl group, an alkenyl group, an alkynyl group, or a halogen atom;
R2 is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe;
Ra, Rb and Rc are independently an alkyl group or an aryl group;
Rd is an alkyl group, an aryl group, an alkoxylalkyl group, —RiSiRaRbRc or a benzyl group, wherein Ri is an alkylene group;
Re is an alkyl group, an allyl group, a benzyl group, an aryl group, an alkoxy group, or —NRgRh, wherein Rg and Rh are independently H, an alkyl group or an aryl group;
R3 is (CH2)n where n is and integer in the range of 0 to 5, —CH2CH(CH3)—, —CH═CH—, —CH═C(CH3)—, or —C≡C—;
R4 is (CH2)p where p is an integer in the range of 4 to 12, —(CHRk1)y1(CHRk2)y2(CHRk3)y3(CHRk4)y4(CHRk5)y5C(Rs1)═C(Rs2)C(Rs3)═C(Rs4)—, —(CHRk1)y1(CHRk2)y2(CHRk3)y3(CHRk4)y4(CHRk5)y5CH(Rs1)CH(Rs2)C(Rs3)═C(Rs4)—, —(CHRk1)y1(CHRk2)y2(CHRk3)y3(CHRk4)y4(CHRk5)y5C(Rs1)═C(Rs2)CH(Rs3)CH(Rs4)—, —(CHRk1)y1(CHRk2)y2(CHRk3)y3(CHRk4)y4(CHRk5)y5CH(Rs1)CH(Rs2)CH(Rs3)CH(Rs4)—, wherein y1 and y2 are 1 and y3, y4 and y5 are independently 0 or 1, Rk1, Rk2, Rk3, Rk4 and Rk5 are independently H, CH3, or OR2a, and Rs1, Rs2, Rs3, and Rs4 are independently H or CH3, wherein R2a is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe; and
R5 is H or OR2b, wherein R2b is H, an alkyl group, an aryl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe; provided that the compound is not dictyostatin 1.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • The present application claims the benefit of U.S. Provisional Patent Application Ser. No. 60/408,503, filed Sep. 6, 2002 and U.S. Provisional Patent Application Ser. No. 60/437,736 filed Jan. 2, 2003, the disclosures of which are incorporated herein by reference.
  • GOVERNMENT INTEREST
  • This invention was made with government support under grant CA 78039 awarded by the National Institutes of Health. The government has certain rights in this invention.
  • BACKGROUND OF THE INVENTION
  • The present invention relates to analogs of discodermolide and dictyostatin-1, intermediates for the synthesis of such analogs and methods of synthesis of such intermediates and analogs.
  • References set forth herein may facilitate understanding of the present invention or the background of the present invention. Inclusion of a reference herein, however, is not intended to and does not constitute an admission that the reference is available as prior art with respect to the present invention.
  • The discovery and development of new chemotherapeutic agents for the treatment of cancer is currently of high importance. Some of the best currently available chemotherapeutic agents are natural products or natural product analogs. For example, Taxol (paclitaxel) is a natural product that is currently being used to treat patients with breast and ovarian cancer among others. A number of analogs of Taxol, including Taxotere (docetaxel), are also powerful anticancer agents.
  • Recently, the natural product (+)-discodermolide and its analogs have shown great promise as anticancer agents. Discodermolide has been shown to have a mechanism of action similar to Taxol but it is active against Taxol-resistant cell lines and it is more water soluble than Taxol. Accordingly, it may have a different and/or broader spectrum of action than Taxol and be easier to formulate and administer. Like Taxol, discodermolide is difficult to synthesize. Some syntheses of discodermolide are described in the following papers: Nerenberg, J. B.; Hung, D. T.; Somers, P. K.; Schreiber, S. L. Total synthesis of the immunosuppressive agent (−)-discodermolide. J. Am. Chem. Soc. 1993, 115, 12621-12622; Smith, A. B., III; Qiu, Y.; Jones, D. R.; Kobayashi, K. Total Synthesis of (−)-Discodermolide. J. Am. Chem. Soc. 1995, 117, 12011-12012; Marshall, J. A.; Johns, B. A. Total synthesis of (+)-discodermolide. J. Org. Chem. 1998, 63, 7885-7892; Paterson, I.; Florence, G. J.; Gerlach, K.; Scott, J. P. Total synthesis of the antimicrotubule agent (+)-discodermolide using boron-mediated aldol reactions of chiral ketones. Angew. Chem., Int. Ed. Eng. 2000, 39, 377-380; Paterson, I.; Florence, G. J. Synthesis of (+)-discodermolide and analogues by control of asymmetric induction in aldol reactions of gamma-chiral (Z)-enals. Tetrahedron Lett. 2000, 41, 6935-6939; Smith, A. B.; Beauchamp, T. J.; LaMarche, M. J.; Kaufman, M. D.; Qiu, Y. P. et al. Evolution of a gram-scale synthesis of (+)-discodermolide. J. Am. Chem. Soc. 2000, 122, 8654-8664.
    Figure US20060270862A1-20061130-C00002
  • Analogs of discodermolide have also been made and tested for activity. For example, see the above references and Paterson, I.; Florence, G. J.; Gerlach, K.; Scott, J. P.; Sereinig, N. A practical synthesis of (+)-discodermolide and analogues: Fragment union by complex aldol reactions. J. Am. Chem. Soc. 2001, 123, 9535-9544; Martello, L. A.; LaMarche, M. J.; He, L.; Beauchamp, T. J.; Smith, A. B. et al. The relationship between taxol and (+)-discodermolide: synthetic analogs and modeling studies. Chemistry Biol. 2001, 8, 843-855; Harried, S. S.; Yang, G.; Strawn, M. A.; Myles, D. C. Total Synthesis of (−)-Discodermolide: An Application of a Chelation-Controlled Alkylation Reaction. J. Org. Chem. 1997, 62, 6098-6099; Paterson, I.; Florence, G. J. Synthesis of (+)-discodermolide and analogues by control of asymmetric induction in aldol reactions of gamma-chiral (Z)-enals. Tetrahedron Lett. 2000, 41, 6935-6939.
  • Unlike Taxol, discodermolide is not readily available in large quantities from natural sources. Accordingly, assuring a sufficient supply of discodermolide is problematic. Simplified analogs that retain high anti-cancer activity but are easier to make are in urgent need.
  • Very recently, an unusual macrolactone natural product dictyostatin 1 has been isolated from two different sponges and a partial structure has been assigned as shown below. See Pettit, G. R.; Cichacz, Z. A. Isolation and structure of dictyostatin 1. In U.S. Pat. No. 5,430,053; 1995; Pettit, G. R.; Cichacz, Z. A.; Gao, F.; Boyd, M. R.; Schmidt, J. M. Isolation and structure of the cancer cell growth inhibitor dictyostatin 1. J. Chem. Soc., Chem. Commun. 1994, 1111-1112. The configurations at C16 and C19 have not yet been assigned in the natural product and the absolute configuration is not known. Dictyostatin shows extremely high potencies against and array of cancer cell lines.
    Figure US20060270862A1-20061130-C00003
  • Recently, dictyostatin has also been shown to stabilize microtubules, like discodermolide and Taxol. See Wright, A. E.; Cummins, J. L.; Pomponi, S. A.; Longley, R. E.; Isbrucker, R. A. Dictyostatin compounds for stabilization of microtubules. In PCT Int. Appl.; WO62239, 2001. Accordingly, dictyostatin 1 and its analogs show great promise as new anticancer agents. There is an urgent need for a synthetic route to make dictyostatin 1 and its analogs in order to fully assign the structure of dictyostatin 1, to produce analogs to study the structure/activity relationship and to identify and produce the best possible drugs in this family.
  • The inventors of the present invention, as one aspect of the present invention, herein set forth a number of analogs of discodermolide, as well as methods and intermediates for the synthesis thereof. The inventors of the present invention, as another aspect of the present invention, herein set forth a family of both closed and open analogs of dictyostatin 1 with methods and intermediates for the synthesis of this family.
  • SUMMARY OF THE INVENTION
  • In one aspect, the present invention provides a compound of the following structure:
    Figure US20060270862A1-20061130-C00004

    wherein R1 is H, an alkyl group, an aryl group, an alkenyl group, an alkynyl group, or a halogen atom;
    R2 is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe;
    Ra, Rb and Rc are independently an alkyl group or an aryl group;
    Rd is an alkyl group, an aryl group, an alkoxylalkyl group, —RiSiRaRbRc or a benzyl group, wherein Ri is an alkylene group;
    Re is an alkyl group, an allyl group, a benzyl group, an aryl group, an alkoxy group, or —NRgRh, wherein Rg and Rh are independently H, an alkyl group or an aryl group;
    R3 is (CH2)n where n is and integer in the range of 0 to 5, —CH2CH(CH3), —CH═CH—, —CH═C(CH3)—, or —C≡C—;
    R4 is (CH2)p where p is an integer in the range of 4 to 12, (CHRk1)y1(CHRk2)y2(CHRk3)y3(CHRy4)y4 (CHRk5)y5C(Rs1)═C(Rs2)C(Rs3)═C(Rs4), —(CHRk1)y1(CHRk2)y2(CHRk3)y3(CHRk4)y4(CHRk5)y5CH(Rs1)CH(Rs2)C(Rs3)═C(Rs4)—, —(CHRk1)y1(CHRk2)y2(CHRk3)y3(CHRk4)y4(CHRk5)y5C(Rs1)═C(Rs2)CH(Rs3)CH(Rs4)—, —(CHRk1)y1(CHRk2)y2(CHRk3)y3(CHRk4)y4(CHRk5)y5CH(Rs1)CH(Rs2)CH(Rs3)CH(Rs4)—, wherein y1 and y2 are 1 and y3, y4 and y5 are independently 0 or 1, Rk1, Rk2, Rk3, Rk4 and Rk5 are independently H, CH3, or OR2a, and Rs1, Rs2, Rs3, and Rs4 are independently H or CH3, wherein R2a is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe; and
    R5 is H or OR2b, wherein R2b is H, an alkyl group, an aryl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe; provided that the compound is not dictyostatin 1.
  • When groups including, but not limited to, —SiRaRbRc, CH2ORd, and/or CORe are set forth as a substituent for more than one group in compounds of the claims and the specification of the present invention (for example, as a substituent of R2, R2a, Rs1, Rs2, Rs3, Rs4 and R5 above), it is to be understood that the groups of those substituents (Ra, Rb, Rc, Rd, and Re in this example), are independently, the same of different within each group and amoung the groups.
  • In one embodiment, the compound has the following stereostructure, or its enantiomer:
    Figure US20060270862A1-20061130-C00005

    wherein R1 is alkenyl; R2 is H; R3 is —CH2CH(CH3) or —CH═C(CH3); and R4 is —(CHRk1)y1(CHRk2)y2(CHRk3)y3(CHRk4)y4(CHRk5)y5C(Rs1)═C(Rs2)C(Rs3)C(Rs4)— wherein y1-y4 are 1, y5 is 0, Rk1 and Rk3 are OH, Rk2 is H, Rk4 is CH3, Rs1, Rs2, Rs3 and Rs4 are H, and R5 is OH.
  • In one embodiment, R1 is —CH═CH2 and R4 is
    Figure US20060270862A1-20061130-C00006
  • In another aspect, the present invention provides a compound of the following structure:
    Figure US20060270862A1-20061130-C00007

    wherein R1 is H, an alkyl group, an aryl group, an alkenyl group, an alkynyl group, or a halogen atom;
    R2 and R2d are independently H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORE;
    Ra, Rb and Rc are independently an alkyl group or an aryl group;
    Rd is an alkyl group, an aryl group, an alkoxylalkyl group, —RiSiRaRbRc or a benzyl group, wherein Ri is an alkylene group;
    Re is an alkyl group, an allyl group, a benzyl group, an aryl group, an alkoxy group, or —NRgRh, wherein Rg and Rh are independently H, an alkyl group or an aryl group;
    R3 is (CH2)n where n is and integer in the range of 0 to 5, —CH2CH(CH3)—, CH═CH—, —CH═C(CH3)—, or —C≡C—;
    R4 is (CH2)p where p is an integer in the range of 4 to 12, —(CHRk1)y1(CHRk2)y2(CHRk3)y3(CHRk4)y4(CHRk5)y5C(Rs1)═C(Rs2)C(Rs3)═C(Rs4), —(CHRk1)y1(CHRk2)y2(CHRk3)y3(CHRk4)y4(CHRk5)y5CH(Rs1)CH(Rs2)C(Rs3)═C(Rs4), —(CHRk1)y1(CHRk2)y2(CHRk3)y3(CHRk4)y4(CHRk5)y5C(Rs1)═C(Rs2)CH(Rs3)CH(Rs4)—, —(CHRk1)y1(CHRk2)y 2(CHRk3)y3(CHRk4)y4(CHRk5)y5CH(Rs1)CH(Rs2)CH(Rs3)CH(Rs4)—, wherein y1 and y2 are 1 and y3, y4 and y5 are independently 0 or 1, Rk1, Rk2, Rk3, Rk4 and Rk5 are independently H, —CH3, or OR2a, and Rs1, Rs2, Rs3, and Rs4 are independently H or CH3, wherein R2a is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe; and
    R5 is H or OR2b, wherein R2b is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe; and
    R10 is H or alkyl.
  • In one embodiment, the compound has the following stereostructure, or its enantiomer
    Figure US20060270862A1-20061130-C00008

    wherein R1 is alkenyl; R2 is H; R2d is H, OC(O)CH3 or OC(O)NRgRh wherein Rg and Rh are independently H, an alkyl group or an aryl group; R3 is CH2CH(CH3) or CH═C(CH3); and R4 is —(CHRk1)y1(CHRk2)y2(CHRk3)y3(CHRk4)y4(CHRk5)y5C(Rs1)═C(Rs2)C(Rs3)═C(Rs4)— wherein y1-y4 are 1, y5 is 0, Rk1 and RK3 are OH, Rk2 is H, Rk4 is CH3, Rs1, Rs2, Rs3 and Rs4 are H, R5 is OH; and R10 is H or alkyl.
  • In another embodiment, R1 is —CH═CH2, and R2d is H, OC(O)CH3 or OC(O)NH2.]
  • In a further aspect, the present invention provides a compound of the following structure:
    Figure US20060270862A1-20061130-C00009

    wherein R1 is H, an alkyl group, an aryl group, an alkenyl group, an alkynyl group, or a halogen atom;
    R2 and R2d are independently H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe;
    Ra, Rb and Rc are independently an alkyl group or an aryl group;
    Rd is an alkyl group, an aryl group, an alkoxylalkyl group, —RiSiRaRbRc or a benzyl group, wherein Ri is an alkylene group;
    Re is an alkyl group, an allyl group, a benzyl group, an aryl group, an alkoxy group, or —NRgRh, wherein Rg and Rh are independently H, an alkyl group or an aryl group;
    R3 is (CH2)n where n is and integer in the range of 0 to 5, —H2CH(CH3)—, —CH═CH—, —CH═C(CH3), or —C≡C—;
    R5 is H or OR2b, wherein R2b is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe;
    R11a and R11b are independently H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, CORe, or R11a and R11b together form a portion of six-membered acetal ring incorporating CRtRu;
    Rt and Ru are independently H, an alkyl group, an aryl group or an alkoxyarly group; and
    R12 is a halogen atom, CH2OR2c, CHO, CO2R10, CH═CHCH2OR2cC, CH═CHCHO, wherein R2c is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe, and R10 is H or alkyl.
  • In one embodiment, the compound has the following stereostructure, or its enantiomer
    Figure US20060270862A1-20061130-C00010

    wherein R1 is alkenyl; R2d is H, OC(O)CH3 or OC(O)NRgRh wherein Rg and Rh are independently H, an alkyl group or an aryl group; R3 is CH2CH(CH3) or CH═C(CH3); R11a and R11b are H or together form a portion of a six-membered acetal ring containing C(H)(p-C6H4OCH3) or C(CH3)2; R12 is a halogen atom, CH2OR2c, CHO, CO2R10, CH═CHCH2OR2c, CH═CHCHO, wherein R2c is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe, and R10 is H or alkyl.
  • In another embodiment, R1 is —CH═CH2, R2d is H, —OC(O)CH3 or —OC(O)NH2, and R12 is —CH2OH, —CHO or —CO2R10.
  • In another aspect, the present invention provides a compound having the following structure:
    Figure US20060270862A1-20061130-C00011

    wherein R2 is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe;
    Ra, Rb and Rc are independently an alkyl group or an aryl group;
    Rd is an alkyl group, an aryl group, an alkoxylalkyl group, —RiSiRaRbRc or a benzyl group, wherein Ri is an alkylene group;
    Re is an alkyl group, an allyl group, a benzyl group, an aryl group, an alkoxy group, or —NRgRh, wherein Rg and Rh are independently H, an alkyl group or an aryl group;
    R3 is (CH2)n where n is and integer in the range of 0 to 5, —H2CH(CH3)—, —CH═CH—, —CH═C(CH3), or —C≡C—;
    R5 is H or OR2b, wherein R2b is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe;
    R11a and R11b are independently H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, CORe, or R11a and R11b together form a portion of six-membered acetal ring containing CRtRu;
    Rt and Ru are independently H, an alkyl group, an aryl group or an alkoxy aryl group;
    R12 is a halogen atom, CH2OR2c, CHO, CO2R10, CH═CHCH2OR2c or CH═CHCHO, CH═CHCO2R10, wherein R2c is H, an alkyl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe, and R10 is H or alkyl; and
    R14a and R14b are independently H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, CORe, or R14a and R14b together form a six-membered ring containing CRvRw, wherein Rv and Rw are independently H, an alkyl group, an aryl group or an alkoxyaryl group.
  • In one embodiment, the compound has the following stereostructure, or its enantiomer
    Figure US20060270862A1-20061130-C00012

    R2 is H; R2d is H, OC(O)CH3 or OC(O)NRgRh wherein Rg and Rh are independently H, an alkyl group or an aryl group; R3 is CH2CH(CH3) or CH═C(CH3); R11a and R11b are H or together form a portion of a six-membered acetal ring containing C(H)(p-C6H4OCH3) or C(CH3)2; R12 is a halogen atom, CH2OR2c, CHO, CO2R10, CH═CHCH2OR2C, CH═CHCHO or CH═CHCO2R10, wherein R2c is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe, and R10 is H or alkyl; and R14a and R14b are H or together form a portion of a six-membered acetal ring containing C(H)(p-C6H4OCH3) or C(CH3)2.
  • In another aspect, the present invention provides a compound having the following formula
    Figure US20060270862A1-20061130-C00013

    wherein R1 is H, an alkyl group, an aryl group, an alkenyl group, an alkynyl group, or a halogen atom;
    R2 is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe;
    Ra, Rb and Rc are independently an alkyl group or an aryl group;
    Rd is an alkyl group, an aryl group, an alkoxylalkyl group, —RiSiRaRbRc or a benzyl group, wherein Ri is an alkylene group;
    Re is an alkyl group, an allyl group, a benzyl group, an aryl group, an alkoxy group, or —NRgRh, wherein Rg and Rh are independently H, an alkyl group or an aryl group;
    R3 is (CH2)n where n is and integer in the range of 0 to 5, —H2CH(CH3), —H═CH—, —CH═C(CH3)—, or —C≡C—;
    R11a and R11b are independently H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, CORe, or R11a and R11b together form a portion of six-membered acetal ring containing CRtRu;
    Rt and Ru are independently H, an alkyl group, an aryl group or an alkoxyarly group;
    R12 is a halogen atom, CH2OR2c, CHO, CO2R10, CH═CHCH2OR2c, CH═CHCHO or CH═CHCO2R10, wherein R2c is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe, and R10 is H or alkyl; and
    R14a and R14b are independently H, an alkyl group, and aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, CORe, or R14a and R14b together form a six-membered ring containing CRvRw, wherein Rv and Rw are independently H, an alkyl group, an aryl group or an alkoxyaryl group.
  • In one embodiment, the compound has the following stereostructure, or its enantiomer
    Figure US20060270862A1-20061130-C00014

    wherein R3 is CH2CH(CH3) or CH═C(CH3); R11a and R11b are H or together form a portion of a six-membered acetal ring containing C(H)(p-C6H4OCH3) or C(CH3)2; R12 is a halogen atom, CH2OR2c, CHO, CO2R10, CH═CHCH2OR2c, CH═CHCHO or CH═CHCO2R10, wherein R2c is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe, and R10 is H or alkyl; and R14a and R14b are H or together form a portion of a six-membered acetal ring containing C(H)(p-C6H4OCH3) or C(CH3)2.
  • In a further aspect, the present invention provides a compound having the following formula
    Figure US20060270862A1-20061130-C00015

    wherein R2 is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe;
    Ra, Rb and Rc are independently an alkyl group or an aryl group;
    Rd is an alkyl group, an aryl group, an alkoxylalkyl group, —RiSiRaRbRc or a benzyl group, wherein Ri is an alkylene group;
    Re is an alkyl group, an allyl group, a benzyl group, an aryl group, an alkoxy group, or —NRgRh, wherein Rg and Rh are independently H, an alkyl group or an aryl group;
    R11a and R11b are independently H, an alkyl group, and aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, CORe, or R11a and R11b together form a portion of six-membered acetal ring containing CRtRu;
    Rt and Ru are independently H, an alkyl group, an aryl group or an alkoxyarly group;
    R12 is a halogen atom, CH2OR2c, CHO, CO2R10, CH═CHCH2OR2c, CH═CHCHO or CH═CHCO2R10, wherein R2c is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORE, and R10 is H or alkyl;
    R16 is H or alkyl; and
    R17 is CH2OR2c, CHO, CO2R10, wherein R2f is H, an alkyl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe.
  • In one embodiment, the compound has the following stereostructure, or its enantiomer
    Figure US20060270862A1-20061130-C00016

    wherein R2 is H, an alkyl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe, R11a and R11b are H or together form a portion of a six-membered acetal ring containing C(H)(p-C6H4OCH3) or C(CH3)2; and R16 is H or alkyl.
  • In still a further aspect, the present invention provides a compound having the following formula
    Figure US20060270862A1-20061130-C00017

    wherein R1 is H, an alkyl group, an aryl group, an alkenyl group, an alkynyl group, or a halogen atom;
    R2, R2d and R2e are independently H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe;
    Ra, Rb and Rc are independently an alkyl group or an aryl group;
    Rd is an alkyl group, an aryl group, an alkoxylalkyl group, —RiSiRaRbRc or a benzyl group, wherein Ri is an alkylene group;
    Re is an alkyl group, an allyl group, a benzyl group, an aryl group, an alkoxy group, or —NRgRh, wherein Rg and Rh are independently H, an alkyl group or an aryl group;
    R3 is (CH2)n where n is and integer in the range of 0 to 5, —H2CH(CH3), —CH═CH—, —CH═C(CH3), or —C≡C—;
    R5 is H or OR2b, wherein R2b is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe;
    q is an integer in the range of 2 to 5;
    R18 is H, and R19 is hydroxy, alkoxy or —SRz, wherein Rz is an alkyl group or an aryl group, or R18 and R19 taken together are ═O.
  • In one embodiment, the compound has the following stereostructure, or its enantiomer
    Figure US20060270862A1-20061130-C00018
  • In one embodiment of the compound, R1 is a CH═CH2 and R3 is (Z)-CH═CH—, or —CH2CH2—.
  • In a further aspect, the present invention provides a compound having the following structure
    Figure US20060270862A1-20061130-C00019

    R11a and R11b are independently H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, CORe, or R11a and R11b together form a portion of six-membered acetal ring containing CRtRu;
    Rt and Ru are independently H, an alkyl group, an aryl group or an alkoxyarly group;
    Ra, Rb and Rc are independently an alkyl group or an aryl group;
    Rd is an alkyl group, an aryl group, an alkoxylalkyl group, —RiSiRaRbRc or a benzyl group, wherein Ri is an alkylene group;
    Re is an alkyl group, an allyl group, a benzyl group, an aryl group, an alkoxy group, or —NRgRh, wherein Rg and Rh are independently H, an alkyl group or an aryl group;
    R20 is CH2OR2g, CHO, CO2R10; wherein R2g is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe, and wherein R10 is H or alkyl; and
    R21 is a halogen atom, CH2OR2c, CHO, CO2R11a, CH═CHCH2OR2c, CH═CHCHO or CH═CHCO2R10, wherein R2c is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe, and wherein R10a is H or alkyl.
  • In one embodiment, the compound has the following stereostructure, or its enantiomer
    Figure US20060270862A1-20061130-C00020

    wherein R11a and R11b are independently H, an alkyl group, and aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, CORe, or R11a and Rb together form a portion of six-membered acetal ring incorporating CRtRu;
    Rt and Ru are independently H, an alkyl, an aryl group or an alkoxyarly group;
    Ra, Rb and Rc are independently an alkyl group or an aryl group;
    Rd is an alkyl group, an aryl group, an alkoxylalkyl group, —RiSiRaRbRc or a benzyl group, wherein Ri is an alkylene group;
    Re is an alkyl group, an allyl group, a benzyl group, an aryl group, an alkoxy group, or —NRgRh, wherein Rg and Rh are independently H, an alkyl group or an aryl group;
    R20 is CH2OR2g, CHO, CO2R10; wherein R2g is H, an alkyl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe, and wherein R10 is H or alkyl; and
    R21 is a halogen atom, CH2OR2c, CHO, CO2R10a, CH═CHCH2OR2c, CH═CHCHO, CH═CHCO2R10 wherein R2c is H, an alkyl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe, and wherein R10a is H or alkyl.
  • In one embodiment, R11a and R11b are H or together form a portion of a six-membered acetal ring containing C(H)(p-C6H4OCH3) or C(CH3)2; R21 is a halogen atom, CH2OR2c, CHO, CO2R10, CH═CHCH2OR2c, CH═CHCHO, wherein R2c is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe, and R10 is H or alkyl.
  • In another embodiment, R1 is CH═CH2, and R21 is CH2OH, CHO or CO2R10.
  • In still another aspect, the present invention provides a compound having the following formula
    Figure US20060270862A1-20061130-C00021

    wherein R13 is H or an alkyl group, R14 is H, an aryl group, an alkoxyaryl group or an alkyl group, and R22 is a halogen atom or —P(Ar)3X, wherein X is a counteranion selected from the groups halide, tetrafluoroborate, hexafluorophosphate and sulfonate, provided that when R13 and R14 are methyl groups, X is not I. In one embodiment, when R13 and R14 are alkyl groups, X is not halogen.
  • In another embodiment, the compound has the following stereostructure, or its enantiomer
    Figure US20060270862A1-20061130-C00022

    wherein R13 is H or an alkyl group, and R14 is H, an aryl group, an alkoxyaryl group, or an alkyl group, an aryl group or an alkoxyarly group, R22 is a halogen atom or —P(Ar)3X, wherein X is a counteranion selected from the groups halide, tetrafluoroborate, hexafluorophosphate and sulfonate, provided that when R13 and R14 are methyl groups, X is not 1.
  • In one embodiment, R13 is H, R14 is aryl, and R22 is P(C6H5)3X. In another embodiment, R14 is C6H4-p-OCH3.
  • In still a further aspect, the present invention provides a process for conversion of a first compound with the formula
    Figure US20060270862A1-20061130-C00023

    wherein R1 is H, an alkyl group, an aryl group, an alkenyl group, an alkynyl group, or a halogen atom;
    R2 is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe;
    R2d is H
    Ra, Rb and Rc are independently an alkyl group or an aryl group;
    Rd is an alkyl group, an aryl group, an alkoxylalkyl group, —RiSiRaRbRc or a benzyl group, wherein Ri is an alkylene group;
    Re is an alkyl group, an allyl group, a benzyl group, an aryl group, an alkoxy group, or —NRgRh, wherein Rg and Rh are independently H, an alkyl group or an aryl group;
    R3 is (CH2)n where n is and integer in the range of 0 to 5, —CH2CH(CH3)—, CH═CH—, —CH═C(CH3)—, or —C≡C—;
    R4 is (CH2)p where p is an integer in the range of 4 to 12, —(CHRk1)y1(CHRk2)y2(CHRk3)y3(CHRk4)y4(CHRk5)y5C(Rs1)═C(Rs2)C(Rs3)═C(Rs4)—, —(CHRk1)y1(CHRk2)y2(CHRk3)y3(CHRy4)y4(CHRk5)y5CH(Rs1)CH(Rs2)C(Rs3)═C(Rs4), —(CHRk1)y1(CHRy2)y2(CHRk3)y3(CHRk4)y4(CHRk5)y5C(Rs1)═C(Rs2)CH(Rs3)CH(Rs4)—, —(CHRk1)y1(CHRk2)y2(CHRk3)y3(CHRk4)y4(CHRk5)y5CH(Rs1)CH(Rs2)CH(Rs3)CH(Rs4)—, wherein y1 and y2 are 1 and y3, y4 and y5 are independently 0 or 1, Rk1, Rk2, Rk3 k, Rk4 and Rk5 are independently H, CH3, or OR2a, and Rs1, Rs2, Rs3, and Rs4 are independently H or CH3, wherein R2a is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe; and
    R5 is H or OR2b, wherein R2b is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe; and
    R10 is H;
    to a second compound with the formula
    Figure US20060270862A1-20061130-C00024

    comprising the step of reacting the first compound under conditions suitable to effect macrolactonization.
  • In one embodiment, the process is for conversion of a compound with the following stereostructure or its enantiomer
    Figure US20060270862A1-20061130-C00025

    wherein R1 is H, an alkyl group, an alkenyl group, an alkynyl group, or a halogen atom;
    R2 is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe;
    R2d is H
    Ra, Rb and Rc are independently an alkyl group or an aryl group;
    Rd is an alkyl group, an aryl group, an alkoxylalkyl group, —RiSiRaRbRc or a benzyl group, wherein Ri is an alkylene group;
    Re is an alkyl group, an allyl group, a benzyl group, an aryl group, an alkoxy group, or —NRgRh, wherein Rg and Rh are independently H, an alkyl group or an aryl group;
    R3 is (CH2)n where n is and integer in the range of 0 to 5, —H2CH(CH3), CH═CH—, —CH═C(CH3)—, or —C—;
    R4 is (CH2)p where p is an integer in the range of 4 to 12, —(CHRk1)y1(CHRk2)y2(CHRk3)y3(CHRk4)y4(CHRk5)y5C(Rs1)═C(Rs2)C(Rs3)═C(Rs4)—, —(CHRk1)y1(CHRk2)y2(CHRk3)y3(CHRk4)y4(CHRk5)y5CH(Rs1)C(Rs2)═C(Rs4)—, —(CHRk1)y1(CHRk2)y2(CHRk3)y3(CHRk4)y4(CHRk5)y5C(Rs1)═C(Rs2)CH(Rs3)CH(Rs5)—, —(CHRk1)y1(CHRk2)y2(CHRk3)y3(CHRk4)y4(CHRk5)y5CH(Rs1)CH(Rs2)CH(Rs3)CH(Rs4)—, wherein y1 and y2 are 1 and y3, y4 and y5 are independently 0 or 1, Rk1, Rk2, Rk3, Rk4 and Rk5 are independently H, —CH3, or OR2a, and Rs1, Rs2, Rs3, and Rs4 are independently H or CH3, wherein R2a is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe;
    R5 is H or OR2b, wherein R2b is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe; and
    R10 is H
    to a second compound with the formula
    Figure US20060270862A1-20061130-C00026
  • In one embodiment of the process, R1 is alkenyl; R3 is CH2CH(CH3) or CH═C(CH3); and R4 is —(CHRk1)y1(CHRk2)y2(CHRk3)y3(CHRk4)y4(CHRk5)y5C(Rs1)═C(Rs2)C(Rs3)═C(Rs4)— wherein y1-y4 are 1, y5 is 0, Rk1 and Rk3 are R2a, Rk2 is H, Rk4 is CH3, Rs1-Rs4 are H, and R5 is OR2b.
  • In another embodiment of the process, R1 is CH═CH2 and R4 is
    Figure US20060270862A1-20061130-C00027
  • In one embodiment of the process, the first compound is converted by reacting the first compound with 2,4,6-trichlorobenzoylchloride.
  • The above general structures for the compounds of the present invention include all stereoisomers thereof (other than the natural compound dictyostatin 1). Moreover, the structures of the compounds of the present invention include the compounds in racemic form, enantiomerically enriched form or enantiomerically pure form.
  • The terms “alkyl”, “aryl” and other groups refer generally to both unsubstituted and substituted groups unless specified to the contrary. In that regard, the groups set forth above can be substituted with a wide variety of substituents to synthesize analogs retaining biological activity. Unless otherwise specified, alkyl groups are hydrocarbon groups and are preferably C1-C15 (that is, having 1 to 15 carbon atoms) alkyl groups, and more preferably C1-C10 alkyl groups, and can be branched or unbranched, acyclic or cyclic. The above definition of an alkyl group and other definitions apply also when the group is a substituent on another group (for example, an alkyl group as a substituent of an alkylamino group or a dialkylamino group). The term “aryl” refers to phenyl or naphthyl. As used herein, the terms “halogen” or “halo” refer to fluoro, chloro, bromo and iodo.
  • The term “alkoxy” refers to —OR, wherein R is an alkyl group. The term “alkenyl” refers to a straight or branched chain hydrocarbon group with at least one double, bond, preferably with 2-15 carbon atoms, and more preferably with 2-10 carbon atoms (for example, —CH═CHR or —CH2CH═CHR; wherein R can be a group including, but not limited to, an alkyl group, an alkoxyalkyl group, an amino alkyl group, an aryl group, or a benzyl group). The term “alkynyl” refers to a straight or branched chain hydrocarbon group with at least one triple bond, preferably with 2-15 carbon atoms, and more preferably with 2-10 carbon atoms (for example, —C≡CR or —CH2—C≡CR; wherein R can be a group including, but not limited to, an alkyl group, an alkoxyalkyl group, an amino alkyl group, an aryl group, or a benzyl group). The terms “alkylene,” “alkenylene” and “alkynylene” refer to bivalent forms of alkyl, alkenyl and alkynyl groups, respectively.
  • The term “trityl” refers to a triphenyl methyl group or —C(Ph)3.
  • Certain groups such as amino and hydroxy groups may include protective groups as known in the art. Preferred protective groups for amino groups include tert-butyloxycarbonyl, formyl, acetyl, benzyl, p-methoxybenzyloxycarbonyl, trityl. Other suitable protecting groups as known to those skilled in the art are disclosed in Greene, T., Wuts, P. G. M., Protective Groups in Organic Synthesis, Wiley (1991), the disclosure of which is incorporated herein by reference.
  • Other aspects of the present invention include the synthesis of the compounds of the preent invention as well as the biological assaying of such compound and the biological activity of such compounds against, for example, cancer (such as breast, prostate cancer and ovarial cancer). For example, in another aspect, the present invention provides a method of treating a patient for cancer, including the step of administering a pharmaceutically effective amount of a biologically active compound of the present invention or a pharmaceutically acceptable salt thereof.
  • The present invention, along with the attributes and attendant advantages thereof, will best be appreciated and understood in view of the following detailed description taken in conjunction with the accompanying drawings.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 illustrates one embodiment of the syntheses of a representative isomeric aldehyde for incorporation of the left part of the configuration of (+)-discodermolide.
  • FIG. 2 illustrates one embodiment of the syntheses of another representative isomeric aldehyde for incorporation of the left part of the configuration of (+)-discodermolide.
  • FIG. 2 illustrates one embodiment of the syntheses of an intermediate for the center part of the configuration of (+)-discodermolide.
  • FIG. 4 illustrates one embodiment of the construction of the right fragment or part of the molecule.
  • FIG. 5 illustrates an embodiment of the synthesis of several discodermolide analogs of the present invention.
  • FIGS. 6A through D illustrate the tubulin polymerization-inducing properties of discodermolide (Figure A), as well as discodermolide analog compounds 40 (Figure B), 41 (Figure C) and 42 (Figure D) of the present invention in comparison to 10 μM paclitaxel (PTX).
  • FIG. 7 illustrates a retrosynthetic analysis of a dictyostatin analog of the present invention.
  • FIG. 8 illustrates an embodiment of the coupling of three fragments of a dictyostatin analog of the present invention.
  • FIG. 9 illustrates the structures of several acyclic compounds of the present invention that were tested for biological activity.
  • FIG. 10 illustrates an embodiment of the synthesis of a lower fragment of a dictyostatin analog of the present invention.
  • FIG. 11 illustrates an embodiment of the synthesis of a macrolactone of the present invention.
  • FIG. 12 illustrates a second representative general scheme for synthesis of stereoisomers and analogs of dictyostatin.
  • FIG. 13 illustrates a summary of an embodiment of the synthesis of the bottom fragment of dictyostatin analogs of the present invention.
  • FIG. 14 illustrates an embodiment of the coupling of the bottom fragment of FIG. 13 with the center fragment of the dictyostatin analogs of the present invention.
  • FIG. 15 illustrates an embodiment of the introduction of the C16 stereocenter and introduction of the top fragment of the dictyostatin analog of the present invention.
  • FIG. 16 illustrates an embodiment of the completion of the synthesis of the dictyostatin analog of the present invention.
  • FIG. 17 illustrates embodiments of representative methods to make analogs of the terminal diene fragment of the dictyostatin analog of the present invention.
  • FIG. 18 illustrates a summary of an embodiment of the synthesis of the bottom fragment of the dictyostatin analog of the present invention.
  • FIG. 19 illustrates an embodiment of the synthesis of two fragments with anti/anti configurations as assigned to dictyostatin 1 at C13-C15.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Synthesis of Simplified Analogs of Discodermolide: The inventors of the present invention hypothesized that active analogs of discodermolide would result after removal of the C14 and C16 methyl groups and the C7 hydroxy group. These deletions greatly simplify the synthesis by allowing the two cis-disubstituted alkenes of analogs 1 and 2 to be made by Wittig-type reactions. A family of simple analogs 1 were shown to have moderate activity. However, by incorporating a lactone in place of the simple ester side chains of 1, the inventors of the present invention have discovered anti-cancer agents of increased activity that are still significantly simpler to make than discodermolide.
    Figure US20060270862A1-20061130-C00028
  • The syntheses of two representative isomeric aldehydes 9a and 9b for, incorporation of the left part of these molecules are shown in FIGS. 1 and 2. he synthesis of the left display bearing the configuration of (+)-discodermolide started with the commercially available methacrolein 3 (FIG. 1). Reaction of 3 with the boron enolate of Evans oxazolidinone 4 gave the corresponding alcohol, which was silylated to give compound 5 in 90% yield. Lactonization followed by the introduction of allyl group was performed by the previously reported method to give 6 in good yield. See Day, B. W.; Kangani, C. O.; Avor, K. S. Convenient syntheses of (2R,3S,4R)-3-(tert-butyidimethylsilanyloxy)-2,4-dimethyl-5-oxopentanoic acid methoxymethyl-amide from methacrolein. Preparation of C1-C7 and C17-C24 fragments of (+)-discodermolide. Tetrahedron Asymmetry 2002, 13, 1161-1165. Lactone opening, oxidation and allylation gave 7. Conversion to the methyl acetal was accomplished by DIBAL (diisobutylaluminum hydride) reduction to the corresponding lactol followed by treatment with camphorsulfonic acid (CSA) in methanol to give a desilylated intermediate, which was protected with methoxymethyl chloride (MOMCl) to give a mixture of anomers 8 (β:α=1:1). These were separable by silica gel flash chromatography. The final left fragment 9a was obtained by hydroboration of the α-anomer 8 with BH3-DMS (borane-dimethysulfide) followed by oxidation with SO3-pyridine.
  • The synthesis of the C4-epi lactone left display 9b started from 1,4-butanediol (FIG. 2). Monoprotection with PMB bromide (PMB is p-methoxybenzyl) was performed with NaH in DMF. After oxidation, reaction of the crude aldehyde 10 with the boron enolate of Evans oxazolidinone 4 gave the corresponding syn-alcohol, which was silylated to give compound 11 in 90% yield. Lithium borohydride reduction followed by oxidation gave the aldehyde 12. A second syn-aldol addition was performed with same Evans oxazolidinone 4 to give the corresponding alcohol, which was protected with MOM chloride to give compound 13. Desilylation of 13 with HF gave the cyclized product 14 in high yield. Conversion to the methyl acetal was easily accomplished by DIBAL reduction to the corresponding lactol followed by treatment with PPTS (pyridinium p-toluene sulfonate) in methanol to give 15 as a 2.5:1 (x[3) mixture of anomers from which the major anomer (a) was isolated by silica gel flash chromatography. The final left fragment 9b was obtained by hydrogenolysis of the PMB protecting group followed by Dess-Martin oxidation.
  • These synthetic routes are flexible and substantially any stereoisomer of the lactone 9 can be made with the appropriate chiral auxiliary and reaction conditions.
  • Center intermediate 21 was prepared as shown in FIG. 3. Oxazolidinone 18 was prepared from (S)-3-hydroxy-2-methylpropionic methyl ester 16 by the known procedure for the preparation of ent-18. See Clark, D. L.; Heathcock, C. H. Studies on the alkylation of chiral enolates: application toward the total synthesis of discodermolide. J. Org. Chem. 1993, 58 5878-5879. Reduction of 18 with lithium borohydride gave the diol, which was protected by anisaldehyde dimethyl acetal 19 to give the acetal 20. Deprotection of the primary TBS group with tetrabutylammonium fluoride (TBAF), iodination, and treatment with triphenylphosphine afforded the phosphonium salt 21 in 72% yield. Phosphonium salts 22 and 23 were also used for Wittig olefination, but the results were unsatisfactory. Smith also encountered difficulties with related Wittig reagents in discodermolide synthesis. See: Smith, A. B.; Beauchamp, T. J.; LaMarche, M. J.; Kaufman, M. D.; Qiu, Y. P. et al. Evolution of a gram-scale synthesis of (+)-discodermolide. J. Am. Chem. Soc. 2000, 122, 8654-8664. These difficulties may result, at least in part, from the hygroscopic nature of the phosphonium salts. In contrast, compound 21 is a white, non-hygroscopic solid and its Wittig reactions were reliable even though no special care was taken with its storage (for example, salt 21 was useful for reactions even after it was stored at room temperature for several months).
  • As shown in FIG. 4, the construction of the right fragment 34 featured aldol reactions. Syn-Aldol reaction of aldehyde 24 with 4 provided 25, which was reduced to a diol and protected with anisaldehyde dimethyl acetal 19 to give 26. Selective opening of the benzylidine ring of 26 with DIBAL gave a primary alcohol, which was oxidized to aldehyde 27. The subsequent anti-aldol condensation using Heathcock's aldol reaction with dimethylphenyl propionate 28 furnished compound 29 as the major product in 73% yield. See Heathcock, C. H.; Pirrung, M. C.; Montgomery, S. H.; Lampe, J. Acyclic stereoselection-13; Aryl esters: reagents for threo-aldolization. Tetrahedron 1981, 37, 4087-4095. The relative configuration of intermediate 29 was confirmed by 13C and 1H NMR analyses of the corresponding acetonide 34. Silylation of the newly formed hydroxyl group of 29, reduction of the aryl ester with DIBAL and Dess-Martin oxidation of the resultant primary alcohol afforded aldehyde 30. The Z-diene moiety was introduced by a two-step procedure developed by Paterson and coworkers using a Nozaki-Hiyama reaction. See Paterson, I.; Schlapbach, A. Studies towards the total synthesis of the marine-derived immunosuppresant discodermolide: stereoselective synthesis of a C9-C24 subunit. Synlett. 1995, 498-500. Addition of the aldehyde 30 and allyl bromide 31 to a suspension of CrCl2 in THF produced an intermediate β-hydroxy silane (not shown), which upon treatment with NaH underwent syn elimination to generate the required Z-diene 32. Selective deprotection of the primary TBS group, iodination, and then treatment with triphenylphosphine gave the phosphonium salt 33 in good yield.
  • The first Wittig reaction of 9a with 21 provided 35 (see FIG. 5). DIBAL reductive cleavage of the acetal followed by Dess-Martin oxidation gave aldehyde 37. Likewise, aldehyde 38 was made from 9b via 36. The second Wittig olefination was accomplished with 38 as an example and phosphonium salt 33 (FIG. 5). Tetrabutylammonium fluoride deprotection followed by carbamoylation using Kocovsky's method (See Kocovsky, P. Carbamates: a method of synthesis and some synthetic applications. Tetrahedron Lett. 1986, 27 5521-5524) afforded the C19 carbamate-containing compound 39. The lactone was built from the methyl acetal in the left fragment by using aqueous 60% acetic acid in THF followed by Dess-Martin oxidation. Deprotected analog 40 containing a free C3 hydroxy group on the lactone was obtained by the removal of MOM group with 4N HCl followed by removal of the PMB protecting groups with DDQ oxidation. Two additional example compounds, 41 and 42, were prepared from the intermediate 39 by using appropriate conditions. All three of these analogs exhibited significant activity, as shown in FIGS. 6A-D and Table 1. Surprisingly, the C3-MOM-protected analogs 41 and 42 showed better microtubule hypernucleation activities than the analog 40 with a free C3-hydroxy group. As can be seen in FIG. 6A, discodermolide is superior to paclitaxel (taxol) in that it causes equivalent microtubule assembly at both lower concentrations and temperatures (the increase in absorbance caused by discodermolide occurs at a time point earlier than that caused by paclitaxel). Additionally, the polymer induced to form by discodermolide is more resistant to cold-induced disassembly than is the paclitaxel-induced polymer. Both analogs 41 (FIG. 6C) and 42 (FIG. 6D) showed these more rapid polymer-inducing and cold-resistant properties, albeit at somewhat lower potencies (for example, higher concentrations of the analogs were necessary for these effects to be detected) than discodermolide. MOM ether lactone 41 was the most potent among these analogs.
  • Table 1 shows microtubule stabilizing, antiproliferative, and paclitaxel-displacing properties of 40-42. Again, the lactone, MOM ether 41 was more potent than the lactol 42 or free hydroxy 40 relatives. The cellular activity of 41 was good, showing a submicromolar 50% growth inhibitory (GI50) concentration. This compound also showed considerable affinity for the paclitaxel binding site on tubulin. A 2-fold molar excess of 41 displaced [3H]paclitaxel from microtubules better than paclitaxel and at almost the same potency as discodermolide.
    TABLE 1
    Microtubule stabilizing, antiproliferative and paclitaxel-displacing properties of
    compounds 40-42 in comparison to (+)-discodermolide (1) and paclitaxel.
    MT
    stabilizing Displacement of
    activity MDA-MB231 GI50 (μM)b 2008 [3H]paclitaxel
    Compound (%)a (breast) PC3 (prostate) (ovarian) (%)c
    discodermolide >100 0.016 ± 0.003 0.067 ± 0.004 0.072 ± 0.005 64 ± 2
    paclitaxel 100 0.0024 ± 0.0016 0.015 ± 0.002 0.0092 ± 0.0016 37 ± 1
    40 11 2.1 ± 1.8 7.5 ± 2.0 5.2 ± 1.0 21 ± 1
    41 27 0.87 ± 0.21 1.8 ± 0.9 0.65 ± 0.25 57 ± 2
    42 11 3.4 ± 0.8 15 ± 3  4.7 ± 0.6 19 ± 2

    aPercent tubulin assembly induced by test agent at 10 μM vs. that caused by 10 μM paclitaxel (100%); single determinations at 30° C.

    bConcentrations at which test agent caused 50% inhibition of cell growth; means (N = 4 over 10 concentrations) ± SD after 72 h of continuous exposure to the agent.

    cPercent displacement by 4 μM test agent of 2 μM [3H]paclitaxel bound to microtubules formed from 2 μM tubulin and 20 μM dideoxyGTP.
  • Macrocycle 43 is a representative example of a dictyostatin analog with an alkyl chain bridging the lactone carbonyl group and the C10/C11 alkene and with two Z-double bonds in the macrocycle. It can also be considered as a macrocyclic analog of discodermolide. This can be synthesized convergently from three components-33, 21 and 44-via sequential Wittig couplings and a macrocyclization (FIG. 7). This design allows the synthesis of substantially any stereoisomer by employing the desired isomer of the relevant precursor—21 or 33.
  • Fragment 45 was synthesized from 1,10-decanediol (not shown) by mono-TBS protection (NaH/TBSCl, 42%) followed by Dess-Martin oxidation (80%). Fragment 21 was prepared as shown in FIG. 3. Fragment 33 was prepared as shown in FIG. 4.
  • The coupling of the three fragments is summarized in FIG. 8. Generation of the ylide from phosphonium salt 21 and NaHDMS followed by addition of aldehyde 44 gave the Wittig product in good yield (75%) provided that the reaction was conducted at high concentration (1M in 21). The formation of the Z-alkene was confirmed by the 10 Hz coupling constant between the vinyl protons. Selective opening of the PMB acetal was accomplished by addition of 3 equiv of DIBAL to give a primary alcohol. This was oxidized to an aldehyde under Dess-Martin conditions. Wittig conditions similar to those above were then deployed to prepare 45 from this aldehyde and phosphonium salt 33.
  • Selective deprotection of 45 was achieved using HF-pyridine and the resulting primary alcohol was oxidized to acid 46. The other TBS group was then removed with TBAF. Using the Yamaguchi protocol, the macrolactone ring was then constructed. PMB deprotection using DDQ provided target product 43, whose protons and carbons were assigned by COSY and HMQC NMR experiments. The location of the macrolactone ring was confirmed by HMBC NMR experiments.
  • Acyclic compounds 47, 48 and 49 were readily made from appropriate synthetic intermediates (45 or 46) in reasonable yields (FIG. 9).
  • These analogs were tested for antiproliferative activity in vitro against two human cancer cell lines (Table 2). Macrolactone 43 and non-cyclized alcohol 47 and ester 48 exhibited similar 50% growth inhibitory concentrations, in the 15-30 μM range. Carboxylic acid 49 was inactive (>50 μM) possibly due to poor cell membrane penetration. The modest activity of these compounds is encouraging given the simplicity and flexibility of their lower chain.
  • We therefore decided to introduce the more complex bottom part of dictyostatin-1 lacking only the C9′-OH group. The synthesis of a lower fragment more closely related to dictyostatin is shown in FIG. 10. Synthesis of the needed aldehyde 51 (FIG. 10) started from the intermediate 25, which was reduced to an alcohol with LiBH4, followed by PMB acetal protection as in FIG. 4. Selective acetal opening produced alcohol 50, which was subjected to Dess-Martin oxidation to give aldehyde 27 (see FIG. 4). Wittig-Homer reaction, and removal of the TBS group with HF-pyridine gave a primary alcohol, which was oxidized to aldehyde 51.
  • Center part Wittig salt 21 (FIG. 3) was reacted with aldehyde 51 to give the (Z)-olefin (FIG. 11). This was followed by selective PMP acetal opening with NaCNBH3-TMSCl to yield a primary alcohol. The aldehyde obtained after Dess-Martin oxidation was again subjected to Wittig reaction with 33 to generate 52. Ester 52 was reduced to the alcohol with DIBAL, followed by Dess-Martin oxidation and application of the Still (Z)-variant of the Wittig reaction to afford (E,Z) doubly unsaturated ester 53. Selective removal of the TBS groups was accomplished by exposure to 3N HCl-MeOH in THF (1:1). The resulting ester was hydrolyzed by using 1N KOH in refluxing in EtOH. Finally, the Yamaguchi lactonization protocol followed by DDQ deprotection gave macrolactone 54, whose structure was confirmed by HMBC and other NMR experiments. No isomerization of either of the dienes or the isolated cis-alkenes was detected.
  • Compound 54 proved to be quite potent in terms of antiproliferative activity against human carcinoma cells (Table 2) showing a 50% growth inhibitory concentration against breast and ovarian cancer cells of about 1 μM. Furthermore compound 54 displaced [3H]paclitaxel stoichiometrically bound to microtubules at about ⅓rd the potency of discodermolide.
    TABLE 2
    Human Cancer Cell Growth Inhibitory and Paclitaxel Displacing
    Properties of Macrolactone Discodermolide Analogs
    GI50(μM)a
    MDA-MB-231 2008 Displacement of
    (breast) (ovary) [3H]paclitaxel (%)c
    43   27 ± 1   16 ± 1 18 ± 5
    47   18 ± 1   22 ± 5 21 ± 2
    48   26 ± 3   19 ± 2 17 ± 1
    49 >50 >50 16 ± 3
    54  1.4 ± 0.1b  1.0 ± 0.1b 27 ± 8
    discodermolide 0.016 ± 0.003b 0.072 ± 0.005b 64 ± 2

    aFifty percent growth inhibitory concentration after 48 or 72b hours of continuous exposure (mean ± standard deviation; N = 4).

    cPercent displacement by 4 μM test agent of 2 μM [3H]paclitaxel bound to microtubules formed from 2 μM tubulin and 20 μM dideoxyGTP (N = 6, means ± SD).
  • A second representative general strategy for the synthesis of stereoisomers and close analogs of dictyostatin is shown in FIG. 12. Again the molecule is dissected such that every stereocenter can be controlled and modified either by starting with an appropriate precursor or through an asymmetric reaction allowing access to both possible isomers.
  • FIG. 13 summarizes the synthesis of the bottom fragment. (S)-Diethyl maleate was reduced and the resulting diol was converted to acetonide 55. Reduction to the aldehyde and standard Evans aldol reaction gave 56. Reduction of this to the aldehyde and Wittig-Homer Emmons reaction gave 57. Removal of the acetonide and silyation gave 58, which was mono-desilylated to 59 and oxidized to aldehyde 60.
  • Coupling of the bottom fragment with the center fragment and elaboration are shown in FIG. 14. Wittig reaction of 21 (FIG. 3) and 60 proceeded smoothly to form 61, which was hydrolyzed and reduced to give 62. After tritylation to 63, DIBAL reduction gave 64. Oxidation and Wittig-Homer reaction produced 65, which was reduced to give 66 and hydrolyzed to acid 67. Activation of 67 as the mixed anhydride preceded conversion to the oxazolidinone 68.
  • Introduction of the C16 stereocenter and introduction of the top fragment are shown in FIG. 15. Evans asymmetric alkylation to 69 followed by removal of the chiral auxiliary by reduction gave 70. Separately, reagent 71 was made from the Evans oxazolindinone by displacement with LiCH2P(O)(OMe)2. Dess-Martin oxidation and Horner-Emmons olefination with 71 then gave 72, which was reduced with NiCl2/NaBH4 to 73. Now reduction with sodium borohydride gave a 2.8/1 mixture of stereoisomers, which could be separated and converted to the final products independently. Silylation to 75 followed by DIBAL reduction gave 76, which was converted to diene 77 as described above. Detriylation then gave 78.
  • Completion of the synthesis is shown in FIG. 16. Dess-Martin oxidation and Still-Gennari olefination gave 79 which was deprotected with DDQ to 80. Saponification then gave the hydroxy acid 81 ready for macrocyclization. Treatment of 81 under the Yamaguchi protocol gave 82, which was finally deprotected to give the target product 83, an isomer of dictyostatin 1 called dictyostatin 5.
  • Representative methods to make analogs of the terminal diene fragment are shown in FIG. 17. Alkene 84 was ozonized to give the aldehyde, which was subjected to a Wittig reaction to give analogs like 85. Alternatively, 84 can be converted to the Z-vinyl iodide 86, which can in turn be coupled with organometallic reagents like phenyl zinc iodide to give 87. This combination of olefination and organometallic and related coupling methods allows access to a wide variety of groups in this position.
  • The versatility of the synthesis is illustrated by the preparations of representative additional fragments that can be used to make dictyostatin, its isomers and its analogs. FIG. 18 summarizes the synthesis of a fully elaborated bottom fragment 92. Acetal 88, readily prepared from (D)-malic acid, was silylated with t-butyidiphenylsilyl chloride (TBDPSCl). Reductive cleavage of the acetal with DIBAL followed by Swem oxidation provided aldehyde 89. Reaction of 89 with the indicated Z-crotyl boronate according to Roush (See: Roush, W. R.; Hoong, L. K.; Palmer, M. A. J.; Straub, J. A.; Palkowitz, A. D. Asymmetric synthesis using tartrate modified allyl boronates. 2. Single and double asymmetric reactions with alkoxy-substituted aldehydes, J. Org. Chem. 1990, 55, 4117-4126) provided 90 in 63% isolated yield. PMB protection, ozonolysis and Wittig-Horner olefination then gave 91. This was converted to the E/Z diene 92 by DIBAL reduction, Dess-Martin oxidation, Still-Gennari olefination and desilylation with HF/pyridine.
  • FIG. 19 shows the synthesis of two fragments with anti/anti configurations as assigned to dictyostatin 1 at C13-C15. Evan anti-aldol reaction of ent-4 and methacrolein (See: Evans, D. A.; Tedrow, J. S.; Shaw, J. T.; Downey, C. W. Diastereoselective magnesium halide-catalyzed anti-aldol reactions of chiral N-acyloxazolidinones. J. Am. Chem. Soc. 2002, 124, 392-393) followed by TFA treatment gave 93 in 78% yield. A minor diastereomer of 93 (about 16/1 ratio) was separated by chromatography. Silylation of 93 followed by hydroboration and oxidation provided alcohol 94 in 75% yield alongside the lactone resulting from cyclization of the terminal hydroxyl group with displacement of the chiral auxiliary (not shown, 10% yield). Silylation of 94 and reductive cleavage of the auxiliary provided 95, which was oxidized to 96 by the Swem method. Related aldehyde 98 was made by Roush allylboration of ent-17 (see 17 in FIG. 3) with the indicated E-crotylborate (mismatched case, 4/1 selectivity) to give 97, followed by reaction with PMBBr and ozonolysis.
  • EXAMPLES
  • (4R)-4-Benzyl-3-[(2R,3R)-3-(tert-butyidimethylsilanyloxy)-2,4-dimethylpent-4-enoyl]oxazolidin-2-one (5). TBDMSOTf (3.44 mL, 15 mmol) was added to a stirred solution of aldol product (3.03 g, 10 mmol) and 1,6-lutidine (2.32 mL, 20 mmol) in CH2Cl2 (20 mL) at −78° C. and the mixture was stirred for 2 h at ambient temperature. The reaction was quenched by the addition of aqueous HCl (0.5 N, 50 mL). The resulting mixture was extracted with CH2Cl2 and dried over MgSO4 followed by the evaporation of solvent under reduced pressure. The product was purified by short column chromatography (hexane/EtOAc 9:1). Crude 5 was used without purification.
  • (2S,3R,4S,5R)-2-Allyl-6-methoxy-4-methoxymethoxy-3,5-dimethyltetrahydropyran (8). Diisobutylaluminum hydride (1.0 M in THF, 3.3 mL, 3.3 mmol) was added dropwise to a stirred solution of 7 (894 mg, 3 mmol) in anhydrous CH2Cl2 (30 mL) under an atmosphere of N2 at −78° C. and the resulting mixture was stirred for an additional 1 h at −78° C. The reaction was quenched by the careful addition of saturated aqueous potassium sodium tartrate (50 mL) and stirred for 3 h at room temperature. Once the organic and aqueous layers had separated, the aqueous layer was extracted with CH2Cl2. The combined organic layer was washed with brine and dried over MgSO4 followed by the evaporation of the solvent under reduced pressure. The crude lactol was used for the next reaction without further purification.
  • A solution of the lactol and CSA (0.3 mmol) in MeOH was stirred for 24 h at room temperature. The reaction mixture was diluted with EtOAc (100 mL) and washed with saturated NaHCO3 (50 mL). The aqueous layer was extracted with EtOAc (50 mL). The combined organic layer was dried over MgSO4. The solvent was removed under reduced pressure and the crude product was used for the next reaction.
  • N,N-Diisopropylethylamine (7.5 mL) and chloromethyl methyl ether (1.13 mL, 15 mmol) were added to a solution of the alcohol in CH2Cl2 (15 mL). The reaction mixture was heated to reflux and stirred overnight. The reaction was quenched with aqueous saturated NaHCO3 (50 mL) followed by washing with brine. The aqueous layer was extracted with CH2Cl2 (2×50 mL). The combined organic layer was dried over MgSO4. The solvent was removed under reduced pressure and the crude product was purified by column chromatography (hexane/EtOAc 7:3) to provide the pure anomers of 8 (β, 33%; α, 32%). {overscore (β)}8: IR (CHCl3) 3053, 2985, 2305, 1422, 1264 cm−1; 1H NMR (300 MHz, CDCl3) δ 6.04 (m, 1H), 5.20-5.10 (m, 2H), 4.81 (d, 1H, J=6.9 Hz), 4.73 (d, 1H, J=2.37 Hz), 4.67 (d, 1H, J=6.8 Hz), 3.62 (m, 2H), 3.55 (s, 3H), 2.47 (m, 1H), 2.30 (m, 1H), 2.16 (m, 1H), 1.85 (m, 1H), 1.03 (d, 3H, J=7.1 Hz), 0.97 (d, 3H, J=6.8 Hz); 13C NMR (75 MHz, CDCl3) 135.1, 116.4, 101.3, 95.9, 81.9, 75.8, 56.4, 55.7, 37.5, 37.3, 33.6, 13.2, 9.9; HRMS (EI) calcd for C13H24O4 244.1596, found 244.1592. {overscore (α)}8: 1H NMR (300 MHz, CDCl3) δ 6.00 (m, 1H), 5.22-5.12 (m, 2H), 4.83 (d, 1H, J=6.9 Hz), 4.69 (d, 1H, J=7.2 Hz), 4.49 (d, 1H, J=1.8 Hz), 3.88 (dt, 1H, J=3.6, 8.8 Hz), 3.53 (t, 2H, J=3.6 Hz), 3.48 (s, 3H), 2.45 (m, 1H), 2.28-2.11 (m, 3H), 1.94 (m, 1H), 1.12 (d, 3H, J=7.3 Hz), 1.00 (d, 3H, J=6.9 Hz); 13C NMR (75 MHz, CDCl3) 135.2, 116.7, 103.4, 95.6, 78.7, 69.6, 55.7, 37.2, 35.8, 33.6, 15.9, 13.5.
  • (2S,3R,4S,5R,6R)-3-(6-Methoxy-4-methoxymethoxy-3,5-dimethyltetrahydropyran-2-yl)propionaldehyde (9a). BH3.Me2S (1 M in THF, 3 mL, 3 mmol) was added to a solution of 8 (488 mg, 2 mmol) in THF (10 mL) at 0° C. with stirring. The mixture was allowed to warm to room temperature and stirred for 3 h. The reaction was quenched with 2N aqueous NaOH (10 mL) followed by H2O2 (30%, 3 mL). After 1 h, the mixture was extracted with CH2Cl2 (3×20 mL). The combined organic layers were dried over anhydrous MgSO4, evaporated and chromatographed (hexane/EtOAc 7:3) to yield 392 mg (75%) of alcohol as a colorless oil: IR (CHCl3) 3103, 2982, 1375, 1240 cm−1; 1H NMR (300 MHz, CDCl3) δ 4.83 (d, 1H, J=6.9 Hz), 4.76 (d, 1H, J=2.4 Hz), 4.69 (d, 1H, J=6.9 Hz), 3.75 (t, 2H, J=5.4 Hz), 3.61 (t, 1H, J=2.7 Hz), 3.57 (s, 3H), 2.58 (br s, 1H), 2.17 (m, 1H), 1.90-1.80 (m, 4H), 1.04 (d, 3H, J=7.1 Hz), 0.97 (d, 3H, J=6.8 Hz); 13C NMR (75 MHz, CDCl3) 101.5, 96.0, 82.0, 75.9, 62.8, 56.6, 55.8, 37.6, 34.0, 29.4, 28.6, 13.4, 9.8; HRMS (EI) calcd for C13H26O5 262.1780, found 262.1792.
  • Pyridinium sulfurtrioxide (477 mg, 3 mmol) was added to a stirred solution of alcohol (262 mg, 1 mmol) and N,N-diisopropylethylamine (0.52 mL, 3 mmol) in anhydrous CH2Cl2 (6 mL) and DMSO (12 mL) at 0° C. The reaction mixture was stirred at the ambient temperature for 1 h. The mixture was diluted with ethyl ether (50 mL) and washed with aqueous HCl (0.5 N, 50 mL) and brine (10 ml). The organic layer was dried over MgSO4, filtered and concentrated under vacuum. Flash silica gel column chromatography filtration (hexane/EtOAc 4:1) to remove SO3-pyridine residue provided the crude aldehyde 9a as a colorless oil which was used without further purification.
  • (4R)-4-Benzyl-3-[(2R,3S)-3-(tert-butyldi methylsilyloxy)-6-(4-methoxybenzyloxy)-2-methylhexanoyl]oxazolidin-2-one (11). Pyridinium sulfur trioxide (7.15 g, 45 mmol) was added to a stirred solution of the mono-PMB-protected alcohol 10 (3.15 g, 15 mmol) and N,N-diisopropylethylamine (8.0 mL, 45 mmol) in anhydrous CH2Cl2 (15 mL) and DMSO (30 mL) at 0° C. The mixture was stirred at ambient temperature for 1 h, diluted with ethyl ether (300 mL) and washed with aqueous HCl (0.5 N, 200 mL), and brine. The separated organic layer was dried over MgSO4. Filtration and concentration provided the crude aldehyde 10 as a colorless oil which was used for the next reaction without further purification.
  • N,N-Diisopropylethylamine (1.9 mL, 11 mmol) was added to a solution of propionyloxaiolidinone (2.33 g, 10 mmol) in anhydrous CH2Cl2 (110 mL) at 0° C., followed by dropwise addition of Bu2BOTf (1.0 M in CH2Cl2, 11 mL, 11 mmol). The solution was stirred for 0.5 h at 0° C. Crude 10 in anhydrous CH2Cl2 (30 mL) was added at −78° C. The mixture was stirred for 10 min at −78° C. followed by an additional 2 h at 0° C. The reaction was quenched by addition of phosphate buffer, pH 7.0 (50 mL). A solution of hydrogen peroxide (30%, 10 mL) in methanol (20 mL) was added and the mixture was allowed to stir for 1 h at 0° C. After separation of organic and aqueous layers, the aqueous layer was extracted with CH2Cl2. The combined organic layers were dried over anhydrous MgSO4, evaporated and chromatographed (hexane/EtOAc 4:1) to yield the aldol adduct (3.83 g, 87%) as a colorless oil: IR (CHCl3) 3472, 2954, 2860, 2252, 1778, 1691, 1383 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.48-7.36 (m, 7H), 7.01 (d, 2H, J=8.7 Hz), 4.58 (m, 1H), 4.33 (s, 2H), 4.18 (br s, 1H), 3.94 (s, 3H), 3.91 (m, 1H), 3.63 (t, 2H, J=6.0 Hz), 3.40 (dd, 1H, J=3.2, 13.3 Hz), 3.37 (br s, 1H), 2.90 (dd, 1H, J=3.8, 13.3 Hz), 1.97-1.59 (m, 5H), 1.40 (d, 3H, J=7.2 Hz); 13C NMR (75 MHz, CDCl3) 177.3, 159.2, 153.1, 135.3, 130.6, 129.5, 129.3, 129.0, 127.4, 113.8, 72.5, 71.5, 69.9, 66.2, 55.2, 42.7, 37.7, 31.3, 26.4, 14.3, 11.1; HRMS (EI) calcd for C25H31NO6 441.2151, found 441.2162.
  • TBDMSOTf (1.7 mL, 7.5 mmol) was added to a stirred solution of the above alcohol (2.20 g, 5 mmol) and 2,6-lutidine (1.2 mL, 10 mmol) in CH2Cl2 (50 mL) at −78° C. and the mixture was stirred for 2 h at ambient temperature. The reaction was quenched by addition of aqueous HCl (0.5 N, 100 mL). The reaction mixture was extracted with CH2Cl2 and dried over MgSO4 followed by the evaporation of the solvent under reduced pressure. The product was purified by column chromatography (hexane/EtOAc 9:1) to yield 11: IR (CHCl3) 3020, 2955, 2858, 1779, 1362, 1211 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.45-7.36 (m, 7H), 7.00 (d, 2H, J=8.7 Hz), 4.70 (m, 1H), 4.56 (s, 2H), 4.27-4.15 (m, 3H), 4.04 (dd, 1H, J=5.4, 6.8), 3.91 (s, 3H), 3.60 (m, 3H), 3.40 (dd, 1H, J=3.0, 13.2 Hz), 2.90 (dd, 1H, J=9.5, 13.2 Hz), 1.80 (br m, 4H), 1.38 (d, 3H, J=6.8 Hz), 1.05 (s, 9H), 0.21 (s, 3H), 0.17 (s, 3H); 13C NMR (75 MHz, CDCl3) 175.4, 159.2, 153.2, 135.5, 130.9, 129.6, 129.3, 129.0, 127.4, 113.8, 72.9, 72.5, 70.2, 66.1, 55.9, 55.3, 42.8, 37.7, 32.1, 26.0, 25.2, 18.2, 12.2, −3.92, −4.65; LRMS (ESI) 578.3 (M+Na).
  • (2S,3S)-3-(tert-Butyidimethylsilanyloxy)-6-(4-methoxybenzyloxy)-2-methylhexan-1-ol. Lithium borohydride (2.0 M in THF, 5 mL, 10 mmol) was added dropwise to a stirred solution of 11 (2.77 g, 5 mmol) and methanol (0.4 mL, 10 mmol) in anhydrous THF (20 mL) under an atmosphere of N2 at 0° C. The mixture was stirred for 20 min at 0° C. and then warmed to ambient temperature. After 3 h at room temperature, the reaction was quenched with aqueous NH4Cl (100 mL) and extracted with CH2Cl2 (3×20 mL). The combined organic layers were dried over anhydrous MgSO4, evaporated and chromatographed (hexane/EtOAc 7:3) to yield the alcohol (1.48 g, 78%) as a colorless oil: IR (CHCl3) 2948, 2856, 2302, 1612, 1265 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.27 (d, 2H, J=8.5 Hz), 7.00 (d, 2H, J=8.5 Hz), 4.43 (s, 2H), 3.78 (s, 3H), 3.67 (dd, 1H, J=8.6, 10.5 Hz), 3.51-3.41 (m, 3H), 2.78 (br s, 1H), 1.94 (m, 1H), 1.72-1.49 (m, 4H), 0.90 (s, 9H), 0.81 (d, 3H, J=7.0 Hz), 0.09 (s, 3H), 0.08 (s, 3H); 13C NMR (75 MHz, CDCl3) 159.2, 130.6, 129.3, 113.8, 75.4, 72.6, 70.1, 65.8, 55.9, 55.3, 39.7, 29.1, 26.6, 25.9, 18.0, 12.1, −4.28, −4.38; LRMS (ESI) 405.2 (M+Na).
  • (4R)-4-Benzyl-3-[(2R,3S,4R,5S)-5-(tert-butyldi methylsilanyloxy)-3-hydroxy-8-(4-methoxybenzyloxy)-2,4-dimethyloctanoyl]oxazolidin-2-one (12). Pyridinium sulfur trioxide (2.38 g, 15 mmol) was added to a stirred solution of the above TBS-protected alcohol (1.91 g, 5 mmol) and N,N-diisopropylethylamine (2.65 mL, 15 mmol) in anhydrous CH2Cl2 (5 mL) and DMSO (10 mL) at 0° C. The mixture was stirred at the ambient temperature for 1 h, diluted with ethyl ether (100 mL), washed with aqueous HCl (0.5 N, 100 mL) and brine, then dried over MgSO4. Filtration and concentration provided the crude aldehyde as a colorless oil which was used without further purification.
  • N,N-Diisopropylethylamine (0.97 mL, 5.5 mmol) was added to a solution of propionyloxazolidinone (1.16 g, 5 mmol) in anhydrous CH2Cl2 (11 mL) at 0° C., followed by dropwise addition of Bu2BOTf (1.0 M in CH2Cl2, 5.5 mL, 5.5 mmol). The solution was stirred for 0.5 h at 0° C. A solution of crude aldehyde 12 from above in anhydrous CH2Cl2 (10 mL) was added at −78° C. The reaction mixture was stirred for 10 min at −78° C. then for 2 h at 0° C. The reaction mixture was quenched with phosphate buffer, pH 7.0 (50 mL). A solution of hydrogen peroxide (30%, 10 mL) in methanol (20 mL) was slowly added and the mixture was stirred for 1 h. After the separation of organic and aqueous layers, the aqueous layer was extracted with CH2Cl2. The combined organic layers were dried over anhydrous MgSO4, evaporated and chromatographed (hexane/EtOAc 4:1) to yield desired compound (2.29 g, 75%) as a colorless oil: IR (CHCl3) 2949, 2855, 2253, 1779, 1692, 1463 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.33-7.19 (m, 7H), 7.01 (d, 2H, J=8.1 Hz), 4.42 (m, 1H), 4.44 (s, 2H), 4.16 (d, 1H, J=5.1 Hz), 4.01 (m, 1H), 3.95 (t, 1H, J=6.3 Hz), 3.85 (m, 1H), 3.79 (s, 3H), 3.43 (br s, 2H), 3.24 (br s, 1H), 3.20 (dd, 1H, J=2.4, 13.5 Hz), 2.77 (dd, 1H, J=9.6, 13.2 Hz), 1.56-1.31 (m, 5H), 1.32 (d, 3H, J=6.9 Hz), 0.95 (d, 3H, J=6.9 Hz), 0.89 (s, 9H), 0.08 (s, 6H); 13C NMR (75 MHz, CDCl3) 177.2, 159.2, 152.7, 135.1, 130.6, 129.5, 129.3, 129.0, 127.5, 113.8, 76.8, 74.2, 72.6, 70.0, 66.1, 55.3, 55.0, 40.6, 38.1, 37.8, 31.3, 25.9, 18.1, 13.2, 7.4, −3.5, −4.6; HRMS (EI) calcd for C34H51NO7Si 613.3435, found 613.3427.
  • (4R)-4-Benzyl-3-[(2R,3S,4R,5S)-5-(tert-butyldimethylsilanyloxy)-8-(4-methoxybenzyloxy)-3-methoxymethoxy-2,4-dimethyloctanoyl]oxazolidin-2-one (13). N,N-Diisopropylethylamine (7.5 mL) and chloromethyl methyl ether (mL, 9 mmol) were added to a solution of the alcohol from above (1.83 g, 3 mmol) in CH2Cl2 (15 mL). The mixture was stirred at reflux overnight. The reaction was quenched with aqueous sat'd NaHCO3 (50 mL) and washed with brine. The aqueous layer was extracted with CH2Cl2 (2×50 mL). The combined organic layer was dried over MgSO4. The solvent was removed under reduced pressure and the crude product was purified by column chromatography (hexane/EtOAc 4:1) to provide the pure product in 92% yield: IR (CHCl3) 3020, 2862, 1781, 1215 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.36-7.27 (m, 7H), 6.94 (d, 2H, J=8.7 Hz), 4.75 (s, 2H), 4.69 (m, 1H), 4.53 (s, 2H), 4.19 (dd, 1H, J=10.2, 15.0 Hz), 3.97 (dd, 1H, J=3.0, 6.6 Hz), 3.84 (br s, 4H), 3.43 (br t, 2H), 3.45 (s, 3H), 3.30 (dd, 1H, J=3.0, 13.2 Hz), 2.77 (dd, 1H, J=9.3, 13.5 Hz), 1.81-1.75 (m, 5H), 1.36 (d, 3H, J=6.9 Hz), 1.02 (d, 3H, J=6.9 Hz), 0.98 (s, 9H), 0.16 (s, 3H), 0.15 (s, 3H); 13C NMR (75 MHz, CDCl3) 175.5, 159.1, 153.0, 135.4, 131.0, 129.5, 129.2, 129.0, 127.4, 113.7, 98.3, 80.3, 76.9, 73.2, 72.3, 70.5, 66.0, 56.3, 55.6, 55.2, 41.7, 40.8, 37.6, 30.6, 26.1, 24.2, 18.3, 14.0, 10.5, −3.9, −4.3; HRMS (EI) calcd for C34H50NO7Si (M−CH2OCH3) 612.3356, found 612.3367 (M−CH2OCH3).
  • 6-[(3R,4S,5S,6S)-3-(4-Methoxybenzyloxy)propyl]-4-methoxymethoxy-3,5-dimethyltetrahydropyran-2-one (14). HF-pyridine (6 mL) was added to a solution of 13 (1.31 g, 2 mmol) in MeOH (20 mL) and pyridine (10 mL) at 0° C. The mixture was stirred at room temperature for 48 h, diluted with EtOAc (100 mL), washed with aqueous HCl (0.5 N, 2×50 mL) and with brine. The aqueous layer was extracted with EtOAc (50 mL). The combined organic layer was dried over anhydrous Na2SO4. The solvent was removed under reduced pressure, and the crude product was purified by column chromatography (hexane/EtOAc 4:1) to provide the pure product in 83% yield: IR (CHCl3) 3020, 2952, 1730, 1513, 1216 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.24 (d, 2H, J=8.7 Hz), 6.87 (d, 2H, J=8.7 Hz), 4.72 (d, 1H, J=7.2 Hz), 4.60 (d, 1H, J=7.2 Hz), 4.48 (m, 1H), 4.43 (s, 2H), 3.80 (s, 3H), 3.50-3.39 (m, 2H), 3.39 (s, 3H), 3.26 (dd, 1H, J=1.1, 7.0 Hz), 2.58 (t, 1H, J=6.9 Hz), 2.03 (d, 1H, J=7.5 Hz), 1.81-1.63 (m, 4H), 1.31 (d, 3H, J=6.7 Hz), 0.91 (d, 3H, J=7.3 Hz); 13C NMR (75 MHz, CDCl3) 174.1, 159.2, 130.5, 129.3, 113.8, 95.5, 82.6, 76.7, 72.6, 69.4, 55.9, 55.3, 40.5, 38.4, 28.6, 26.0, 14.4, 11.9, −3.9; HRMS (EI) calcd for C20H30O6 366.2042, found 366.2050.
  • 2-Methoxy-6-[(3R,4S,5S,6S)-3-(4-methoxybenzyloxy)propyl]-4-methoxymethoxy-3,5-dimethyltetrahydropyran (15). Diisobutylaluminum hydride (1.0 M in THF, 2.2 mL, 2.2 mmol) was added dropwise to a stirred solution of 14 (732 mg, 2 mmol) in anhydrous CH2Cl2 (20 mL) under an atmosphere of N2 at −78° C. and the mixture was stirred for 1 h at −78° C. The reaction was quenched by the careful addition of aqueous sat'd potassium sodium tartrate (50 mL) and stirring for 3 h at room temperature. Once the organic and aqueous layers separated, the aqueous layer was extracted with CH2Cl2. The combined organic layer was washed with brine and dried over MgSO4 followed by the evaporation of solvent under reduced pressure. The crude lactol obtained was used without further purification.
  • A solution of the lactol and PPTS (0.2 mmol) in MeOH was stirred for 15 h at room temperature. The reaction mixture was diluted with EtOAc (100 mL) and washed with sat'd aqueous NaHCO3 (50 mL). The aqueous layer was extracted with EtOAc (50 mL). The combined organic layer was dried over MgSO4. The solvent was removed under reduced pressure, and the crude product was purified by column chromatography (hexane/EtOAc 7:3) to provide the pure product each anomer 15 (β, 64%; α, 26%). β-15: IR (CHCl3) 3020, 2858, 2299, 1514, 1216 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.35 (d, 2H, J=8.7 Hz), 6.95 (d, 2H, J=8.7 Hz), 4.76 (d, 2H, J=3.0 Hz), 4.52 (s, 2H), 4.37 (d, 1H, J=4.6 Hz), 4.09 (m, 1H), 3.89 (s, 2H), 3.57 (m, 2H), 3.47 (s, 3H), 3.32 (t, 1H, J=5.7 Hz), 1.89-1.71 (m, 6H), 1.16 (d, 3H, J=7.2 Hz), 1.07 (d, 3H, J=7.9 Hz); 13C NMR (75 MHz, CDCl3) 159.2, 130.7, 129.3, 113.8, 103.2, 96.3, 82.0, 72.6, 69.9, 69.3, 55.7, 55.3, 39.1, 38.0, 27.1, 26.5, 16.0, 13.1; HRMS (EI) calcd for C21H34O6 382.2353, found 382.2355. α-15: 1H NMR (300 MHz, CDCl3) δ 7.34 (d, 2H, J=8.7 Hz), 6.95 (d, 2H, J=8.7 Hz), 4.72 (s, 2H), 4.70 (d, 1H, J=2.8 Hz), 4.52 (s, 2H), 4.09 (br m, 4H), 3.67 (br s, 1H), 3.56 (m, 5H), 3.44 (s, 3H), 2.08-1.54 (m, 6H), 1.11 (d, 3H, J=3.0 Hz), 1.08 (d, 3H, J=2.9 Hz).
  • (2S,3S,4S,5R,6R)-3-(6-Methoxy-4-methoxymethoxy-3,5-dimethyltetrahydropyran-2-yl)propionaldehyde (9b). A mixture of PMB ether 15 (458 mg, 1.2 mmol) and palladium (10% Pd/C, 5 mg) was stirred in EtOAc (12 mL) for 3 h at room temperature under an H2 atmosphere (balloon), filtered and concentrated to yield the debenzylated alcohol which was used without further purification.
  • The crude alcohol in CH2Cl2 (12 mL) was treated with Dess-Martin periodinane (636 mg, 1.5 mmol) at room temperature. The reaction was quenched with saturated aqueous NaHCO3 (20 mL). The aqueous layer was extracted with CH2Cl2 (10 mL×2) and the combined extracts were dried over anhydrous MgSO4. Filtration and concentration followed by short flash column chromatography (hexane/EtOAc 4:1) provided 274 mg (88%) of the crude aldehyde as a colorless oil which was used without further purification: 1H NMR (500 MHz, CDCl3) δ 9.80 (s, 1H), 4.67 (dd, 2H, J=7.0, 12.5 Hz), 4.27 (d, 1H, J=4.5 Hz), 3.99 (dd, 1H, J=3.5, 4.0 Hz), 3.36 (s, 6H), 3.24 (t, 1H, J=6.0 Hz), 2.61 (m, 1H), 2.52 (m, 1H), 1.83 (m, 3H), 1.68 (m, 1H), 1.05 (d, 3H, J=7.0 Hz), 1.01 (d, 3H, J=7.5 Hz).
  • (2S,3R,4S)-5-(tert-Butyldimethylsilanyloxy)-2,4-dimethylpentane-1,3-diol. MeOH (0.51 mL) and LiBH4 (2.0 M in THF, 6.2 mL, 12.4 mmol) were added dropwise to a stirred solution of aldol product 18 [22] (5.38 g, 12.3 mmol) in THF (50 mL) at 0° C. After stirring for 1 h at 0° C., saturated aqueous sodium potassium tartrate (70 mL) was added. The mixture was allowed to warm room temperature and extracted with CH2Cl2 (2×50 mL). The combined organic layer was washed with brine (40 mL), dried over anhydrous MgSO4, concentrated and flash column chromatographed (hexane/EtOAc 4:1) to yield 2.99 g (92%) of the desired product as a colorless oil: IR (CHCl3) 3409, 2958, 2927, 2853, 2878, 1469, 1385, 1361, 1252, 1082, 838, 773 cm−1; 1H NMR (300 MHz, CDCl3) δ 4.45 (br s, 1H), 3.54 (br s, 1H), 1.92 (m, 1H), 1.83 (m, 1H), 1.06 (d, 3H, J=6.98 Hz), 1.00 (s, 9H), 0.84 (d, 3H, J=6.88 Hz), 0.19 (s, 6H); 13C NMR (75 MHz, CDCl3) 79.3, 69.7, 67.5, 37.4, 36.6, 25.9, 18.1, 12.8, 8.9, −5.5, −5.6; LRMS (EI) 263 (M+H); HRMS (EI) calcd for C13H30O3Si 263.2042, found 263.2042; [α]20 D+35.5 (c 0.85, CHCl3).
  • (2S)-tert-Butyl-{(4R,5S)-2-[2-(4-methoxyphenyl)-5-methyl[1,3]dioxan-4-yl]propoxy}dimethylsilane (20). A solution of the above diol (2.8 g, 10.7 mmol), p-anisaldehyde dimethylacetal (2.0 mL, 11.7 mmol) and PPTS (0.27 g, 1.1 mmol) in benzene was heated to reflux for 3 h. The solvent was evaporated under reduced pressure and the residue was purified by column chromatography (hexane/EtOAc 9:1) to give 20 (2.6 g, 6.8 mmol) in 64% yield: IR (CHCl3) 2955, 2927, 2853, 1617, 1518, 1459, 1382, 1157, 1101, 1033, 826 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.49 (m, 2H), 6.98 (m, 2H), 5.50 (m, 2H), 3.89 (s, 3H), 3.82 (dd, 1H, J=10.9, 4.9 Hz), 3.76 (dd, 2H, J=8.1, 2.8 Hz), 1.87 (m, 1H), 1.71 (m, 1H), 1.23 (d, 3H, J=7.6 Hz), 1.00 (d, 3H, J=6.5 Hz); 13C NMR (75 MHz, CDCl3) 160.1, 132.1, 127.6, 113.8, 113.7, 101.9, 80.1, 74.3, 65.2, 64.3, 55.5, 37.4, 30.0, 26.3, 26.2, 18.7, 12.4, 11.3, −5.0, −5.1; LRMS (EI) 323, 207, 187, 157, 145, 121, 75; HRMS (EI) calcd for C21H36O4Si1 323.1678 (M−tBu), found 323.1694 (M−tBu); [α]20 D −33.6 (c 1.24, CHCl3).
  • (2S)-2-[(4R,5S)-2-(4-Methoxyphenyl)-5-methyl[1,3]dioxan-4-yl]propan-1-ol. TBAF (11.0M in THF, 22 mL, 22 mmol) was added to a solution of 20 (2.8 g, 7.3 mmol) in THF (70 mL) at room temperature and the mixture was stirred for 2 h. The mixture was diluted with ethyl ether (100 mL) and brine. The organic layer was dried over MgSO4. Filtration and concentration followed by flash column chromatography (hexane/EtOAc 7:3) provided alcohol (1.95 g, 7.2 mmol) as a yellow oil: IR (CHCl3) 3428, 2964, 2930, 2835, 1614, 1512, 1463, 1391, 1249, 1098, 1027 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.38 (m 2H), 6.87 (m, 2H), 5.48 (s, 1H), 4.11 (dd, 2H, J=4.6, 4.5 Hz), 3.75 (s, 3H), 3.73 (m, 2H), 3.52 (apparent t, 1H, J=11.1 Hz), 2.08 (m, 1H), 2.00 (m, 1H), 1.04 (d, 3H, J=7.1 Hz), 0.77 (d, 3H, J=6.7 Hz); 13C NMR (75 MHz, CDCl3) 160.0, 131.5, 127.4, 113.6, 101.6, 83.4, 73.9, 66.3, 55.2, 36.8, 30.4, 11.9, 9.9; LRMS (EI) 266, 207, 177, 153, 135, 77; HRMS (EI) calcd for C15H22O4 266.1518, found 266.1517; [α]20 D −4.8 (c 0.67, CHCl3).
  • (2S)-{2-[(4R,5S)-2-(4-Methoxyphenyl)-5-methyl[1,3]dioxan-4-yl]propyltriphenyl-□5-phosphane iodide (21). I2 (4.48 g, 17.6 mmol) was added at 0° C. to a solution of the alcohol from above (2.35 g, 8.82 mmol) in CH2Cl2 (110 mL) containing imidazole (1.32 g, 19.4 mmol) and triphenylphosphine (4.63 g, 17.6 mmol). The resulting slurry was stirred for 1 h and quenched with saturated aqueous Na2S2O3(10 mL). The organic layer was separated and washed with water (20 mL), brine and dried over anhydrous MgSO4. The solvent was evaporated under reduced pressure and the residue was purified by column chromatography (hexane/EtOAc 9:1) to give the pure iodide.
  • The iodide was quickly dissolved in benzene (44 mL), PPh3 was added (11.5 g, 44.1 mmol) and the mixture heated to reflux for 36 h. The reaction mixture was cooled to room temperature and anhydrous ethyl ether (50 mL) was added, whereupon a white solid precipitated. Filtration followed by washing of the solid with ethyl ether (10 mL) provided the phosphonium salt (4.5 g) as a white foam: IR (CHCl3) 3054, 2961, 2909, 1611, 1515, 1435, 1246, 1107, 993, 752 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.55 (m, 9H), 7.37 (m, 6H), 7.21 (m, 2H), 6.65 (m, 1H), 5.41 (s, 1H), 3.95 (d, 1H, J=10.2 Hz), 3.68 (d, 2H, J=12.3 Hz), 3.54 (s, 3H), 3.26 (m, 1H), 1.85 (m, 1H), 1.46 (apparent d, 1H, J=6.5 Hz), 0.78 (d, 3H, J=6.8 Hz), 0.44 (d, 3H, J=6.6 Hz); 13C NMR (75 MHz, CDCl3) 160.0, 135.2, 135.1, 133.7, 133.5, 131.1, 130.5, 130.4, 127.9, 119.0, 117.9, 113.5, 101.9, 82.3, 82.1, 73.2, 55.5, 30.7, 29.2, 25.3, 15.7, 10.4 HRMS (EI) calcd for C33H36O3P 511.2402, found 511.2428; [α]20 D +31.9 (c 0.78, CHCl3).
  • (4R,5R)-tert-Butyl-{3-[2-(4-methoxyphenyl)-5-methyl[1,3]dioxan-4-yl]propoxy}dimethylsilane (26). Lithium borohydride (2.0 M in THF, 25 mL, 50 mmol) was added dropwise to a stirred solution of 25 (8.70 g, 20 mmol) and MEOH (1.61 mL, 40 mmol) in anhydrous THF (100 mL) under an atmosphere of N2 at 0° C. The mixture was stirred for 20 min at 0° C. and then warmed to ambient temperature. After 2 h at room temperature, the reaction was quenched with aqueous NH4Cl (100 mL) and extracted with CH2Cl2 (3×10 mL). The combined organic layers were dried over anhydrous MgSO4, evaporated and chromatographed (hexane/EtOAc 7:3) to yield 4.97 g (95%) of the diol as a colorless oil.
  • A solution of the diol (2.62 g, 10 mmol), anisaldehyde dimethyl acetal (2.00 g, 11.0 mmol), and PPTS (0.1 equiv) in benzene was stirred for 15 h at reflux. The reaction mixture was quenched with aqueous sat'd NaHCO3 (50 mL) followed by washing with water. The aqueous layer was extracted with ethyl ether (2×50 mL). The combined organic layer was dried over MgSO4. The solvent was removed under reduced pressure and the residue was purified by column chromatography (hexane/EtOAc 7:3) to provide the pure 26 in 72% yield: IR (CHCl3) 2992, 1742, 1373, 1240 cm−1; 1H NMR (500 MHz, CDCl3) δ 7.43 (d, 2H, J=8.4 Hz), 6.88 (d, 2H, J=8.4 Hz), 5.45 (s, 1H), 4.08 (dd, 2H, J=10.5, 29.9 Hz), 3.90 (br s, 1H), 3.80 (s, 3H), 3.67 (m, 2H), 1.67-1.50 (m, 5H), 1.17 (d, 3H, J=7.0 Hz), 0.90 (s, 9H), 0.06 (s, 6H); 13C NMR (75 MHz, CDCl3) 159.9, 131.6, 127.4, 113.6, 101.7, 79.7, 73.9, 63.1, 55.3, 31.8, 29.3, 28.7, 26.0, 18.4, 11.1, −5.1; LRMS (ESI) 402.68 (M+Na).
  • (4R,5S)-4-{(1S,2Z)-5-[(2S,3R,4S,5R,6R)-6-Methoxy-4-methoxymethoxy-3,5-dimethyltetrahydropyran-2-yl]-1-methylpent-2-enyl}-2-(4-methoxyphenyl)-5-methyl[1,3]dioxane (35). NaHMDS (1.0 M in THF, 1.1 mL, 1.1 mmol) was slowly added to a solution of the salt 21 (701 mg, 1.1 mmol) in dry THF (2.2 mL) at 0° C. The resulting red solution was stirred at room temperature for 20 min. The mixture was cooled to −78° C. and a solution of the aldehyde 9a (260 mg, 1 mmol) in THF (1 mL×2) was added dropwise. The mixture was stirred for 20 min at −78° C. and then warmed to room temperature. After 4 h at room temperature, the mixture was quenched with saturated NH4Cl (10 mL) and extracted with CH2Cl2 (3×10 mL). The combined organic layers were dried over anhydrous MgSO4, evaporated and chromatographed (hexane/ether 9:1) to yield 329 mg (67%) of 35 as a colorless oil: IR (CHCl3) 2922, 2866, 2628, 2350, 1740, 1516 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.45 (d, 2H, J=8.4 Hz), 6.88 (d, 2H, J=8.4 Hz), 5.46 (m, 2H), 5.30 (t, 1H, J=9.9 Hz), 4.77 (d, 1H, J=6.9 Hz), 4.70 (d, 1H, J=1.8 Hz), 4.63 (d, 1H, J=6.9 Hz), 4.06 (br d, 1H, J=2.1 Hz), 3.80 (s, 3H), 3.54 (m, 3H), 3.43 (s, 3H), 3.32 (s, 3H), 2.77 (m, 1H), 2.31 (dd, 2H, J=7.5, 14.7 Hz), 1.79-1.55 (m, 4H), 1.22 (d, 3H, J=6.6 Hz), 1.02 (d, 3H, J=7.2 Hz), 0.96 (d, 3H, J=6.9 Hz), 0.86 (d, 3H, J=6.9 Hz); 3C NMR (75 MHz, CDCl3) δ 159.7, 133.4, 131.8, 130.1, 127.3, 113.3, 101.5, 101.3, 95.9, 83.5, 82.0, 75.2, 73.9, 56.4, 55.7, 55.2, 37.6, 34.2, 33.6, 33.0, 30.0, 23.4, 16.1, 13.3, 11.2, 9.9; HRMS (EI) calcd for C27H40O6 460.2824, found 460.2846.
  • (4R,5S)-4-{(1S,2Z)-5-[(2S,3S,4S,5R,6R)-6-Methoxy-4-methoxymethoxy-3,5-dimethyltetrahydropyran-2-yl]-1-methylpent-2-enyl}-2-(4-methoxyphenyl)-5-methyl[1,3]dioxane (36). NaHMDS (1.0 M in THF, 1.1 mL, 1.1 mmol) was slowly added to a solution of the salt 21 (0.70 g, 1.1 mmol) in dry THF (2 mL) at 0° C. The resulting red solution was stirred at room temperature for 20 min. The mixture was cooled to −78° C. and a solution of the aldehyde 9b (260 mg, 1 mmol) in THF (1 mL) was added dropwise. The mixture was stirred for 20 min at −78° C. and then warmed to room temperature. After 4 h at room temperature, the reaction was quenched with saturated NH4Cl (10 mL) and extracted with CH2Cl2 (3×10 mL). The combined organic layers were dried over anhydrous MgSO4, evaporated and chromatographed (hexane/ether 9:1) to yield 329 mg (67%) of 36 as a colorless oil: IR (CHCl3) 2922, 2866, 2628, 2350, 1740, 1516 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.48 (d, 2H, J=9.0 Hz), 6.91 (d, 2H, J=9.0 Hz), 5.46 (m, 2H), 5.32 (t, 1H, J=9.6 Hz), 4.73 (s, 2H), 4.31 (d, 1H, J=5.4 Hz), 4.07 (br s, 2H), 3.81 (s, 1H), 3.57 (dd, 1H, J=1.8, 9.6 Hz), 3.45 (s, 3H), 3.43 (s, 3H), 3.20 (t, 1H, J=6.6 Hz), 2.77 (m, 1H), 2.31-2.17 (m, 2H), 1.90-1.62 (m, 4H), 1.24-0.98 (m, 12H); 13C NMR (75 MHz, CDCl3) δ 159.7, 133.7, 131.7, 129.4, 127.2, 113.4, 102.8, 101.5, 96.6, 83.5, 82.2, 73.9, 70.0, 55.7, 55.2, 39.8, 38.4, 33.6, 30.0, 29.9, 24.3, 15.9, 15.5, 13.2, 11.1; HRMS (EI) calcd for C27H40O6 (M−HOCH3) 460.2824, found 460.2846.
  • (2R,3S,4S,5Z)-3-(4-Methoxybenzyloxy)-8-[(2S,3R,4S,5R,6R)-6-methoxy-4-methoxymethoxy-3,5-dimethyltetrahydropyran-2-yl]-2,4-dimethyloct-5-enal (37). DIBAL (1.0 M in hexane, 2.1 mL, 2.1 mmol) was added dropwise to a solution of the acetal 35 (329 mg, 0.67 mmol) in dry CH2Cl2 (6.7 mL) at 0° C. After stirring for 2 h, the reaction was quenched with saturated aqueous sodium tartrate (20 mL) followed by vigorously stirring for several hours. The aqueous phase was extracted with CH2Cl2 (3×10 mL) and the combined organic layers were washed with brine (10 mL). The residue obtained after drying over MgSO4 and evaporation under vacuum was dissolved in anhydrous CH2Cl2 (6 mL) and DMSO (12 mL), treated with N,N-diisopropylethylamine (0.52 mL, 3 mmol), cooled to 0° C. and treated with pyridinium sulfur trioxide (477 mg, 3 mmol). The reaction mixture was stirred at ambient temperature for 1 h, diluted with ethyl ether (50 mL) and washed with aqueous HCl (0.5 N, 50 mL) and brine (10 ml). The separated organic layer was dried over MgSO4. Filtration and concentration followed by short flash column chromatography (hexane/EtOAc 4:1) provided the crude aldehyde 37 (270 mg, 0.55 mmol) as a colorless oil which was used without further purification.
  • (2R,3S,4S,5Z)-3-(4-Methoxybenzyloxy)-8-[(2S,3S,4S,5R,6R)-6-methoxy-4-methoxymethoxy-3,5-dimethyltetrahydropyran-2-yl]-2,4-dimethyloct-5-enal (38). DIBAL (1.0 M in hexane, 2.1 mL, 2.1 mmol) was added dropwise to a solution of the acetal 36 (329 mg, 0.67 mmol) in dry CH2Cl2 (6.7 mL) at 0° C. After the mixture was stirred for 2 h, the reaction was quenched with saturated aqueous sodium tartrate (20 mL) followed by vigorous stirring for several hours. The aqueous phase was extracted with CH2Cl2 (3×10 mL) and the combined organic layers were washed with brine (10 mL). After drying over MgSO4 and evaporation under vacuum, the residue was used for the next reaction without further purification. The crude alcohol in CH2Cl2 (13 mL) was treated with Dess-Martin periodinane (340.8 mg, 0.80 mmol). After the reaction was complete, the mixture was quenched with saturated NaHCO3 (20 mL). The aqueous layer was extracted with CH2Cl2 (2×10 mL) and the combined extracts were dried over anhydrous MgSO4. Filtration and concentration followed by short flash column chromatography filtration (hexane/EtOAc 9:1) provided crude aldehyde 38 (267 mg, 81%) as a colorless oil which was used without further purification.
  • (2S,3R)-6-(tert-Butyldimethylsilanyloxy)-3-(4-methoxybenzyloxy)-2-methylhexanal (27). DIBAL (1.0 M in THF, 15 mL, 15 mmol) was added dropwise to a stirred solution of 26 (1.90 g, 5 mmol) in anhydrous CH2Cl2 (50 mL) under an atmosphere of N2 at 0° C. and the mixture was stirred for 1 h at 0° C. The reaction was quenched by the careful addition of aqueous sat'd potassium sodium tartrate (100 mL) and stirring for 3 h at room temperature. Once the aqueous and organic layers separated, the aqueous layer was extracted with CH2Cl2. The combined organic layer was washed with brine and dried over MgSO4 followed by the evaporation of the solvent under reduced pressure. The crude alcohol (1.56 g, 4.1 mmol) was used without further purification.
  • Pyridinium sulfur trioxide (2.38 g, 15 mmol) was added to a stirred solution of the crude alcohol from above and diisopropylethylamine (2.6 mL, 15 mmol) in anhydrous CH2Cl2 (10 mL) and DMSO (20 mL) at 0° C. The mixture was stirred at ambient temperature for 1 h. After the reaction was complete, the mixture was diluted with ethyl ether (100 mL) and washed with aqueous HCl (0.5 N, 100 mL) and brine (100 ml). The separated organic layer was dried over MgSO4. Filtration and concentration followed by short flash column chromatography filtration (hexane/EtOAc 4:1) to remove SO3-pyridine provided the crude aldehyde 27 as a colorless oil which was used without further purification.
  • (2R,3R,4R,5R)-8-(tert-Butyldi methylsilanyloxy)-3-hydroxy-5-(4-methoxybenzyloxy)-2,4-dimethyloctanoic acid, 2,6-dimethylphenyl ester (29). LDA (2M in THF, 3.1 mL, 6.2 mmol) was added to a solution of 2,6-dimethylphenoxy propionate (1.10 g, 6.2 mmol) in anhydrous THF (12.4 mL) at −78° C., followed by stirring for 1 h at −78° C. The crude aldehyde 27 (4.1 mmol) from above dissolved in anhydrous THF (10 mL) was added slowly at −78° C. After 2 h at room temperature, the mixture was quenched with saturated aqueous NH4Cl (10 mL) and extracted with CH2Cl2 (3×10 mL). The combined organic layer was dried over anhydrous MgSO4, evaporated and chromatographed (hexane/EtOAc 4:1) to yield 29 (1.67 g, 2.99 mmol) as a colorless oil: IR (CHCl3). 3120, 2857, 1744, 1514, 1216, 1099 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.26 (d, 2H, J=8.4 Hz), 6.87 (d, 2H, J=8.4 Hz), 4.62 (d, 1H, J=11.1 Hz), 4.40 (d, 1H, J=11.1 Hz), 4.06 (d, 1H, J=6.8 Hz), 3.79 (s, 3H), 3.66-3.61 (m, 3H), 2.89 (m, 1H), 2.19 (s, 6H), 1.86 (m, 2H), 1.55 (m, 3H), 1.27 (d, 3H, J=6.8 Hz), 1.01 (d, 3H, J=6.9 Hz), 0.93 (s, 9H), 0.07 (s, 6H); 13C NMR (75 MHz, CDCl3) δ 173.5, 159.2, 148.0, 130.0, 129.9, 129.4, 128.4, 125.6, 113.8, 83.5, 70.7, 62.9, 55.1, 44.0, 35.6, 28.7, 26.6, 25.8, 18.2, 16.3, 14.2, 5.7, −5.3; HRMS (EI) calcd for C32H50O6Si 558.3377, found 558.3392.
  • (2S,3S,4R,5R)-3,8-Bis-(tert-butyidimethylsilanyloxy)-5-(4-methoxybenzyloxy)-2,4-dimethyloctan-1-ol. TBDMSOTf (0.68 mL, 3 mmol) was added to a stirred solution of 29 (1.11 g, 2 mmol) and 2,6-lutidine (0.69 mL, 6 mmol) in CH2Cl2 (20 mL) at −78° C. The mixture was stirred for 2 h at ambient temperature. The reaction was quenched by the addition of aqueous HCl (0.5 N, 50 mL). The reaction mixture was extracted with CH2Cl2, dried over MgSO4 and the solvent was removed under reduced pressure. Short column chromatography (hexane/EtOAc 4:1) provided the crude product.
  • DIBAL (1.0 M in THF, 6 mL, 6 mmol) was added dropwise to a stirred solution of the TBS-protected aryl ester (1.90 g, 2 mmol) from above in anhydrous CH2Cl2 (20 mL) under an atmosphere of N2 at 0° C. and the mixture was stirred for additional 1 h at 0° C. The reaction was quenched by the careful addition of aqueous sat'd potassium sodium tartrate (50 mL). The mixture was stirred for 3 h at room temperature. Once the aqueous and organic layers had separated, the aqueous layer was extracted with CH2Cl2 (20 mL). The combined organic layer was washed with brine and dried over MgSO4 followed by the evaporation of the solvent under reduced pressure. The residue was purified by column chromatography (EtOAc/hexane/EtOAc 3:7) to give pure (997 mg, 1.8 mmol): IR (CHCl3) 3125, 1544, 1289, 1065 cm−1, 1H NMR (300 MHz, CDCl3) δ 7.28 (d, 2H, J=8.7 Hz), 6.89 (d, 2H, J=8.7 Hz), 4.51 (d, 1H, J=11.1), 4.41 (d, 1H, J=10.8 Hz), 3.83 (d, 3H), 3.79 (m, 1H), 3.64 (m, 4H), 3.36 (m, 1H), 2.45 (br s, 1H), 1.93 (m, 2H), 1.63 (m, 4H), 1.00 (d, 2H, J=7.0 Hz), 0.92 (s, 24H), 0.14 (s, 6H), 0.13 (s, 6H); 3C NMR (75 MHz, CDCl3) δ 159.1, 130.6, 129.4, 113.6, 80.3, 76.0, 71.2, 65.2, 63.0, 55.1, 39.1, 39.0, 29.0, 27.1, 26.1, 25.9, 18.2, 14.5, 11.6, −3.6, −3.9, −5.3; LRMS (ESI) 576.8 (M+Na).
  • (2R,3S,4R,5R)-3,8-Bis-(tert-butyidimethylsilanyloxy)-5-(4-methoxybenzyloxy)-2,4-dimethyloctanal (30). Pyridinium sulfurtrioxide (858 mg, 5.4 mmol) was added to a stirred solution of alcohol (997 mg, 1.8 mmol) from above and diisopropylethylamine (0.94 mL, 5.4 mmol) in anhydrous CH2Cl2 (3.6 mL) and DMSO (7.2 mL) at 0° C. The mixture was stirred at ambient temperature for 1 h. After the reaction was complete, the mixture was diluted with ethyl ether (50 mL) and washed with aqueous HCl (0.5N, 50 mL) and brine (10 ml). The organic layer was dried over MgSO4. Filtration and concentration followed by short flash column chromatography filtration (hexane/EtOAc 4:1) to remove SO3-pyridine provided the crude aldehyde 30 as a colorless oil which was used without further purification: 1H NMR (300 MHz, CDCl3) δ 9.69 (s, 1H), 7.22 (d, 2H, J=8.6 Hz), 6.85 (d, 2H, J=8.6 Hz), 4.45 (d, 1H, J=11.1 Hz), 4.28 (d, 1H, J=11.1 Hz), 3.94 (dd, 1H, J=5.5, 4.0 Hz), 3.79 (s, 3H), 3.60 (t, 2H, J=6.0 Hz), 3.40-3.34 (m, 1H), 2.66-2.58 (m, 1H), 1.92-1.84 (m, 1H), 1.67-1.59 (m, 2H), 1.55-1.45 (m, 2H), 1.02 (d, 3H, J=7.0 Hz), 0.98 (d, 3H, J=7.0 Hz), 0.89 (s, 9H), 0.86 (s, 9H), 0.05 (s, 3H), 0.04 (s, 9H).
  • (1R,2R,3S,4S,5Z)-1-{3-(tert-Butyldimethylsilanyloxy)-1-[3-(tert-butyldi methylsilanyloxy)propyl]-2,4-dimethylocta-5,7-dienyloxymethyl}-4-methoxybenzene (32). CrCl2 (1.09 g, 9.0 mmol) was added to a stirred solution of the crude aldehyde (1.8 mmol) from above and 1-bromoallyl trimethylsilane 31 (578 mg, 5.4 mmol) in anhydrous THF (18 mL) under an atmosphere of N2 at room temperature. The mixture was stirred for 14 h at ambient temperature, then diluted with ethyl ether followed by filtration through Celite. After the evaporation of the solvent under reduced pressure, the residue was purified by short silica gel column chromatography (CH2Cl2). The resulting residue was used without further purification.
  • The above product in THF (50 mL) was cooled to 0° C. and NaH (95% w/w, 207 mg, 9.0 mmol) was added in one portion. The ice bath was removed after 15 min and the mixture was stirred for 2 h at ambient temperature. The reaction mixture was cooled to 0° C., quenched with H2O (10 mL) and extracted with ethyl ether (2×50 mL). The combined organic layer was washed with brine and dried over MgSO4 followed by the evaporation of the solvent under reduced pressure. The residue was purified by column chromatography (hexane/EtOAc 4:1) to give pure 32 (622 mg, 1.2 mmol): IR (CHCl3) 2954, 2931, 2857, 1608, 1513, 1463, 1251, 1098, 1047 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.35 (d, 2H, J=8.6 Hz), 6.96 (d, 2H, J=8.6 Hz), 6.41 (ddd, 1H, J=16.7, 11.0, 10.1 Hz), 6.04 (dd, 1H, J=11.1, 11.0 Hz), 5.57 (dd, 1H, J=10.1, 16.8 Hz), 5.20 (d, 1H, J=16.7 Hz), 5.06 (d, 1H, J=10.1 Hz), 4.51 (d, 1H, J=11.3 Hz), 4.35 (d, 1H, J=11.3 Hz), 3.81 (s, 3H), 3.63-3.57 (m, 3H), 3.28 (dt, 1H, J=5.5, 5.5 Hz), 2.70 (ddq, 1H, J=10.3, 6.9, 3.2 Hz), 1.73-1.58 (m, 3H), 1.50-1.44 (m, 2H), 0.94 (d, 3H, J=6.9 Hz), 0.93-0.91 (m, 21H), 0.06 (s, 6H), 0.05 (s, 6H); 13C NMR (75 MHz, CDCl3) 159.1, 134.8, 132.5, 131.0, 129.5, 128.9, 117.1, 113.7, 78.9, 76.6, 70.7, 63.2, 55.2, 40.0, 36.4, 31.6, 28.7, 26.2, 26.0, 18.9, 18.5, 18.3, 10.9, −3.3, −3.4, −5.3; LRMS (EI) 576, 519, 467, 387, 357, 293, 225, 121; HRMS (EI) calcd for C29H51O4Si2 519.3326, found 519.3332; [α]20 D −18.8° (c 0.75, CHCl3).
  • (2R)-2-{(4R,5S,6R)-6-[3-(tert-Butyldimethylsilanyloxy)propyl]-2,2,5-trimethyl[1,3]dioxan-4-yl}-propionic acid, 2,6-dimethylphenyl ester (34). A mixture of PMB ether 29 (55.8 mg, 0.1 mmol) and palladium (10% Pd/C, 5 mg) in EtOAc (10 mL) was stirred at room temperature under an H2 atmosphere (balloon) for 3 h. The mixture was filtered and concentrated to yield the diol which was used without further purification. A solution of the crude diol, dimethyl dimethyl acetal (12.4 mg, 0.12 mmol) and PPTS (0.1 equiv.) in benzene was stirred for 5 h at 65° C. The reaction was quenched with aqueous sat'd NaHCO3 (50 mL) followed by washing with water. The aqueous layer was extracted with ethyl ether (2×50 mL). The combined organic layer was dried over MgSO4. The solvent was removed under reduced pressure and the crude product was purified by column chromatography (hexane/EtOAc 9:1) to provide the pure 34 in 52% yield: IR (CHCl3) 2855, 1742, 1510, 1091 cm−1; 1H NMR (500 MHz, CDCl3) δ 7.03 (br s, 3H), 4.20 (dd, 1H, J=1.7, 10.2 Hz), 3.91 (m, 1H), 3.64 (m, 2H), 2.91 (dq, 1H, J=10.2, 6.9 Hz), 2.16 (s, 6H), 1.59-1.32 (m, 6H), 1.41 (s, 3H), 1.39 (s, 3H), 1.24 (d, 3H, J=4.2 Hz), 0.92 (br s, 12H), 0.06 (s, 6H); 13C NMR (75 MHz, CDCl3) 173.7, 148.2, 130.3, 128.5, 125.8, 99.1, 75.2, 73.0, 63.1, 42.3, 31.8, 29.9, 29.3, 28.8, 26.0, 19.5, 18.4, 16.4, 12.9, 4.54, −5.19; HRMS (EI) calcd for C27H46O5Si 478.3115, found 463.2889 (M−CH3).
  • (1S,2S,3R,6Z,8S,9S,10S,11Z)-[3,9-Bis-(4-methoxybenzyloxy)-14-[(2S,3S,4S,5R,6R)-6-methoxy-4-methoxymethoxy-3,5-dimethyltetrahydropyran-2-yl]-2,8,10-trimethyl-1-[(1S,2Z)-1-methylpenta-2,4-dienyl)tetradeca-6,11-dienyloxy]-tert-butyldimethylsilane. NaHMDS (1.0 M in THF, 0.54 mL, 0.54 mmol) was added slowly to a solution of the salt 33 (475.9 mg, 0.54 mmol) in dry THF (1.08 mL) at 0° C. The mixture was cooled to −78° C. and a solution of the aldehyde 38 (267 mg, 0.54 mmol) in THF (0.54 mL×2) was added dropwise. The mixture was stirred for 20 min at −78° C. and then warmed to room temperature. After 4 h at room temperature the mixture was quenched with saturated aqueous NH4Cl (10 mL) and extracted with CH2Cl2 (3×10 mL). The combined organic layer was dried over anhydrous MgSO4, evaporated and chromatographed (hexane/EtOAc 9:1) to yield the desired compound (257 mg, 0.28 mmol) as a colorless oil: IR (CHCl3) 2920, 2861, 2620, 1740, 1520 cm−1, 1H NMR (300 MHz, CDCl3) 7.38 (m, 4H), 6.96 (m, 4H), 6.47 (ddd, 1H, J=16.8, 11.0, 10.1 Hz), 6.04 (t, 1H, J=11.1 Hz), 5.57 (t, 1H, J=10.5 Hz), 5.49-5.12 (m, 6H), 4.75 (d, 2H, J=2.1 Hz), 4.67-4.33 (m, 5H), 4.07 (m, 1H), 3.65 (dd, 1H, J=3.3, 6.0 Hz), 3.47 (br s, 7H), 3.35 (dd, 1H, J=4.5, 4.7 Hz), 3.27 (t, 1H, J=6.6 Hz), 3.15 (dd, 1H, J=4.5, 6.9 Hz), 2.77 (m, 3H), 2.18 (m, 2H), 1.91 (m, 2H), 1.74-1.62 (m, 4H), 1.11-0.99 (m, 18H), 0.12 (s, 6H); 3C NMR (75 MHz, CDCl3) 159.2, 159.0, 134.7, 133.7, 132.9, 132.5, 131.4, 131.0, 129.6, 129.1, 128.9, 128.6, 117.3, 113.8, 113.7, 103.0, 96.5, 88.0, 82.1, 78.8, 74.9, 70.8, 69.7, 55.8, 55.3, 40.0, 39.5, 38.3, 35.6, 35.4, 31.3, 30.3, 26.4, 24.2, 23.7, 19.0, 18.8, 18.6, 17.3, 15.7, 13.2, 11.0, −3.1, −3.2; LRMS (ESI) 942.5 (M+Na).
  • Carbamic acid, (1S,2S,3R,6Z,8S,9S,10S,11Z)-3,9-bis-(4-methoxybenzyloxy)-14-[(2S,3S,4S,5R,6R)-6-methoxy-4-methoxymethoxy-3,5-dimethyltetrahydropyran-2-yl]-2,8,10-trimethyl-1-[(1S,2Z)-1-methylpenta-2,4-dienyl]tetradeca-6,11-dienyl ester (39). The above compound (128.5 mg, 0.14 mmol) in THF (4 mL) was treated with TBAF (1.0 M in THF, 0.40 mL, 0.40 mmol) and the mixture was stirred at room temperature for 48 h. The mixture was diluted with ethyl ether (30 mL) and washed with water (10 mL). After drying over MgSO4 and evaporation under vacuum, the resulting alcohol was used without further purification.
  • A solution of the alcohol in CH2Cl2 (8 mL) at 0° C. was treated with trichloroacetylisocyanate (0.05 mL, 0.42 mmol) and stirred at room temperature. After 30 min, the solution was concentrated under reduced pressure and the residue was taken up in MeOH (4 mL). K2CO3 (50 mg) was added to this solution and the mixture was stirred at room temperature for 3 h at room temperature. The mixture was diluted with EtOAc (30 mL). The organic layer was washed with brine. The aqueous layer was extracted with EtOAc, and the combined extracts were dried over anhydrous Na2SO4. Filtration and concentration followed by flash column chromatography (hexane/EtOAc 3:2) provided carbamate 39 (84.9 mg, 72%) as a yellow oil: IR (CHCl3) 3100, 3019, 2430, 2286, 1720, 1524 cm−1; 1H NMR (300 MHz, CDCl3) 7.41 (m, 4H), 7.01 (m, 4H), 6.46 (ddd, 1H, J=16.8, 11.0, 10.1 Hz), 6.11 (t, 1H, J=10.8 Hz), 5.62 (t, 1H, J=10.5 Hz), 5.53-5.18 (m, 6H), 4.94 (t, 1H, J=6.0 Hz), 4.84 (br s, 2H), 4.81 (d, 2H, J=2.1 Hz), 4.71-4.45 (m, 4H), 4.39 (d, 1H, J=5.1 Hz), 4.12 (m, 1H), 3.37 (m, 2H), 3.19 (dd, 1H, J=4.5, 6.9 Hz), 2.90 (m, 3H), 2.27 (m, 2H), 1.91 (m, 2H), 1.74-1.61 (m, 4H), 1.11-0.99 (m, 18H); 13C NMR (75 MHz, CDCl3) 159.2, 159.0, 157.1, 133.7, 133.3, 132.8, 132.2, 131.4, 130.9, 129.8, 129.6, 129.1, 128.5, 117.8, 113.8, 113.7, 102.9, 96.5, 88.0, 82.1, 78.4, 78.1, 74.9, 70.5, 69.8, 55.8, 55.7, 55.3, 39.5, 38.2, 37.7, 35.8, 35.4, 34.3, 30.6, 30.3, 24.2, 23.6, 18.9, 17.8, 17.4, 15.7, 13.2, 9.8; LRMS (ESI) 888.4 (M+K).
  • Carbamic acid, (1S,2S,3R,6Z,8S,9S,10S,11Z)-3,9-bis-(4-methoxybenzyloxy)-14-[(2S,3S,4S,5R)-4-methoxymethoxy-3,5-dimethyl-6-oxotetrahydropyran-2-yl]-2,8,10-trimethyl-1-[(1S,2Z)-1-methylpenta-2,4-dienyl]tetradeca-6,11-dienyl ester. A solution of 39 (42.4 mg, 0.05 mmol) in THF (0.5 mL) and 60% aqueous acetic acid (2.5 mL) was stirred at 70° C. for 4 h. After the reaction was complete by TLC, the mixture was neutralized slowly with saturated aqueous K2CO3 and diluted with EtOAc (20 mL). The aqueous phase was extracted with EtOAc (2×10 mL). The combined organic layers were dried over MgSO4 and evaporated under reduced pressure. The crude lactol was used for the next reaction without further purification.
  • Dess-Martin periodinane reagent (31.8 mg, 0.075 mmol) was added to a solution of the lactol in CH2Cl2 (5 mL). The resultant solution was stirred for 1 h and quenched by the simultaneous addition of saturated aqueous Na2S2O3 (5 mL) and saturated aqueous NaHCO3. The aqueous layer was extracted with CH2Cl2 (2×10 mL) and the combined extracts were dried over anhydrous MgSO4. Filtration and concentration followed by flash column chromatography (hexane/EtOAc 8:2) provided 28.3 mg (68%) of the lactone as a colorless oil: IR (CHCl3) 2992, 2361, 2332, 1742, 1374, 1242, 1047 cm−1; 1H NMR (300 MHz, CDCl3) 7.40 (m, 4H), 7.03 (m, 4H), 6.51 (ddd, 1H, J=16.8, 11.1, 10.0 Hz), 6.11 (t, 1H, J=11.1 Hz), 5.67 (t, 1H, J=10.8 Hz), 5.52-5.18 (m, 7H), 4.94 (t, 1H, J=6.0 Hz), 4.87-4.47 (m, 9H), 3.94 (br s, 4H), 3.93 (s, 3H), 3.54 (s, 3H), 3.40 (m, 2H), 3.19 (dd, 1H, J=4.5, 6.9 Hz), 2.87 (m, 2H), 2.72 (m, 2H), 2.32-1.89 (m, 7H), 1.45 (d, 3H, J=6.6 Hz), 1.15-01.02 (m, 15H); 13C NMR (75 MHz, CDCl3) 174.2, 159.2, 159.0, 156.9, 133.6, 133.5, 133.4, 132.2, 131.3, 130.9, 129.8, 129.6, 129.5, 129.2, 128.7, 127.8, 113.8, 113.7, 95.4, 88.0, 82.7, 79.9, 78.4, 76.5, 75.0, 55.9, 55.7, 55.3, 40.5, 38.6, 37.7, 36.0, 35.4, 34.3, 31.3, 30.6, 23.9, 23.6, 23.5, 19.0, 17.8, 14.4, 12.1, 9.8; LRMS (ESI) 872.4 (M+K).
  • Carbamic acid, (1S,2S,3R,6Z,8S,9S,10S,11Z)-3,9-dihydroxy-14-[(2S,3S,4S,5R)-4-hydroxy-3,5-dimethyl-6-oxotetrahydropyran-2-yl]-2,8,10-trimethyl-1-[(1S,2Z)-1-methylpenta-2,4-dienyl]tetradeca-6,11-dienyl ester (40). A solution of the above lactone (2.83 mg, 0.005 mmol) in THF (2 mL) was treated with aqueous 4N HCl (1 mL). The flask was fitted with a glass stopper and the resulting solution was stirred at room temperature for 48 h. Saturated aqueous K2CO3 was added dropwise followed by EtOAc. The aqueous layer was extracted with EtOAc and the combined extracts were dried over MgSO4. Filtration and concentration followed by simple short flash column chromatography (EtOAc/hexane/ether 3:2) provided the crude MOM-deprotected compound. A solution of PMB ether in CH2Cl2 (2 mL) at 0° C. was treated with NaHCO3 (4.2 mg, 0.5 mmol). After 1 h, the mixture was diluted with CH2Cl2 and washed with water. The aqueous layer was extracted with CH2Cl2 and the combined extracts were dried over anhydrous MgSO4. Filtration and concentration followed by flash column chromatography (EtOAc/hexane 3:2) provided carbamate 40 (1.1 mg, 0.002 mmol) as a colorless oil: IR (CHCl3) 2995, 2937, 2323, 1755, 1449, 1374, 1242, 1049 cm−1; 1H NMR (500 MHz, CDCl3) 6.63 (ddd, 1H, J=16.8, 11.0, 10.1 Hz), 6.06 (t, 1H, J=11.0 Hz), 5.46-5.32 (m, 5H), 5.25 (d, 1H, J=17 Hz), 5.14 (d, 1H, J=10.0 Hz), 4.94 (t, 1H, J=6.0 Hz), 4.74 9m, 1H), 4.60 (br s, 1H), 4.54 (m, 1H), 3.65 (m, 1H), 3.38 (d, 1H, J=5.0 Hz), 3.27 (t, 1H, J=6.0 Hz), 3.00 (m, 1H), 2.78 (m, 1H), 2.63 (m, 2H), 2.18 (m, 1H), 2.01 (m, 1H), 1.83 (m, 1H), 1.77 (m, 1H), 1.35 (d, 3H, J=7.0), 1.01-0.93 (m, 15H); 13C NMR (125 MHz, CDCl3) 174.2, 157.3, 133.6, 132.9, 129.6, 128.9, 125.0, 121.4, 118.0, 95.5, 82.6, 79.7, 79.2, 72.8, 55.9, 40.5, 39.9, 38.6, 35.4, 34.7, 31.6, 23.8, 19.2, 18.2, 17.7, 15.7, 14.8, 12.0; LRMS (ESI) 571.4 (M+Ka); HRMS (ESI) calcd for C31H51NO7Na 588.3303, found 588.3336 (M+K); [α]20 D +34.0 (c 0.05, CHCl3).
  • Carbamic acid, (1S,2S,3R,6Z,8S,9S,10S,11Z)-3,9-dihydroxy-14-1(2S,3S,4S,5R)-4-methoxymethoxy-3,5-dimethyl-6-oxotetrahydropyran-2-yl]-2,8,10-trimethyl-1-[(1S,2Z)-1-methylpenta-2,4-dienyl]tetradeca-6,11-dienyl ester (41). Carbamate 39 (8.49 mg, 0.01 mmol) was subjected to the lactonization procedure described above. The removal of the PMB protecting group was accomplished by treating with NaHCO3 and DDQ. Flash chromatography (EtOAc/hexane 3:2) provided 41 (2.9 mg, 49% overall 3 steps) as a colorless oil: IR (CHCl3) 3404, 2362, 1749, 1373, 1241, 1049 cm−1; 1H NMR (500 MHz, CDCl3) 6.62 (ddd, 1H, J=16.8, 11.0, 10.1 Hz), 6.04 (t, 1H, J=11.0 Hz), 5.48-5.33 (m, 5H), 5.24 (d, 1H, J=17 Hz), 5.13 (d, 1H, J=10.0 Hz), 4.77-4.60 (br m, 5H), 4.48 (m, 1H), 3.65 (m, 1H), 3.41 (s, 3H), 3.28 (d, 1H, J=7.0 Hz), 3.23 (t, 1H, J=5.5 Hz), 3.02 (m, 1H), 2.62 (m, 2H), 2.25-2.18 (m, 3H), 2.04 (m, 2H), 1.90 (m, 1H), 1.88 (m, 1H), 1.83 (m, 1H), 1.77-1.67 (m, 2H), 1.51 (m, 2H), 1.34 (d, 3H, J=7.0), 1.02-0.92 (m, 15H); 13C NMR (125 MHz, CDCl3) 174.1, 157.3, 133.6, 132.9, 132.2, 129.6, 128.9, 125.0, 121.4, 118.0, 95.5, 82.6, 79.7, 79.2, 72.8, 55.9, 40.5, 39.8, 38.6, 35.4, 34.9, 34.7, 31.6, 23.8, 19.2, 18.2, 17.2, 15.7, 14.8, 12.0; LRMS (ESI) 616.3 (M+Na); HRMS (ESI) calcd for C33H55NO8Na 616.3825, found 616.3829 (M+Na); [α]20 D +59.0 (c 0.1, CHCl3).
  • Carbamic acid, (1S,2S,3R,6Z,8S,9S,1S,11Z)-3,9-dihydroxy-14-[(2S,3S,4S,5R,6R)-6-methoxy-4-methoxymethoxy-3,5-dimethyltetrahydropyran-2-yl]-2,8,10-trimethyl-1-[(1S,2Z)-1-methylpenta-2,4-dienyl]tetradeca-6,11-dienyl ester (42). Carbamate 39 (4.25 mg, 0.005 mmol) was subjected to the deprotection procedure of PMB described in the preparation of 40. Flash chromatography (EtOAc/hexane 3:2) of the crude product provided 42 (2.8 mg, 92%) as a colorless oil: IR (CHCl3) 3115, 2749, 2328, 1676, 1508, 1215 cm−1; 1H NMR (300 MHz, CDCl3) 6.76 (ddd, 1H, J=16.8, 11.0, 10.1 Hz), 6.18 (t, 1H, J=10.8 Hz), 5.70-5.46 (m, 5H), 5.35 (d, 1H, J=16.8 Hz), 4.49 (dd, J=4.5, 6.6 Hz), 4.82 (d, 2H, J=2.4 Hz), 4.73 (br s, 2H), 4.43 (d, 1H, J=5.1 Hz), 4.15, (m, 1H), 3.78 (m, 1H), 3.56 (s, 3H), 3.54 (s, 3H), 3.36 (t, 2H, J=6.9 Hz), 3.14 (m, 1H), 2.76 (m, 2H), 2.35-2.18 (m, 6H), 2.00-1.60 (m, 7H), 1.22 (d, 3H, J=7.2 Hz), 1.16-1.12 (m, 12H), 1.07 (d, 3H, J=7.2 Hz); 13C NMR (125 MHz, CDCl3) 157.3, 133.7, 133.5, 132.2, 132.0, 130.0, 128.7, 118.0, 109.6, 103.0, 96.5, 82.1, 79.7, 79.1, 72.7, 55.8, 39.9, 39.5, 38.3, 35.5, 35.0, 34.8, 34.6, 30.2, 29.8, 24.3, 18.1, 17.7, 15.7, 15.4, 14.2, 13.2, 8.1; LRMS (ESI) 632.4 (M+Na); HRMS (ESI) calcd for C33H55NO8Na 632.4138, found 632.4139; [α]20 D +21.6 (c 0.25, CHCl3).
  • (12S,13S,14S,19R,20R,21R,22S)-21-(tert-Butyldimethylsilanyloxy)-13,19-bis-(4-methoxybenzyloxy)-12,14,20,22-tetramethylhexacosa-10,15,23,25-tetraen-1-(tert-Butyldimethylsilanyl)-ol (45). NaHMDS (1.0 M in THF, 0.45 mL, 0.45 mmol) was slowly added to a solution of the salt 21 (322 mg, 1.1 mmol) in dry THF (0.3 mL) at 0° C. The resulting red solution was stirred at room temperature for 20 min. The mixture was cooled to −78° C. and a solution of the aldehyde 44 (120 mg, 0.42 mmol) in THF (0.1 mL×2) was added dropwise. The mixture was stirred for 20 min at −78° C. and then warmed to room temperature. After 4 h, the mixture was quenched with saturated NH4Cl (5 mL) and extracted with ethyl ether (3×10 mL). The combined organic layers were dried over anhydrous MgSO4, evaporated and the residue was column chromatographed (hexane/ether 9:1) to yield 163 mg (75%) as a colorless oil: IR (CHCl3) 2928, 2854, 1617, 1517, 1462, 1390, 1249, 1114, 1035, 833, 726 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.44-7.38 (m, 2H), 6.90-6.85 (m, 2H), 5.42 (s, 1H), 5.39 (ddd, J=11.7, 10.2, 7.2 Hz, 1H), 5.24 (apparent t, J=10.2 Hz, 1H), 4.08-4.00 (m, 2H), 3.79 (s, 3H), 3.61 (t, J=6.5 Hz, 2H), 3.54 (dd, J=10.5, 1.9 Hz, 1H), 2.69 (dd, J=16.1, 9.2 Hz, 1H), 2.04 (apparent d, J=6.6 Hz, 2H), 1.71-1.68 (m, 1H), 1.54-1.50 (m, 3H), 1.27 (br, 1H), 1.20 (d, J=6.9 Hz, 3H), 0.91 (s, 9H), 0.06 (s, 6H); 13C NMR (75 MHz, CDCl3) δ 160.0, 133.3, 132.6, 132.5, 132.1, 130.7, 128.9, 128.7, 127.6, 113.7, 101.8, 83.9, 74.2, 63.6, 55.5, 33.9, 33.3, 30.4, 30.1, 30.0, 29.9, 29.8, 29.7, 28.0, 26.3, 26.2, 18.7, 16.3, 11.5, −4.9; LRMS (API-ES) 541(M+Na)+, 493, 431, 365, 295, 251; [α]20 D +26.0 (c 0.90, CHCl3).
  • To a solution of 164 mg (0.325 mmol) of the above acetal in dry CH2Cl2 (2.0 mL) DIBAL (1.0 M in hexane, 0.95 mL, 0.96 mmol) at 0° C. was added dropwise. After 2 h, the mixture was quenched with saturated sodium potassium tartrate solution (20 mL) followed by vigorously stirring for 4 h. The aqueous phase was extracted with CH2Cl2 (3×10 mL) and the combined organic layers were washed with brine (10 mL). After drying over MgSO4 and evaporation under vacuum, flash column chromatography (hexane/ether 9:1) provided 115 mg (70%) of alcohol as a colorless oil: IR (CHCl3) 3430, 2928, 2855, 1613, 1514, 1463, 1249, 1098, 1038, 835, 776 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.28-7.25 (m, 2H), 6.88-6.85 (m, 2H), 4.59 (d, J=10.8 Hz, 11H), 4.47 (d, J=10.8 Hz, 11H), 3.80 (s, 3H), 3.65-3.50 (m, 4H), 3.36 (dd, J=5.9, 3.9 Hz, 1H), 2.86-2.78 (m, 11H), 2.11-2.01 (m, 2H), 1.98-1.95 (m, 1H), 1.77 (br, 1H), 1.50 (br, 3H), 1.27 (br, 1H), 0.97 (apparent t, J=7.1 Hz, 6H), 0.90 (s, 9H), 0.05 (s, 6H); 13C NMR (75 MHz, CDCl3) δ 159.2, 132.8, 131.1, 130.0, 129.5, 113.8, 84.5, 73.7, 66.4, 63.5, 55.3, 37.6, 34.6, 33.0, 29.8, 29.7, 29.64, 29.61, 29.5, 27.7, 26.1, 25.9, 18.8, 18.4, 11.7, −5.1; LRMS (EI) 541 (M+Na)+, 462, 375, 325, 255, 207, 122; HRMS (EI) calcd for C27H47O4Si1 463.3254 (M−tBu)+, found 463.3254; [α]20 D +25.9 (c 0.48, CHCl3).
  • The above alcohol (94 mg, 0.18 mmol) in CH2Cl2 (2 mL) was treated with Dess-Martin periodinane (120 mg, 0.27 mmol). After 2 h, the mixture was quenched with saturated NaHCO3 (20 mL). The aqueous layer was extracted with ethyl ether (10 mL×2) and the combined extracts were dried over anhydrous MgSO4. Filtration and concentration followed by short flash column chromatography (hexane/EtOAc 9:1) to remove the residue from Dess-Martin reagent provided 78 mg (83%) of the crude aldehyde as a colorless oil which was used for the next reaction without further purification. NaHMDS (1.0 M in THF, 0.15 mL, 0.15 mmol) was slowly added to a solution of the salt 33 (140 mg, 0.17 mmol) in dry THF (0.15 mL) at 0° C. The resulting red solution was stirred at room temperature for 20 min. The mixture was cooled to −78° C. and a solution of the aldehyde above (69 mg, 0.13 mmol) in THF (0.05 mL×2) was added dropwise. The mixture was stirred for 20 min at −78° C. and then warmed to room temperature. After 4 h at room temperature, the mixture was quenched with saturated NH4Cl (2 mL) and extracted with ethyl ether (3×5 mL). The combined organic layers were dried over anhydrous MgSO4, evaporated and the residue was purified by column chromatography (hexane/ether 9:1) to yield 45 (111 mg, 65% for 2 steps) as a colorless oil: IR (CHCl3) 2926, 1612, 1513, 1462, 1361, 1250, 1173, 1098, 836, 774 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.40-7.33 (m, 4H), 6.99-6.93 (m, 4H), 6.49 (ddd, J=16.8, 10.8, 10.7 Hz, 1H), 6.06 (apparent t, J=11.0 Hz, 1H), 5.59 (d, J=10.5 Hz, 1H), 5.51 (d, J=9.8 Hz, 1H), 5.44-5.31 (m, 3H), 5.23 (d, J=16.8 Hz, 1H), 5.13 (d, J=10.1 Hz, 1H), 4.68-4.57 (m, 3H), 4.42 (d, J=11.3 Hz, 1H), 3.90 (s, 6H), 3.69 (t, J=6.5 Hz, 2H), 3.37-3.36 (m, 1H), 3.14 (q, J=3.7 Hz, 1H), 2.82-2.71 (m, 2H), 2.08-2.00 (m, 4H), 1.78-1.77 (m, 2H), 1.71-1.58 (m, 6H), 1.36 (br, 11H), 1.11 (d, J=6.7 Hz, 6H), 1.04-1.00 (m, 24H), 0.15 (s, 12H); 3C NMR (75 MHz, CDCl3) δ 159.3, 159.1, 134.8, 133.8, 132.5, 132.0, 131.5, 131.1, 129.9, 129.7, 129.2, 129.1, 128.6, 117.3, 113.8, 113.7, 88.1, 79.1, 74.8, 71.0, 63.4, 55.4, 40.1, 36.6, 35.8, 35.3, 33.0, 31.5, 30.0, 29.8, 29.7, 29.6, 27.7, 26.4, 26.1, 23.8, 19.0, 18.9, 18.5, 17.6, 11.1, −3.2, −3.3, −5.1; LRMS (EI) 890(M−tBu)+, 866; HRMS (EI) calcd for C54H89O6Si2 865.5258(M−tBu)+, found 865.5225; [α]20 D +20.5 (c 0.60, CHCl3).
  • (12S,13S,14S,19R,20R,21R,22S)-21-(tert-Butyldimethylsilanyloxy)-13,19-bis-(4-methoxybenzyloxy)-12,14,20,22-tetramethylhexacosa-10,15,23,25-tetraenoic acid (46). To a solution of TBS ether 45 (93 mg, 0.098 mmol) in THF (2 ml) was slowly added HF-pyridine in pyridine (4 ml, prepared by slow addition of 1.2 ml pyridine to 0.3 ml HF-pyridine complex followed by dilution with 3 ml THF). The mixture was stirred overnight at room temperature and quenched with sat'd NaHCO3 (20 ml). The aqueous layer was separated and extracted with CH2Cl2 (3×10 ml). The combined organic layer was washed with sat'd CuSO4 (3×20 ml), dried over MgSO4, and concentrated. Flash column chromatography (EtOAc/Hexane 1:4) afforded 64 mg (78%) of the alcohol: IR (CHCl3) 3429, 2928, 2855, 1694, 1612, 1513, 1462, 1250, 1173, 1038, 836, 773 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.40-7.33 (m, 4H), 6.99-6.95 (m, 4H), 6.49 (ddd, J=16.8, 10.6, 10.5 Hz, 1H), 6.05 (apparent t, J=11.0 Hz, 1H), 5.58 (d, J=10.9 Hz, 1H), 5.52 (d, J=9.7 Hz, 1H), 5.46-5.34 (m, 2H), 5.23 (d, J=16.8 Hz, 1H), 5.14 (d, J=10.1 Hz, 1H), 4.68-4.57 (m, 3H), 4.45-4.41 (m, 1H), 3.89 (s, 3H), 3.88 (s, 3H), 3.72-3.67 (m, 2H), 3.37-3.36 (m, 2H), 3.14 (q, J=3.6 Hz, 1H), 2.80-2.70 (m, 2H), 2.08-1.99 (m,4H), 1.78-1.77 (m, 2H), 1.71-1.58 (m, 6H), 1.36 (br, 11H), 1.10 (d, J=6.6 Hz, 6H), 1.02 (d, J=2.6 Hz, 6H), 1.00 (s, 9H), 0.15 (s, 6H); 13C NMR (75 MHz, CDCl3) δ 159.3, 159.1, 134.8, 134.8, 133.8, 132.5, 132.1, 131.5, 131.1, 129.9, 129.7, 129.2, 129.1, 128.6, 117.3, 113.8, 113.7, 88.1, 79.0, 76.7, 74.8, 71.0, 63.1, 55.4, 40.1, 36.6, 35.8, 35.4, 32.9, 31.4, 30.0, 29.7, 29.63, 29.56, 27.6, 26.4, 25.9, 23.8, 19.0, 18.9, 18.6, 17.6, 11.1, −3.2, −3.3; LRMS (API-ES) 871 (M+K)+, 445, 364, 338; [α]20 D +27.0 (c 0.24, CHCl3).
  • The above alcohol (0.213 g, 0.26 mmol) in CH2Cl2 (10 mL) was treated with Dess-Martin periodinane (160 mg, 0.38 mmol). After 2 h, the mixture was quenched with saturated NaHCO3 (10 mL). The aqueous layer was extracted with ethyl ether (10 mL×2) and the combined extracts were dried over anhydrous MgSO4. Filtration and concentration followed by short flash column chromatography (hexane/EtOAc 8:2) to remove the residue from Dess-Martin reagent provided the aldehyde as a colorless oil which was used for the next reaction without further purification. A solution of the above aldehyde in 1 ml of THF and 0.5 ml of H2O was treated with 0.74 ml (1.48 mmol) of a 2M solution of 2-methyl-2-butene in THF, 0.11 g (0.77 mmol) of NaH2PO4—H2O and 0.087 g (0.77 mmol) of NaClO2. The reaction mixture was stirred for 2 h, diluted with 20 ml of 1N HCl and extracted with CH2Cl2 (2×20 ml). The combined organic layers were dried over MgSO4, concentrated in vacuo and the residue was chromatographed on SiO2 (EtOAc/hexane 1:3) to yield 192 mg (89% for 2 steps) of the acid 46 as a viscous oil: IR (CHCl3) 3398, 2929, 2855, 1710, 1612, 1513, 1249, 1040 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.46-7.40 (m, 4H), 7.05-6.99 (m, 4H), 6.55 (ddd, J=16.8, 10.6, 10.3 Hz, 1H), 6.12 (apparent t, J=11.0 Hz, 1H), 5.66 (d, J=10.6 Hz, 1H), 5.58 (d, J=11.0 Hz, 1H), 5.52-5.37 (m, 3H), 5.30 (d, J=16.8 Hz, 1H), 5.21 (d, J=10.0 Hz, 1H), 4.74-4.64 (m, 3H), 4.52-4.48 (m, 1H), 3.95 (s, 6H), 3.72 (dd, J=6.2, 3.3 Hz, 1H), 3.43 (dd, J=10.5, 5.9 Hz, 1H), 3.21 (q, J=3.8 Hz, 1H), 2.87-2.77 (m, 3H), 2.49 (t, J=7.4 Hz, 2H), 2.15-2.09 (m, 4H), 1.87-1.70 (m, 5H), 1.43 (br, 11H), 1.17 (d, J=6.8 Hz, 6H), 1.10 (d, J=3.0 Hz, 6H), 1.07 (s, 9H), 0.22 (s, 6H); 13C NMR (75 MHz, CDCl3) δ 180.06 159.3, 159.1, 134.8, 133.8, 132.5, 132.1, 131.5, 131.1, 129.8, 129.7, 129.2, 129.1, 128.6, 117.3, 113.8, 113.7, 88.1, 79.0, 76.7, 74.8, 70.9, 55.4, 40.1, 36.6, 35.8, 35.4, 34.2, 31.4, 29.9, 29.5, 29.3, 29.2, 27.6, 26.4, 24.8, 23.8, 19.0, 18.9, 18.6, 17.6, 11.1, −3.2, −3.3; LRMS (API-ES) 846 (M), 845 (M−H); [α]20 D +24.5 (c 0.38, CHCl3).
  • (1S,13S,14S,15S,20R,21R,22R)-14,20-Dihydroxy-13,15,21-trimethyl-22-(1-methylpenta-2,4-dienyl)-oxacyclodocosa-11,16-dien-2-one (43). To 46 (146 mg, 0.17 mmol) in THF (2 mL) was added TBAF (1.0 M in THF, 1.72 mL, 1.72 mmol) and the mixture was stirred at room temperature for 48 h. The reaction mixture was diluted with ethyl ether (30 mL) and was washed with water (10 mL). After drying over MgSO4 and evaporation under vacuum, the crude was chromatographed on SiO2 (EtOAc/hexane 1:4) to yield 72 mg (57%) of the acid as a colorless oil: IR (CHCl3) 3467, 2927, 2854, 1710, 1612, 1513, 1460, 1248, 1174, 1036 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.39-7.32 (m, 4H), 6.98-6.93 (m, 4H), 6.69 (ddd, J=16.7, 10.7, 10.6 Hz, 1H), 6.18 (apparent t, J=10.9 Hz, 1H), 5.57 (d, J=10.4 Hz, 1H), 5.50 (d, J=10.9 Hz, 1H), 5.46-5.37 (m, 3H), 5.30 (d, J=17.2 Hz, 1H), 5.19 (d, J=10.1 Hz, 1H), 4.69-4.58 (m, 3H), 4.47-4.44 (m, 1H), 3.89 (s, 3H), 3.87 (s, 3H), 3.58-3.56 (m, 2H), 3.16 (q, J=3.5 Hz, 1H), 2.86-2.79 (m, 2H), 2.73-2.70 (m, 1H), 2.42 (t, J=7.3 Hz, 2H), 2.14-2.02 (m, 4H), 1.91-1.89 (m, 1H), 1.81-1.71 (m, 4H), 1.37 (br, 11H), 1.12 (d, J=6.6 Hz, 6H), 1.07 (d, J=6.9 Hz, 3H), 1.01 (d, J=6.6 Hz, 3H); 13C NMR (75 MHz, CDCl3) δ 179.6 159.3, 159.1, 135.6, 134.1, 132.4, 132.0, 131.4, 130.4, 130.1, 129.9, 129.6, 129.3, 128.3, 117.9, 113.9, 113.8, 88.1, 83.0, 78.2, 74.9, 71.0, 55.4, 36.7, 36.2, 36.1, 35.4, 34.1, 30.6, 29.9, 29.8, 29.5, 29.3, 29.2, 27.6, 24.8, 23.7, 19.0, 17.7, 17.5, 6.9; LRMS (API-ES) 755.5 (M+Na)+, 866; [α]20 D +31.3 (c 0.64, CHCl3).
  • A solution of above hydroxy acid (60 mg, 0.081 mmol) in THF (1 ml) was treated at 0° C. with Et3N (0.068 ml, 0.49 mmol) and 2,4,6-trichlorobenzoyl chloride (0.064 ml, 0.41 mmol). The reaction mixture was stirred at 0° C. for 30 min and then added to a 4-DMAP (41 ml, 0.81 mmol, 0.02 M solution in toluene) at 25° C. and stirred for overnight. The reaction mixture was concentrated, EtOAc (10 mL) was added and the crude was washed with 1N HCl (2×10 ml), dried over MgSO4. Purification by flash column chromatography (EtOAc/hexane 1:9) furnished macrolactone (33 mg, 57%) as a colorless oil: IR (CHCl3) 2926, 2855, 1730, 1612, 1513, 1459, 1248, 1174, 1109 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.41-7.35 (m, 4H), 6.98-6.94 (m, 4H), 6.55 (ddd, J=16.5, 10.9, 10.6 Hz, 1H), 6.06 (apparent t, J=10.8 Hz, 1H), 5.66 (apparent t, J=10.0 Hz, 1H), 5.48-5.29 (m, 4H), 5.24 (d, J=6.9 Hz, 1H), 5.16 (d, J=10.3 Hz, 11H), 5.01 (dd, J=7.5, 3.5 Hz, 1H), 4.66-4.53 (m, 3H), 4.43 (d, J=10.6 Hz, 1H), 3.89 (s, 3H), 3.85 (s, 3H), 3.20-3.18 (m, 1H), 3.13 (d, J=9.6 Hz, 11H), 2.97-2.89 (m, 1H), 2.76-2.64 (m, 2H), 2.37-2.19 (m, 3H), 2.04-1.98 (m, 4H), 1.78-1.57 (m, 4H), 1.38 (br, 11H), 1.16-1.10 (m, 9H), 0.99 (d, J=6.6 Hz, 3H); 13C NMR (75 MHz, CDCl3) δ 173.4 159.6, 159.3, 134.4, 133.7, 132.5, 131.8, 131.4, 131.0, 130.2, 130.0, 129.9, 129.4, 129.3, 118.0, 114.2, 114.0, 89.0, 80.6, 76.6, 75.7, 72.0, 55.6, 38.3, 37.5, 36.1, 34.9, 34.7, 31.7, 30.0, 29.6, 29.0, 28.8, 28.7, 27.2, 25.1, 24.5, 20.0, 18.8, 17.4, 10.4; HRMS (EI) calcd for C46H66O6 714.4859, found 714.4848; [α]20 D +5.8 (c 0.39, CHCl3).
  • The above macrolactone (12 mg, 16. μmol) was dissolved in CH2Cl2 (2 ml)—H2O (0.2 ml) and DDQ (12 mg, 53 μmol) was added at 0° C. After 1 h of stirring at 0° C., the reaction mixture was quenched by adding sat'd NaHCO3 (5 ml). The organic phase was washed by sat'd NaHCO3 solution (3×20 ml) and brine, dried over MgSO4 and concentrated. Purification by flash column chromatography (EtOAc/hexane 1:4) furnished macrolactone (6.8 mg, 85%) as a colorless oil: IR (CHCl3) 3434, 2926, 2854, 2359, 2341, 1731, 1651, 1505, 1456, 1377, 1261, 1107, 965, 905, 803 cm−1; 1H NMR (300 MHz, CDCl3) δ 6.75 (dt, J=16.8, 10.9 Hz, 1H), 6.13 (t, J=10.9 Hz, 1H), 5.60-5.56(m, 1H), 5.54-5.46 (m, 2H), 5.42-5.30 (m, 3H), 5.25 (d, J=10.1 Hz, 1H), 5.08 (dd, J=8.9; 2.6 Hz, 1H), 3.49 (ddd, J=9.5, 7.4, 2.8 Hz, 1H), 3.37 (dd, J=7.3, 4.3 Hz, 1H), 3.18-3.05 (m, 1H), 2.86-2.74 (m, 2H), 2.43-2.30 (m, 3H), 2.23-2.05 (m, 2H), 1.84 (br, 9H), 1.42 (br, 9H), 1.23 (d, J=6.8 Hz, 3H), 1.19 (d, J=6.9 Hz, 3H), 1.17 (d, J=6.9 Hz, 3H), 1.11 (d, J=6.7 Hz, 3H); 13C NMR (75 MHz, CDCl3) δ 173.4, 134.6, 132.4, 131.9, 130.5, 129.9, 129.8, 129.1, 117.9, 80.1, 76.4, 72.9, 40.0, 36.9, 35.4, 34.8, 34.6, 34.5, 29.0, 28.6, 28.4, 28.3, 28.1, 26.9, 24.8, 24.2, 18.9, 18.8, 17.1, 9.6; HRMS (EI) calcd for C30H49O3 456.3603 (M−OH)+, found 456.3622; [α]20 D +29.0 (c 0.10, CHCl3).
  • (12S,13S,14S,19R,20R,21R,22S)-12,14,20,22-Tetramethylhexacosa-10,15,23,25-tetraene-1,13,19,21-tetraol (47). The protected alcohol 45 (54 mg, 57 μmol) was dissolved in CH2Cl2 (3 ml)-H2O (0.3 ml) and DDQ (39 mg, 0.17 mmol) was added at 0° C. After 1 h of stirring at 0° C., the reaction mixture was quenched by adding sat'd NaHCO3 (10 ml). The organic phase was washed by sat'd NaHCO3 solution (3×20 ml) and brine, dried over MgSO4 and concentrated. Purification by flash column chromatography (EtOAc/hexane 1:9) furnished the diol (20 mg, 53%) as a colorless oil: IR (CHCl3) 3434, 2958, 2924, 2853, 2362, 1463, 1382, 1246, 1095, 1021, 832, 773 cm−1; 1H NMR (300 MHz, CDCl3) δ 6.58 (ddd, J=16.7, 10.7, 10.6 Hz, 1H), 6.05 (apparent t, J=11.0 Hz, 11H), 5.62 (t, J=10.3 Hz, 1H), 5.55-5.45 (m, 1H), 5.41-5.27 (m, 3H), 5.21 (d, J=7.6 Hz, 1H), 5.14 (d, J=10.2 Hz, 11H), 3.74-3.72 (m, 1H), 3.65-3.63 (m, 1H), 3.60 (t, J=6.6 Hz, 2H), 3.20 (dd, J=6.1, 5.4 Hz, 1H), 2.96-2.91 (m, 1H), 2.69-2.56 (m, 2H), 2.17-1.95 (m, 4H), 1.60-1.51 (m, 8H), 1.27 (br, 11H), 1.03 (d, J=7.0 Hz, 3H), 0.98 (d, J=6.7 Hz, 3H), 0.97 (d, J=6.7 Hz, 3H), 0.92 (s, 11H), 0.90 (s, 10H), 0.10 (s, 3H), 0.08 (s, 3H), 0.06 (s, 6H); LRMS (API-ES) 729.5 (M+Na)+, 557.5, 413, 243; [α]2 D +48.0 (c 0.025, CHCl3).
  • To an above solution (20 mg, 28 μmol) in THF (1 mL) was added TBAF (1.0 M in THF, 0.28 mL, 0.28 mmol) and the mixture was stirred at room temperature for 2 h. The reaction mixture was diluted with EtOAc (10 mL) and was washed with water (10 mL). After drying over MgSO4 and evaporation under vacuum, the crude was chromatographed on SiO2 (EtOAc/hexane 1:3) to yield 11 mg (83%) of the alcohol 47 as a colorless oil: IR (CHCl3) 3378, 2925, 2853, 2359, 1651, 1455, 1377, 1056, 971, 903 cm−1; 1H NMR (300 MHz, CDCl3)
    Figure US20060270862A1-20061130-P00900
    {tilde over (□)}□
    Figure US20060270862A1-20061130-P00901
    ddd, J=16.8, 10.6, 10.5 Hz, 1H), 6.21 (apparent t, J=11.0 Hz, 1H), 5.54-5.46 (m, 1H), 5.43-5.36 (m, 2H), 5.31-5.18 (m, 4H), 3.84 (dd, J=7.3, 4.6 Hz, 1H), 3.65 (apparent t, J=6.6 Hz, 2H), 3.46 (d, J=9.3 Hz, 1H), 3.22 (t, J=5.6 Hz, 1H), 2.86-2.78 (m, 1H), 2.72-2.59 (m, 2H), 2.23-2.02 (m, 4H), 1.71-1.53 (m, 8H), 1.29-1.26 (m, 13H), 1.01-0.91 (m, 12H); 13C NMR (75 MHz, CDCl3) δ 134.5, 133.8, 132.2, 132.1, 131.9, 131.3, 128.9, 119.1, 80.6, 79.1, 76.1, 63.2, 53.6, 37.6, 36.4, 35.5, 35.1, 34.5, 32.9, 29.8, 29.6, 29.5, 29.4, 27.7, 25.8, 24.2, 18.1, 16.7, 15.3, 4.5; LRMS (API-ES) 517 (M+K)+, 501 (M+Na)+, 479 (M+H)+, 461 (M+H−H2O)+, 443; [α]20 D +43.3 (c 0.18, CHCl3).
  • (12S,13S,14S,19R,20R,21R,22S)-13,19,21-Trihydroxy-12,14,20,22-tetramethylhexacosa-10,15,23,25-tetraenoic acid methyl ester (48). To an acid 46 (34 mg, 40 μmol) in DMF (3 ml) K2CO3 (0.017 g, 0.12 mmol) and MeI (0.009 ml, 0.06 mmol) were added and stirred for 1 h at room temperature. The reaction mixture was quenched by H2O (1 ml) and extracted with EtOAc (3×5 ml) and washed with brine (5 ml). The organic phase was dried over MgSO4 and evaporated and the residue was used as crude without no further purification (36 mg, 85%): IR (CHCl3) 2928, 2855, 1740, 1613, 1513, 1462, 1301, 1248, 1172, 1038, 836, 773 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.30-7.24 (m, 4H), 6.90-6.86 (m, 4H), 6.39 (ddd, J=16.9, 10.7, 10.6 Hz, 1H), 5.96 (apparent t, J=11.0 Hz, 1H), 5.50 (d, J=10.3 Hz, 1H), 5.42 (d, J=10.7 Hz, 1H), 5.36-5.21 (m, 3H), 5.14 (d, J=16.8 Hz, 1H), 5.04 (d, J=9.9 Hz, 1H), 4.59-4.47 (m, 3H), 4.39-4.31 (m, 1H), 3.81 (s, 3H), 3.80 (s, 3H), 3.67 (s, 3H), 3.57 (dd, J=5.9, 3.3 Hz, 1H), 3.28-3.26 (m, 1H), 3.05 (q, J=3.7 Hz, 1H), 2.71-2.61 (m, 3H), 2.30 (t, J=7.5 Hz, 2H), 1.99-1.90 (m, 4H), 1.68-1.59 (m, 5H), 1.26 (br, 11H), 1.01 (d, J=6.6 Hz, 6H), 0.91 (br, 15H), 0.05 (s, 6H); 13C NMR (75 MHz, CDCl3) δ 177.4, 159.3, 159.1, 134.8, 133.8, 132.5, 132.1, 131.5, 131.1, 129.8, 129.7, 129.2, 129.1, 128.6, 117.3, 113.8, 113.7, 88.1, 79.0, 55.4, 51.6, 40.0, 36.5, 35.8, 35.4, 34.2, 31.4, 29.9, 29.8, 29.5, 29.4, 29.3, 27.6, 26.4, 25.1, 23.7, 19.0, 18.6, 17.6, 11.1, −3.2, −3.3; LRMS (API-ES) 883.6 (M+Na)+; [α]20 D +24.7 (c 1.6, CHCl3).
  • The above ester (41 mg, 47 μmol) was dissolved in CH2Cl2 (2 ml)—H2O (0.4 ml) and DDQ (32 mg, 0.14 mmol) was added at 0° C. and was followed same procedure for 43. Purification by flash column chromatography (EtOAc/Hexane 1:8) furnished the diol (25 mg, 84%) as a colorless oil: IR (CHCl3) 3487, 2924, 2850, 1741, 1602, 1463, 1367, 1249, 838, 761 cm−1; 1H NMR (300 MHz, CDCl3) δ 6.59 (ddd, J=16.8, 10.8, 10.6 Hz, 1H), 6.04 (apparent t, J=11.0 Hz, 1H), 5.62 (t, J=10.1 Hz, 1H), 5.54-5.46 (m, 1H), 5.41-5.30 (m, 3H), 5.23 (d, J=17.9 Hz, 1H), 5.14 (d, J=10.2 Hz, 1H), 3.75-3.71 (m, 1H), 3.68 (s, 3H), 3.65-3.63 (m, 1H), 3.20 (t, J=5.8 Hz, 1H), 2.96-2.90 (m, 1H), 2.69-2.58 (m, 2H), 2.31 (t, J=7.6 Hz, 2H), 2.17-1.95 (m, 5H), 1.62-1.52 (m, 5H), 1.29 (br, 11H), 1.03 (d, J=6.9 Hz, 3H), 0.98 (d, J=6.7 Hz, 3H), 0.97 (d, J=6.7 Hz, 3H), 0.93 (s, 12H), 0.10 (s, 3H), 0.08 (s, 3H); 3C NMR (75 MHz, CDCl3) δ 174.3, 134.4, 133.7, 132.0, 131.4, 129.4, 128.8, 118.1, 114.3, 79.0, 78.7, 71.2, 51.4, 42.5, 35.8, 35.7, 35.5, 34.4, 34.1, 29.7, 29.3, 29.2, 29.1, 27.6, 26.1, 24.9, 24.3, 19.2, 18.3, 17.9, 15.0, 14.1, 9.5, −3.7, −3.9; LRMS (API-ES) 643.5 (M+Na)+, 471.4; [α]20 D +41.6 (c 0.74, CHCl3).
  • To an above solution (25 mg, 40 μmol) in THF (2 mL) was added TBAF (1.0 M in THF, 0.12 mL, 0.12 mmol) and the mixture was stirred at room temperature for 2 h. The reaction mixture was diluted with EtOAc (10 mL) and was washed with water (10 mL). After drying over MgSO4 and evaporation under vacuum, the crude was chromatographed on SiO2 (EtOAc/hexane 1:3) to yield 8.5 mg (93%) of the ester 48 as a colorless oil: IR (CHCl3) 3444, 2952, 2925, 2847, 1734, 1451, 1379, 1237, 1197, 1451, 967 cm−1; 1H NMR (300 MHz, CDCl3)
    Figure US20060270862A1-20061130-P00900
    {tilde over (□)}□
    Figure US20060270862A1-20061130-P00901
    ddd, J=16.8, 10.7, 10.6 Hz, 1H), 6.20 (apparent t, J=10.7 Hz, 1H), 5.54-5.46 (m, 1H), 5.43-5.36 (m, 2H), 5.31-5.18 (m, 4H), 3.83 (dd, J=9.0, 3.9 Hz, 1H), 3.67 (s, 3H), 3.46 (dd, J=9.3, 2.0 Hz, 1H), 3.22 (apparent t, J=5.4 Hz, 1H), 2.85-2.78 (m, 1H), 2.72-2.59 (m, 2H), 2.31 (t, J=7.4 Hz, 3H), 2.20-1.95 (m, 3H), 1.74-1.59 (m, 6H), 1.29 (br, 12H), 1.01-0.93 (m, 12H); 13C NMR (75 MHz, CDCl3) δ 174.3, 134.4, 133.7, 131.9, 131.3, 128.7, 118.9, 80.5, 79.1, 76.0, 51.4, 37.7, 36.4, 35.5, 35.1, 34.5, 34.1, 30.0, 29.3, 29.2, 29.1, 27.6, 24.9, 24.2, 22.7, 17.9, 16.7, 15.1, 4.5; LRMS (API-ES) 529 (M+Na)+, 507, 489, 471, 453; [α]20 D +27.3 (c 0.43, CHCl3).
  • (12S,13S,14S,19R,20R,21R,22S)-13,19,21-Trihydroxy-12,14,20,22-tetramethylhexacosa-10,15,23,25-tetraenoic acid (49). To an above solution 48 (8.0 mg) in THF—H2O (0.3 ml, 0.1 ml each) was added LiOHH2O (1.3 mg) and the solution was warmed to 60° C. After stirring 6 h, 1N HCl (1 ml) was added and reaction mixture was extracted with CH2Cl2 (10 ml×2). After drying over MgSO4 and evaporation under vacuum, the crude was chromatographed on SiO2 (EtOAc/hexane 1:3) to yield 6.3 mg (81%) of the 49 as a colorless oil: IR (CHCl3) 3412, 2964, 2921, 2850, 2658, 1710, 1459, 1404, 1268, 971, 903 cm−1; 1H NMR (300 MHz, CDCl3) δ 6.64 (ddd, J=16.7, 10.6, 10.0 Hz, 1H), 6.20 (apparent t, J=10.9 Hz, 1H), 5.54-5.46 (m, 1H), 5.42-5.36 (m, 2H), 5.32-5.18 (m, 4H), 3.88-3.84 (m, 1H), 3.48 (d, J=9.2 Hz, 1H), 3.23 (apparent t, J=5.7 Hz, 1H), 2.86-2.76 (m, 1H), 2.69-2.60 (m, 2H), 2.34 (t, J=7.4 Hz, 2H), 2.21-2.04 (m, 5H), 1.70-1.62 (m, 5H), 1.28 (br, 13H), 1.00 (d, J=6.8 Hz, 3H), 0.98 (d, J=6.9 Hz, 3H), 0.94 (d, J=6.7 Hz, 6H); 13C NMR (75 MHz, CDCl3) δ 180.6, 134.4, 133.8, 132.1, 132.0, 132.0, 131.3, 128.8, 119.1, 80.7, 79.2, 76.2, 37.6, 35.5, 34.6, 29.8, 29.7, 29.5, 29.2, 29.1, 29.0, 27.6, 24.8, 24.2, 22.8, 18.1, 16.7, 15.4, 14.2, 4.5; LRMS (API-ES) 515.3 (M+Na)+, 493 (M+H)+, 475 (M+H—H2O)+, 457 (M+H−H2O)+, 242; [α]20 D +33.0 (c 0.23, CHCl3).
  • (4R,5R)-5-(4-Methoxybenzyloxy)-4-methyl-8-oxooct-2-enoic acid ethyl ester (51). To a cooled (0° C.) stirred suspension of NaH (2.27 g, 11.3 mmol, 60% dispersion in mineral oil) in THF (130 ml) was added dropwise a solution of triethyl phosphonoacetate (2.27 ml, 11.4 mmol) over 10 min period. The mixture was brought to room temperature with a water bath (30 min) and then cooled back to 0° C. and the aldehyde from 50 (3.43 g, 9.0 mmol) in THF (10 ml) was added. The resulting mixture was stirred for 1 h at 0° C. then pH7 phosphate buffer solution (30 ml) and diethyl ether (100 ml) were added. The mixture was allowed to warm to room temperature and the phase was separated. The organic phase was washed with sat'd NH4Cl solution (30 ml) and brine (30 ml), dried with MgSO4, filtered and concentrated to give oily crude product. Purification by flash chromatography (EtOAc/hexane 1:4) afforded pure ester (3.82 g, 94%): IR (CHCl3) 2954, 2928, 2855, 1720, 1513, 1250, 1034, 835 cm−1; 1H NMR (300 MHz, CDCl3)
    Figure US20060270862A1-20061130-P00900
    {tilde over (□)}□9-7.25 m, 2H), 7.00 (dd, J=15.7, 7.5 Hz, 1H), 6.91-6.84 (m, 2H), 5.83 (d, J=15.7 Hz, 1H), 4.47 (dd, J=14.6, 11.1 Hz, 2H), 4.19 (q, J=7.1 Hz, 2H), 3.81 (s, 3H), 3.64-3.52 (m, 2H), 3.37-3.33 (m, 1H), 2.64 (dd, J=13.2, 6.5 Hz, 1H), 1.42-1.68 (m, 4H), 1.30 (t, J=7.1 Hz, 3H), 1.09 (d, J=6.7 Hz, 3H), 0.89 (s, 9H), 0.04 (s, 6H); 13C NMR (75 MHz, CDCl3) δ 166.7, 159.3, 151.2, 130.8, 130.4, 129.5, 121.3, 113.9, 81.8, 71.6, 63.1, 60.3, 55.3, 39.8, 28.9, 27.7, 26.1, 18.5, 15.1, 14.4, −5.1; LRMS(API-ES) 489.1 (M+K)+, 435, 263, 204; [α]20 D +6.4 (c 0.43, CHCl3).
  • To a solution of above TBS ether (0.324 g, 0.72 mmol) in THF (5 ml) was slowly added HF-pyridine in pyridine (8 ml, prepared by slow addition of 2.4 ml pyridine to 0.6 ml HF-pyridine complex followed by dilution with 5 ml THF). The mixture was stirred overnight at room temperature and quenched with sat'd NaHCO3 (20 ml). The aqueous layer was separated and extracted with CH2Cl2 (3×10 ml). The combined organic layer was washed with sat'd CuSO4 (3×20 ml), dried over MgSO4, and concentrated. Flash column chromatography (EtOAc/hexane 1:3) afforded 0.203 g (84%) of the alcohol: IR (CHCl3) 1715, 1612, 1514, 1249, 1180, 1035 cm−1; 1H NMR (300 MHz, CDCl3)
    Figure US20060270862A1-20061130-P00900
    {tilde over (□)}□7-7.24 m, 2H), 6.98 (dd, J=15.8, 7.5 Hz, 1H), 6.88-6.85 (m, 2H), 5.83 (d, J=15.8 Hz, 1H), 4.47 (dd, J=14.6, 11.1 Hz, 2H), 4.17 (q, J=7.1 Hz, 2H), 3.79 (s, 3H), 3.59-3.56 (m, 2H), 3.37-3.33 (m, 1H), 2.71-2.65 (m, 1H), 2.15 (br, 1H), 1.77-1.40 (m, 4H), 1.28 (t, J=7.1 Hz, 3H), 1.08 (d, J=6.8 Hz, 3H); 13C NMR (75 MHz, CDCl3) δ 166.7, 159.3, 150.8, 130.4, 129.6, 121.4, 113.9, 81.9, 71.7, 62.8, 60.4, 55.3, 39.5, 28.9, 27.8, 15.2, 14.4; LRMS(API-ES) 375.1 (M+K)+, 359.1 (M+Na)+, 241, 225; [α]20 D +12.0 (c 0.15, CHCl3).
  • The above alcohol (0.203 g, 0.61 mmol) in CH2Cl2 (6 mL) was treated with Dess-Martin periodinane (0.38 g, 0.90 mmol). After 2 h, the mixture was quenched with saturated NaHCO3 (10 mL). The aqueous layer was extracted with ethyl ether (10 mL×2) and the combined extracts were dried over anhydrous MgSO4. Filtration and concentration followed by short flash column chromatography (hexane/EtOAc 3:1) to remove the residue from Dess-Martin reagent provided 0.146 g (72%) of the crude aldehyde 51 as a colorless oil which was used for the next reaction without further purification: 1H NMR (300 MHz, CDCl3) δ 9.68 (s, 1H), 7.27-7.21 (m, 2H), 6.99 (dd, J=15.8, 7.3 Hz, 1H), 6.88-6.85 (m, 2H), 5.84 (d, J=15.8 Hz, 1H), 4.48 (d, J=11.0 Hz, 1H), 4.34 (d, J=11.0 Hz, 1H), 4.18 (q, J=7.1 Hz, 2H), 3.79 (s, 3H), 3.41-3.31 (m, 1H), 2.73-2.63 (m, 1H), 2.55-2.40 (m, 1H), 1.90-1.78 (m, 1H), 1.71-1.61 (m, 1H), 1.28 (t, J=7.1 Hz, 3H), 1.27-1.22 (m, 2H), 1.10 (d, J=6.8 Hz, 3H).
  • (4R,5R,10S,11S,12S,17R,18R,19R,20S)-19-(tert-Butyldimethylsilanyloxy)-5,11,17-tris-(4-methoxybenzyloxy)-4,10,12,18,20-pentamethyltetracosa-2,8,13,21,23-pentaenoic acid ethyl ester (52). NaHMDS (1.0 M in THF, 0.49 mL, 0.49 mmol) was slowly added to a solution of the salt 21 (0.35 g, 0.55 mmol) in dry THF (0.50 mL) at 0° C. The resulting red solution was stirred at room temperature for 20 min. The mixture was cooled to −78° C. and a solution of the aldehyde 51 (146 mg, 0.44 mmol) in THF (0.1 mL) was added dropwise. The mixture was stirred for 20 min at −78° C. and then warmed to room temperature. After 4 h at room temperature, the mixture was quenched with saturated NH4Cl (2 mL) and extracted with ethyl ether (3×10 mL). The combined organic layers were dried over anhydrous MgSO4, evaporated and the residue was flash column chromatographed (hexane/EtOAc 9:1) to yield (183 mg, 74%) as a colorless oil: IR (CHCl3) 2962, 2850, 1716, 1614, 1515, 1249, 1179, 1035 cm−1; 1H NMR (300 MHz, CDCl3)
    Figure US20060270862A1-20061130-P00900
    {tilde over (□)}□6-7.49 m, 2H), 7.41-7.36 (m, 2H), 7.12 (dd, J=15.8, 7.5 Hz, 1H), 7.00-6.97 (m, 4H), 5.95 (d, J=15.8 Hz, 1H), 5.56 (s, 1H), 5.52-5.39 (m, 2H), 4.53 (d, J=3.0 Hz, 2H), 4.33 (q, J=7.1 Hz, 2H), 4.22-4.14 (m, 2H), 3.93 (s, 3H), 3.91 (s, 3H), 3.69 (dd, J=9.6, 1.9 Hz, 1H), 3.46-3.40 (m, 1H), 2.88-2.80 (m, 1H), 2.75-2.67 (m, 1H), 2.36-2.14 (m, 2H), 1.84-1.81 (m, 1H), 1.67-1.55 (m, 2H), 1.43 (t, J=7.1 Hz, 3H), 1.32 (d, J=6.9 Hz, 3H), 1.15 (d, J=6.8 Hz, 3H), 1.03 (d, J=6.8 Hz, 3H); 13C NMR (75 MHz, CDCl3) δ 166.8, 159.8, 159.3, 151.3, 134.0, 131.8, 130.9, 129.7, 129.5, 127.4, 121.1, 113.9, 113.5, 101.7, 83.9, 81.5, 74.0, 71.7, 60.3, 55.4, 39.7, 33.8, 31.5, 30.1, 23.9, 16.1, 15.0, 14.4, 11.2; LRMS (API-ES) 605.3 (M+K)+, 589.3 (M+Na)+, 567.3 (M+H)+; [α]20 D +30.0 (c 0.01, CHCl3).
  • Trimethylsilyl chloride (0.24 ml, 1.9 mmol) was added dropwise to a stirred mixture containing above acetal (0.177 g, 0.31 mmol), NaCNBH3 (0.12 g, 1.9 mmol) and 4 Å molecular sieve in acetonitrile (6 ml) at 0° C. The reaction mixture was stirred for 1 h at 0° C. and filtered through Celite, poured into 1N HCl (10 ml). The aqueous phase was extracted by CH2Cl2 (2×20 ml), dried (MgSO4), filtered and concentrated. The residue was purified by column chromatography on silica gel (EtOAc/hexane 1:3) to yield the alcohol (0.121 g, 68%) as a colorless oil: IR (CHCl3) 3467, 2962, 2931, 2873, 1716, 1612, 1514, 1462, 1248, 1179, 1035 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.26-7.24 (m, 4H), 6.98 (dd, J=15.7, 7.6 Hz, 1H), 6.91-6.85 (m, 4H), 5.83 (d, J=15.7 Hz, 1H), 5.48 (dd, J=10.8, 9.6 Hz, 1H), 5.41-5.33 (m, 1H), 4.58 (d, J=12.7 Hz, 1H), 4.46 (d, J=11.9 Hz, 3H), 4.19 (q, J=7.2 Hz, 2H), 3.81 (s, 3H), 3.79 (s, 3H), 3.63-3.49 (m, 2H), 3.39-3.29 (m, 2H), 2.78 (dd, J=15.7, 6.8 Hz, 1H), 2.61 (dd, J=13.1, 6.6 Hz, 1H), 2.23-2.17 (m, 1H), 2.09-1.94 (m, 3H), 1.58-1.44 (m, 2H), 1.29 (t, J=7.1 Hz, 3H), 1.06 (d, J=6.8 Hz, 3H), 0.96 (d, J=6.8 Hz, 3H), 0.94 (d, J=6.9 Hz, 3H); 13C NMR (75 MHz, CDCl3) δ 166.8, 159.2, 151.1, 133.6, 131.1, 130.7, 129.5, 129.1, 128.7, 121.3, 114.0, 113.9, 113.8, 84.3, 81.6, 73.9, 71.6, 66.2, 60.4, 55.4, 39.7, 37.7, 34.8, 31.5, 23.8, 18.7, 15.1, 14.4, 11.5; LRMS (API-ES) 591.2 (M+Na)+, 569.3 (M+H)+, 551; [α]20 D +37.2 (c 0.39, CHCl3).
  • The same procedure for 45 was used with above alcohol (0.088 g, 0.16 mmol) to yield 64 mg (42% for 2 steps) of the 52 by flash column chromatography (EtOAc/hexane 1:5): IR (CHCl3) 2956, 2932, 2857, 1717, 1612, 1513, 1462, 1301, 1248, 1172, 1037, 835, 773 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.29-7.23 (m, 6H), 6.98 (dd, J=15.9, 7.6 Hz, 1H), 6.89-6.85 (m, 6H), 6.40 (ddd, J=16.8, 10.6, 10.5 Hz, 1H), 5.96 (apparent t, J=10.9 Hz, 1H), 5.83 (d, J=15.8 Hz, 1H), 5.47 (apparent q, J=10.6 Hz, 11H), 5.36-5.24 (m, 4H), 5.15 (d, J=16.8 Hz, 1H), 5.05 (d, J=9.9 Hz, 1H), 4.58-4.32 (m, 6H), 4.20 (q, J=7.1 Hz, 2H), 3.81 (s, 6H), 3.80 (s, 3H), 3.58-3.57 (m, 1H), 3.30-3.28 (m, 2H), 3.06-3.04 (m, 1H), 2.71-2.65 (m, 2H), 2.62-2.56 (m, 2H), 2.19-2.17 (m, 2H), 2.06-1.88 (m, 4H), 1.76-1.42 (m, 6H), 1.30 (t, J=7.1 Hz, 3H), 1.07-0.97 (m, 12H), 0.92 (s, 9H), 0.06 (s, 6H); 13C NMR (75 MHz, CDCl3) δ 166.7, 159.2, 159.1, 151.2, 134.8, 133.8, 132.9, 132.5, 131.4, 130.7, 129.5, 129.2, 128.9, 128.6, 121.2, 117.3, 113.8, 113.7, 88.0, 81.6, 78.9, 74.9, 71.6, 70.9, 60.3, 55.4, 40.0, 39.7, 36.5, 35.5, 35.4, 31.6, 31.4, 26.3, 23.7, 20.8, 19.0, 18.8, 18.6, 17.2, 15.0, 14.4, 11.1, −3.2; LRMS (API-ES) 1034.2 (M+K)+, 1017.6 (M+Na)+, 995.7 (M+H)+; [α]20 D +40.7 (c 4.09, CHCl3).
  • (6R,7R,12S,13S,14S,19R,20R,21R,22S)-21-(tert-Butyldimethylsilanyloxy)-7,13,19-tris-(4-methoxybenzyloxy)-6,12,14,20,22-pentamethylhexacosa-2,4,10,15,23,25-hexaenoic acid methyl ester (53). To the above ester 52 (64 mg. 64 μmol) in CH2Cl2 (2 ml) was added DIBAL-H (0.16 ml, 0.16 mmol, 1.0 M solution in hexane) at −78° C. dropwise and then warmed up to 0° C. and stirred for 1 h. The reaction mixture was quenched by EtOAc (2 ml) and sat'd sodium potassium tartrate solution (20 mL) followed by vigorously stirring for 4 h. The aqueous phase was extracted with CH2Cl2 (3×10 mL) and the combined organic layers were washed with brine (10 mL). After drying over MgSO4 and evaporation under vacuum, flash column chromatography (hexane/EtOAc 3:1) provided 47 mg of alcohol (77%) as a colorless oil: IR (CHCl3) 3429, 2956, 2857, 2360, 1613, 1513, 1463, 1248, 1037, 835 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.43-7.37 (m, 6H), 7.03-6.98 (m, 6H), 6.53 (ddd, J=16.8, 10.6, 10.5 Hz, 1H), 6.10 (apparent t, J=11.0 Hz, 1H), 5.81-5.77 (m, 2H), 5.64 (d, J=10.4 Hz, 1H), 5.57 (d, J=10.9 Hz, 1H), 5.18 (d, J=10.1 Hz, 1H), 4.72-4.45 (m, 6H), 4.21 (q, J=3.2 Hz, 2H), 3.94 (s, 6H), 3.93 (s, 3H), 3.71 (dd, J=5.6, 3.0 Hz, 1H), 3.41 (dd, J=10.5, 5.2 Hz, 1H), 3.33 (dd, J=11.1, 6.4 Hz, 1H), 3.19 (dd, J=12.1, 5.9 Hz, 1H), 2.36-2.19 (m, 2H), 2.12-2.02 (m, 3H), 1.87-1.59 (m, 5H), 1.16-1.13 (m, 9H), 1.07-1.05 (m, 6H), 1.04 (s, 9H), 0.19 (s, 6H); 13C NMR (75 MHz, CDCl3) δ 159.3, 159.2, 159.1, 135.1, 134.8, 133.9, 132.7, 132.6, 131.5, 131.1, 129.7, 129.5, 129.3, 129.2, 128.8, 128.6, 117.4, 114.1, 113.8, 88.1, 82.5, 79.0, 74.9, 71.5, 70.9, 65.2, 64.0, 55.4, 40.0, 39.4, 36.6, 35.6, 35.4, 31.4, 29.9, 26.4, 23.9, 23.7, 19.1, 18.8, 18.7, 17.3, 16.1, 11.1, −3.1, −3.2; LRMS (API-ES) 991.6 (M+K)+, 975.6 (M+Na)+; [α]20 D +38.3 (c 1.05, CHCl3).
  • The above alcohol (47 mg, 49 μmol) in CH2Cl2 (2 mL) was treated with Dess-Martin periodinane (31 mg, 73 μmol). After 2 h, the mixture was quenched with saturated NaHCO3 (5 mL). The aqueous layer was extracted with ethyl ether (5 mL×2) and the combined extracts were dried over anhydrous MgSO4. Filtration and concentration followed by short flash column chromatography filtration (hexane/EtOAc 3:1) to remove the residue from Dess-Martin reagent provided crude aldehyde as a colorless oil which was used for the next reaction without further purification. To a stirred solution of bis(2,2,2-trifluoroethyl)-(methoxycarbonylmethyl) phosphate (0.013 ml, 59 μmol), 18-crown-6 (0.065 g, 0.25 mmol) in THF (1 ml) cooled to −78° C. was added dropwise potassium bis(trimethylsilyl)amide (0.12 ml, 59 μmol, 0.5M solution in toluene). Thereafter the above aldehyde in THF (1 ml) was added and the solution was stirred for 6 h at −78° C. The reaction mixture was quenched by addition of a sat'd NH4Cl solution (1 ml) and diluted with diethyl ether (10 ml). The layer was separated and organic phase was washed with brine (10 ml) and dried with MgSO4, filtered, and concentrated. The residue was purified by flash chromatography (EtOAc/Hexane 1:5), yielding 40 mg of (E,Z)-doubly unsaturated ester 53 (85% for 2 steps): IR (CHCl3) 2956, 2856, 1717, 1612, 1513, 1462, 1301, 1248, 1173, 1037, 820 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.53 (dd, J=15.4, 11.3 Hz, 1H), 7.43-7.37 (m, 6H), 7.03-6.98 (m, 6H), 6.68 (dd, J=11.4, 11.3 Hz, 1H), 6.53 (ddd, J=17.0, 10.7, 10.4 Hz, 1H), 6.19 (dd, J=15.4, 7.6 Hz, 1H), 6.09 (apparent t, J=11.2 Hz, 1H), 5.73 (d, J=11.4 Hz, 1H), 5.64 (d, J=10.3 Hz, 1H), 5.56 (d, J=11.0 Hz, 1H), 5.45-5.41 (m, 3H), 5.28 (d, J=15.3 Hz, 1H), 5.18 (d, J=10.0 Hz, 1H), 4.71-4.45 (m, 6H), 3.94 (s, 6H), 3.93 (s, 3H), 3.87 (s, 3H), 3.70 (dd, J=6.1, 3.2 Hz, 1H), 3.44-3.38 (m, 1H), 3.19 (dd, J=6.9, 4.2 Hz, 1H), 2.85-2.77 (m, 3H), 2.34-2.31 (m, 2H), 2.08-2.04 (m, 3H), 1.84-1.55 (m, 5H), 1.20 (d, J=6.8 Hz, 3H), 1.14 (d, J=6.9 Hz, 3H), 1.12 (d, J=8.2 Hz, 3H), 1.07-1.01 (m, 15H), 0.16 (s, 6H); 13C NMR (75 MHz, CDCl3) δ 166.9, 159.1, 158.9, 147.6, 145.6, 134.7, 133.7, 132.7, 132.4, 131.3, 130.9, 130.8, 129.5, 129.4, 129.0, 128.9, 128.4, 126.4, 117.2, 115.4, 113.7, 87.9, 82.1, 78.8, 74.7, 71.4, 70.8, 55.2, 53.4, 51.1, 40.0, 36.4, 35.4, 35.2, 31.4, 31.3, 29.7, 26.2, 23.7, 23.6, 18.9, 18.6, 18.5, 17.1, 15.4, 10.9, −3.3, −3.4; LRMS (API-ES) 1045.5 (M+K)+, 1029.5 (M+Na)+; [α]20 D +35.3 (c 0.96, CHCl3).
  • (7R,8R,13S,14S,15S,20R,21R,22R,23S)-8,14,20-Trihydroxy-7,13,15,21-tetramethyl-22-(1-methylpenta-2,4-dienyl)-oxacyclodocosa-3,5,11,16-tetraen-2-one (54). To a stirred solution of protected alcohol 53 (33 mg, 33 μmol) in THF (1 ml) at 0° C. was added 2 ml of 3 N HCl (prepared by adding 25 ml of conc. HCl to 75 ml MeOH). After 6 h, the reaction mixture was diluted with EtOAc (5 ml) and H2O (5 ml) and the organic phase was separated and aqueous phase was extracted with EtOAc (2×5 ml). The combined organic phase was washed with sat'd NaHCO3 (10 ml), dried with MgSO4, concentrated and the residue was purified by flash chromatography (EtOAc/Hexane 1:4) to yield 19 mg (21 μmol) of product (63%): IR (CHCl3) 3491, 2958, 2869, 1716, 1612, 1513, 1456, 1248, 1036 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.40 (dd, J=15.3, 11.5 Hz, 1H), 7.29-7.22 (m, 6H), 6.92-6.84 (m, 6H), 6.61 (ddd, J=17.7, 10.7, 10.4 Hz, 1H), 6.54 (dd, J=11.5, 11.4 Hz, 1H), 6.10 (apparent t, J=11.0 Hz, 1H), 6.06 (dd, J=15.1, 7.7 Hz, 1H), 5.59 (d, J=11.3 Hz, 1H), 5.49 (d, J=10.4 Hz, 1H), 5.41 (d, J=10.6 Hz, 1H), 5.37-5.30 (m, 3H), 5.21 (d, J=17.0 Hz, 1H), 5.11 (d, J=10.2 Hz, 1H), 4.58-4.34 (m, 6H), 3.81 (s, 3H), 3.80 (s, 3H), 3.79 (s, 3H), 3.73 (s, 3H), 3.47-3.44 (m, 2H), 3.31-3.25 (m 1H), 3.05 (dd, J=7.3, 4.0 Hz, 1H), 2.80-2.69 (m, 2H), 2.66-2.61 (m, 1H), 2.20-2.15 (m, 2H), 2.05-1.91 (m, 3H), 1.85-1.76 (m, 1H), 1.72-1.61 (m, 2H), 1.57-1.47 (m, 2H), 1.09 (d, J=6.8 Hz, 3H), 1.00 (apparent q, J=7.1 Hz, 9H), 0.91 (d, J=6.7 Hz, 3H); 13C NMR (75 MHz, CDCl3) δ 167.1, 159.3, 159.1, 147.7, 145.7, 136.8, 134.1, 132.8, 132.5, 131.4, 132.8, 132.5, 131.4, 130.9, 130.4, 130.2, 129.6, 129.3, 128.3, 126.6, 118.0, 115.6, 114.0, 113.9, 88.1, 83.1, 82.2, 75.0, 71.5, 71.0, 55.4, 51.2, 40.1, 36.7, 36.3, 35.9, 35.4, 31.6, 30.6, 29.9, 23.9, 23.8, 19.0, 17.6, 15.6; LRMS (API-ES) 915.5 (M+Na)+; [α]20 D +41.1 (c 0.45, CHCl3).
  • To the stirred solution of above ester (19 mg, 21 μmol) in EtOH (1 ml) was added 1N aqueous KOH solution (0.056 ml) and the mixture was refluxed gently until the ester disappeared (about 6 h) as determined by TLC. The ethanolic solution was concentrated and then diluted with EtOAc (2 ml). After the solution was acidified to pH3 with 1N HCl solution, organic phase was separated and aqueous phase was extracted with EtOAc (2×5 ml). The combined organic phase were dried with MgSO4, concentrated and used as crude without further purification. The same procedure for 43 was used with above acid compound to yield 14 mg (79% for 2 steps) of the macrolactone product by flash column chromatography (EtOAc/hexane 1:3): IR (CHCl3) 2961, 2869, 1708, 1612, 1513, 1462, 1248, 1174, 1076, 1036, 820, 755 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.41-7.38 (m, 7H), 7.03-6.96 (m, 6H), 6.73-6.57 (m, 1H), 6.67 (apparent t, J=11.2 Hz, 1H), 6.31 (dd, J=15.8, 6.4 Hz, 1H), 6.12 (apparent t, J=11.0 Hz, 1H), 5.69 (d, J=11.1 Hz, 1H), 5.53-5.40 (m, 3H), 5.34-5.19 (m, 4H), 4.70-4.47 (m, 6H), 3.94 (s, 6H), 3.89 (s, 3H), 3.43-3.38 (m, 1H), 3.26-3.16 (m 2H), 3.08-3.03 (m, 1H), 2.87-2.86 (m, 1H), 2.78-2.73 (m, 2H), 2.22-2.19 (m, 2H), 2.07-2.05 (m, 3H), 1.93-1.55 (m, 5H), 1.23 (d, J=6.9 Hz, 3H), 1.19 (d, J=7.2 Hz, 9H), 1.07 (d, J=6.6 Hz, 3H); 13C NMR (75 MHz, CDCl3) δ 166.3, 159.2, 158.9, 145.4, 143.6, 134.0, 133.0, 132.2, 131.4, 130.8, 130.7, 129.9, 129.53, 128.48, 129.43, 129.4, 129.3, 129.2, 129.0, 126.2, 117.7, 116.9, 113.8, 113.6, 88.0, 83.2, 75.2, 71.7, 71.2, 55.2, 39.4, 38.4, 37.0, 35.6, 34.3, 31.7, 25.4, 24.9, 19.7, 18.6, 17.2, 15.4, 10.0; LRMS (API-ES) 899.5 (M+K)+, 883.5 (M+Na)+; [α]20 D +40.4 (c 0.47, CHCl3).
  • The same procedure for 43 was used with above lactone (14 mg, 16 μmol) to yield 3.7 mg (46%) of the product 54 by flash column chromatography (EtOAc/hexane 1:2): IR (CHCl3) 3411, 2964, 2926, 2872, 1692, 1637, 1435, 1182, 999, 962 cm−1; 1H NMR (600 MHz, CDCl3) δ 7.27 (ddt, J=15.7, 11.2, 1.2 Hz, 1H), 6.61 (ddt, J=16.8, 1.1, 10.7 Hz, 1H), 6.53 (apparent t, J=11.3 Hz, 1H), 6.02 (dd, J=15.4, 6.7 Hz, 1H), 6.01 (apparent t, J=11.0 Hz, 1H), 5.56 (d, J=11.5 Hz, 1H), 5.43 (dd, J=10.8, 9.1 Hz, 1H), 5.39-5.36 (m, 1H), 5.33 (apparent t, J=10.6 Hz, 1H), 5.30-5.23 (m, 2H), 5.19 (dt, J=16.8, 0.9 Hz, 1H), 5.10 (d, J=10.1 Hz, 1H), 5.00 (dd, J=7.8, 3.2 Hz, 1H), 3.67 (ddd, J=11.7, 5.8, 4.6 Hz, 1H), 3.41 (ddd, J=8.9, 6.0, 2.4 Hz, 1H), 3.31 (dd, J=7.0, 5.0 Hz, 1H), 3.06-3.00 (m, 1H), 2.68-2.61 (m, 2H), 2.41 (dd, J=13.7, 6.8 Hz, 1H), 2.20-2.11 (m, 2H), 1.82 (dt, J=7.2, 3.2 Hz, 1H), 1.77-1.71 (m, 2H), 1.41-1.35 (m, 2H), 1.32-1.25 (m, 2H), 1.11 (d, J=6.9 Hz, 3H), 1.06 (d, J=6.9 Hz, 3H), 1.03 (d, J=7.0 Hz, 3H), 1.02 (d, J=7.0 Hz, 3H), 1.01 (d, J=6.7 Hz, 3H); 13C NMR (125 MHz, CDCl3) δ 166.5, 145.8, 143.5, 133.8, 132.3, 132.1, 131.5, 130.0, 129.8, 129.7, 127.0, 117.9, 117.2, 79.3, 73.1, 72.7, 42.8, 40.4, 36.7, 35.0, 34.9, 34.6, 33.7, 24.7, 24.0, 18.7, 17.8, 17.3, 15.3, 9.9; HRMS (EI) calcd for C31H47O4 482.3396 (M−OH)+, found 482.3416; [α]20 D +19.2 (c 0.24, CHCl3).
  • 4(R)-Benzyl-3-[4-(2,2-dimethyl-[1,3(S)]dioxolan-4-yl)-3(S)-hydroxy-2(R)-methyl-butyryl]-oxazolidin-2-one (56). Diisopropylethylamine (13 ml) was added to a solution of propionyloxazolidinone (13.1 g) in anhydrous CH2Cl2 (250 ml) at 0° C., followed by dropwise addition of nBu2BOTf (1.0M in CH2Cl2, 68 ml). The solution was stirred for 1 h at 0° C. A solution of crude aldehyde from 55 (8.9 g) prepared before in anhydrous CH2Cl2 (10 ml) was added slowly at −78° C. After addition, the reaction mixture was warmed to 0° C. and stirred for 1 h then quenched with pH7 phosphate buffer (20 mL). A solution of hydrogen peroxide (30%, 40 mL) in MeOH (80 mL) was added at 0° C. and the mixture was allowed to stir for 1 h. The reaction mixture was extracted with CH2Cl2 (50 mL×2) and dried over MgSO4 followed by flash chromatography (EtOAc/hexane 1:1) to yield 20.7 g of product (98%): IR (CHCl3) 3434, 2956, 2929, 2858, 1724, 1472, 1463, 1257, 1097, 836, 775 cm−1; 1H NMR (300 MHz, CDCl3)
    Figure US20060270862A1-20061130-P00900
    7.33 m, 3H), 7.22 (m 2H), 4.72 (ddd, J=10.5, 6.9, 3.2 Hz, 1H), 4.35 (m, 1H), 4.23 (m, 3H), 4.12 (dd, J=8.5, 6.5 Hz, 1H), 3.82 (ddd, J=10.2, 7.0, 3.2 Hz, 1H), 3.61 (t, J=7.7 Hz, 1H), 3.25 (dd, J=13.4, 3.3 Hz, 1H), 2.82 (dd, J=13.4, 9.4 Hz, 1H), 1.80 (ddd, J=14.2, 9.7, 4.6 Hz, 1H), 1.68 (ddd, J=10.8, 7.8, 3.0 Hz, 1H), 1.43 (s, 3H), 1.38 (s, 3H), 1.30 (d, J=7.0 Hz, 3H); 13C NMR (75 MHz, CDCl3) δ 176.9, 152.9, 134.9, 129.3, 128.8, 127.3, 108.6, 73.4, 69.5, 68.5, 66.0, 54.9, 42.4, 37.5, 26.8, 25.6, 14.1, 10.8; [α]20 D −28.1 (c 4.1, CHCl3).
  • 6-(2,2-Dimethyl-[1,3(S)]dioxolan-4-yl)-5(S)-hydroxy-4(R)-methyl-hex-2-enoic acid ethyl ester (57). To a solution of RED-A1 (4.6 ml) in THF (100 ml) at −78° C. was added aldol product 56 (5.39 g) in THF (10 ml) slowly over 10 min. The evolution of gas could be seen as the solution was stirred for 10˜15 min at −78° C. The reaction was then warmed to −50° C. and stirred between −55 and −40° C. for 1 h. The reaction was quenched at −50° C. with 100 ml of EtOAc and 10 ml of MeOH and then poured into a mixture of sat'd Rochelle salt (30 ml) and Et2O (60 ml) and stirred at −20° C. for 10 min. The aqueous layer froze as a gel. The ether layer was separated and the aqueous layer rinsed quickly with Et2O (2×30 ml). The combined organic extracts were dried over MgSO4 and concentrated in vacuo. The crude aldehyde was taken immediately on to the Wittig reaction. To a 200 ml of dry THF was added 3.26 ml of triethylphosphonoacetate, followed by 1.86 g of potassium tert-butoxide. The mixture was stirred at room temperature for 10 min before cooling to −78° C. The crude aldehyde was added in 20 ml of THF and stirred overnight while warming to room temperature. The mixture was poured into 30 ml of brine, extracted with Et2O (3×40 ml), dried over MgSO4 and concentrated in vacuo. Flash silica gel chromatography (hexane/EtOAc 3:2) provided 2.02 g (52% for 2 steps) of pure product as an colorless oil: 1H NMR (300 MHz, CDCl3)
    Figure US20060270862A1-20061130-P00900
    6.88 (dd, J=15.8, 8.0 Hz, 1H), 5.83 (d, J=15.8 Hz, 1H), 4.28 (m, 1H), 4.14 (q, J=7.1 Hz, 1H), 4.03 (dd, J=8.1, 6.0 Hz, 1H), 3.76 (m, 1H), 3.53 (t, J=8.0 Hz, 1H), 2.48 (brs, 1H), 2.41 (m, 1H), 1.72-1.56 (m, 2H), 1.37 (s, 3H), 1.31 (s, 3H),1.24 (t, J=7.1 Hz, 3H), 1.07 (d, J=6.8 Hz, 3H); 13C NMR (75 MHz, CDCl3) δ 166.4, 150.3, 121.6, 108.6, 73.5, 71.3, 69.3, 60.2, 42.8, 37.2, 25.8, 25.5, 14.5, 14.1.
  • 5(S),7(S),8-Tris-(tert-butyl-dimethyl-silanyloxy)-4(R)-methyl-oct-2-enoic acid ethyl ester (58). To a stirred solution of conjugated ester 57 (1.73 g) in MeOH (20 ml) was added Dowex HCR-W2 ion-exchange resin (2.0 g, activated by aqueous 1N HCl for 24 h then filtered, MeOH as eluent) and stirred for 24 h. The resin was filtered and filtrate was concentrated and dried for 2 h in vacuo. The triol was then used in next step without further purification. To a stirred solution of triol and 2,6-lutidine (3.3 mL, 28.6 mmol) in CH2Cl2 (30 mL) at 0° C. was added TBDMSOTf (5.1 mL, 22.2 mmol) and the reaction mixture was stirred for 1 h at 0° C. The reaction mixture was quenched by the addition of water (25 mL). The reaction mixture was extracted by CH2Cl2 and dried over MgSO4 followed by the evaporation of the solvent under reduced pressure. The residue was purified by short column chromatography (hexane/EtOAc 9:1) whereupon the 58 (2.96 g, 81% for 2 steps) was obtained: 1H NMR (300 MHz, CDCl3)
    Figure US20060270862A1-20061130-P00900
    7.04 (dd, J=15.9, 6.7 Hz, 1H), 5.75 (dd, J=15.9, 1.5 Hz, 1H), 4.16 (dq, J=1.3, 7.1 Hz, 2H), 3.84 (quint, J=3.6 Hz, 1H), 3.71 (m, 1H), 3.49 (dd, J=10.1, 5.4 Hz, 1H), 3.36 (dd, J=10.1, 5.8 Hz, 1H), 2.48 (m, 1H), 1.59-1.40 (m, 2H), 1.25 (t, J=7.1 Hz, 3H), 0.99 (d, J=6.8 Hz, 3H), 0.85 (m, 27H), 0.056 (s, 3H), 0.049 (s, 3H), 0.04 (s, 3H), 0.02 (s, 3H), 0.01 (s, 6H); 13C NMR (75 MHz, CDCl3) δ 166.6, 151.7, 120.8, 72.8, 71.4, 68.0, 60.0, 42.2, 39.5, 25.9, 25.7, 18.3, 18.1, 14.2, 13.3, −3.0, −3.6, −4.2, −4.5, −5.4.
  • 5(5),7(5)-Bis-(tert-butyl-dimethyl-silanyloxy)-8-hydroxy-4(R)-methyl-oct-2-enoic acid ethyl ester (59): To a solution of TBS ether 58 (7.4 g, 12.9 mmol) in THF (10 ml) was slowly added HF-pyridine in pyridine (40 ml, prepared by slow addition of 12 ml pyridine to 3 ml HF-pyridine complex followed by dilution with 25 ml THF). The mixture was stirred overnight at room temperature and quenched with sat'd NaHCO3 (100 ml). The aqueous layer was separated and extracted with Et2O (3×50 ml). The combined organic layers were washed with sat'd CuSO4 (3×50 ml), dried over MgSO4, and concentrated. Flash column chromatography (EtOAc/Hexane 1:4) afforded 3.86 g (65%) of the alcohol 59: IR (CHCl3) 3492, 2956, 2930, 2857, 1722, 1472, 1367, 1256, 1092, 1039, 836, 775 cm−1; 1H NMR (300 MHz, CDCl3)
    Figure US20060270862A1-20061130-P00900
    7.01 dd, J=15.9, 6.7 Hz, 1H), 5.75 (dd, J=15.9, 1.5 Hz, 1H), 4.15 (dq, J=1.2, 7.2 Hz, 2H), 3.75 (m, 1H), 3.56 (m, 1H), 3.40 (m, 1H), 2.44 (m, 1H), 1.85 (t, J=5.9 Hz, 1H), 1.61 (ddd, J=11.5, 6.4, 5.0 Hz, 1H), 1.50 (ddd, J=13.0, 7.2, 5.8 Hz, 1H), 1.25 (t, J=7.1 Hz, 3H), 0.99 (d, J=6.9 Hz, 3H), 0.86 (s, 9H), 0.85 (s, 9H), 0.60 (s, 6H), 0.34 (s, 3H), 0.02 (s, 3H); 13C NMR (75 MHz, CDCl3) δ 166.5, 151.1, 121.1, 72.8, 71.0, 66.9, 60.1, 41.8, 38.7, 25.8, 18.0, 14.2, 13.3, −4.2, −4.3.
  • 5(S),7(S)-Bis-(tert-butyl-dimethyl-silanyloxy)-4(R)-methyl-8-oxo-oct-2-enoic acid ethyl ester (60). The alcohol 59 (3.86 g, 8.34 mmol) in CH2Cl2 (20 mL) was treated with Dess-Martin periodinane (5.3 g, 12.5 mmol). After 1 h, the mixture was quenched with saturated NaHCO3 (50 mL). The aqueous layer was extracted with ethyl ether (20 mL×2) and the combined extracts were dried over anhydrous MgSO4. Filtration and concentration followed by short flash column chromatography (hexane/EtOAc 4:1) to remove the residue from Dess-Martin reagent provided the aldehyde as a colorless oil: 1H NMR (300 MHz, CDCl3)
    Figure US20060270862A1-20061130-P00900
    9.53 (s, 1H), 7.02 dd, J=15.9, 6.6 Hz, 1H), 5.77 (dd, J=15.9, 1.4 Hz, 1H), 4.15 (dq, J=1.0, 7.2 Hz, 2H), 4.07 (ddd, J=6.4, 4.8, 1.4 Hz, 1H), 3.84 (ddd, J=8.6, 6.8, 4.4 Hz, 1H), 2.52 (m, 1H), 1.66 (m, 2H), 1.25 (t, J=7.1 Hz, 3H), 0.99 (d, J=6.9 Hz, 3H), 0.89 (s, 9H), 0.86 (s, 9H), 0.07 (s, 3H), 0.05 (s, 3H), 0.03 (s, 6H).
  • 5(S),7(S)-Bis-(tert-butyl-dimethyl-silanyloxy)-10(S)-[2-(4-methoxy-benzyl)-5(S)-methyl-[1,3(R)]dioxan-4-yl]-4(R)-methyl-undeca-2,8-dienoic acid ethyl ester (61). NaHMDS (1.0 M in THF, 12.3 mL, 12.3 mmol) was slowly added to a solution of the salt 21 (8.72 g, 13.7 mmol) in dry THF (13.7 mL) at 0° C. The resulting red solution was stirred at room temperature for 20 min. The mixture was cooled to −78° C. and a solution of the aldehyde 60 (5.03 g, 10.9 mmol) in THF (2.0 mL) was added dropwise. The mixture was stirred for 20 min at −78° C. and then warmed to room temperature. After 4 h at room temperature, the mixture was quenched with saturated NH4Cl (20 mL) and extracted with ethyl ether (3×30 mL). The combined organic layers were dried over anhydrous MgSO4, evaporated and the residue was flash column chromatographed (hexane/EtOAc 9:1) to yield 61(5.65 g, 75%) as a colorless oil: IR (CHCl3) 2957, 2929, 2856, 1720, 1650, 1617, 1518, 1463, 1370, 1250, 1158, 1073, 1032, 836, 774 cm−1; 1H NMR (300 MHz, CDCl3) δ7.34 m, 2H), 6.99 (dd, J=15.8, 6.9 Hz, 1H), 6.82 (m, 2H), 5.72 (dd, J=15.8, 1.5 Hz, 1H), 5.36 (s, 1H), 5.32 (dd, J=11.1, 8.6 Hz, 1H), 5.18 (t, J=10.8 Hz, 1H), 4.55 (ddd, J=12.6, 8.6, 4.1 Hz, 1H), 4.12 (m, 2H), 3.99 (d, J=7.2, 2.1 Hz, 1H), 3.91 (m, 1H), 3.77 (s, 3H), 3.52 (dd, J=9.3, 2.1 Hz, 1H), 2.64 (m, 1H), 2.37 (m, 1H), 1.64 (m, 1H), 1.46 (m, 2H), 1.22 (t, J=7.1 Hz, 3H), 1.15 (d, J=6.9 Hz, 3H), 0.93 (d, J=6.8 Hz, 6H), 0.86 (s, 18H), 0.06 (s, 3H), 0.05 (s, 3H), 0.04 (s, 3H), 0.02 (s, 3H); 13C NMR (75 MHz, CDCl3) δ 166.5, 159.7, 151.9, 133.8, 132.7, 131.3, 120.8, 113.4, 101.7, 83.6, 73.8, 71.9, 66.4, 60.0, 55.1, 43.6, 42.9, 34.2, 29.8, 26.0, 25.9, 18.1, 15.6, 14.2, 13.5, 11.2, −3.0, −3.8, −4.1, −4.5; HRMS (ESI) calcd for C36H66O7Si2K 729.3984 (M+K)+, found 729.4013; [α]20 D −8.7 (c 6.8, CHCl3).
  • 5(S),7(S)-Bis-(tert-butyl-dimethyl-silanyloxy)-10(S)-[2-(4-methoxy-benzyl)-5(S)-methyl-[1,3(R)]dioxan-4-yl]-4(R)-methyl-undeca-2,8-dien-1-ol (62). To the stirried solution of ester 61 (3.13 g, 4.53 μmol) in EtOH (20 ml), THF (2 ml) was added 1N aqueous KOH solution (45 ml) and the mixture was refluxed gently until the ester disappeared (about 6 h) as determined by TLC. The ethanolic solution was concentrated and then diluted with EtOAc (50 ml). After the solution was acidified to pH 3 with 1N HCl solution, organic phase was separated and aqueous phase was extracted with EtOAc (2×10 ml). The combined organic phase were dried with MgSO4, concentrated and used as crude in next step without further purification. The carboxylic acid was treated with NEt3 (1.5 ml) and ethyl chloroformate (0.67 ml) in dry THF (50 ml) at −10° C. After 15 min, the mixture was warmed to 0° C. and a solution of NaBH, (1.2 g) in H2O (10 ml) were added. After 4 h, the reaction was quenched by addition of sat'd Rochelle salt solution and Et2O. The layers were separated and the organic layer was washed with H2O, sat'd NaHCO3 solution and brine, dried with MgSO4. Rotary evaporation and silica column chromatography (hexane/EtOAc 4:1) gave product 62 (1.79 g, 61%) as a colorless oil: IR (CHCl3) 3433, 2957, 2929, 2856, 1617, 1518, 1462, 1388, 1250, 1074, 836, 773 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.38 (m, 2H), 6.84 (m, 2H), 5.63 (dd, J=15.7, 6.2 Hz, 1H), 5.48 (dt, J=16.0, 5.6 Hz, 1H), 5.37 (t, J=10.6 Hz, 1H), 4.59 (m, 1H), 3.99 (m, 2H), 3.93 (m, 1H), 3.87 (m, 2H), 3.77 (s, 3H), 3.49 (dd, J=9.6, 2.0 Hz, 1H), 2.68 (m, 1H), 2.31 (m, 1H), 1.79 (brs, 1H), 1.64 (m, 1H), 1.44 (m, 2H), 1.15 (d, J=6.9 Hz, 3H), 0.92 (d, J=6.9 Hz, 3H), 0.88 (m, 21H), 0.09 (s, 3H), 0.06 (s, 3H), 0.05 (s, 6H); 3C NMR (75 MHz, CDCl3) δ 159.6, 134.4, 134.3, 132.4, 131.5, 129.1, 127.4, 113.4, 101.5, 83.5, 73.8, 72.8, 66.5, 63.7, 55.2, 42.2, 34.1, 29.8, 26.1, 25.9, 18.14, 18.10, 15.5, 15.2, 11.3, −2.9, −4.1, −4.2; HRMS (ESI) calcd for C36H64O6Si2Na 671.4139 (M+Na)+, found 671.4141; [α]20 D −14.0 (c 1.5, CHCl3).
  • 4-[4(S),6(S)-Bis-(tert-butyl-dimethyl-silanyloxy)-[(S),7(R)-dimethyl-10-trityloxy-deca-2,8-dienyl]-2-(4-methoxy-benzyl)-5(S)-methyl-[1,3(R)]dioxane (63). To a solution of alcohol 62 (0.105 g) in pyridine (1.6 ml) was added trityl chloride (0.094 g) and DMAP (0.041 g). The mixture was then refluxed for 18 h, cooled to ambient temperature and added to a solution of sat'd CuSO4 (20 ml). The mixture was extracted with Et2O (2×20 ml), washed sat'd CuSO4 (2×20 ml). The organic layer was separated, dried (MgSO4), filtered, and concentrated in vacuo. Flash column chromatography (EtOAc/hexane 1:9) provided product 63 (0.142 g, 99%) as a pale yellow oil: IR (CHCl3) 2956, 2926, 2855, 1616, 1517, 1462, 1378, 1249, 1073, 835, 773, 705 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.63 (m, 6H), 7.51 (m, 2H), 7.40 (m, 9H), 6.93 (m, 2H), 5.91 (dd, J=15.7, 6.5 Hz, 1H), 5.66 (dt, J=15.5, 5.2 Hz, 1H), 5.55 (m, 1H), 5.53 (s, 1H), 5.39 (t, J=10.2 Hz, 1H), 4.78 (dt, J=3.1, 8.9 Hz, 1H), 4.10 (m, 3H), 3.80 (s, 3H), 3.70 (m, 3H), 2.85 (m, 1H), 2.45 (m, 1H), 1.78 (m, 1H), 1.65 (m, 2H), 1.31 (d, J=6.9 Hz, 3H), 1.08 (m, 24H), 0.28 (s, 3H), 0.27 (s, 3H), 0.25 (s, 6H); 13C NMR (75 MHz, CDCl3) δ 159.5, 146.8, 144.3, 135.0, 134.1, 132.4, 131.3, 128.6, 127.8, 127.6, 127.3, 127.1, 126.7, 126.3, 113.3, 101.5, 86.6, 83.4, 73.8, 72.7, 66.6, 65.0, 55.0, 43.5, 42.8, 34.2, 29.9, 26.1, 25.9, 18.1, 15.7, 14.5, 11.3, −2.9, −3.8, −4.1, −4.3; HRMS (ESI) calcd for C55H78O6Si2K 929.4969 (M+K)+, found 929.5008; [α]20 D −7.3 (c 1.1, CHCl3).
  • 7(S),9(S)-Bis-(tert-butyl-dimethyl-silanyloxy)-3-(4-methoxy-benzyloxy)-2(S)(S),10(R)-trimethyl-13-trityloxy-trideca-5,11-dien-1-ol (64). To the PMB acetal 63 (3.75 g. 4.21 μmol) in CH2Cl2 (20 ml) was added DIBAL-H (21 ml, 21 mmol, 1.0 M solution in hexane) at −78° C. dropwise and then warmed up to 0° C. and stirred for 1 h. The reaction mixture was quenched by EtOAc (10 ml) and sat'd sodium potassium tartrate solution (50 mL) followed by vigorously stirring for 4 h. The aqueous phase was extracted with CH2Cl2 (3×20 mL) and the combined organic layers were washed with brine (30 mL). After drying over MgSO4 and evaporation under vacuum, flash column chromatography (hexane/EtOAc 4:1) provided 64 (2.78 g, 74%) as a colorless oil: IR (CHCl3) 3434, 2956, 2928, 2856, 1612, 1514, 1471, 1249, 1073, 836, 774, 706 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.48 (m, 6H), 7.29 (m, 11H), 6.84 (m, 2H), 5.84 (dd, J=15.7, 6.2 Hz, 1H), 5.57 (dt, J=15.7, 5.4 Hz, 1H), 5.44 (t, J=8.7 Hz, 2H), 4.63 (m, 1H), 4.53 (d, J=10.9 Hz, 1H), 4.46 (d, J=10.9 Hz, 1H), 3.94 (m, 1H), 3.80 (s, 3H), 3.57 (d, J=4.8 Hz, 2H), 3.48 (m, 1H), 3.31 (m, 2H), 2.80 (m, 1H), 2.42 (m, 1H), 1.84 (m, 2H), 1.55 (ddd, J=14.2, 10.1, 1.9 Hz, 1H), 1.40 (ddd, J=13.9, 8.6, 2.0 Hz, 1H), 1.07 (d, J=6.8 Hz, 3H), 0.97 (m, 12H), 0.93 (s, 9H), 0.87 (d, J=7.0 Hz, 3H), 0.16 (s, 3H), 0.15 (s, 3H), 0.11 (s, 3H), 0.10 (s, 3H); 13C NMR (75 MHz, CDCl3) δ 159.0, 144.3, 134.0, 133.7, 131.5, 130.9, 129.3, 128.6, 127.9, 127.7, 127.2, 126.8, 126.5, 113.6, 86.7, 84.0, 73.9, 73.0, 66.2, 65.8, 65.1, 55.2, 42.3, 42.2, 38.0, 35.1, 26.0, 25.9, 18.5, 18.2, 18.1, 14.8, 12.0, −2.9, −4.0, −4.19, −4.23; HRMS (ESI) calcd for C55H80O6Si2K 931.5125 (M+K)+, found 931.5152; [α]20 D −21.4 (c 0.52, CHCl3).
  • 9(S),11(S)-Bis-(tert-butyl-dimethyl-silanyloxy)-5(R)-(4-methoxy-benzyloxy)-4(S),6(S),12(R)-trimethyl-15-trityloxy-pentadeca-2,7,13-trienoic acid ethyl ester (65). The alcohol 64 (2.01 g, 2.25 μmol) in CH2Cl2 (20 mL) was treated with Dess-Martin periodinane (1.43 g, 3.4 μmol). After 1 h, the mixture was quenched with saturated NaHCO3 (20 mL). The aqueous layer was extracted with ethyl ether (25 mL×2) and the combined extracts were dried over anhydrous MgSO4. Filtration and concentration followed by short flash column chromatography filtration (hexane/EtOAc 3:1) to remove the residue from Dess-Martin reagent provided crude aldehyde as a colorless oil which was used for the next reaction without further purification. To a stirred solution of triethyl phosphonoacetate (0.51 ml, 2.6 □mol) in THF (20 ml) cooled to −78° C. was added dropwise potassium tert-butoxide (0.29 g, 2.50 mol) and stirred for 30 min. Thereafter the above aldehyde in THF (5 ml) was added and the solution was stirred for 1 h at −78° C., then 2 h at 0° C. The reaction mixture was quenched by addition ‘of a sat’ d NH4Cl solution (5 ml) and diluted with diethyl ether (20 ml). The layer was separated and organic phase was washed with brine (20 ml) and dried with MgSO4, filtered, and concentrated. The residue was purified by flash chromatography (EtOAc/Hexane 1:9), yielding 2.01 g of unsaturated ester 65 (93% for 2 steps): IR (CHCl3) 2956, 2929, 2856, 1718, 1650, 1612, 1514, 1448, 1250, 1180, 1074, 836, 774, 706 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.55 (m, 6H), 7.34 (m, 11H), 7.09 (dd, J=15.8, 7.1 Hz, 1H), 6.89 (m, 2H), 5.89 (dd, J=15.7, 5.8 Hz, 1H), 5.78 (d, J=15.8 Hz, 1H), 5.66 (dt, J=6.0, 15.7 Hz, 1H), 5.45 (m, 2H), 4.66 (m, 1H), 4.51 (m, 2H), 4.23 (m, 2H), 3.99 (m, 1H), 3.83 (s, 3H), 3.66 (d, J=5.3 Hz, 2H), 3.29 (t, J=4.7 Hz, 1H), 2.79 (m, 1H), 2.65 (m, 1H), 2.49 (m, 1H), 1.60 (m, 1H), 1.48 (m, 1H), 1.33 (t, J=7.1 Hz, 3H), 1.12 (d, J=6.7 Hz, 3H), 1.11 (d, J=6.6 Hz, 3H), 1.06 (d, J=6.9 Hz, 3H), 1.01 (s, 9H), 1.00 (s, 9H), 0.20 (s, 3H), 0.19 (s, 3H), 0.17 (s, 3H), 0.15 (s, 3H); 13C NMR (75 MHz, CDCl3) δ 166.5, 158.9, 152.2, 144.3, 134.3, 133.9, 131.0, 130.5, 129.3, 128.6, 127.6, 126.7, 126.4, 120.2, 113.5, 107.0, 86.6, 85.5, 73.4, 72.8, 66.3, 65.1, 59.9, 55.1, 42.2, 38.9, 35.2, 26.0, 25.9, 18.2, 18.1, 14.6, 14.2, 13.7, −3.0, −4.1, −4.2, −4.3; HRMS (ESI) calcd for C59H84O7Si2K 999.5393 (M+K)+, found 999.5387; [α]20 D +4.6 (c 3.1, CHCl3).
  • 9(S),11 (S)-Bis-(tert-butyl-dimethyl-silanyloxy)-5(R)-(4-methoxy-benzyloxy)-4(S),6(S),12(R)-trimethyl-15-trityloxy-pentadeca-7,13-dienoic acid ethyl ester (66). To a stirred solution of unsaturated ester 65 (2.02 g, 2.100 mol) in MeOH (10 ml), THF (1 ml) at 0° C. was added 0.25 g of NiCl2.6H2O then portionwise NaBH4 (0.16 g). After 1 h, the reaction mixture was evaporated and filtered with Celite using Et2O as an eluent (5 ml). The organic phase was concentrated and the residue was purified by flash chromatography (EtOAc/Hexane 1:9) to yield 1.96 g (2.04 μmol) of product 66 (97%) as a colorless oil: IR (CHCl3) 2956, 2929, 2856, 1735, 1613, 1514, 1479, 1448, 1374, 1249, 1174, 1072, 836, 773, 706 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.53 (m, 6H), 7.33 (m, 11H), 6.84 (m, 2H), 5.81 (dd, J=15.7, 6.1 Hz, 1H), 5.65 (m, 1H), 5.45 (m, 2H), 4.65 (m, 1H), 4.56 (d, J=10.9 Hz, 1H), 4.45 (d, J=10.9 Hz, 1H), 4.14 (q, J=7.1 Hz, 2H), 3.96 (m, 1H), 3.80 (s, 3H), 3.62 (m, 2H), 3.14 (m, 1H), 2.79 (m, 1H), 2.43 (m, 1H), 2.23 (m, 1H), 1.72 (m, 2H), 1.54 (m, 3H), 1.28 (t, J=7.1 Hz, 3H), 1.06 (d, J=6.7 Hz, 3H), 1.01 (d, J=6.9 Hz, 3H), 0.97 (s, 18H), 0.93 (d, J=6.4 Hz, 3H), 0.17 (s, 3H), 0.154 (s, 31H), 0.151 (s, 3H), 0.14 (s, 3H); 13C NMR (75 MHz, CDCl3) δ 173.6, 158.8, 144.4, 134.6, 133.6, 132.1, 131.2, 129.1, 128.7, 127.7, 126.8, 126.4, 103.4, 86.6, 86.1, 73.8, 72.8, 66.5, 65.2, 60.0, 55.1, 42.8, 42.3, 35.4, 35.1, 32.3, 29.4, 26.0, 25.9, 18.4, 18.1, 14.6, 14.2, 13.9, −2.9, −4.0, −4.1; HRMS (ESI) calcd for C59H86O7Si2K 1001.5549 (M+K)+, found 1001.5586; [α]20 D −9.8 (c 0.95, CHCl3).
  • 4(R)-Benzyl-3-[9(S),11(S)-bis-(tert-butyl-dimethyl-silanyloxy)-5(R)-(4-methoxy-benzyloxy)-4(R),6(S),12(S)-trimethyl-15-trityloxy-pentadeca-7,13-dienoyl]-oxazolidin-2-one (68). To the stirred solution of ester 66 (1.61 g, 1.670 mol) in EtOH (20 ml), THF (2 ml) was added 1N aqueous KOH solution (17 ml) and the mixture was refluxed gently until the ester disappeared (about 6 h) as determined by TLC. The ethanolic solution was concentrated and then diluted with EtOAc (20 ml). After the solution was acidified to pH3 with 1N HCl solution, organic phase was separated and aqueous phase was extracted with EtOAc (2×10 ml). The combined organic phase were dried with MgSO4, concentrated and used as crude without further purification. A solution of the above acid and Et3N (0.47 ml) in dry THF (17 ml) was cooled to −78° C., treated dropwise with pivaloyl chloride (0.25 ml), stirred in the cold for 1 h, and warmed to 0° C. prior to the addition of the (S)-oxazolidinone 4 (0.30 g) and LiCl (0.21 g). This reaction, mixture was stirred overnight at room temperature and diluted with water (10 ml). The separated aqueous phase was extracted with ether (2×10 ml) and the combined organic phase were dried and evaporated and flash column chromatography (EtOAc/hexane 1:4) gave the product 68 (1.52 g, 83%) as a colorless oil: IR (CHCl3) 2956, 2856, 1785, 1701, 1612, 1513, 1449, 1385, 1249, 1074, 910, 836, 774, 734, 706 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.47 (m, 6H), 7.30 (m, 10H), 7.24 (m, 6H), 6.78 (m, 2H), 5.75 (dd, J=15.7, 6.2 Hz, 1H), 5.54 (dt, J=15.5, 5.5 Hz, 1H), 5.41 (m, 2H), 4.62 (m, 2H), 4.55 (d, J=11.0 Hz, 1H), 4.42 (d, J=11.1 Hz, 1H), 4.16 (m, 2H), 3.91 (m, 1H), 3.75 (s, 3H), 3.56 (m 2H), 3.30 (dd, J=13.4, 3.2 Hz, 1H), 3.15 (dd, J=6.7, 2.2 Hz, 1H), 2.85 (m, 2H), 2.77 (m, 2H), 2.37 (m, 1H), 1.78 (m, 2H), 1.61 (m, 3H), 1.44 (m, 3H), 1.01 (d, J=6.7 Hz, 3H), 0.96 (d, J=7.1 Hz, 3H), 0.92 (m, 21H), 0.12 (s, 3H), 0.10 (s, 3H), 0.09 (s, 6H); 13C NMR (75 MHz, CDCl3) δ 173.1, 158.7, 153.2, 144.3, 135.3, 134.7, 133.6, 132.5, 131.2, 129.3, 129.1, 128.8, 128.6, 127.6, 127.2, 126.7, 126.2, 113.4, 86.5, 85.8, 73.7, 72.7, 66.4, 65.9, 65.1, 55.0, 42.8, 42.4, 37.8, 35.5, 34.9, 33.5, 28.7, 26.0, 25.9, 18.2, 18.1, 14.5, 13.9, −2.9, −4.0, −4.2; HRMS (ESI) calcd for C67H91NO8Si2K 1132.5920 (M+K)+, found 1132.5874; [α]20 D +14.8 (c 0.61, CHCl3).
  • 4(R)-Benzyl-3-[9(S),11 (S)-bis-(tert-butyl-dimethyl-silanyloxy)-5(R)-(4-methoxy-benzyloxy)-2(S),4(S),6(S),12(R)-tetra methyl-15-trityloxy-pentadeca-7,13-dienoyl]-oxazolidin-2-one (69). NaHMDS (1.0 M in THF, 1.68 ml) was added at −78° C. to a solution of 68 (1.67 g) in THF (4 ml). After 30 min, the reaction mixture was treated with MeI (0.29 ml) at −78° C., stirred for an additional 4 h, quenched with sat'd aqueous NH4Cl, and extracted with ether (2×10 ml). The combined organic layers were dried (MgSO4), concentrated and purified by flash column chromatography (EtOAc/hexane 1:9) to give product 69 (1.05 g, 62%) as a colorless oil: IR (CHCl3) 2957, 2929, 2856, 1783, 1697, 1513, 1449, 1385, 1249, 1074, 836, 774, 705 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.49 (m, 6H), 7.29 (m, 16H), 6.80 (m, 2H), 5.79 (dd, J=15.6, 6.2 Hz, 1H), 5.56 (dt, J=15.6, 5.7 Hz, 1H), 5.42 (m, 2H), 4.62 (m, 2H), 4.56 (d, J=11.3 Hz, 1H), 4.37 (d, J=11.1 Hz, 1H), 4.17 (m, 1H), 4.05 (m, 1H), 3.92 (m, 1H), 3.77 (s, 3H), 3.58 (d, J=5.2 Hz, 1H), 3.27 (m, 1H), 3.08 (dd, J=6.3, 2.5 Hz, 1H), 2.77 (m, 2H), 2.38 (m, 1H), 1.76 (m, 1H), 1.64 (m, 2H), 1.46 (m, 4H), 1.10 (d, J=6.7 Hz, 3H), 1.00 (d, J=6.3 Hz, 3H), 0.98 (d, J=6.7 Hz, 3H), 0.93 (m, 21H), 0.14 (s, 3H), 0.11 (s, 6H), 0.10(s, 3H); 13C NMR (75 MHz, CDCl3) δ 177.3, 158.7, 152.8, 144.4, 135.3, 134.9, 133.6, 132.3, 131.3, 129.4, 128.9, 128.8, 128.6, 127.6, 127.2, 126.7, 126.3, 113.5, 86.6, 86.5, 74.0, 72.8, 66.5, 65.8, 65.2, 43.0, 42.5, 37.8, 35.4, 35.3, 33.0, 26.3, 26.0, 25.9, 18.3, 18.1, 17.4, 14.5, 14.2, −2.9, −4.0, −4.1, 4.2; HRMS (ESI) calcd for C68H93NO8Si2K 1146.6077 (M+K)+, found 1146.6079; [α]20 D +16.70 (c 1.1, CHCl3).
  • 9(S),11(S)-Bis-(tert-butyl-dimethyl-silanyloxy)-5(R)-(4-methoxy-benzyloxy)-2(5),4(S),6(S),12(R)-tetramethyl-15-trityloxy-pentadeca-7,13-dien-1-ol (70). To a stirred solution of 69 (0.41 g, 0.37 mmol) in THF (1.5 ml) at 0° C. was added MeOH (0.015 ml) and LiBH4 (0.81 ml, 2.0 M soln in THF) dropwise. After stirring 2 h at 0° C., saturated sodium potassium tartrate (10 ml) was added dropwise. The reaction mixture was warmed to room temperature and extracted with CH2Cl2 (10 ml×2). The combined organic layer were washed with brine (10 ml) and dried over anhydrous MgSO4, evaporated and the residue was chromatographed (hexane/EtOAc 4:1) to yield 70 (0.30 g, 87%) as a colorless oil: IR (CHCl3) 3400, 2956, 2928, 2856, 1613, 1514, 1449, 1377, 1249, 1074, 836, 774, 706 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.48 (m, 6H), 7.29 (m, 11H), 6.84 (m, 2H), 5.78 (dd, J=15.7, 6.0 Hz, 1H), 5.58 (dt, J=15.7, 5.2 Hz, 1H), 5.46 (m, 1H), 5.35 (m, 1H), 4.59 (t, J=9.5, Hz, 1H), 4.48 (q, J=10.9 Hz, 2H), 3.92 (m, 1H), 3.79 (s, 3H), 3.57 (d, J=5.5 Hz, 2H), 3.25 (m 2H), 3.03 (t, J=4.5 Hz, 1H), 2.75 (m 1H), 2.41 (m, 1H), 1.75 (m, 1H), 1.55 (m, 2H), 1.32 (m, 2H), 1.17 (m, 2H), 1.07 (d, J=6.7 Hz, 3H), 0.97 (d, J=6.8 Hz, 3H), 0.94 (s, 9H), 0.91 (m, 12H), 0.72 (d, J=6.6 Hz, 3H), 0.13 (s, 3H), 0.12 (s, 3H), 0.09 (s, 3H), 0.07 (s, 3H); 13C NMR (75 MHz, CDCl3) δ 158.9, 144.4, 134.4, 133.4, 131.5, 131.4, 129.1, 128.7, 127.7, 126.8, 126.5, 113.6, 87.6, 86.8, 74.1, 73.0, 68.9, 66.5, 65.4, 55.2, 42.7, 42.4, 37.1, 35.0, 33.1, 26.0, 25.9, 18.9, 18.1, 15.8, 14.9, 14.7, −2.8, −4.0, −4.06, −4.10; HRMS (ESI) calcd for C58H86O6Si2K 973.6301 (M+K)+, found 973.6264; [α]20 D −31.7 (c 1.3, CHCl3).
  • {3(R)-[2-(4-Methoxy-phenyl)-5(S)-methyl-[1 (S),3]dioxan-4-yl]-2-oxo-butyl}-phosphonic acid dimethyl ester (71). n-Butyllithium (4.5 ml, 1.6 M solution in hexane) was added dropwise to a stirred solution of dimethyl methanephosphonate (0.77 ml) in THF (7 ml) at −78° C. After 1 h, a solution of the known weinreb amide (Smith, A. B. et al. J. Am. Chem. Soc. 2000, 122, 8654-8664) (0.46 g) in THF (0.5 ml) was added. After 30 min, the reaction was then allowed to warm to 0° C. and quenched by pouring into brine (100 ml) and extracted with EtOAc (2×50 ml). The combined extracts were washed with brine (50 ml), dried over MgSO4 and concentrated in vacuo. Flash silica gel column chromatography (EtOAc) gave the desired product 71 (0.47 g, 85%) as a colorless oil: 1H NMR (300 MHz, CDCl3) δ 7.38 (m, 2H), 6.89 (m, 2H), 5.50 (s, 1H), 4.14 (dd, J=11.3, 4.7, Hz, 1H), 4.06 (dd, J=10.0, 2.7 Hz, 1H), 3.80 (s, 3H), 3.77 (s, 3H), 3.74 (s, 3H), 3.59 (t, J=11.1 Hz, 1H), 3.42 (d, J=14.5 Hz, 1H), 3.34 (d, J-=14.5 Hz, 1H), 3.20 (d, J=14.5 Hz, 1H), 3.13 (d, J=14.5 Hz, 1H), 3.02 (dq, J=2.8, 7.0 Hz, 1H), 2.06 (m, 1H), 1.26 (d, J=7.0 Hz, 3H), 0.85 (d, J=6.7 Hz, 3H); 13C NMR (75 MHz, CDCl3) δ 202.5, 159.5, 130.4, 126.9, 113.1, 100.5, 82.1, 72.4, 54.9, 52.6, 48.6, 39.3, 37.6, 30.6, 11.6, 8.7
  • 13(S),15(S)-Bis-(tert-butyl-dimethyl-silanyloxy)-2-[2-(4-methoxy-benzyl)-5(S)-methyl-[1,3(S)]dioxan-4-yl]-9(R)-(4-methoxy-benzyloxy)-6(S),8(S),10(S),16(R)-tetramethyl-19-trityloxy-nonadeca-4,11,17-trien-3-one (72). The alcohol 70 (0.30 g, 0.32 μmol) in CH2Cl2 (10 mL) was treated with Dess-Martin periodinane (0.20 g, 0.47 μmol). After 1 h, the mixture was quenched with saturated NaHCO3 (10 mL). The aqueous layer was extracted with ethyl ether (10 mL×2) and the combined extracts were dried over anhydrous MgSO4. Filtration and concentration followed by short flash column chromatography filtration (hexane/EtOAc 4:1) to remove the residue from Dess-Martin reagent provided crude aldehyde as a colorless oil which was used for the next reaction without further purification. A mixture of ketophosphonate 71 (0.14 g) and Ba(OH)2 (0.043 g, activated by heating to 100° C. for 1-2 h before use) in THF (2 ml) was stirred at room temperature for 30 min. A solution of the above aldehyde in wet THF (2 ml+2×1 ml washings, 40:1 THF/H2O) was then added and stirred for overnight. The reaction mixture was diluted with Et2O (10 ml) and washed with sat'd NaHCO3 (10 ml) and brine (10 ml). The organic solution was dried (MgSO4) and the solvent was evaporated in vacuo. The residue was chromatographed (hexane/EtOAc 4.5:1) to yield 72 (0.34 g, 90%) as a colorless oil: IR (CHCl3) 2957, 2929, 2855, 1615, 1515, 1461, 1249, 1076, 1036, 835, 774 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.47 (m, 6H), 7.38 (m, 2H), 7.28 (m, 12H), 6.89 (m, 2H), 6.78 (m, 2H), 6.22 (d, J=15.6 Hz, 1H), 5.74 (dd, J=15.7, 6.2 Hz, 1H), 5.57 (m, 1H), 5.45 (s, 1H), 5.38 (m, 2H), 4.60 (m, 1H), 4.52 (d, J=11.0 Hz, 1H), 4.33 (d, J=11.0 Hz, 1H), 4.12 (dd, J=11.2, 4.5 Hz, 1H), 3.90 (m, 2H), 3.81 (s, 3H), 3.76 (s, 3H), 3.55 (m, 3H), 3.04 (m, 1H), 2.92 (m, 1H), 2.75 (m, 1H), 2.36 (m, 1H), 2.25 (quint, J=7.2 Hz, 1H), 2.02 (m, 1H), 1.71 (m, 1H), 1.56˜1.33 (m, 4H), 1.25 (d, J=6.9 Hz, 3H), 0.96 (d, J=7.8 Hz, 3H), 0.95 (d, J=7.1 Hz, 3H), 0.92 (m, 211H), 0.85 (d, J=7.3 Hz, 3H); 13C NMR (75 MHz, CDCl3) δ 201.1, 159.7, 158.8, 153.1, 144.3, 134.6, 133.6, 132.4, 131.2, 131.0, 129.1, 128.6, 127.7, 127.2, 126.8, 126.3, 126.0, 113.5, 113.4, 100.7, 86.6, 85.7, 82.8, 73.8, 72.8, 66.4, 65.2, 55.2, 47.0, 42.8, 42.4, 40.4, 35.5, 34.2, 32.8, 32.2, 26.0, 25.9, 19.2, 18.4, 18.3, 18.1, 14.5, 14.4, 12.4, 10.7, −2.9, −4.0, −4.1; HRMS (ESI) calcd for C74H104O9Si2K 1231.6856 (M+K)+, found 1231.6850; [α]20 D +22.8 (c 0.88, CHCl3).
  • 13(S),15(S)-Bis-(tert-butyl-dimethyl-silanyloxy)-2-[2-(4-methoxy-benzyl)-5(S)-methyl-[1,3(S)]dioxan-4-yl]-9(R)-(4-methoxy-benzyloxy)-6(S),8(S),10(S),16(R)-tetramethyl-19-trityloxy-nonadeca-11,17-dien-3-one (73). To a stirred solution of unsaturated ketone 72 (0.34 g, 0.29 μmol) in MeOH (4 ml), THF (0.5 ml) at 0° C. was added 0.034 g of NiCl2, 6H2O then portionwise NaBH4 (0.022 g). After 1 h, the reaction mixture was evaporated and filtered with Celite using Et2O as a eluent (5 ml). The organic phase was concentrated and the residue was purified by flash chromatography (EtOAc/Hexane 1:4) to yield 0.31 g of product 73 (89%) as a colorless oil: IR (CHCl3) 2956, 2929, 2855, 1713, 1614, 1515, 1461, 1249, 1075, 1036, 835, 774, 706 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.47 (m, 6H), 7.29 (m, 13H), 6.87(m, 2H), 6.80 (m, 2H), 5.75 (dd, J=15.7, 6.1 Hz, 1H), 5.55 (m, 1H), 5.45 (s, 1H), 5.38 (m, 2H), 4.60 (m, 1H), 4.48 (d, J=10.9 Hz, 1H), 4.36 (d, J=10.9 Hz, 1H), 4.13 (dd, J=11.2, 4.4 Hz, 1H), 3.93 (m, 2H), 3.79 (s, 3H), 3.76 (s, 3H), 3.55 (m, 2H), 2.99 (m, 2H), 2.70 (m, 2H), 2.45 (t, J=7.0 Hz, 1H), 2.36 (m, 1H), 2.02 (m, 1H), 1.75 (m, 1H), 1.63 (m, 1H), 1.49 (m, 2H), 1.37 (m, 3H), 1.23 (d, J=7.1 Hz, 3H), 1.02 (d, J=6.7 Hz, 3H), 0.95 (d, J=7.0 Hz, 3H), 0.91 (m, 21H), 0.81 (d, J=6.8 Hz, 3H), 0.80 (d, J=6.7 Hz, 3H), 0.12 (s, 3H), 0.09 (s, 6H), 0.08 (s, 3H); 13C NMR (75 MHz, CDCl3) δ 211.9, 159.8, 158.8, 144.6, 144.4, 134.9, 133.4, 132.3, 131.8, 131.5, 131.0, 129.0, 128.9, 128.7, 127.7, 127.6, 127.2, 126.8, 126.7, 126.3, 113.5, 100.8, 87.4, 86.7, 83.1, 74.0, 72.9, 66.6, 65.2, 55.22, 55.18, 48.3, 43.1, 42.5, 41.6, 38.3, 35.5, 32.7, 31.5, 31.3, 29.6, 26.1, 26.0, 19.0, 18.5, 18.1, 14.5, 14.1, 12.1, 9.7, −2.9, −4.0, −4.1, −4.2; HRMS (ESI) calcd for C74H108O9Si2K 1233.7013 (M+K)+, found 1233.7036; [α]20 D +3.0 (c 1.7, CHCl3).
  • 13(S),15(S)-Bis-(tert-butyl-dimethyl-silanyloxy)-2-[2-(4-methoxy-benzyl)-5(S)-methyl-[1,3(S)]dioxan-4-yl]-9(R)-(4-methoxy-benzyloxy)-6(S),8(S),10(S),16(R)-tetramethyl-19-trityloxy-nonadeca-11,17-dien-3-ol (74). To a solution of 73 (0.27 g) in MeOH (4 ml) was added NaBH4 (0.013 g) at 0° C. After stirring for 2 h at 0° C., the reaction mixture was evaporated and water (5 ml) was added. The reaction mixture was extracted with ether (2×20 ml) and washed with brine (10 ml), dried over MgSO4 and concentrated in vacuo. The residue was purified by flash chromatography (EtOAc/Hexane 1:4.5) to yield 0.19 g of major product 74 (71%) and 0.069 g (25%) of minor product as a colorless oil: (major isomer) IR (CHCl3) 3533, 2956, 2929, 2855, 1614, 1515, 1462, 1250, 1072, 1036, 835, 774, 734 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.51 (m, 6H), 7.43 (m, 2H), 7.30 (m, 11H), 6.92 (m, 2H), 6.84 (m, 2H), 5.78 (dd, J=15.6, 6.1 Hz, 1H), 5.61 (m, 1H), 5.57 (s, 1H), 5.43 (m, 2H), 4.65 (m, 1H), 4.55 (d, J=11.0 Hz, 1H), 4.45 (d, J=10.8 Hz, 1H), 4.18 (dd, J=11.2, 4.5 Hz, 1H), 3.95 (m, 1H), 3.84 (s, 3H), 3.82 (m, 1H), 3.79 (s, 3H), 3.74 (m, 1H), 3.59 (m, 2H), 3.06 (m, 2H), 2.78 (m, 1H), 2.41 (m, 1H), 2.19 (m, 1H), 1.81 (m, 2H), 1.56 (dd, J=13.8, 8.1 Hz, 3H), 1.44 (m, 3H), 1.34 (m, 3H), 1.08 (d, J=7.0 Hz, 6H), 0.99 (d, J=7.2 Hz, 3H), 0.96 (m, 18H), 0.90 (d, J=6.7 Hz, 3H), 0.82 (d, J=6.6 Hz, 6H), 0.16 (s, 3H), 0.14 (s, 6H), 0.13 (s, 3H); 13C NMR (75 MHz, CDCl3) δ 160.0, 158.8, 144.6, 144.4, 134.9, 133.4, 132.3, 131.5, 130.7, 129.0, 128.9, 128.7, 127.7, 127.6, 127.2, 126.8, 126.7, 126.3, 113.7, 113.5, 89.0, 87.5, 86.7, 76.7, 74.0, 73.1, 72.8, 66.6, 65.2, 55.2, 55.1, 43.1, 42.5, 41.8, 37.4, 35.5, 34.4, 32.9, 32.4, 30.4, 30.1, 26.0, 25.9, 19.2, 18.5, 18.1, 14.5, 14.1, 11.9, 5.7, −2.9, −4.0, −4.1, −4.2; HRMS (ESI) calcd for C74H108O9Si2K 1235.7169 (M+K)+, found 1235.7149; [α]20 D +3.5 (c 0.6, CHCl3).
  • 5,15(S),17(S)-Tris-(tert-butyl-dimethyl-silanyloxy)-11(R)-(4-methoxy-benzyloxy)-3(S)-[2-(4-methoxy-phenyl)-ethoxy]-2(S),4(R),8(S),10(S),12(S),18(R)-hexamethyl-21-trityloxy-heneicosa-13,19-dien-1-ol (76). To a stirred solution of 74 (0.19 g, 0.16 mmol) and 2,6-lutidine (0.037 mL, 0.32 mmol) in CH2Cl2 (16 mL) at 0° C. was added TBDMSOTf (0.055 mL, 0.24 mmol) and the reaction mixture was stirred for 2 h at ambient temperature. The reaction mixture was quenched by the addition of water (5 mL). The reaction mixture was extracted by CH2Cl2 and dried over MgSO4 followed by the evaporation of the solution under reduced pressure. The residue was purified by short column chromatography (hexane/EtOAc 9:1). To a stirred solution of TBS protected acetal (0.20 g, 0.15 mmol) in anhydrous CH2Cl2 (3 mL), under an atmosphere of N2 at 0° C. was added diisobutylaluminum hydride (1.0 M in THF, 1.5 mL, 1.5 mmol) dropwise, and the reaction mixture was stirred for additional 1 h at 0° C. The reaction mixture was quenched by the careful addition of aqueous sat'd potassium sodium tartrate solution (10 mL). The reaction mixture was stirred for 3 h at room temperature. The organic layer was separated, and the water layer was extracted by CH2Cl2 (20 mL). The combined organic layer was washed with brine and dried over MgSO4 followed by the evaporation of the organic solution under reduced pressure. The residue was purified by column chromatography (EtOAc/hexane 1:4) whereupon the pure compound 76 (0.19 g, 91% for 2 steps) was obtained: IR (CHCl3) 3466, 2955, 2928, 2856, 1613, 1514, 1462, 1249, 1072, 1037, 835, 773 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.52 (m, 6H), 7.30 (m, 13H), 6.94 (m, 2H), 6.85 (m, 2H), 5.79 (dd, J=15.7, 6.3 Hz, 1H), 5.59 (dt, J=15.7, 5.9 Hz, 1H), 5.44 (m, 2H), 4.67 (m, 1H), 4.60 (s, 2H), 4.57 (d, J=11.1 Hz, 1H), 4.44 (d, J=10.9 Hz, 1H), 3.97 (m, 1H), 3.91 (m, 1H), 3.85 (s, 3H), 3.79 (s, 3H), 3.68 (m, 2H), 3.60 (d, J=5.6 Hz, 11H), 3.52 (dd, J=6.6, 4.3 Hz, 1H), 3.07 (m, 2H), 2.97 (brs, 1H), 2.80 (dd, J=14.5, 6.7 Hz, 1H), 2.40 (m, 1H), 2.02 (m, 1H), 1.95 (ddd, J=9.6, 6.9, 4.0 Hz, 1H), 1.81 (m, 1H), 1.71 (m, 1H), 1.56 (m, 3H), 1.47 (m, 3H), 1.33 (m, 2H), 1.19 (d, J=7.0 Hz, 3H), 1.08 (d, J=6.7 Hz, 6H), 1.00 (s, 9H), 0.97 (m, 21H), 0.90 (d, J=6.7 Hz, 3H), 0.82 (d, J=6.4 Hz, 3H), 0.17 (s, 3H), 0.15 (s, 3H), 0.14 (s, 3H), 0.137 (s, 3H), 0.133 (s, 3H), 0.127 (s, 3H); 13C NMR (75 MHz, CDCl3) δ 159.3, 158.8, 144.6, 144.4, 135.0, 133.6, 132.5, 131.4, 130.6, 129.2, 129.0, 128.9, 128.7, 127.7, 127.6, 126.8, 126.7, 126.3, 113.9, 113.5, 87.4, 86.7, 85.9, 75.3, 74.0, 73.6, 72.8, 66.6, 65.2, 65.1, 55.2, 55.1, 43.2, 42.5, 42.0, 41.5, 37.0, 35.6, 33.4, 32.9, 31.9, 30.1, 26.08, 26.05, 25.98, 19.4, 18.4, 18.1, 15.8, 14.4, 13.9, 10.0, −2.9, −3.7, −3.9,−4.1, 4.2, −4.4; HRMS (ESI) calcd for C80H124O9Si3K 1351.8190 (M+K)+, found 1351.8134; [α]20 D −6.1 (c 0.48, CHCl3).
  • 7,17(S),19(S)-Tris-(tert-butyl-dimethyl-silanyloxy)-5(S),13(R)-bis-(4-methoxy-benzyloxy)-4(S),6(S),10(R),12(S),14(S),20(S)-hexamethyl-23-trityloxy-tetracosa-1,3,15,21-tetraen (77). The alcohol 76 (0.17 g, 0.13 μmol) in CH2Cl2 (5 ml) was treated with Dess-Martin periodinane (0.081 g, 0.21 mol). After 1 h, the mixture was quenched with saturated NaHCO3 (5 ml). The aqueous layer was extracted with ethyl ether (5 ml×2) and the combined extracts were dried over anhydrous MgSO4. Filtration and concentration followed by short flash column chromatography filtration (hexane/EtOAc 4.5:1) to remove the residue from Dess-Martin reagent provided crude aldehyde as a colorless oil which was used for the next reaction without further purification. To a stirred solution of the above crude aldehyde and 1-bromoallyl trimethylsilane (160 mg, 0.65 mmol) in anhydrous THF (3 ml) under an atmosphere of N2 at room temperature was added CrCl2 (0.13 g, 1.1 mmol) and the mixture was stirred for additional 14 h at ambient temperature. The reaction mixture was diluted with hexane followed by filtration through celite. After the evaporation of the solvent under reduced pressure, the residue was purified by short silica gel column chromatography using EtOAc/hexane (1:9). The foregoing product in THF (3 ml) was cooled to 0° C. and NaH (95% w/w, 64 mg, 2.56 mmol) was added in one portion. The ice bath was removed after 15 min and the mixture was stirred for 2 h at ambient temperature. The reaction mixture was cooled to 0° C., quenched with H2O (5 ml), extracted with ethyl ether (5 ml×2). The combined organic layer was washed with brine and dried over MgSO4 followed by the evaporation of the organic solution under reduced pressure. The residue was purified by column chromatography (hexane/EtOAc 9:1) whereupon the pure compound 77 (122 mg, 72% for 3 steps) was obtained: IR (CHCl3) 2955, 2928, 2856, 1613, 1514, 1462, 1249, 1072, 1039, 835, 773, 705 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.47 (m, 6H), 7.28 (m, 13H), 6.89 (m, 2H), 6.79 (m, 2H), 6.61 (ddd, J=16.8, 10.7, 10.6 Hz, 1H), 6.04 (t, J=10.8 Hz, 1H), 5.73 (dd, J=15.6, 6.3 Hz, 1H), 5.61 (t, J=10.4 Hz, 1H), 5.58 (m, 1H), 5.37 (m, 2H), 5.20 (d, J=16.8 Hz, 1H), 5.11 (d, J=10.1 Hz, 1H), 4.54 (m, 3H), 4.50 (d, J=11.0 Hz, 1H), 4.37 (d, J=10.8 Hz, 1H), 3.90 (m, 1H), 3.82 (s, 3H), 3.76 (s, 3H), 3.62 (m, 1H), 3.54 (d, J=5.3 Hz, 1H), 3.35 (dd, J=7.7, 3.1 Hz, 1H), 3.00 (m, 2H), 2.73 (m, 1H), 2.31 (m, 1H), 1.69 (m, 4H), 1.43 (m, 8H), 1.14 (d, J=6.8 Hz, 3H), 1.00 (d, J=7.1 Hz, 3H), 0.96 (s, 9H), 0.92 (s, 3H), 0.91 (s, 3H), 0.89 (m, 6H), 0.83 (d, J=6.6 Hz, 3H), 0.72 (d, J=6.4 Hz, 3H), 0.11 (s, 3H), 0.10 (s, 3H), 0.08 (s, 3H), 0.07 (s, 6H); 13C NMR (75 MHz, CDCl3) δ 159.0, 158.8, 144.6, 144.4, 135.0, 134.6, 133.7, 133.4, 132.6, 132.4, 131.5, 131.4, 129.1, 129.0, 128.98, 128.94, 128.7, 127.7, 126.8, 126.3, 117.2, 113.7, 113.5, 87.3, 86.7, 84.3, 75.0, 74.0, 72.9, 72.8, 66.6, 65.2, 55.2, 55.1, 43.2, 42.6, 42.0, 40.6, 35.7, 35.3, 33.2, 32.8, 32.3, 30.1, 26.1, 26.0, 19.4, 18.8, 18.3, 18.2, 18.1, 14.4, 14.0, 13.9, −2.9, −3.6, −3.9, −4.1, −4.2, −4.4; [α]20 D +2.5 (c 1.2, CHCl3).
  • 7(S),9(S),19-Tris-(tert-butyl-dimethyl-silanyloxy)-13(R),21(S)-bis-(4-methoxy-benzyloxy)-6(R),12(S),14(S),16(S),20(R),22(S)-hexamethyl-hexacosa-2,4,10,23,25-pentaenoic acid methyl ester (79). A solution of 77 (18.6 mg) in CH2Cl2 (0.2 ml) was cooled to −78° C. and B-chlorocatecholborane (0.25 M in CH2Cl2, 0.17 ml) was added. The solution was stirred at −78° C. for 1 h followed by treatment with sat'd aqueous NaHCO3 (1 ml). The resulting reaction mixture was then diluted with CH2Cl2 (10 ml) and H2O (3 ml). The layers were separated and the aqueous layer was further extracted with CH2Cl2 (2×5 ml). The combined organic layers were washed with brine, dried over MgSO4 and concentrated under vacuum. The residue was purified by flash chromatography (hexane/EtOAc 4;1) on silica gel to yield 78 (9.4 mg) as a colorless oil. The alcohol 78 (20 mg, 0.018 μmol) in CH2Cl2 (0.5 mL) was treated with Dess-Martin periodinane (12 mg, 0.028 μmol). After 1 h, the mixture was quenched with saturated NaHCO3 (1 ml). The aqueous layer was extracted with ethyl ether (3 ml×2) and the combined extracts were dried over anhydrous MgSO4. Filtration and concentration followed by short flash column chromatography filtration (hexane/EtOAc 4.5:1) to remove the residue from Dess-Martin reagent provided crude aldehyde as a colorless oil which was used for the next reaction without further purification. To a stirred solution of bis(2,2,2-trifluoroethyl)-(methoxycarbonylmethyl) phosphate (0.005 ml, 0.024 □mol), 18-crown-6 (0.024 g, 0.09 mmol) in THF (0.5 ml) cooled to −78° C. was added dropwise potassium bis(trimethylsilyl)amide (0.044 ml, 0.022 □mol, 0.5M solution in toluene). Thereafter the above aldehyde in THF (0.5 ml) was added and the solution was stirred for 6 h at −78° C. The reaction mixture was quenched by addition of a sat'd NH4Cl solution (1 ml) and diluted with diethyl ether (5 ml). The layers were separated and organic phase was washed with brine (5 ml) and dried with MgSO4, filtered, and concentrated. The residue was purified by flash chromatography (EtOAc/Hexane 1:9) yielding 17 mg of (E,Z)-doubly unsaturated ester 79 (82% for 2 steps): IR (CHCl3) 2956, 2929, 2856, 1720, 1613, 1514, 1462, 1249, 1173, 1075, 836, 773 cm−1; 1H NMR (300 MHz, CDCl3) δ 7.22 (m, 5H), 6.82 (m, 4H), 6.55 (ddd, J=16.8, 10.8, 10.8 Hz, 1H), 6.38 (t, J=11.4 Hz, 1H), 6.05 (dd, J=15.4, 6.2 Hz, 1H), 5.98 (t, J=11.0 Hz, 1H), 5.55 (t, J=10.5 Hz, 1H), 5.48 (d, J=11.5 Hz, 1H), 5.31 (m, 2H), 5.14 (d, J=16.8 Hz, 1H), 5.05 (d, J=10.1 Hz, 1H), 4.54 (m, 1H), 4.49 (m, 3H), 4.31 (d, J=10.9 Hz, 11H), 3.87 (m, 1H), 3.77 (s, 3H), 3.75 (s, 3H), 3.68 (s, 3H), 3.57 (m, 11H), 3.29 (dd, J=7.7, 3.1 Hz, 1H), 2.94 (m, 2H), 2.68 (m, 1H), 2.48 (m, 1H), 1.65 (m, 3H), 1.43-1.28 (m, 6H), 1.20 (m, 2H), 1.08 (d, J=6.8 Hz, 3H), 0.96 (d, J=6.9 Hz, 3H), 0.94 (d, J=6.1 Hz, 3H), 0.90 (s, 9H), 0.86 (m, 21H), 0.81 (d, J=6.7 Hz, 3H), 0.71 (d, J=6.4 Hz, 3H), 0.06 (s, 3H), 0.05 (s, 3H), 0.04 (s, 3H), 0.02 (s, 6H); 13C NMR (75 MHz, CDCl3) δ 166.9, 159.0, 158.8, 147.7, 146.9, 145.8, 134.6, 133.5, 132.7, 132.5, 131.6, 131.4, 129.1, 128.9, 128.8, 128.7, 128.4, 127.9, 127.7, 127.3, 126.4, 117.2, 114.9, 113.7, 113.6, 87.6, 84.3, 77.2, 74.9, 74.2, 72.9, 72.7, 66.4, 55.3, 55.2, 50.9, 43.1, 42.5, 42.1, 40.6, 35.8, 35.3, 33.6, 33.2, 32.9, 18.14, 18.11, 14.6, 13.9, 9.3, −2.9, −3.6, −3.9, −4.1, −4.4; HRMS (ESI) calcd for C67H114O9Si3K 1185.7408 (M+K)+, found 1185.7464; [α]20 D −12.6 (c 0.75, CHCl3).
  • 8(S),10(S),14(R),20-Tetrahydroxy-7(S),13(S),15(S),17(R),21(S)-pentamethyl-22(S)-(1 (S)-methyl-penta-2,4-dienyl)-oxa-cyclodocosa-3,5,11-trien-2-one (83). The ester 79 (8.5 mg, 7.4 μmol) was dissolved in CH2Cl2 (1 ml)-H2O (0.05 ml) and DDQ (5.0 mg, 22 μmol) was added at 0° C. After 1 h of stirring at 0° C., the reaction mixture was quenched by adding sat'd NaHCO3 (5 ml). The organic phase was washed by sat'd NaHCO3 solution (3×10 ml) and brine, dried over MgSO4 and concentrated. Purification by flash column chromatography (EtOAc/hexane 1:4.5) furnished diol (6.4 mg, 95%) as a colorless oil. To the stirried solution of the above diol (6.4 mg, 7.06 μmol) in EtOH (0.7 ml) was added 1N aqueous KOH solution (0.07 ml) and the mixture was refluxed gently until the ester disappeared (about 7 h) as determined by TLC. The ethanolic solution was concentrated and then diluted with ether (4 ml). After the solution was acidified to pH3 with 1N HCl solution, organic phase was separated and aqueous phase was extracted with EtOAc (2×2 ml). The combined organic phase were dried with MgSO4, concentrated and used as crude without further purification. A solution of above dihydroxy acid in THF (0.5 ml) was treated at 0° C. with Et3N (0.006 ml, 43 μmol) and 2,4,6-trichlorobenzoyl chloride (0.0055 ml, 35 μmol). The reaction mixture was stirred at 0° C. for 30 min and then added to a 4-DMAP (3.5 ml, 0.02 M solution in toluene) at 25° C. and stirred for overnight. The reaction mixture was concentrated, EtOAc (5 mL) was added and the crude was washed with 1N HCl (2×5 ml), dried over MgSO4. Purification by flash column chromatography (EtOAc/hexane 1:9) furnished macrolactone (3.0 mg, 49% for 2 steps) as a colorless oil. To a stirred solution of the above macrolactone (2.7 mg, 3.1 μmol) in MeOH (0.5 ml) at 0° C. was added 0.5 ml of 3 N HCl (prepared by adding 25 ml of conc. HCl to 75 ml MeOH). After 2 h at room temperature, the reaction mixture was diluted with EtOAc (2 ml) and H2O (2 ml) and the organic phase was separated and aqueous phase was extracted with EtOAc (2×2 ml). The combined organic phase was washed with sat'd NaHCO3 (5 ml), dried with MgSO4, concentrated and the residue was purified by flash chromatography (EtOAc/Hexane 1:1) to yield 83 (1.2 mg, 73%): IR (CHCl3) 3400, 2960, 2926, 2854, 1693, 1635, 1599, 1461, 1378, 1277, 1183, 1075, 964 cm−1; 1H NMR (500 MHz, CD3OD) δ 7.34 (dd, J=15.3, 11.3 Hz, 1H), 6.64 (ddd, J=16.9, 10.5, 10.3 Hz, 1H), 6.57 (t, J=11.4 Hz, 1H), 5.96 (t, J=10.9 Hz, 1H), 5.95 (dd, J=15.3, 8.3 Hz, 1H), 5.48 (t, J=10.0 Hz, 1H), 5.47 (d, J=11.6 Hz, 1H), 5.38 (dd, J=11.1, 8.9 Hz, 11H), 5.27 (t, J=10.5 Hz, 1H), 5.16 (d, J=16.9 Hz, 1H), 5.08 (d, J=10.2 Hz, 1H), 5.02 (dd, J=8.0, 3.5 Hz, 1H), 4.65 (dt, J=3.1, 8.4 Hz, 1H), 3.72 (ddd, J=9.0, 6.3, 2.8 Hz, 1H), 3.25 (ddd, J=10.2, 7.4, 2.8 Hz, 1H), 3.16 (dd, J=5.4, 3.4 Hz, 1H), 3.06 (dd, J=16.3, 8.3 Hz, 1H), 2.72 (ddd, J=10.2, 6.7, 6.6 Hz, 1H), 2.36 (dd, J=14.7, 7.2 Hz, 1H), 1.86 (dt, J=6.6, 3.1 Hz, 1H), 1.81 (ddd, J=10.5, 6.8, 3.7 Hz, 1H), 1.69 (m, 2H), 1.58 (m, 1H), 1.47 (ddd, J=13.8, 9.5, 3.5 Hz, 1H), 1.37 (m, 1H), 1.25 (m, 1H), 1.17 (m, 1H), 1.13 (m, 1H), 1.09 (d, J=6.8 Hz, 3H), 1.03 (d, J=6.9 Hz, 6H), 0.98 (d, J=6.7 Hz, 3H), 0.87 (d, J=6.7 Hz, 3H), 0.76 (d, J=6.4 Hz, 3H); 13C NMR (125 MHz, CDCl3) δ 166.3, 147.2, 145.3, 134.39, 134.37, 132.5, 132.3, 130.0, 127.6, 117.8, 116.5, 80.0, 75.4, 74.9, 72.0, 66.2, 43.2, 41.5, 40.7, 40.6, 35.6, 35.4, 35.0, 33.0, 31.2, 30.4, 20.4, 18.1, 17.3, 16.2, 12.4, 10.2; LRMS (ESI) calcd for C32H52O6571.3 (M+K)+, found 571.3; [α]20 D +32.6 (c 0.10, MeOH).
  • (Z)-(4R,5S,6S,7S)-tert-butyl-{6-(3,4-dimethoxybenzyloxy)-4-[4-(4,4,5,5,6,6,7,7,8,8,9,9,10,10,11,11,11-heptadecafluoroundecyloxy)-benzyloxy]-5,7-dimethylundec-8-enyloxy}dimethylsilane (85). A mixture solution of 84 (0.40 g, 0.39 mmol) in MeOH (8.0 ml) and CH2Cl2 (5.3 ml) was cooled to −78° C. and treated with a stream of ozone for 10 min. The reaction mixture was treated with dimethysulfide (2.0 ml) and pyridine (32 μl) and stirred for 3.0 h at ambient temperature. The reaction mixture was concentrated and diluted with Et2O (80 ml). The organic layer was washed with saturated aqueous CuSO4 (2×20 ml) and brine (20 ml), dried over MgSO4, filtered and concentrated. At ambient temperature, a suspension of propyltriphenylphosphonium bromide (0.383 g 98% purity, 0.97 mmol) in THF (15.0 ml) was added NaN(TMS)2 (1.0 M solution in THF, 0.98 ml) at ambient temperature. After stirring 1 h, this solution was cooled to −78° C. Then the crude residue in THF (2.0 ml) was introduced, and the resultant mixture was stirred for 3 h at −78° C. and was allowed to warm to ambient temperature for 12 h. The reaction mixture was quenched with saturated aqueous NaHCO3 (20 ml) and extracted with Et2O (2×40 ml). The combined extracts were washed with brine (20 ml), dried over MgSO4, filtered and concentrated. Flash chromatography (10% AcOEt/hexane) afforded 85 (0.08 g, 26% yield): 1H-NMR (500 MHz, CDCl3) δ 0.06 (s, 6H), 0.91 (s, 9H), 0.93 (t, J=7.6 Hz, 3H), 1.00 (d, J=6.8 Hz, 3H), 1.06 (d, J=6.8 Hz, 3H), 1.47 (m, 2H), 1.59 (m, 1H), 1.66 (m, 1H), 1.83 (m, 1H), 1.94 (m, 2H), 2.11 (m, 2H), 2.33 (m, 2H), 2.66 (m, 1H), 3.35 (m, 1H), 3.60 (t, J=6.3 Hz, 2H), 3.88 (s, 6H), 4.04 (t, J=5.8 Hz, 2H), 4.38 (d, J=11.3 Hz, 1H), 4.48 (d, J=10.9 Hz, 1H), 4.53 (d, J=10.9 Hz, 1H), 4.59 (d, J=11.3 Hz, 1H), 5.35 (dt, J=7.0, 10.3 Hz, 1H), 5.42 (dd, J=10.3, 10.1 Hz, 1H), 6.82-7.02 (m, 4H), 7.21-7.28 (m, 2H); 13C NMR (125 MHz, CDCl3) 6-5.2, 10.3, 14.5, 18.4, 18.9, 20.6, 20.9, 25.9, 26.9, 28.0 (t, J=22.5 Hz), 29.0, 34.9, 39.3, 55.8, 55.9, 63.2, 66.4, 70.8, 74.7, 79.4, 84.2, 110.5, 110.9, 114.3, 119.9, 106-122 (m), 129.6, 131.1, 131.3, 131.5, 131.6, 132.0, 148.4, 148.9, 158.2; IR (thin film/NaCl) cm−1 2933, 2858, 1729, 1612, 1515, 1465, 1242, 1153, 1032, 834; MS (EI) m/z 1060 (M+), 1035, 1003, 909, 567; [∝]D 20 +2.37° (c 0.590, CHCl3).
  • (4R,5S,6S,7S)-tert-butyl-{6-(3,4-dimethoxybenzyloxy)-4-[4-(4,4,5,5,6,6,7,7,8,8,9,9,10,10,11,11,12,12,13,13,13-heneicosafluorotridecyloxy)benzyloxy]-5,7-dimethyl-non-8-enyloxy}dimethylsilane (84). A solution of the alcohol (0.26 g, 0.57 mmol) and 1-bromomethyl-4-(4,4,5,5,6,6,7,7,8,8,9,9,10,10,11,11,12,12,13,13,13-heneicosafluorotridecyloxy)benzene (0.50 g, 0.66 mmol) in THF (6.0 ml) was cooled to −40° C. and tBuOK (1.0 M solution in THF, 0.70 ml) was added. The reaction mixture was stirred for 3.0 h. Then tBuOK (1.0 M solution in THF, 0.40 ml) was added again and the reaction solution was allowed to warm to ambient temperature for 9 h. The reaction mixture was quenched with saturated aqueous NaHCO3 (20 ml) and extracted with Et2O (2×40 ml). The combined extracts were washed with brine, (20 ml), dried over MgSO4, filtered and concentrated. Flash chromatography (10% AcOEt/hexane) afforded 84 (0.326 g, 50% yield): 1H-NMR (300 MHz, CDCl3) δ 0.55 (s, 6H), 0.91 (s, 9H), 1.06 (apparent d, J=6.8 Hz, 3H), 1.49 (m, 2H), 1.64 (m, 2H), 1.93 (m, 1H), 2.07 (m, 2H), 2.21-2.48 (m, 3H), 3.38 (m, 2H), 3.61 (t, J=6.1 Hz, 2H), 3.85 (s, 3H), 3.87 (s, 3H), 4.00 (t, J=5.7 Hz, 2H), 4.39 (d, J=11.2 Hz, 1H), 4.47 (d, J=10.8 Hz, 1H), 4.49 (d, J=11.2 Hz, 1H), 4.56 (d, J=10.8 Hz, 1H), 5.00 (m, 2H), 5.92 (m, 1H), 6.79-6.91 (m, 4H), 7.27 (apparent d, J=8.4 Hz, 2H); 13C NMR (75 MHz, CDCl3) δ 10.5, 14.0, 17.3, 18.3, 20.6, 22.7, 25.9, 26.9, 28.0 (t, J=22.2 Hz), 29.0, 38.5, 42.1, 55.7, 55.8, 63.1, 66.3, 70.9, 74.3, 79.9, 83.7, 110.9, 111.1, 114.3, 114.6, 119.9, 105-124 (m), 129.5, 131.6, 131.9, 141.2, 148.5, 148.9, 158.2; IR (thin film/NaCl) cm−1 2928, 2858, 1611, 1515, 1242, 1154; MS (EI) m/z 1132 (M+), 1075, 981, 817, 667, 465; [∝]D 20 +3.15° (c 1.11, CHCl3).
  • (Z)-(4R,5S,6S,7S)-tert-Butyl-(6-(3,4-dimethoxy-benzyloxy)-4-14-(4,4,5,5,6,6,7,7,8,8,9,9,10,10,11,11,12,12,13,13,13-heneicosafluorotridecyloxy)benzyloxy]-9-iodo-5,7-dimethylnon-8-enyloxy}dimethylsilane (86). A mixture of 84 (0.326 g, 0.28 mmol) in MeOH (8.0 ml) and CH2Cl2 (6.0 ml) was cooled to −78° C. and treated with a stream of ozone for 5 min. The reaction mixture was treated with dimethylsulfide (1.5 ml) and pyridine (23 μl) and stirred for 3.0 h at ambient temperature. The reaction mixture was concentrated and diluted with Et2O (80 ml). The organic layer was washed with saturated aqueous CuSO4 (2×20 ml) and brine (20 ml), dried over MgSO4, filtered and concentrated. At ambient temperature, a suspension of (iodomethyl)triphenylphosphonium iodide (0.213 g, 0.40 mmol) in THF (3.0 ml) was added NaN(TMS)2 (1.0 M solution in THF, 0.40 ml). After stirring 0.5 h, this solution was cooled to −78° C. Then HMPA (0.13 ml) and the crude residue in THF (2.0 ml) were introduced, and the resultant mixture was stirred for 20 min at −78° C. and stirred at ambient temperature for 0.5 h. The reaction mixture was quenched with saturated aqueous NaHCO3 (20 ml) and extracted with Et2O (2×40 ml). The combined extracts were washed with brine (20 ml), dried over MgSO4, filtered and concentrated. Flash chromatography (10% AcOEt/hexane) afforded (Z)-(4R,5S,6S,7S)-tert-butyl-{6-(3,4-dimethoxy-benzyloxy)-4-[4-(4,4,5,5,6,6,7,7,8,8,9,9,10,10,11,11,12,12,13,13,13-heneicosafluorotridecyloxy)benzyloxy]-9-iodo-5,7-dimethylnon-8-enyloxy}dimethylsilane (0.119 g, 33% yield): 1H-NMR (300 MHz, CDCl3) δ 0.04 (s, 6H), 0.89 (s, 9H), 1.03 (d, J=6.9 Hz, 3H), 1.04 (d, J=6.9 Hz, 3H), 1.23-1.77 (m, 5H), 2.04 (m, 2H), 2.23 (m, 2H), 2.70 (m, 1H), 3.44 (m, 2H), 3.59 (t, J=6.3 Hz, 2H), 3.86 (s, 3H), 3.87 (s, 3H), 4.03 (t, J=5.8 Hz, 2H), 4.40 (d, J=11.3 Hz, 1H), 4.53 (s, 2H), 4.57 (d, J=11.3 Hz, 1H), 6.15 (d, J=7.3 Hz, 1H), 6.28 (dd, J=7.3, 9.0 Hz, 1H), 6.80-6.89 (m, 4H), 7.22-7.30 (m, 2H); 13C NMR (75 MHz, CDCl3) 6-5.2, 10.2, 14.2, 17.2, 18.4, 20.7, 22.8, 26.0, 27.3, 28.0 (t, J=22.1 Hz), 29.8, 31.7, 40.2, 42.9, 51.4, 55.9, 56.0, 63.3, 66.4, 71.1, 75.4, 78.9, 82.2, 84.1, 107-122 (m), 110.9, 111.2, 114.4, 120.1, 129.5, 131.6, 131.8, 143.2, 148.6, 148.9, 158.2; IR (thin film/NaCl) cm−1 2956, 2859, 1727, 1611, 1514, 1467, 1243, 1154, 856; MS (EI) m/z 1201 (M+−C4H9), 1107, 667; [∝]D 20 +4.04° (c 1.01, CHCl3).
  • (Z)-(4R,5S,6S,7S)-tert-butyl-{6-(3,4-dimethoxybenzyloxy)-4-[4-(4,4,5,5,6,6,7,7,8,8,9,9,10,10,11,11,12,12,13,13,13-heneicosafluorotridecyloxy)benzyloxy]-5,7-dimethyl-9-phenyl-non-8-enyloxy}dimethylsilane (87). To a solution of (Z)-(4R,5S,6S,7S)-tert-butyl-{6-(3,4-dimethoxy-benzyloxy)-4-[4-(4,4,5,5,6,6,7,7,8,8,9,9,10,10,11,11,12,12,13,13,13-heneicosafluorotridecyloxy)benzyloxy]-9-iodo-5,7-dimethylnon-8-enyloxy}dimethylsilane (0.120 g, 0.09 mmol) and Pd(PPh3)4 (0.011 g, 0.01 mmol) in THF (1.0 ml) was added PhZnI (0.5 M solution in THF, 0.97 ml) at ambient temperature. After stirring for 24 h, the reaction mixture was quenched with saturated aqueous NaHCO3 (20 ml) and extracted with Et2O (2×40 ml). The combined extracts were washed with brine (20 ml), dried over MgSO4, filtered and concentrated. Flash chromatography (10% AcOEt/hexane) afforded 87 (0.0726 g, 63% yield): 1H-NMR (300 MHz, CDCl3) δ 0.09 (s, 6H), 0.95 (s, 9H), 1.07 (d, J=6.8 Hz, 3H), 1.19 (d, J=6.7 Hz, 3H), 1.27-1.59 (m, 4H), 1.85 (m, 1H), 2.13 (m, 2H), 2.35 (m, 2H), 3.11 (m, 2H), 3.39 (brt, J=5.2 Hz, 1H), 3.57 (t, J=6.2 Hz, 2H), 3.84 (s, 3H), 3.91 (s, 3H), 4.02 (d, J=11.3 Hz, 1H), 4.08 (t, J=5.8 Hz, 2H), 4.32 (d, J=11.3 Hz, 1H), 4.52 (d, J=10.6 Hz, 1H), 4.67 (d, J=10.6 Hz, 1H), 5.83, (dd, J=11.5, 11.5 Hz, 1H), 6.53 (d, J=11.5 Hz, 1H), 6.86-7.37 (m, 9H); 13C NMR (75 MHz, CDCl3) δ −5.2, 10.2, 14.2, 18.4, 18.9, 20.7, 22.8, 26.1, 27.3, 27.8 (t, J=21.7 Hz), 28.9, 31.7, 35.6, 39.7, 55.8, 56.0, 63.3, 66.4, 71.2, 75.2, 79.9, 84.6, 107-122 (m), 110.9, 111.2, 114.3, 120.0, 126.7, 128.3, 128.7, 128.9, 129.3, 131.9, 135.3, 137.8, 148.5, 148.9, 158.1; IR (thin film/NaCl) cm−1, 2926, 2857, 1727, 1515, 1464, 1242, 1154, 1031; MS (EI) m/z 1208 (M+), 1151, 1066, 667; [∝]D 20 +3.73° (c 0.805, CHCl3).
  • [2R]-Butane-1,2,4-triol. To a dry 1 L two-necked flask equipped with a pressure-equalizing addition funnel, a magnetic stirring bar and a reflux condenser was added THF (200 mL), B(OMe)3 (100 mL), and (R)-(+)-malic acid (40.0 g, 0.30 mol). To this solution was added dropwise BH3—SMe2 (100 mL, 1.0 mol) over 2 h in a water bath as instantaneous H2 evolution occurred throughout the addition. After stirring for 20 h at rt, MeOH (200 mL) was added dropwise, and the resulting solution was filtered through a glass frit funnel charged with Celite to remove any solids. The clear, yellow filtrate was concentrated in vacuo to give a yellow oil. The residue was dissolved in MeOH (100 mL) and concentrated in vacuo. This was repeated 5 times giving 26.9 g of the triol (85%). The spectral data matched that of the known compound.
  • [2R]-2-(4-Methoxyphenyl)-[1,3]dioxan-4-yl]methanol (88). A solution of the triol (5.0 g, 47.1 mmol), p-anisaldehyde (9.62 g, 70.7 mmol), PPTS (0.12 g, 0.047 mmol) in benzene (100 mL) was refluxed for 10 h with the azeotropic removal of H2O. NaHCO3 (0.20 g, 2.4 mmol) was added to the solution and concentrated in vacuo. The crude mixture was purified by flash chromatography (20% to 50% EtOAc in hexanes) to give 88 (65%). 1H NMR (300 MHz, CDCl3)-7.41 (d, J=8.4 Hz, 2H), 6.90 (d, J=8.4 Hz, 2H), 5.49 (s, 1H), 4.28 (ddd, J=11.1, 5.1, 1.3 Hz, 1H), 4.00-3.95 (m, 2H), 3.79 (s, 3H), 3.67-3.64 (m, 2H), 2.04 (br s, 1H), 1.91 (dq, J=12.4, 5.1 Hz, 1H), 1.46-1.41 (m, 1H); 13C NMR (75 MHz, CDCl3)—160.1, 132.0, 127.4, 113.6, 101.2, 76.6, 66.5, 65.6, 55.3, 26.9.
  • [2R]-tert-Butyl-[2-(4-methoxyphenyl)-[1,3]dioxan-4-ylmethoxy]diphenylsilane. A solution of alcohol 88 (8.13 g, 36.2 mmol), imidazole (3.9 g, 57.3 mmol), and TBDPSCl (10.2 mL, 39.7 mmol) in DMF (75 mL) was stirred overnight at room temperature under argon. A mixture of H2O (250 mL) and EtOAc (150 mL) was added and the layers were separated. The aqueous layer was extracted with EtOAc (2×100 mL). The organic layers were combined and washed with H2O (2×100 mL), brine (100 mL), dried over MgSO4, filtered and concentrated in vacuo. The crude oil was purified by flash chromatography (20% EtOAc in hexanes) to yield 15.9 g (95%) of the silyl ether as a clear yellow oil. 1H NMR (300 MHz, CDCl3) δ 7.69-7.66 (m, 4H), 7.41-7.34 (m, 8H), 6.86 (d, J=8.8 Hz, 2H), 5.46 (s, 1H), 4.28 (dd, J=11.0, 4.2 Hz, 1H), 4.02-3.96 (m, 2H), 3.86-3.83 (m, 1H), 3.79 (s, 3H), 3.67 (dd, J=10.2, 5.6 Hz, 1H), 1.84 (dq, J=12.2, 4.9 Hz, 1H), 1.65-1.61 (m, 1H), 1.05 (s, 9H); 13C NMR (75 MHz, CDCl3) δ 160.0, 135.7, 133.4, 130.0, 129.6, 128.2, 128.0, 113.4, 101.0, 77.5, 66.9, 66.0, 55.3, 28.1, 26.8, 19.3.
  • 4-(tert-Butyldiphenylsilanyloxy)-3-(4-methoxybenzyloxy)-butan-1-ol. To a stirred solution of the acetal (5.0 g, 10.8 mmol) in toluene (20 mL) at −78° C. was added slowly via cannula DibalH in toluene (33.0 mL, 1M). The reaction mixture was maintained at −78° C. for 12 h and quenched by slow addition to a vigorously stirred saturated solution of Rochelle salt in H2O (70 mL). The emulsion was stirred until two layers formed (1 h). The aqueous layer was extracted with CH2Cl2 (4×15 mL) and the organic layers were combined, dried over MgSO4, filtered and concentrated in vacuo. The crude oil was purified by flash chromotaography (20% to 50% EtOAc in hexanes) providing 4.01 g (80%) of the corresponding alcohol as a clear, yellow oil. 1H NMR (300 MHz, CDCl3) δ 7.69-7.66 (m, 4H), 7.43-7.38 (m, 6H), 7.20 (d, J=8.5 Hz, 2H), 6.86 (d, J=8.5 Hz, 2H), 4.60 (d, J=11.2 Hz, 1H), 4.41 (d, J=11.2 Hz, 1H), 3.79 (s, 3H), 3.77-3.66 (m, 5H), 1.82-1.79 (m, 2H), 1.05 (s, 9H); 13C NMR (75 MHz, CDCl3) δ 159.3, 135.5, 133.2, 129.4, 129.0, 128.0, 127.6, 113.8, 78.3, 72.1, 66.4, 60.2, 55.2, 34.2, 26.8, 19.1.
  • 4-(tert-Butyidiphenylsilanyloxy)-3-(4-methoxybenzyloxy)-butyraldehyde (89). DMSO (0.9 mL, 12.8 mmol) was added, dropwise, to a stirred solution of oxalyl chloride (0.5 mL, 6.0 mmol) in CH2Cl2 (8.0 mL) at −78° C. under argon. The reaction mixture was stirred for 5 min then a solution of the alcohol (2.04 g, 4.41 mmol) in CH2Cl2 (27.0 mL) was added dropwise. After 1 h at −78° C., Et3N (3.15 mL, 22.4 mmol) was added slowly via syringe, the mixture was stirred for 5 min then warmed to room temperature. The reaction mixture was diluted with CH2Cl2 (30 mL), washed with ice cold 0.5 M HCl (50 mL) then H2O (40 mL) and the layers were separated. The aqueous layers were combined and extracted with CH2Cl2 (2×40 mL) and the combined organic layers were dried over MgSO4, filtered, and concentrated in vacuo. The intermediate aldehyde 89 was used in the following reaction without purification: 1H NMR (300 MHz, CDCl3) δ 9.76 (s, 1H), 7.67-7.63 (m, 4H), 7.44-7.34 (m, 6H), 7.16 (d, J=8.5 Hz, 2H), 6.83 (d, J=8.5 Hz, 2H), 4.52 (d, J=11.1 Hz, 1H), 4.41 (d, J=11.1 Hz, 1H), 4.03-3.99 (m, 1H), 3.81 (s, 3H), 3.79-3.74 (m, 2H), 3.68-3.63 (m, 1H), 2.67 (dd, J=6.1, 1.9 Hz, 1H), 1.04 (s, 9H).
  • [3R,4R,6R]-7-(tert-Butyldiphenylsilanyloxy)-6-(4-methoxybenzyloxy)-3-methylhept-1-en-4-ol (90). A solution of (R,R)-diisopropyl tartrate (Z)-crotylboronate (15.0 mmol) was added to 4 Å powdered molecular sieves (0.170 g) in toluene (8.4 mL) under argon and the mixture was stirred for 20 min at room temperature. The mixture was cooled to −78° C. and a solution of the aldehyde 89 (2.0 g, 4.4 mmol) in toluene (5.0 mL) was added dropwise via cannula. The resulting mixture was maintained at −78° C. for 3 h and then treated with NaBH4 (0.072 g, 1.75 mmol) in EtOH (2.0 mL) and warmed to 0° C. The reaction mixture was treated with 1N NaOH (30 mL) and stirred vigorously for 30 min, followed by separation of the organic layer. The aqueous layer was extracted with CH2Cl2 (5×55 mL) and the combined organic layers were dried over MgSO4, filtered, and concentrated in vacuo. The crude oil was purified by flash chromatography (5% to 20% Et2O in hexanes) providing 1.43 g (63% 2 seps) of alcohol, a clear oil: [α]20 D=+0.32 (c 1.8, CHCl3); 1H NMR (300 MHz, CDCl3) δ 7.67 (d, J=7.0 Hz, 2H), 7.45-7.35 (m, 6H), 7.20 (d, J=8.4 Hz, 2H), 6.84 (d, J=8.4 Hz, 2H), 5.73 (ddd, J=18.2, 11.0, 7.4 Hz, 1H0, 5.03 (dd, J=11.0, 1.7 Hz, 1H), 5.02 (dd, J=18.2, 1.7 Hz, 1H), 4.59 (d, J=11.3 Hz, 1H), 4.40 (d, J=11.3 Hz, 1H), 3.79 (s, 3H), 3.89-3.75 (m, 2H), 3.68-3.65 (m, 2H), 2.18 (sext, J=6.8 Hz, 1H), 1.84-1.55 (m, 2H), 1.05 (s, 9H), 1.01 (d, J=6.8 Hz, 3H); 13C NMR (75 MHz, CDCl3) 6159.0, 140.6, 135.6, 133.5 (2C), 130.3, 131.0, 127.6, 129.6, 114.4, 113.6, 72.2, 83.1, 64.1, 71.3, 55.2, 40.8, 37.2, 26.8, 19.2, 14.4; IR (thin film) 3056, 2989, 2859, 1513, 1426, 1248, 1112, 1077 cm1; LRMS (EI) 517 (M−H), 435, 333, 303, 255, 241, 223, 199, 135, 121 m/z.
  • [2R,4R,5R]-[2,4-bis-(4-Methoxybenzyloxy)-5-methylhept-6-enyloxy]tert-butyidiphenyl silane. A mixture of NaH (1.8 g, 7.23 mmol) in THF (5 mL) was cooled to 0° C. then DMF (5 mL), the alcohol (1.25 g, 2.41 mmol) in THF (5 mL), and PMBBr (1.14 g, 6.03 mmol) were added. The reaction mixture was warmed to room temperature and stirred for 48 h. The resulting mixture was poured into a pH 7 phosphate buffer and diluted with ether (85 mL). The organic layer was separated and washed with pH 7 buffer (3×55 mL), dried over K2CO3, filtered and concentrated in vacuo. The resulting crude yellow oil was purified by flash chromatography (5% to 10% EtOAc in hexanes) providing 0.985 g (64%) of the PMB ether a clear, yellow tinted oil: [α]20 D=+0.31 (c 1.8, CHCl3); 1H NMR (300 MHz, CDCl3) δ 7.69-7.65 (m, 4H), 7.39-7.37 (m, 6H), 7.22-7.19 (m, 4H), 6.86-6.84 (m, 4H), 5.73 (ddd, J=17.8, 9.8, 7.0 Hz, 1H), 5.04-4.99 (m, 2H), 4.64 (d, J=11.2 Hz, 1H), 4.49 (d, J=11.1 Hz, 1H),4.30 (d, J=11.2 Hz, 1H), 4.18 (d, J=11.1 Hz, 1H), 3.78 (s, 6H), 3.81-3.52 (m, 4H), 2.60-2.56 (m, 1H), 1.57-1.51 (m, 2H), 1.05 (s, 9H), 1.0 (d, J=6.9 Hz, 3H); 13C NMR (75 MHz, CDCl3) δ 159.0, 140.6, 135.6, 133.5 (2C), 130.3, 131.0, 127.6, 129.6, 114.4, 113.6, 72.2, 83.1, 64.1, 75.4, 55.2, 40.8, 37.2, 26.8, 19.2, 14.4, −5.3; IR (thin film) 3055, 2985, 1422, 1280, 247 cm−1; LRMS (EI) 581.34 (M−C4H7), 579.34, 522.34, 444, 326, 383, 323, 339, 301, 255, 137, 122 m/z.
  • [2R,3R,5R]-6-(tert-Butyldiphenylsilanyloxy)-3,5-bis-(4-methoxybenzyloxy)-2-methylhexanal. To a solution of MeOH (30 mL), CH2Cl2 (10 mL) and a few drops of pyridine was added the PMB ether (800 mg, 1.25 mmol) and the mixture was cooled to −78° C. Ozone was bubbled through the reaction mixture until a slight purple color was seen. Excess DMS (6.0 mL) was added to the solution and allowed to warm to RT. After 3 h, the mixture was concentrated in vacuo. The yellow residue was dissolved in h hexanes (60 mL) and washed with H2O (2×40 mL) and brine (20 mL). The organic layer was dried over MgSO4, filtered and concentrated in vacuo to give the intermediate aldehyde (800 mg, 1.25 mmol) that was used in the following step without further purification: 1H NMR (300 MHz, CDCl3) δ 9.70 (d, J=2.0, 1H), 7.69-7.65 (m, 4H), 7.39-7.37 (m, 6H), 7.22-7.19 (m, 4H), 6.86-6.84 (m, 4H), 4.64 (d, J=11.2 Hz, 1H), 4.49 (d, J=11.1 Hz, 1H), 4.30 (d, J=11.2 Hz, 1H), 4.18 (d, J=11.1 Hz, 1H), 3.78 (s, 6H), 3.81-3.52 (m, 4H), 2.60-2.56 (m, 1H), 1.57-1.51 (m, 2H), 1.05 (s, 9H), 1.0 (d, J=6.9 Hz, 3H).
  • [4R,5R,7R]-8-(tert-Butyldiphenylsilanyloxy)-5,7-bis-(4-methoxybenzyloxy)-4-methyloct-2-enoic acid ethyl ester (91). To a stirred suspension of NaH (36 g, 1.56 mmol) in toluene (10 mL) at 0° C. and under argon was added a solution of 2-(diethoxyphosphoryl)propionic acid ethyl ester (0.47 mL, 1.77 mmol) in toluene (0.40 mL) dropwise. The reaction mixture was warmed to rt for 30 min then recooled to 0° C. The intermediate aldehyde (0.5 g, 1.3 mmol) in THF (10 mL) was added dropwise and the reaction mixture was stirred at 0° C. for 2 h. The solution was quenched by addition of pH 7 buffer (5 mL) and diluted with Et2O (12 mL). The emulsion was warmed to rt and the layers were separated. The organic layer was washed with a saturated solution of NH4Cl (10 mL) and the aqueous layers were combined and extracted with Et2O (3×20 ml). The organic layers were combined, dried over MgSO4, filtered and concentrated in vacuo. The crude mixture was purified by flash chromatography (0% to 20% EtOAc in hexanes) to give 623 mg (70% 2 steps) of 91 a yellow oil: 1H NMR (300 MHz, CDCl3) δ 7.69-7.65 (m, 4H), 7.39-7.37 (m, 4H), 7.21-7.18 (m, 4H), 6.85-6.83 (m, 4H), 6.98 (dd, J=15.7, 7.6 Hz, 1H), 5.82 (dd, J=15.7, 1.0 Hz, 1H), 4.64 (d, J=11.2 Hz, 1H), 4.50 (d, J=11.1 Hz, 1H), 4.36 (d, J=11.2 Hz, 1H), 4.20 (d, J=11.1 Hz, 1H), 3.78 (s, 6H), 3.81-3.52 (m, 4H), 2.60-2.56 (m, 1H), 1.57-1.51 (m, 2H), 1.05 (s, 9H), 1.0 (d, J=6.8 Hz, 3H); 13C NMR (75 MHz, CDCl3) δ 166.3, 158.9, 150.5, 131.0, 130.0, 129.2, 129.1, 121.1, 113.5, 78.4, 75.9, 71.7, 71.2, 64.1, 59.9, 55.0, 38.9, 34.1, 25.7, 18.1, 14.4, 14.1, −5.5; IR (thin film) 3055, 2956, 2933, 2908, 2857, 1705, 1243, 1097 cm−1; HRMS (EI) cald for C43H54O7Si 710.3628, found 371.3627.
  • [4R,5R,7R]-8-(tert-Butyldiphenylsilanyloxy)-5,7-bis-(4-methoxybenzyloxy)-4-methyloct-2-en-1-ol. To a stirred solution of ester 91 (600 mg, 0.84 mmol) in CH2Cl2 (6 mL) at −40° C. under argon was added slowly over 10 min via syringe DibalH in toluene (9 mL, 1M). After 30 min at −40° C., the reaction mixture was quenched by slow addition of MeOH (0.6 mL) and warmed to rt. The reaction mixture was poured into a vigorously stirred solution of saturated Rochelle salt in H2O (8 mL) and EtOAc (12 mL) and stirred overnight; The aqueous layer was separated and extracted with EtOAc (3×5 mL), dried over MgSO4, filtered and concentrated in vacuo. The crude mixture was purified by flash chromatography (20% EtOAc in hexanes) to produce 450 mg (80%) of the alcohol, a clear liquid: 1H NMR (300 MHz, CDCl3) δ 7.70-7.66 (m, 4H), 7.39-7.35 (m, 6H), 7.21-7.18 (m, 4H), 6.85-6.82 (m, 4H), 5.68-5.65 (m, 2H), 4.64 (d, J=11.1 Hz, 1H), 4.51 (d, J=11.0 Hz, 1H), 4.46 (d, J=11.1 Hz, 1H), 4.24 (d, J=11.0 Hz, 1H), 4.10-4.06 (m, 2H), 3.78 (s, 6H), 3.81-3.52 (m, 4H), 2.60-2.56 (m, 1H), 1.57-1.51 (m, 2H), 1.05 (s, 9H), 1.03 (d, J=6.8 Hz, 3H); 13C NMR (75 MHz, CDCl3) δ 159.0, 135.6, 135.1, 133.5 (2C), 131.1, 130.3, 130.0, 127.6, 129.4, 129.6, 83.1, 75.2, 72.4, 72.2, 64.4, 63.7, 55.2, 34.7, 26.8, 19.2, 15.4; IR (thin film) 3295, 3045, 2958, 2941, 2910, 2857, 1241, 1097 cm−1; HRMS (EI) cald for C41H52O6Si 668.3599, found 668.3596.
  • [2R,3R,5R]-8-(tert-Butyldiphenylsilanyloxy)-5,7-bis-(4-methoxybenzyloxy)-4-methyl oct-2-enal. To a solution of the alcohol (400 mg, 0.59 mmol) in CH2Cl2 (2 mL) was added Dess-Martin periodane (330 mg, 0.78 mmol) and the reaction mixture was stirred for 30 min. The reaction mixture was diluted with Et2O (10 mL) and poured into a stirring solution of saturated Na2S2O3 (5 mL) and saturated NaHCO3 (5 mL). The layers were separated and the organic layer was washed with saturated NaHCO3 (3×5 mL), dried over MgSO4, filtered and concentrated in vacuo to give the intermediate aldehyde which was used in the next reaction without further purification: 1H NMR (300 MHz, CDCl3) δ 9.50 (d, 7.8 Hz, 1H), 7.69-7.64 (m, 4H), 7.39-7.37 (m, 7H), 7.20-7.14 (m, 4H), 6.84-6.79 (m, 4H), 6.10 (dd, J=7.8, 17.0 Hz, 1H), 4.64 (d, J=11.2 Hz, 1H), 4.49 (d, J=11.1 Hz, 1H), 4.30 (d, J=11.2 Hz, 1H), 4.18 (d, J=11.1 Hz, 1H), 3.78 (s, 6H), 3.81-3.52 (m, 4H), 2.60-2.56 (m, 1H), 1.57-1.51 (m, 2H), 1.05 (s, 9H), 1.00 (d, J=6.9 Hz, 3H).
  • [4R,5R,7R]-10-(tert-butyldiphenysilanyloxy)-7,9-bis(4-methoxybenzyloxy)-6-methyldeca-2,4-dienoicacid methyl ester. To a stirred solution of [bis-(2,2,2-trifluoroethoxy)phosphoryl]acetic acid methyl ester (210 mg, 0.65 mmol) in THF (12 mL) at −78° C. under argon was added dropwise KHMDS in toluene (1.4 mL, 0.5 M). The reaction mixture was warmed to −40° C. for 1 h then cooled to −78° C. and the intermediate aldehyde (400 mg, 0.59 mmol) in THF (0.5 mL) was added dropwise. After 3 h at −78° C., the solution was warmed to 0° C. and quenched by addition of a saturated solution of NH4Cl (5 mL) and diluted with Et2O (5 mL). The aqueous layer was separated and extracted with diethyl ether (5×3 mL). The combined organic phases were washed with brine (5 mL), dried over MgSO4, filtered and concentrated in vacuo. The crude mixture was purified by flash chromatography (10% to 30% EtOAc in hexanes), yielding 220 mg (65% 2 steps) of conjugated ester, a clear oil: 1H NMR (300 MHz, CDCl3) δ 7.69-7.65 (m, 4H), 7.39-7.37 (m, 7H), 7.20-7.16 (m, 4H), 6.85-6.79 (m, 4H), 6.64 (t, J=11.2 Hz, 1H), 6.22 (ddd, J=15.4, 6.8 Hz, 1H), 5.87 (d, J=11.2 Hz, 1H), 5.04-4.99 (m, 2H), 4.64 (d, J=11.2 Hz, 1H), 4.49 (d, J=11.1 Hz, 1H), 4.30 (d, J=11.2 Hz, 1H), 4.18 (d, J=11.1 Hz, 1H), 3.78 (s, 6H), 3.81-3.52 (m, 4H), 2.60-2.56 (m, 1H), 1.57-1.51 (m, 2H), 1.05 (s, 9H), 1.00 (d, J=6.9 Hz, 3H); 13C NMR (75 MHz, CDCl3) δ 166.9, 147.3, 145.4, 135.6, 133.5 (2C), 130.3, 129.6, 126.7, 115.5, 113.5, 78.4, 75.9, 71.7, 71.8, 64.1, 55.0, 51.1, 39.7, 34.1, 26.8, 19.2, 14.1; IR (CH2Cl2) 3048, 2987, 2931, 2875, 2822, 1715, 15251, 1423, 1250, 1110 cm−1; HRMS (EI) cald for 722.3642, found 722.3640 m/z.
  • [6R,7S,9R]-10-Hydroxy-7,9-bis-(4-methoxybenzyloxy)-6-methyldeca-2,4-dienoic acid Methyl Ester (92). To a solution of the TBDPS ether (100 mg, 0.14 mmol) in THF 1 ml) was slowly added HF-pyridine in pyridine (1.5 ml, prepared by slow addition of 0.45 ml pyridine to 0.1 ml HF-pyridine complex followed by dilution with 0.94 ml THF) at 0° C. The mixture was warmed to room temperature and stirred overnight at room temperature. The reaction mixture was slowly quenched with saturated NaHCO3 (5 mL) and the aqueous layer was separated and extracted with CH2Cl2 (5×2 mL). The combined organic layers were washed with saturated CuSO4 (2 mL), dried over MgSO4, filtered and concentrated in vacuo. The crude product was purified by flash chromatography (25% EtOAc in hexanes) affording 50 mg (75%) of alcohol 92: [c]20D=+8.56 (c 0.1, CHCl3); 1H NMR (300 MHz, CDCl3) δ 7.53 (dd, J=15.0, 11.4 Hz, 1H), 7.35-7.30 (m, 4H), 6.97-6.93 (m, 6H), 6.65 (dd, J=11.3, 1.3 Hz, 1H), 6.23 (dd, J=15.0, 6.9 Hz, 1H), 5.69 (d, J=11.3 Hz, 1H), 4.64 (d, J=10.9 Hz, 1H), 4.59 (d, J=11.2 Hz, 1H), 4.44 (d, J=11.2 Hz, 1H), 4.34 (d, J=10.9 Hz, 1H), 3.88 (s, 3H), 3.82 (s, 3H), 2.77-2.73 (m, 2H), 3.67-3.64 (m, 1H), 3.54 (dd, J=3.6, 11.5 Hz, 1H), 2.95-2.89 (m, 1H), 1.90-1.81 (m, 2H), 1.19 (d, J=6.7 Hz, 3H); 13C NMR (75 MHz, CDCl3) δ 166.9, 159.0, 147.3, 145.4, 130.1, 130.3, 129.7, 126.7, 115.5, 113.6, 75.9, 78.5, 71.1, 63.9, 55.0, 51.0, 38.4, 33.7, 14.1; IR (thin film) 3315, 3055, 2986, 2858, 1710, 1513, 1423, 1247, 1105 cm−1; LRMS (EI) 484, 469, 349, 425, 223, 121 m/z; HRMS (EI) calcd for C28H36O7 484.2582, found 484.2579.
  • (R,R)-diisopropyl tartrate (Z)-crotylboronate. An oven-dried 1 L three-neck round bottom flask equipped with a magnetic stir bar and a −100° C. thermometer was charged with 206 mL of anhydrous THF and KOtBu (28.2 g, 250 mmol). This mixture was flushed with Ar and cooled to −78° C., then cis-2-butene (23 mL, 250 mmol), condensed from a gas lecture bottle into a rubber-stoppered round bottom flask immersed in a −78° C. dry ice-acetone bath, was poured into the reaction mixture. n-BuLi (100 mL, 2.5 M in hexane) was then added dropwise via cannula over 1.5 h. After completion of the addition, the cooling bath was removed and the reaction mixture was allowed to warm to −20 to −25° C. for 30 min before being recooled to −78° C. Triisopropylborate (57.8 mL, 250 mmol) was added drop-wise via cannula to the (Z)-crotylpotassium solution over 2 h. After addition, the reaction mixture was maintained at −78° C. for 30 min and then rapidly poured into a 2 L separatory funnel containing 470 mL of 1 N HCl saturated with NaCl. The aqueous layer was adjusted to pH 1 by using 1 N HCl (100-150 mL), and then a solution of (R,R)-diisopropyl tartrate (52.8 g, 250 mmol) in 88 mL of Et2O was added. The phases were separated, and the aqueous layer was extracted with additional Et2O (4×120 mL). The combined extracts were dried over MgSO4 for 1 h then vacuum filtered through a fritted glass funnel under Ar blanket into an oven-dried round-bottom flask. The filtrate was concentrated in vacuo, and pumped to constant weight at under vacuum. Anhydrous toluene (170 mL) was added to the flask make a 1M solution.
  • [4S,3S,2R]-4-Benzyl-3-(3-hydroxy-2,4-dimethylpent-4-enoyl)oxazolidin-2-one (93). Oxazolidinone 4 (10.0 g, 43.1 mmol) was treated with MgCl2 (0.20 g, 2.2 mmol), NaSbF6 (1.7 g, 6.5 mmol), Et3N (6.03 mL, 86.2 mmol), methacrolein (2.67 mL, 25.9 mmol) and TMSCl (3.92 mL, 32.3 mmol) in EtOAc (50 mL) and allowed to stir under Ar at rt for 24 h. The yellow-green slurry was filtered through a plug of silica gel with Et2O (1 L). GC analysis of the solution gave the isomeric composition of the TMS ether in a 16:1 ratio with its diastereomers. The ether was concentrated in vacuo, and MeOH (86 mL) and TFA (1 mL) was added. The reaction mixture was stirred for 30 min and concentrated to give a yellow which was purified by flash chromatography (10% acetone in hexanes) to yield 5.02 g of alcohol 93 (78% 2 steps). Data matches known literature.32 [α]20 D=+0.06 (c 0.1, CHCl3); 1H NMR (300 MHz, CDCl3) δ 7.39-7.31 (m, 5H), 5.08 (s, 1H), 5.02 (s, 1H), 4.77-4.75 (m, 1H), 4.27-4.22 (m, 4H), 3.35 (dd, J=13.5, 3.2 Hz, 1H), 2.83 (dd, J=13.5, 9.5 Hz, 1H), 2.75 (br s, 1H), 1.86 s (3H), 1.19 (d, J=6.7 Hz, 3H); 13C NMR (75 MHz, CDCl3) δ 175.6, 152.9, 145.0, 135.7, 129.3, 128.9, 127.1, 114.5, 80.1, 65.6, 55.5, 41.9, 38.4, 16.1, 14.7; IR (thin film) 3300, 3057, 2931, 2857, 1781, 1702, 1422, 1384, 1271, 1209, 1079 cm−1; HRMS (EI) cald for C17H21NO4 303.1582, found 303.1581.
  • [4R,2S,3R]-4-Benzyl-3-[3-(tert-butyldimethylsilanyloxy)-2,4-dimethylpent-4-enoyl]oxazolidin-2-one. To a stirred solution of alcohol 93 (20.24 g, 66.72 mmol) in CH2Cl2 (170 mL) at 0° C. under argon was added 2,6-lutidine (9.3 mL, 79.85 mmol) and TBSOTf (16.1 mL, 73.4 mmol). After 3 h at 0° C. the reaction mixture was quenched with MeOH (34 mL) then concentrated to dryness. The residue was taken up in Et2O (225 mL) and washed with a saturated solution of NH4Cl (2×50 mL). The aqueous layers were combined and extracted with Et2O (2×20 mL) and the combined organic layers were dried over MgSO4 and concentrated in vacuo. Flash chromatography of the crude mixture (10% to 20% EtOAc in hexanes) gave 26.5 g (95%) of the silyl ether as a clear oil: [α]20 D=+0.07 (c 0.15, CHCl3); 1H NMR (300 MHz, CDCl3) δ 7.40-7.29 (m, 5H), 5.00 (s, 1H), 4.98 (s, 1H), 4.72 (dq, J=7.0, 3.2 Hz, 1H), 4.51 (d, J=9.6 Hz, 1H), 4.21-4.18 (m 3H), 3.49 (dd, J=13.3, 3.2 Hz, 1H), 2.64 (dd, J=13.3, 10.2 Hz, 1H), 1.80 (s, 3H), 1.03 (d, J=7.0 Hz, 3H), 0.9 (s, 9H), 0.11 (s, 3H), 0.09 (s, 3H); 13C NMR (75 MHz, CDCl3) δ 175.9, 153.1, 145.0, 135.7, 129.3, 128.9, 127.2, 114.6, 79.4, 65.8, 55.5, 41.9, 38.4, 25.8, 18.1, 16.1, 14.7, −4.7, −5.1; IR (thin film) 3047, 2938, 2854, 1779, 1702, 1429, 1380, 1271, 1210, 1082 cm−1; LRMS (EI) 417, 402, 360, 290, 234, 185, 117; HRMS (EI) calcd for C23H35NO4Si 417.2335, found 417.2345.
  • [4S,2S,3R]-4-Benzyl-3-[3-(tert-butyldimethylsilanyloxy)-5-hydroxy-2,4-dimethylpentanoyl]oxazolidin-2-one (94). A stirred solution of 9-BBN in THF (29 mL, 0.5 M) was treated with the alkene (5.0 g, 11.97 mmol) in THF (29 mL). The reaction mixture was stirred at rt for 24 h, then treated sequentially with 1:1 EtOH-THF (29 mL), pH 7 buffer (29 mL) and 30% aq. H2O2 (14.5 mL) and stirred for 12 h at rt. The mixture was extracted with diethyl ether (3×20 mL). The combined organic layers were washed with H2O (15 mL) and saturated aqueous NaCl (15 mL) then dried over MgSO4, filtered and concentrated in vacuo. Purification of the mixture by flash chromatography (20% EtOAc in hexanes) gave 94 as a clear oil (3.91 g, 75%): [α]20 D=+0.24 (c 0.05, CHCl3); 1H NMR (300 MHz, CDCl3) δ 7.33-7.23 (m, 5H), 4.69-4.60 (m, 1H), 4.21 (dd, J=6.8, 3.8 Hz, 1H), 4.17-4.04 (m, 3H), 3.77-3.60 (m 2H), 3.44 (dd, J=13.1, 3.2 Hz, 1H), 2.60 (dd, J=13.1, 10.7 Hz, 1H), 2.50 (br s, 1H), 2.02-1.85 (m, 1H), 1.23 (d, J=6.9 Hz, 3H), 1.00 (d, J=7.0 Hz, 3H), 0.9 (s, 9H), 0.14 (s, 3H), 0.09 (s, 3H); 13C NMR (75 MHz, CDCl3) δ 174.7, 153.1, 135.4, 129.3, 129.0, 127.3, 66.1, 65.4, 55.8, 43.9, 38.3, 37.0, 26.7, 26.0, 18.2, 16.4, 12.4, −4.2, −4.8; IR (thin film) 3538, 2927, 1780, 1700, 1273, 1201 cm−1; HRMS (EI) cald for C23H37NO5Si 435.2461, found 435.2460.
  • (4S)-[2R,3R,4R]-Benzyl-3-[3,5-bis(tert-butyldi methylsilanyloxy)-2,4-dimethylpentanoyl]oxazolidin-2-one. To a stirred solution of alcohol 94 (2.6 g, 5.97 mmol) in CH2Cl2 (15 mL) at 0° C. under argon was added 2,6-lutidine (0.83 mL, 7:14 mmol) and TBSOTf (1.44 mL, 6.57 mmol). After 3 h at 0° C. the reaction mixture was quenched with MeOH (3 mL) then concentrated to dryness. The residue was taken up in Et2O (20 mL) and washed with a saturated solution of NH4Cl (2×5 mL). The aqueous layers were combined and extracted with Et2O (2×5 mL) and the combined organic layers were dried over MgSO4 and concentrated in vacuo. Flash chromatography of the crude mixture (10% to 20% EtOAc in hexanes) gave 3.01 g (95%) of the silyl ether as a clear oil: [α]20 D=+0.24(c 0.05, HCl3); 1H NMR (300 MHz, CDCl3) δ 7.33-7.22 (m, 5H), 4.69-4.61 (m, 1H), 4.24 (dd, J=3.7, 7.0 Hz, 1H), 4.13-4.11 (m, 3H), 3.76 (dd, J=6.0, 10.3 Hz, 1H), 3.48-3.42 (m, 2H), 2.60 (dd, J=10.3, 13.1 Hz, 1H), 2.01-1.87 (m, 1H), 1.17 (d, J=7.0 Hz, 3H), 0.976 (d, J=7.0 Hz, 3H), 0.90 (s, 9H), 0.88 (s, 9H), 0.11 (s, 3H), 0.08 (s, 3H), 0.05 (s, 6H); 13C NMR (75 MHz, CDCl3) δ 175.5, 153.1, 135.7, 129.3, 129.0, 127.3, 74.4, 66.0, 64.9, 55.7, 43.2, 39.9, 38.5, 26.1, 26.0, 18.3, 14.4, 13.5, −4.0, −4.6, −5.4 (2C); IR (thin film) 3057, 2952, 2860, 1781, 1699, 1382, 1259, 1100 cm−1; LRMS (EI) 492 (M−C4H9), 377, 374, 199, 177, 115; HRMS (EI) calcd for C25H42NO5Si2 492.3306, found 492.3301.
  • [2R,3R,4R]-3,5-bis(tert-Butyldi methylsilanyloxy)-2,4-dimethylpentan-1-ol (95). To a stirred solution of silyl ether (5 g, 9.09 mmol) in THF (50 mL) at 0° C. were added MeOH (1.14 mL, 27.3 mmol) and LiBH4 in THF (14 mL, 2M) under argon. The solution was stirred at 0° C. for 30 min and then quenched by the addition of a saturated solution of Rochelle salt in H2O (60 mL) and stirred for 10 min at 0° C. The mixture was poured into CH2Cl2 (100 mL) and stirred vigorously until 2 layers appeared (2 h). The aqueous layer was separated extracted with of CH2Cl2 (40 mL). The combined organic layers were washed with brine (40 mL), dried (MgSO4), filtered and concentrated in vacuo. Flash chromatography (30% EtOAc in hexanes) gave 2.32 g (65%) of alcohol 95 as a colorless oil: [α]20 D=+10.0 (c 1.2, CHCl3); 1H NMR (300 MHz, CDCl3) δ 3.67 (t, J=5.2 Hz, 1H), 3.55-3.50 (m, 3H), 3.39 (dd, J=10.0, 6.5 Hz, 1H), 2.98 (br s, 1H), 1.87-1.77 (m, 2H), 0.92 (d, J=7.0 Hz, 3H), 0.86 (d, J=7.0 Hz, 3H), 0.83 (s, 9H), 0.81 (s, 9H), 0.03 (s, 3H), 0.01 (s, 3H), −0.03 (s, 6H); 13C NMR (75 MHz, CDCl3) δ 800.2, 66.0, 65.2, 39.7, 38.8, 25.9, 25.6, 18.3, 18.0, 14.9, 14.7, −5.4, −5.3, −4.1; IR (thin film) 3330, 2930, 2858, 1471, 1250, 1023 cm−1; HRMS (EI) calcd for C19H44O3Si2 376.2859, found 376.2858.
  • [2R,3R,4R]-3,5-bis-(tert-Butyldimethylsilanyloxy)-2,4-dimethylpentanal (96). Following the procedure for 4-(tert-butyldiphenylsilanyloxy)-3-(4-methoxybenzyloxy)- butyraldehyde 89, 96 can be prepared in a similar manner.
  • (2R)-3-(tert-Butyldimethylsilanyloxy)-2-methylpropionic acid methyl ester. A solution of alcohol (10.0 g, 84.6 mmol), imidazole (9.2 g, 133.9 mmol), and TBSCl (19.1 g, 126.9 mmol) in DMF (150 mL) was stirred overnight at room temperature under argon. A mixture of H2O (500 mL) and EtOAc (300 mL) was added and the layers were separated. The aqueous layer was extracted with EtOAc (2×200 mL). The organic layers were combined and washed with H2O (2×200 mL), brine (200 mL), dried over MgSO4, filtered and concentrated in vacuo. The crude oil was purified by flash chromatography (20% EtOAc in hexanes) to yield 17.7 g (90%) of the silyl ether as a clear yellow oil: The spectral data matched that of the known compound. 1H NMR (300 MHz, CDCl3) δ 3.62 (dd, J=6.7, 7.0 Hz, 1H), 3.50 (dd, J=6.7, 7.0 Hz, 1H), 3.50 (m, 3), 2.48 (sext, J=7.0 Hz, 1H), 0.97 (d, J=7.0 Hz, 3H), 0.71 (s, 9H), −0.1 (s, 6H); 13C NMR (75 MHz, CDCl3) δ 175.4, 65.2, 51.5, 42.5, 25.7, 18.2, 13.4, −5.5.
  • [2R]-3-(tert-Butyldimethylsilanyloxy)-2-methylpropan-1-ol. To a stirred solution of silyl ether (5.0 g, 21.5 mmol) in CH2Cl2 (125 mL) at −40° C. under argon was added slowly over 1.5 h via cannula DibalH in toluene (100 mL, 1M). After 30 min at −40° C., the reaction mixture was quenched by slow addition of MeOH (15 mL) and warmed to RT. The reaction mixture was poured into a vigorously stirred solution of saturated Rochelle salt (200 mL) and EtOAc (300 mL) and stirred overnight. The aqueous layer was separated and extracted with EtOAc (3×50 mL), dried (MgSO4) and concentrated in vacuo. The crude mixture was purified by flash chromatography (20% EtOAc in hexanes) to produce 3.46 g (79%) of the alcohol, a clear liquid. The spectral data matched that of the known compound: 1H NMR (300 MHz, CDCl3) δ 3.73 (dd, J=4.5, 9.8, Hz, 1H), 3.57 (m, 3H), 2.80 (br s, 1H), 1.99-1.86 (m, 1H), 0.89 (s, 9H), 0.82 (d, J=7.0 Hz, 3H), 0.06 (d, 6H); 13C NMR (75 MHz, CDCl3) δ 68.8, 68.4, 37.0, 25.8, 18.1, 13.0, −5.6.
  • [2R]-3-(tert-Butyidimethylsilanyloxy)-2-methyl propionaldehyde (ent-17). DMSO (3.0 mL, 42.6 mmol) was added, dropwise, to a stirred solution of oxalyl chloride (1.5 mL, 19.9 mmol) in CH2Cl2 (80 mL) at −78° C. under argon. The reaction mixture was stirred for 5 min then a solution of alcohol (3.0 g, 14.7 mmol) in CH2Cl2 (25 mL) was added dropwise. After 1 h at −78° C., Et3N (10.5 mL, 74.7 mmol) was added slowly via syringe, the mixture was stirred for 5 min then warmed to room temperature. The reaction mixture was diluted with CH2Cl2 (25 mL), washed with ice cold 0.5 M HCl (50 mL) then H2O (30 mL) and the layers were separated. The aqueous layers were combined and extracted with CH2Cl2 (2×30 mL) and the combined organic layers were dried over MgSO4, filtered, and concentrated in vacuo. The intermediate aldehyde ent-17 was used in the following reaction without purification: 1H NMR (300 MHz, CDCl3) δ 9.75 (6, 1.5Hζ, 1H), 3.86-3.82 (m, 2H), 2.53-2.50 (m, 1H), 1.10 (d, J=7.0 Hz, 3H), 0.88 (s, 9H), 0.05 (s, 6H).
  • [3S,4S,5S]-2,4-dimethyl-1-[(tert-butyidimethylsilyl)oxy]-hexene-5-en-3-ol (97). A solution of (R,R)-diisopropyl tartrate (E)-crotylboronate (22.1 mmol) was added to 4 Å powdered molecular sieves (0.025 g) in toluene (1 mL) under argon and the mixture was stirred for 20 min at room temperature. The mixture was cooled to −78° C. and a solution of the aldehyde ent-17 (3.0 g, 14.7 mmol) in toluene (8 mL) was added dropwise via syringe. The resulting mixture was maintained at −78° C. for 3 h and then treated with NaBH4 (0.106 g, 2.6 mmol) in EtOH (4 mL) and warmed to 0° C. The reaction mixture was treated with 1N NaOH (40 mL) and stirred vigorously for 30 min, followed by separation of the organic layer. The aqueous layer was extracted with CH2Cl2 (5×80 mL) and the combined organic layers were dried over MgSO4, filtered, and concentrated in vacuo. The crude oil was purified by flash chromatography (5% to 25% Et2O in hexanes) providing 2.87 g (65%) of 97, a clear, yellow tinted oil: 1H NMR (300 MHz, CDCl3) δ 5.91 (ddd, J=8.8, 12.0, 15.8 Hz, 1H), 5.05 (dd, J=1.8, 12.0 Hz, 1H), 5.04 (dd, J=1.8, 15.8 Hz, 1H), 3.81 (s, 1H), 3.73 (dd, J=4.2, 9.8 Hz, 1H), 3.60 (dd, J=8.2, 9.8 Hz 1H), 3.37 (dd, J=3.0, 4.8 Hz, 1H), 2.39-2.29 (m, 1H), 1.83-1.70 (m, 1H), 1.09 (d, J=6.9 Hz, 3H), 0.89 (s, 9H), 0.81 (d, J=6.9 Hz, 3H), 0.06 (s, 6H); 3C NMR (75 MHz, CDCl3) δ 139.9, 114.9, 80.2, 68.7, 41.2, 37.5, 25.8 [2C], 18.1, 17.7, 13.4, −5.6, −5.7.
  • [2S,3S,4S]-tert-Butyl-[3-(4-methoxybenzyloxy)-2,4-dimethylhex-5-enyloxy]dimethylsilane. A mixture of NaH (2.9 g, 11.6 mmol) in THF (5 mL) was cooled to 0° C. then DMF (5 mL), alcohol 97 (1.0 g, 3.87 mmol) in THF (5 mL), and PMBBr (1.8 g, 9.7 mmol) were added. The reaction mixture was warmed to room temperature and stirred for 48 h. The resulting mixture was poured into a pH 7 phosphate buffer and diluted with ether (90 mL). The organic layer was separated and washed with pH 7 buffer (3×60 mL), dried over K2CO3, filtered and concentrated in vacuo. The resulting crude yellow oil was purified by flash chromatography (5% to 10% EtOAc in hexanes) providing 1.39 g (75%) of the PMB ether as a clear, yellow tinted oil: [α]20 D=+0.06 (c 1.3, CHCl3); 1H NMR (300 MHz, CDCl3) δ 7.31 (d, J=8.6 Hz, 2H), 6.90 (d, J=8.6 Hz, 2H), 5.93 (ddd, J=15.6, 12.0, 8.3 Hz, 1H), 5.05 (d, J=12 Hz, 1H), 5.04 (d, J=15.61H), 3.89 (s, 3H), 3.29 (dd, J=4.6, 2.9 Hz, 1H), 2.51-2.49 (m, 1H), 1.91-1.86 (m, 1H), 1.01 (d, J=6.9 Hz, 3H), 0.08 (s, 6H); 13C NMR (75 MHz, CDCl3) δ 158.9, 142.1, 131.2, 129.2, 114.0, 113.6, 82.8, 74.1, 65.7, 55.7, 38.4, 37.3, 25.9, 18.2, 15.6, 11.2, −5.2; IR (thin film) 3057, 2957, 2857, 1612, 1513, 1246, 1085 cm−1; LRMS (EI) 323 (M−C4H7), 321, 271, 255, 186, 122 m/z.
  • [2S,3S,4S]-5-(tert-Butyldimethylsilanyloxy)-3-(4-methoxybenzyloxy)-2,4-dimethyl pentanal (98). Following the procedure for [2R,3R,5R]-6-(tert-Butyldiphenylsilanyloxy)-3,5-bis-(4-methoxybenzyloxy)-2-methylhexanal, 98 can be prepared in a similar manner.
  • (R,R)-Diisopropyl tartrate (E)-crotylboronate. An oven-dried 1 L three-neck round bottom flask equipped with a magnetic stir bar and a −100° C. thermometer was charged with 206 mL of anhydrous THF and KOtBu (28.2 g, 250 mmol). This mixture was flushed with Ar and cooled to −78° C., then trans-2-butene (23 mL, 250 mmol), condensed from a gas lecture bottle into a rubber-stoppered round bottom flask immersed in a −78° C. dry ice-acetone bath, was poured into the reaction mixture. n-BuLi (100 mL, 2.5 M in hexane) was then added dropwise via cannula over 1.5 h. After completion of the addition, the cooling bath was removed and the reaction mixture was allowed to warm to an internal temperature of −50° C. for 15 min then immediately recooled to −78° C. Triisopropylborate (57.8 mL, 250 mmol) was added drop-wise via cannula to the (E)-crotylpotassium solution over 2 h. After addition, the reaction mixture was maintained at −78° C. for 30 min and then rapidly poured into a 2L separatory funnel containing 470 mL of 1 N HCl saturated with NaCl. The aqueous layer was adjusted to pH 1 by using 1 N HCl (100-150 mL), and then a solution of (R,R)-diisopropyl tartrate (52.8 g, 250 mmol) in 88 mL of Et2O was added. The phases were separated, and the aqueous layer was extracted with additional Et2O (4×120 mL). The combined extracts were dried over MgSO4 for 1 h then vacuum filtered through a fritted glass funnel under Ar blanket into an oven-dried round-bottom flask. The filtrate was concentrated in vacuo, and pumped to constant weight at under vacuum. Anhydrous toluene (170 mL) was added to the flask make a 1M solution.
  • Biology
  • General. Tubulin without microtubule-associated proteins was prepared from fresh bovine brains[32] The normoisotopic and tritiated forms of paclitaxel and normoisotopic docetaxel were provided by the Drug Synthesis and Chemistry Branch, National Cancer Institute. (+)-Discodermolide was from Novartis Pharmaceutical Corporation. Ca2+- and Mg2+-free RPMI-1640 culture medium were from GIBCO/BRL-Life Technologies. Fetal bovine serum (FBS) was from Hyclone. Cell lines were obtained from American Type Culture Collection (Manassas, Va.).
  • Tubulin Polymerization[32] Tubulin assembly was followed in a Beckman-Coulter 7400 spectrophotometer, equipped with an electronic Peltier temperature controller, reading absorbance (turbidity) at 350 nm. Reaction mixtures (0.25 mL final volume) contained tubulin (final concentration 10 μM; 1 mg/mL), monosodium glutamate (0.8 M from a stock solution adjusted to pH 6.6 with HCl), DMSO (final volume 4% v/v), and differing concentrations of test agent where indicated. Reaction mixtures without test agent were cooled to 0° C. and added to cuvettes held at 0.25-0.5° C. in the spectrophotometer. Test agent in DMSO was then rapidly mixed in the reaction mixture. Each run contained one positive control (paclitaxel, 10 μM final concentration) and one negative control (DMSO only). Baselines were established at 0.25-2.5° C. and temperature was rapidly raised to 30° C. (in approximately 1 min) and held there for 20 min. The temperature was then rapidly lowered back to 0.25-2.5° C.
  • Cell Growth Inhibition[34] Cells were plated (500-2000 cells/well depending on the cell line) in 96-well microplates, allowed to attach and grow for 48 h, then treated with vehicle (4% DMSO, a concentration that allowed doubling times of 24 h or less) or test agent (50, 10, 2, 0.4 and 0.08 μM for the new agents; 0.001, 0.005, 0.010, 0.020 and 0.100 μM for paclitaxel and discodermolide) for the given times. One plate consisted of cells from each line used for a time zero cell number determination. The other plates in a given determination contained eight wells of control cells, eight wells of medium and each agent concentration tested in quadruplicate. Cell numbers were obtained spectrophotometrically (absorbance at 490 nm minus that at 630 nm) in a Dynamax plate reader after treatment with 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) using phenazine methanesulfonate as the electron acceptor. After initial screening with the above 5-fold dilutions, fifty percent growth inhibitory concentration (GI50) values were determined for each agent by repeating the screen using 2-fold dilutions (five concentrations) centered on the initial estimated GI50 concentration, again in quadruplicate.
  • Paclitaxel binding site inhibition assay. [34] A stock solution of [3H]paclitaxel (26.8 μM, 16.2 Ci/mmol), obtained from the NCI, was prepared in 37% (v/v) DMSO. The test agents were prepared in 25% (v/v) DMSO-0.75 M monosodium glutamate (prepared from a 2M stock solution adjusted to pH 6.6 with HCl). The radiolabeled paclitaxel and test agents, as indicated in terms of final concentrations, were mixed in equal volumes and warmed to 37° C. A reaction mixture (50 μL) containing 0.75 M monosodium glutamate, 4.0 μM tubulin, and 40 μM ddGTP (a non-hydrolyzable analog of GTP) was prepared and incubated at 37° C. for 30 min to preform microtubules. An equivalent volume of drug mixture with [3H]paclitaxel was added to preformed polymer and incubated for 30 min at 37° C. Bound [3H]paclitaxel was separated from free drug by centrifugation of the reaction mixtures at 14000 rpm for 20 min at room temperature. Non-specific binding was determined by addition of a 12-fold excess of docetaxel. Radioactive counts from the supernatant (50 μL) were determined by scintillation spectrometry. Bound [3H]paclitaxel was calculated from the following: total paclitaxel added to each reaction mixture minus the paclitaxel present in the supernatant (free drug). The % bound values were normalized to the control values with no inhibitor added.
  • The foregoing description and accompanying drawings set forth the preferred embodiments of the invention at the present time. Various modifications, additions and alternative designs will, of course, become apparent to those skilled in the art in light of the foregoing teachings without departing from the scope of the invention. The scope of the invention is indicated by the following claims rather than by the foregoing description. All changes and variations that fall within the meaning and range of equivalency of the claims are to be embraced within their scope.

Claims (26)

1.-3. (canceled)
4. A compound of the following structure
Figure US20060270862A1-20061130-C00029
wherein R1 is H, an alkyl group, an aryl group, an alkenyl group, an alkynyl group, or a halogen atom;
R2 and R2d are independently H, an alkyl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe;
Ra, Rb and Rc are independently an alkyl group or an aryl group;
Rd is an alkyl group, an aryl group, an alkoxylalkyl group, —RiSiRaRbRc or a benzyl group, wherein Ri is an alkylene group;
Re is an alkyl group, an allyl group, a benzyl group, an aryl group, an alkoxy group, or —NRgRh, wherein Rg and Rh are independently H, an alkyl group or an aryl group;
R3 is (CH2)n where n is and integer in the range of 0 to 5, —H2CH(CH3)—, —CH═CH—, —CH═C(CH3)—, or —C═C—;
R4 is (CH2)p where p is an integer in the range of 4 to 12, —(CHRk1)y1(CHRk2)y2(CHRk3)y3(CHRk4)y4(CHRk5)y5C(Rs1)═C(Rs2)C(Rs3)═C(Rs4)—, (CHRk1)y1(CHRk2)y2(CHRk3)y3(CHRy4)y4(CHRk5)y5CH(Rs1)CH(Rs2)C(Rs3)C(Rs4), —(CHRk1)y1(CHRk2)y2(CHRk3)y3(CHRy4)y4(CHRk5)y5C(Rs1)═C(Rs2)CH(Rs3)CH(Rs4)—, —(CHRk1)y1(CHRk2)y2(CHRk3)y3(CHRk4)y4(CHRk5)y5CH(Rs1)CH(Rs2)CH(Rs3)CH(Rs4)—, wherein y1 and y2 are 1 and y3, y4 and y5 are independently 0 or 1, Rk1, Rk2, Rk3, Rk4 and Rk5 are independently H, —CH3, or OR2a, and Rs1, Rs2, Rs3, and Rs4 are independently H or CH3, wherein R2a is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe; and
R5 is H or OR2b, wherein R2b is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe; and
R10 is H or alkyl.
5. The compound of claim 4 with the following stereostructure, or its enantiomer
Figure US20060270862A1-20061130-C00030
wherein R1 is alkenyl; R2 is H; R2d is H, OC(O)CH3 or OC(O)NRgRh wherein Rg and Rh are independently H, an alkyl group or an aryl group; R3 is CH2CH(CH3) or CH═C(CH3);
and R4 is —(CHRk1)y1(CHRk2)y2(CHRk3)y3(CHRk4)y4(CHRk5)y5C(Rs1)═C(Rs2)C(Rs3)═C(Rs4)— wherein y1-y4 are 1, y5 is 0, Rk1 and Rk3 are OH, Rk2 is H, Rk4 is CH3, Rs1, Rs2, Rs3 and Rs4 are H, R5 is OH; and R10 is H or alkyl.
6. The compound of claim 5 wherein R1 is —CH═CH2, and R2d is H, OC(O)CH3 or OC(O)NH2.
7. A compound of the following structure:
Figure US20060270862A1-20061130-C00031
wherein R1 is H, an alkyl group, an aryl group, an alkenyl group, an alkynyl group, or a halogen atom;
R2 and R2d are independently H, an alkyl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe;
Ra, Rb and Rc are independently an alkyl group or an aryl group;
Rd is an alkyl group, an aryl group, an alkoxylalkyl group, —RiSiRaRbRc or a benzyl group, wherein Ri is an alkylene group;
Re is an alkyl group, an allyl group, a benzyl group, an aryl group, an alkoxy group, or —NRgRh, wherein Rg and Rh are independently H, an alkyl group or an aryl group;
R3 is (CH2)n where n is and integer in the range of 0 to 5, —H2CH(CH3)—, —CH═CH—, —CH═C(CH3)—, or —C≡C—;
R5 is H or OR2b, wherein R2b is H, an alkyl group, an aryl group, a benzyl group, a trityl group, SiRaRbRc, CH2ORd, or CORe;
R11a and R11b are independently H, an alkyl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, CORe, or R11a and R11b together form a portion of six-membered acetal ring incorporating CRtRu;
Rt and Ru are independently H, an alkyl group, an aryl group or an alkoxyaryl group; and
R12 is a halogen atom, CH2OR2c, CHO, CO2R10, CH═CHCH2OR2c, CH═CHCHO, wherein R2c is H, an alkyl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe, and R10 is H or alkyl.
8. A compound of claim 7 with the following stereostructure, or its enantiomer
Figure US20060270862A1-20061130-C00032
wherein R1 is alkenyl; R2d is H, OC(O)CH3 or OC(O)NRgRh wherein Rg and Rh are independently H, an alkyl group or an aryl group; R3 is CH2CH(CH3) or CH═C(CH3);
R11a and R11b are H or together form a portion of a six-membered acetal ring containing C(H)(p-C6H4OCH3) or C(CH3)2; R12 is a halogen atom, CH2OR2c, CHO, CO2R10, CH═CHCH2OR2c, CH═CHCHO, wherein R2c is H, an alkyl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe, and R10 is H or alkyl.
9. The compound of claim 8 wherein R1 is —CH═CH2, R2d is H, —OC(O)CH3 or —OC(O)NH2, and R12 is —CH2OH, —CHO or —CO2R10.
10. A compound having the following structure:
Figure US20060270862A1-20061130-C00033
wherein R2 is H, an alkyl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe;
Ra, Rb and Rc are independently an alkyl group or an aryl group;
Rd is an alkyl group, an aryl group, an alkoxylalkyl group, —RiSiRaRbRc or a benzyl group, wherein Ri is an alkylene group;
Re is an alkyl group, an allyl group, a benzyl group, an aryl group, an alkoxy group, or —NRgRh, wherein Rg and Rh are independently H, an alkyl group or an aryl group;
R3 is (CH2)n where n is and integer in the range of 0 to 5, —CH2CH(CH3)—, —CH═CH—, —CH═C(CH3)—, or —C≡C—;
R5 is H or OR2b, wherein R2b is H, an alkyl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe;
R11a and R11b are independently H, an alkyl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, CORe, or R11a and R11b together form a portion of six-membered acetal ring containing CRtRu;
Rt and Ru are independently H, an alkyl group, an aryl group or an alkoxyarly group;
R12 is a halogen atom, CH2OR2c, CHO, CO2R10, CH═CHCH2OR2c or CH═CHCHO, CH═CHCO2R10, wherein R2c is H, an alkyl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe, and R10 is H or alkyl; and
R14a and R14b are independently H, an alkyl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, CORe, or R14a and R14b together form a six-membered ring containing CRvRw, wherein Rv and Rw are independently H, an alkyl group, an aryl group or an alkoxyaryl group.
11. The compound of claim 10 with the following stereostructure, or its enantiomer
Figure US20060270862A1-20061130-C00034
R2 is H; R3 is CH2CH(CH3) or CH═C(CH3); R11a and R11b are H or together form a portion of a six-membered acetal ring containing C(H)(p-C6H4OCH3) or C(CH3)2; R12 is a halogen atom, CH2OR2c, CHO, CO2R10, CH═CHCH2OR2c, CH═CHCHO or CH═CHCO2R10, wherein R2c is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe, and R10 is H or alkyl; and R14a and R14b are H or together form a portion of a six-membered acetal ring containing C(H)(p-C6H4OCH3) or C(CH3)2.
12. A compound having the following formula
Figure US20060270862A1-20061130-C00035
wherein R1 is H, an alkyl group, an aryl group, an alkenyl group, an alkynyl group, or a halogen atom;
R2 is H, an alkyl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe;
Ra, Rb and Rc are independently an alkyl group or an aryl group;
Rd is an alkyl group, an aryl group, an alkoxylalkyl group, —RiSiRaRbRc or a benzyl group, wherein Ri is an alkylene group;
Re is an alkyl group, an allyl group, a benzyl group, an aryl group, an alkoxy group, or —NRgRh, wherein Rg and Rh are independently H, an alkyl group or an aryl group;
R3 is (CH2)n where n is and integer in the range of 0 to 5, —H2CH(CH3)—, CH═CH—, —CH═C(CH3)—, or —C≡C—;
R11a and R11b are independently H, an alkyl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, CORe, or R11a and R11b together form a portion of six-membered acetal ring containing CRtRu;
Rt and Ru are independently H, an alkyl group, an aryl group or an alkoxyaryl group;
R12 is a halogen atom, CH2OR2c, CHO, CO2R10, CH═CHCH2OR2c, CH═CHCHO or CH═CHCO2R10, wherein R2c is H, an alkyl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe, and R10 is H or alkyl; and
R14a and R14b are independently H, an alkyl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, CORe, or R14a and R14b together form a six-membered ring containing CRvRw, wherein Rv and Rw are independently H, an alkyl group, an aryl group or an alkoxyaryl group.
13. The compound of claim 12 with the following stereostructure, or its enantiomer
Figure US20060270862A1-20061130-C00036
wherein R3 is CH2CH(CH3) or CH═C(CH3); R11a and R11b are H or together form a portion of a six-membered acetal ring containing C(H)(p-C6H4OCH3) or C(CH3)2; R12 is a halogen atom, CH2OR2c, CHO, CO2R10, CH═CHCH2OR2c, CH═CHCHO or CH═CHCO2R10, wherein R2c is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe, and R10 is H or alkyl; and R14a and R14b are H or together form a portion of a six-membered acetal ring containing C(H)(p-C6H4OCH3) or C(CH3)2.
14. A compound having the following formula
Figure US20060270862A1-20061130-C00037
wherein R2 is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe;
Ra, Rb and Rc are independently an alkyl group or an aryl group;
Rd is an alkyl group, an aryl group, an alkoxylalkyl group, —RiSiRaRbRc or a benzyl group, wherein Ri is an alkylene group;
Re is an alkyl group, an allyl group, a benzyl group, an aryl group, an alkoxy group, or —NRgRh, wherein Rg and Rh are independently H, an alkyl group or an aryl group;
R11a and R11b are independently H, an alkyl group, and aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, CORe, or R11a and R11b together form a portion of six-membered acetal ring containing CRtRu;
Rt and Ru are independently H, an alkyl group or an aryl group;
R12 is a halogen atom, CH2OR2c, CHO, CO2R10, CH═CHCH2OR2c, CH═CHCHO or CH═CHCO2R10, wherein R2c is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe, and R10 is H or alkyl;
R16 is H or alkyl; and
R17 is CH2OR2f, CHO, CO2R10, wherein R2f is H, an alkyl group, an aryl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe.
15. The compound of claim 14 with the following stereostructure, or its enantiomer
Figure US20060270862A1-20061130-C00038
wherein R2 is H, an alkyl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe, R11a and R11b are H or together form a portion of a six-membered acetal ring containing C(H)(p-C6H4OCH3) or C(CH3)2.
16. A compound having the following formula
Figure US20060270862A1-20061130-C00039
wherein R1 is H, an alkyl group, an aryl group, an alkenyl group, an alkynyl group, or a halogen atom;
R2, R2d and R2e are independently H, an alkyl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe;
Ra, Rb and Rc are independently an alkyl group or an aryl group;
Rd is an alkyl group, an aryl group, an alkoxylalkyl group, —RiSiRaRbRc or a benzyl group, wherein Ri is an alkylene group;
Re is an alkyl group, an allyl group, a benzyl group, an aryl group, an alkoxy group, or —NRgRh, wherein Rg and Rh are independently H, an alkyl group or an aryl group;
R3 is (CH2)n where n is and integer in the range of 0 to 5, —CH2CH(CH3), CH═CH—, —CH═C(CH3)—, or —C≡C—;
R5 is H or OR2b, wherein R2b is H, an alkyl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe;
q is an integer in the range of 2 to 5;
R18 is H, and R19 is hydroxy, alkoxy or —SRz, wherein Rz is an alkyl group or an aryl group, or R18 and R19 taken together are ═O.
17. The compound of claim 16 with the following stereostructure, or its enantiomer
Figure US20060270862A1-20061130-C00040
18. The compound of claim 17 where wherein R1 is a CH═CH2 and R3 is (Z)-CH═CH—, or —CH2CH2—.
19. A compound having the following structure
Figure US20060270862A1-20061130-C00041
R11a and R11b are independently H, an, alkyl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, CORe, or R11a and R11b together form a portion of six-membered acetal ring containing CRtRu;
Rt and Ru are independently H, an alkyl group, an aryl group or an alkoxyarly group;
Ra, Rb and Rc are independently an alkyl group or an aryl group;
Rd is an alkyl group, an aryl group, an alkoxylalkyl group, —RiSiRaRbRc or a benzyl group, wherein Ri is an alkylene group;
Re is an alkyl group, an allyl group, a benzyl group, an aryl group, an alkoxy group, or —NRgRh, wherein Rg and Rh are independently H, an alkyl group or an aryl group;
R20 is CH2OR2g, CHO, CO2R10; wherein R2g is H, an alkyl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe, and wherein R10 is H or alkyl; and
R21 is a halogen atom, CH2OR2c, CHO, CO2R10a, CH═CHCH2OR2c, CH═CHCHO or CH═CHCO2R10, wherein R2c is H, an alkyl group, a benzyl group, a trityl group, —SiRaRbRc, CH2ORd, or CORe, and wherein R10a is H or alkyl.
20. The compound of claim 19 with the following stereostructure, or its enantiomer
Figure US20060270862A1-20061130-C00042
21. The compound of claim 20 where R11a and R11b are H or together form a portion of a six-membered acetal ring containing C(H)(p-C6H4OCH3) or C(CH3)2.
22. The compound of claim 21 wherein R1 is CH═CH2, and R21 is CH2OH, CHO or CO2R10.
23. A compound having the following formula
Figure US20060270862A1-20061130-C00043
wherein R13 is H or an alkyl group, R14 is H, an alkyl group, an aryl group or an alkoxyaryl group, and R22 is a halogen atom or —P(Ar)3X, wherein X is a counteranion selected from the groups halide, tetrafluoroborate, hexafluorophosphate and sulfonate, provided that when R13 and R14 are methyl groups, X is not I.
24. A compound of claim 23 provided that when R13 and R14 are alkyl groups, X is not halogen.
25. The compound of claim 23 with the following stereostructure, or its enantiomer
Figure US20060270862A1-20061130-C00044
wherein R13 is H or an alkyl group, and R14 is H, an alkyl group, an aryl group or an alkoxyaryl group, R22 is a halogen atom or —P(Ar)3X, wherein X is a counteranion selected from the groups halide, tetrafluoroborate, hexafluorophosphate and sulfonate, provided that when R13 and R14 are methyl groups, X is not I.
26. The compound of claim 25 wherein R13 is H, R14 is aryl, and R22 is P(C6H5)3X.
27. The compound of claim 25 wherein R14 is C6H4-p-OCH3.
28.-33. (canceled)
US11/497,746 2002-09-06 2006-08-02 Analogs of discodermolide and dictyostatin-1, intermediates therefor and methods of synthesis thereof Abandoned US20060270862A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US11/497,746 US20060270862A1 (en) 2002-09-06 2006-08-02 Analogs of discodermolide and dictyostatin-1, intermediates therefor and methods of synthesis thereof
US12/235,720 US7968736B2 (en) 2002-09-06 2008-09-23 Analogs of discodermolide and dictyostatin-1, intermediates therefor and methods of synthesis thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US40850302P 2002-09-06 2002-09-06
US43773603P 2003-01-02 2003-01-02
US10/655,916 US7122686B2 (en) 2002-09-06 2003-09-05 Analogs of discodermolide and dictyostatin-1, intermediates therefor and methods of synthesis thereof
US11/497,746 US20060270862A1 (en) 2002-09-06 2006-08-02 Analogs of discodermolide and dictyostatin-1, intermediates therefor and methods of synthesis thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/655,916 Division US7122686B2 (en) 2002-09-06 2003-09-05 Analogs of discodermolide and dictyostatin-1, intermediates therefor and methods of synthesis thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/235,720 Continuation US7968736B2 (en) 2002-09-06 2008-09-23 Analogs of discodermolide and dictyostatin-1, intermediates therefor and methods of synthesis thereof

Publications (1)

Publication Number Publication Date
US20060270862A1 true US20060270862A1 (en) 2006-11-30

Family

ID=31981594

Family Applications (3)

Application Number Title Priority Date Filing Date
US10/655,916 Expired - Fee Related US7122686B2 (en) 2002-09-06 2003-09-05 Analogs of discodermolide and dictyostatin-1, intermediates therefor and methods of synthesis thereof
US11/497,746 Abandoned US20060270862A1 (en) 2002-09-06 2006-08-02 Analogs of discodermolide and dictyostatin-1, intermediates therefor and methods of synthesis thereof
US12/235,720 Expired - Fee Related US7968736B2 (en) 2002-09-06 2008-09-23 Analogs of discodermolide and dictyostatin-1, intermediates therefor and methods of synthesis thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/655,916 Expired - Fee Related US7122686B2 (en) 2002-09-06 2003-09-05 Analogs of discodermolide and dictyostatin-1, intermediates therefor and methods of synthesis thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
US12/235,720 Expired - Fee Related US7968736B2 (en) 2002-09-06 2008-09-23 Analogs of discodermolide and dictyostatin-1, intermediates therefor and methods of synthesis thereof

Country Status (3)

Country Link
US (3) US7122686B2 (en)
AU (1) AU2003263090A1 (en)
WO (1) WO2004022552A1 (en)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7321046B2 (en) 2002-09-06 2008-01-22 University Of Pittsburgh Analogs of dictyostatin, intermediates therefor and methods of synthesis thereof
AU2003263090A1 (en) 2002-09-06 2004-03-29 University Of Pittsburgh Analogs of discodermolide and dictyostatin-1, intermediates therefor and methods of synthesis thereof
WO2005068451A2 (en) * 2003-12-31 2005-07-28 Cambridge University Technical Services Limited Methods for the synthesis of dictyostatin and derivatives and analogues thereof, stereochemical characterisation, novel dictyostatin compounds and uses thereof and synthetic intermediates
JP2008500370A (en) * 2004-05-27 2008-01-10 ユニヴァーシティ オブ ピッツバーグ Dicutiostatin analogs, intermediates thereof, and methods for their synthesis
WO2006091728A2 (en) * 2005-02-24 2006-08-31 The Trustees Of The University Of Pennsylvania Microtubule stabilizing compounds and methods of their use

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5430053A (en) * 1994-04-19 1995-07-04 Arizona Board Of Regents Acting On Behalf Of Arizona State University Isolation and structure of dictyostatin 1

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2400896A1 (en) * 2000-02-24 2001-08-30 Harbor Branch Oceanographic Institution, Inc. Novel compositions and uses of dictyostatin compounds
MY127609A (en) * 2000-08-07 2006-12-29 Novartis Ag Process for preparing discodermolide and analogues thereof
GB0101599D0 (en) * 2001-01-22 2001-03-07 Novartis Ag Organic compounds
AU2003263090A1 (en) * 2002-09-06 2004-03-29 University Of Pittsburgh Analogs of discodermolide and dictyostatin-1, intermediates therefor and methods of synthesis thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5430053A (en) * 1994-04-19 1995-07-04 Arizona Board Of Regents Acting On Behalf Of Arizona State University Isolation and structure of dictyostatin 1

Also Published As

Publication number Publication date
AU2003263090A1 (en) 2004-03-29
WO2004022552A1 (en) 2004-03-18
US20040186165A1 (en) 2004-09-23
US7968736B2 (en) 2011-06-28
US20090036691A1 (en) 2009-02-05
US7122686B2 (en) 2006-10-17

Similar Documents

Publication Publication Date Title
US6989450B2 (en) Synthesis of epothilones and related analogs
Clive et al. Total synthesis of both (+)-compactin and (+)-mevinolin. A general strategy based on the use of a special titanium reagent for dicarbonyl coupling
US6031133A (en) Synthetic techniques and intermediates for polyhydroxy, dienyl lactones and mimics thereof
US7968736B2 (en) Analogs of discodermolide and dictyostatin-1, intermediates therefor and methods of synthesis thereof
KR20040062418A (en) Compounds Which Mimic the Chemical and Biological Properties of Discodermolide
RU2528393C9 (en) Antitumour compounds
US5072002A (en) Synthesis of cholesterol-lowering agents
JP4357114B2 (en) 24-Hydroxyvitamin D derivative
CA2435371A1 (en) Process for preparing intermediates for the manufacture of discodermolide and discodermolide analogues
US7485631B2 (en) Phorboxazole compounds and methods of their preparation
EP0113881B1 (en) Antihypercholesterolemic compounds
KR20040032898A (en) Compounds Which Mimic the Chemical and Biological Properties of Discodermolide
EP1087958B1 (en) Process to manufacture simvastatin and intermediates
US7339066B1 (en) Intermediates for the synthesis of polypropionate antibiotics
JPH0643360B2 (en) 4-Hydroxy-2-cyclopentenones and pharmaceutical composition
US8030503B2 (en) Process for the preparation of epothilones
US7321046B2 (en) Analogs of dictyostatin, intermediates therefor and methods of synthesis thereof
KR20200073228A (en) Method for producing prostaglandin derivatives
Jung Total synthesis of dictyostatin analogs for anticancer agents
WO2003080567A2 (en) Intermediates for the synthesis of discodermolide and related analogues and methods for their preparation
Ruijter Design and synthesis of ratjadone analogues
Sminia et al. DESIGN AND SYNTHESIS OF RATJADONE ANALOGUES
JPH10251183A (en) Synthetic intermediate useful for synthesizing a-ring moiety of vitamin d derivative, its production and usage thereof
WO2004009574A1 (en) Synthesis of discodermolide
AU2005249480A1 (en) Analogs of dictyostatin, intermediates therefor and methods of synthesis thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: PITTSBURGH, UNIVERSITY OF, PENNSYLVANIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CURRAN, DENNIS P.;SHIN, YOUSEUNG;CHOY, NAKYEN;AND OTHERS;REEL/FRAME:018789/0024

Effective date: 20040524

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION