CN101652383B - Hepatitis c virus inhibitors - Google Patents

Hepatitis c virus inhibitors Download PDF

Info

Publication number
CN101652383B
CN101652383B CN200780049956.9A CN200780049956A CN101652383B CN 101652383 B CN101652383 B CN 101652383B CN 200780049956 A CN200780049956 A CN 200780049956A CN 101652383 B CN101652383 B CN 101652383B
Authority
CN
China
Prior art keywords
compound
alkyl
preparation
hcv
solution
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related
Application number
CN200780049956.9A
Other languages
Chinese (zh)
Other versions
CN101652383A (en
Inventor
A·X·王
B·Z·郑
S·丹德里
Q·赵
P·M·斯科拉
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.)
Bristol Myers Squibb Co
Original Assignee
Bristol Myers Squibb Co
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
Priority claimed from US11/939,780 external-priority patent/US7888464B2/en
Application filed by Bristol Myers Squibb Co filed Critical Bristol Myers Squibb Co
Publication of CN101652383A publication Critical patent/CN101652383A/en
Application granted granted Critical
Publication of CN101652383B publication Critical patent/CN101652383B/en
Expired - Fee Related legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Hepatitis C virus inhibitors having the general formula (I) are disclosed. Compositions comprising the compounds and methods for using the compounds to inhibit HCV are also disclosed.

Description

Hepatitis C virus inhibitors
The cross reference of related application
The application requires in the 60/866th, 130 series number U.S. Provisional Application No. of submission on November 16th, 2006.
Invention field
The application openly relates generally to antiviral compound, more specifically relate to inhibition by the compound of NS3 proteolytic enzyme (being also referred to as " serine protease " herein) function of hepatitis C virus (HCV) coding, comprise the composition of this compounds and the method for inhibition NS3 proteolytic enzyme function.
Background of invention
HCV is main human pathogen, and it worldwide infects roughly 5 times that estimate 100,017,000 people-1 type human immunodeficiency virus infection number.In these HCV infected individuals quite a few develops into the serious hepatopathy that carries out, and comprises liver cirrhosis and hepatocellular carcinoma.
At present, the most effective HCV therapy adopts the combination of alpha-interferon and ribavirin, produces lasting effectiveness in 40% patient.Nearest clinical effectiveness shows that as monotherapy, (pegylated) alpha-interferon of Pegylation is better than unmodified alpha-interferon.But even use the experimental therapy scheme of the combination that comprises Pegylation alpha-interferon and ribavirin, quite a few patient's virus load does not continue to reduce yet.Therefore, developing the effective therapy that is used for the treatment of the HCV infection is the obvious long demand.
HCV is positive chain RNA virus.Based on the comparison of deduced amino acid and the extensive similarity in 5 ' the untranslated district, HCV has been classified as the independent class in the flaviviridae.All members of flaviviridae wrap to be contained by the encode genomic virion of positive chain RNA of all known viruse specific proteins of the single translation of not interrupting open reading frame.
In whole HCV genome, in Nucleotide and amino acid sequence coded, find significant diversity.Characterized at least six kinds of oligogene types, and described more than 50 kinds of hypotypes.The distribution on global difference of the oligogene type of HCV, and the clinical importance of the gene diversity of HCV unpredictable still are although may make many researchs aspect the influence in genotype to pathogeny and treatment.
Strand HCV rna gene group is about 9500 length of nucleotides and has the single open reading frame (ORF) that are about 3000 amino acid whose single big polymeric protein codings.In the cell that infects, this polymeric protein in a plurality of positions by cell and virus protease division and produce structural and unstructuredness (NS) albumen.Under the situation of HCV, realize the generation of ripe Nonstructural Protein (NS2, NS3, NS4A, NS4B, NS5A and NS5B) by two kinds of virus proteases.First kind is considered to split in the punishment of NS2-NS3 contact; Serine protease in second kind of N-stub area that is included in NS3 also mediates all follow-up divisions in NS3 downstream, has both comprised that cis (in NS3-NS4A division site) also comprised trans (for all the other NS4A-NS4B, NS4B-NS5A, NS5A-NS5B site).NS4A albumen seems the performance multi-functional---and serve as the cofactor of NS3 proteolytic enzyme, and may help the film location of NS3 and other rdrp virus component.It is necessary that the mixture formation of NS3 albumen and NS4A looks like the processing event, thereby improve the proteolysis efficient at all sites place.This NS3 albumen also shows ribonucleoside triphosphote enzyme and rna helicase enzymic activity.NS5B participates in the RNA dependent form RNA polymerase that HCV copies.
The application's disclosure provides and can suppress NS3 proteolytic enzyme, for example with the peptides of the function of the NS3 proteolytic enzyme of NS4A proteolytic enzyme associating.In addition, the application's disclosure has been described and has been given combination therapy to the patient, can will effectively suppress The compounds of this invention and one or both other compound Combined Preparation with anti-HCV activity of HCV NS3 proteolytic enzyme thus.
Summary of the invention
Aspect first, the application's disclosure provides formula (I) compound
Figure G2007800499569D00021
Or its pharmacologically acceptable salt, wherein
N is 1,2 or 3;
R 1Be selected from hydroxyl and-NHSO 2R 7
R 2Be selected from hydrogen, alkenyl, alkyl, cyano group alkyl, cycloalkyl and haloalkyl;
R 3Be selected from alkenyl, alkyl, aryl, arylalkyl, cycloalkyl, (cycloalkyl) alkyl, heterocyclic radical and heterocyclic radical alkyl;
R 4Be-OR 8
R 5Be selected from alkenyl, alkoxyalkyl, alkoxy carbonyl alkyl, alkyl, arylalkyl, carboxyalkyl, cyano group alkyl, cycloalkyl, (cycloalkyl) alkyl, halogenated alkoxy alkyl, haloalkyl, hydroxyalkyl, (NR aR b) alkyl and (NR aR b) carbonylic alkyl;
R 6Be selected from alkoxy carbonyl, alkyl, alkyl-carbonyl, alkyl sulphonyl, cycloalkyl oxy carbonyl, cycloalkyl, halo alkoxy carbonyl, haloalkyl, halogenated alkyl carbonyl, (NR aR b) carbonyl and (NR aR b) alkylsulfonyl; Perhaps
R 6Be selected from phenyl and 5 or 6 yuan of partially or completely unsaturated rings, described ring is chosen wantonly and is contained 1,2,3 or 4 heteroatoms that is selected from nitrogen, oxygen and sulphur; Wherein each described ring is optional is independently selected from following substituting group by 1,2,3 or 4 and replaces: alkoxyl group, alkoxy carbonyl, alkyl, alkyl-carbonyl, alkyl sulfenyl, carboxyl, cyano group, cycloalkyl, cycloalkyl oxy, halogen, haloalkyl, halogenated alkoxy ,-NR gR h, (NR jR k) carbonyl, (NR jR k) alkylsulfonyl and oxo base;
R 7Be selected from alkyl, aryl, cycloalkyl, (cycloalkyl) alkyl, heterocyclic radical and-NR cR d
R 8Be selected from alkoxyalkyl, alkyl, alkyl-carbonyl, arylalkyl, cycloalkyl, (cycloalkyl) alkyl, naphthene base carbonyl, halogenated alkoxy alkyl, haloalkyl, (NR eR f) carbonyl and-P (O) (OR ') 2R wherein eAnd R fBe independently selected from hydrogen, alkyl and arylalkyl; Perhaps R eAnd R fThe nitrogen-atoms that connects with them forms 5 or 6 yuan of monocyclic heterocycles, and described heterocycle is optional to be contained one and be selected from O, NR xOther heteroatoms with S; R wherein xBe selected from hydrogen and alkyl; And wherein R ' is selected from hydrogen and alkyl;
R aAnd R bBe independently selected from hydrogen, alkoxyl group, alkoxyalkyl, alkyl, aryl, arylalkyl, cycloalkyl, halogenated alkoxy alkyl, haloalkyl, heterocyclic radical and heterocyclic radical alkyl;
R cAnd R dBe independently selected from alkoxyl group, alkyl, aryl, arylalkyl, cycloalkyl, (cycloalkyl) alkyl, heterocyclic radical and heterocyclic radical alkyl; Perhaps R cAnd R dThe nitrogen-atoms that connects with them forms 5 or 6 yuan of monocyclic heterocycles;
R gAnd R hBe independently selected from hydrogen, alkoxyalkyl, alkoxy carbonyl, alkyl, alkyl-carbonyl, arylalkyl and haloalkyl; And
R jAnd R kBe independently selected from hydrogen, alkyl, aryl, arylalkyl and heterocyclic radical; Wherein the aryl moiety of aryl, arylalkyl and heterocyclic radical are optional is replaced by 1 or 2 substituting group that is independently selected from alkoxyl group, alkyl and halogen.
Aspect first, the application's disclosure provides formula (II) compound
Figure G2007800499569D00041
Or its pharmacologically acceptable salt, wherein
N is 1,2 or 3;
R 2Be selected from hydrogen, alkenyl, alkyl, cyano group alkyl, cycloalkyl and haloalkyl;
R 3Be selected from alkenyl, alkyl, aryl, arylalkyl, cycloalkyl, (cycloalkyl) alkyl, heterocyclic radical and heterocyclic radical alkyl;
R 5Be selected from alkenyl, alkoxyalkyl, alkoxy carbonyl alkyl, alkyl, arylalkyl, carboxyalkyl, cyano group alkyl, cycloalkyl, (cycloalkyl) alkyl, halogenated alkoxy alkyl, haloalkyl, hydroxyalkyl, (NR aR b) alkyl and (NR aR b) carbonylic alkyl;
R 6Be selected from alkoxy carbonyl, alkyl, alkyl-carbonyl, alkyl sulphonyl, cycloalkyl oxy carbonyl, cycloalkyl, halo alkoxy carbonyl, haloalkyl, halogenated alkyl carbonyl, (NR aR b) carbonyl and (NR aR b) alkylsulfonyl; Perhaps
R 6Be selected from phenyl and 5 or 6 yuan of partially or completely unsaturated rings, described ring is chosen wantonly and is contained 1,2,3 or 4 heteroatoms that is selected from nitrogen, oxygen and sulphur; Wherein each described ring is optional is independently selected from following substituting group by 1,2,3 or 4 and replaces: alkoxyl group, alkoxy carbonyl, alkyl, alkyl-carbonyl, alkyl sulfenyl, carboxyl, cyano group, cycloalkyl, cycloalkyl oxy, halogen, haloalkyl, halogenated alkoxy ,-NR gR h, (NR jR k) carbonyl, (NR jR k) alkylsulfonyl and oxo base;
R 7Be selected from alkyl, aryl, cycloalkyl, (cycloalkyl) alkyl, heterocyclic radical and-NR cR d
R 8Be selected from alkoxyalkyl, alkyl, alkyl-carbonyl, arylalkyl, cycloalkyl, (cycloalkyl) alkyl, naphthene base carbonyl, halogenated alkoxy alkyl, haloalkyl, (NR eR f) carbonyl and-P (O) (OR ') 2R wherein eAnd R fBe independently selected from hydrogen, alkyl and arylalkyl; Perhaps R eAnd R fThe nitrogen-atoms that connects with them forms 5 or 6 yuan of monocyclic heterocycles, and described heterocycle is optional to be contained one and be selected from O, NR xOther heteroatoms with S; R wherein xBe selected from hydrogen and alkyl; And wherein R ' is selected from hydrogen and alkyl;
R aAnd R bBe independently selected from hydrogen, alkoxyl group, alkoxyalkyl, alkyl, aryl, arylalkyl, cycloalkyl, halogenated alkoxy alkyl, haloalkyl, heterocyclic radical and heterocyclic radical alkyl;
R cAnd R dBe independently selected from alkoxyl group, alkyl, aryl, arylalkyl, cycloalkyl, (cycloalkyl) alkyl, heterocyclic radical and heterocyclic radical alkyl; Perhaps R cAnd R dThe nitrogen-atoms that connects with them forms 5 or 6 yuan of monocyclic heterocycles;
R gAnd R hBe independently selected from hydrogen, alkoxyalkyl, alkoxy carbonyl, alkyl, alkyl-carbonyl, arylalkyl and haloalkyl; And
R jAnd R kBe independently selected from hydrogen, alkyl, aryl, arylalkyl and heterocyclic radical; Wherein the aryl moiety of aryl, arylalkyl and heterocyclic radical are optional is replaced by 1 or 2 substituting group that is independently selected from alkoxyl group, alkyl and halogen.
In first embodiment aspect second of the application's disclosure, provide formula (II) compound or pharmaceutically acceptable salt thereof, wherein
N is 1;
R 2Be selected from alkenyl, alkyl and haloalkyl;
R 3Be selected from alkenyl, alkyl, aryl, arylalkyl, cycloalkyl, (cycloalkyl) alkyl, heterocyclic radical and heterocyclic radical alkyl;
R 5Be selected from alkenyl, alkyl and arylalkyl;
R 6Be selected from alkoxy carbonyl, cycloalkyl oxy carbonyl, halo alkoxy carbonyl, (NR aR b) carbonyl; Perhaps
R 6Be phenyl, described phenyl is optional to be replaced by 1 or 2 substituting group that is independently selected from alkoxyl group and halogen; And
R 7It is cycloalkyl.
In second embodiment aspect second of the application's disclosure, provide formula (II) compound or pharmaceutically acceptable salt thereof, wherein
N is 1;
R 2Be selected from alkenyl, alkyl and haloalkyl;
R 3Be selected from aryl and heterocyclic radical;
R 5Be selected from alkenyl, alkyl and arylalkyl;
R 6Be selected from alkoxy carbonyl, cycloalkyl oxy carbonyl, halo alkoxy carbonyl, (NR aR b) carbonyl; Perhaps
R 6Be phenyl, described phenyl is optional to be replaced by 1 or 2 substituting group that is independently selected from alkoxyl group and halogen; And
R 7It is cycloalkyl.
R 7It is cycloalkyl.
In the 3rd embodiment aspect second of the application's disclosure, provide formula (II) compound or pharmaceutically acceptable salt thereof, wherein
N is 1;
R 2Be selected from alkenyl, alkyl and haloalkyl;
R 3It is aryl;
R 5Be selected from alkenyl, alkyl and arylalkyl;
R 6Be selected from alkoxy carbonyl, cycloalkyl oxy carbonyl, halo alkoxy carbonyl, (NR aR b) carbonyl; Perhaps
R 6Be phenyl, described phenyl is optional to be replaced by 1 or 2 substituting group that is independently selected from alkoxyl group and halogen; And
R 7It is cycloalkyl.
Aspect the 3rd of the application's disclosure, composition is provided, described composition comprises formula (I) compound or pharmaceutically acceptable salt thereof and pharmaceutically acceptable carrier.In first embodiment aspect the 3rd, composition also comprises at least a additional compounds with whose anti-HCV activity.In second embodiment aspect the 3rd, at least a additional compounds is Interferon, rabbit or ribavirin.In the 3rd embodiment aspect the 3rd, Interferon, rabbit is selected from interferon alpha 2B, Pegylation (pegylated) interferon alpha, compound (consensus) Interferon, rabbit/interferon alpha 2A, and lymphoblastoid interferon-tau (tau).
In the 4th embodiment aspect the 3rd, the application's disclosure provides composition, described composition comprises formula (I) compound or pharmaceutically acceptable salt thereof, pharmaceutically acceptable carrier, and at least a additional compounds with whose anti-HCV activity, wherein said at least a additional compounds be selected from interleukin-22, interleukin 6, interleukin 12, promotion 1 type helper cell reply the compound of development, RNA interfering, sense-rna, Imiquimod (Imiqimod), ribavirin, inosine 5 '-monophosphate dehydrogenase inhibitor, Symmetrel and Rimantadine.
In the 5th embodiment aspect the 3rd, the application's disclosure provides composition, described composition comprises formula (I) compound or pharmaceutically acceptable salt thereof, pharmaceutically acceptable carrier, and at least a additional compounds with whose anti-HCV activity, wherein said at least a additional compounds effectively suppresses to be selected from the function of following target: the HCV metalloprotease, the HCV serine protease, the HCV polysaccharase, the HCV helicase, HCV NS4B albumen, HCV enters (entry), HCV assembles (assembly), HCV deviates from (egress), HCV NS5A albumen, and be used for the treatment of the IMPDH that HCV infects.
Aspect the 4th of the application's disclosure, the method that HCV infects among the treatment patient is provided, described method comprises formula (I) compound or pharmaceutically acceptable salt thereof that gives described patient treatment significant quantity.In first embodiment aspect the 4th, described method also is included in before giving construction (I) compound or pharmaceutically acceptable salt thereof, give at least a additional compounds with whose anti-HCV activity afterwards or simultaneously.In second embodiment aspect the 4th, described at least a additional compounds is Interferon, rabbit or ribavirin.In the 3rd embodiment aspect the 4th, Interferon, rabbit is selected from interferon alpha 2B, glycol interferon alpha, Interferon alfacon-1, interferon alpha 2A and lymphoblastoid interferon-tau.
In the 4th embodiment aspect the 4th, the application's disclosure provides the method that HCV infects among the treatment patient, described method comprises formula (I) compound or pharmaceutically acceptable salt thereof that gives described patient treatment significant quantity, and before giving construction (I) compound or pharmaceutically acceptable salt thereof, give at least a additional compounds with whose anti-HCV activity afterwards or simultaneously, wherein said at least a additional compounds is selected from interleukin-22, interleukin 6, interleukin 12, promote 1 type helper cell to reply the compound of development, RNA interfering, sense-rna, Imiquimod, ribavirin, inosine 5 '-monophosphate dehydrogenase inhibitor, Symmetrel and Rimantadine.
In the 5th embodiment aspect the 4th, the application's disclosure provides the method that HCV infects among the treatment patient, described method comprises formula (I) compound or pharmaceutically acceptable salt thereof that gives described patient treatment significant quantity, and before giving construction (I) compound or pharmaceutically acceptable salt thereof, give at least a additional compounds with whose anti-HCV activity afterwards or simultaneously, wherein said at least a additional compounds effectively suppresses to be selected from the function of following target: the HCV metalloprotease, the HCV serine protease, the HCV polysaccharase, the HCV helicase, HCV NS4B albumen, HCV enters, the HCV assembling, HCV deviates from, HCV NS5A albumen, and be used for the treatment of the IMPDH that HCV infects.
Aspect the 5th of the application's disclosure, composition is provided, described composition comprises formula (I) compound or pharmaceutically acceptable salt thereof, a kind of, two kinds, three kinds, four kinds or five kinds of additional compounds with whose anti-HCV activity, and pharmaceutically acceptable carrier.In first embodiment aspect the 5th, composition comprises three kinds or four kinds of additional compounds with whose anti-HCV activity.In second embodiment aspect the 5th, composition comprises one or both additional compounds with whose anti-HCV activity.
Aspect the 5th of the application's disclosure, the method that HCV infects among the treatment patient is provided, described method comprises formula (I) compound or pharmaceutically acceptable salt thereof that gives described patient treatment significant quantity, and before giving construction (I) compound or pharmaceutically acceptable salt thereof, give a kind of, two kinds, three kinds, four kinds or five kinds of additional compounds with whose anti-HCV activity afterwards or simultaneously.In first embodiment aspect the 6th, described method comprises and gives three kinds or four kinds of additional compounds with whose anti-HCV activity.In second embodiment aspect the 6th, described method comprises and gives the additional compounds that one or both have the whose anti-HCV activity.
The other side of the application's disclosure can comprise the suitable combination of embodiment disclosed herein.
Other side and embodiment can be referring to descriptions provided herein.
Explanation at the specification sheets of the application's disclosure should be consistent with rule and the principle of chemical bond.In some cases, may remove hydrogen atom to hold substituting group in any given position.
Should be appreciated that the compound that the application's disclosure comprises is to be moderately stable those compounds as medicine the time.
All patents, patent application and the reference quoted in specification sheets all are incorporated herein by reference.Under contradictory situation, will be as the criterion with the application's disclosure (comprising definition).
As used in this manual, following term has the meaning of appointment:
As used herein, singulative " ", " one ", and " being somebody's turn to do " comprise plural implication, unless indicate clearly in addition in the text.
Term " alkenyl " as used herein means the straight or branched group of 2-6 the carbon atom that contains at least one carbon-to-carbon double bond.
Term " alkoxyl group " means the alkyl that is connected in parent molecular moiety by Sauerstoffatom as used herein.
Term " alkoxyalkyl " means the alkyl that is replaced by 1,2 or 3 alkoxyl group as used herein.
Term term " alkoxy carbonyl " means the alkoxyl group that is connected in parent molecular moiety by carbonyl as used herein.
Term " alkoxy carbonyl alkyl " means the alkyl that is replaced by 1,2 or 3 alkoxy carbonyl as used herein.
Term " alkyl " means the group derived from the straight or branched stable hydrocarbon that contains 1-6 carbon atom as used herein.
Term " alkyl-carbonyl " means the alkyl that is connected in parent molecular moiety by carbonyl as used herein.
Term " alkyl sulfenyl " means the alkyl that is connected in parent molecular moiety by sulphur atom as used herein.
Term " alkyl sulphonyl " means the alkyl that is connected in parent molecular moiety by alkylsulfonyl as used herein.
Term " aryl " means phenyl as used herein, or one of them or two bicyclic condensed loop systems that ring is phenyl.Bicyclic condensed loop systems is made up of the phenyl that condenses in 4-to 6-unit's aromatics or non-aromatic carbocyclic ring.The aryl of the application's disclosure can be connected in parent molecular moiety by any commutable carbon atom in the group.The representative example of aryl includes but not limited to indanyl, indenyl, naphthyl, phenyl and tetralyl.The aryl of the application's disclosure can be chosen wantonly and is independently selected from following substituting group by 1,2,3,4 or 5 and replace: alkoxyl group, alkoxy carbonyl, alkyl, second aryl, cyano group, halogen, halogenated alkoxy, haloalkyl, heterocyclic radical, heterocyclic radical alkyl, hydroxyl, hydroxyalkyl, nitro ,-NR xR y, (NR xR y) alkoxyl group, (NR xR y) alkyl, (NR xR y) carbonyl and oxo base; Wherein the heterocyclic radical of second aryl, heterocyclic radical and heterocyclic radical alkyl part can be chosen wantonly by 1,2,3,4 or 5 and be independently selected from following substituting group replacement: alkoxyl group, alkyl, cyano group, halogen, halogenated alkoxy, haloalkyl, hydroxyl and nitro.
Term " arylalkyl " means the alkyl that is replaced by 1,2 or 3 aryl as used herein.
Term " carbonyl " as used herein, mean-C (O)-.
Term " carboxyl " means-CO as used herein 2H.
Term " carboxyalkyl " means by the alkyl of 1,2 or 3 carboxyl substituted as used herein.
Term " cyano group " means-CN as used herein.
Term " cyano group alkyl " means the alkyl that is replaced by 1,2 or 3 cyano group as used herein.
Term " cycloalkyl " as used herein, mean have 3-14 carbon atom and 0 heteroatomic saturated monocycle, two ring or tricyclic hydrocarbon loop systems.The representative example of cycloalkyl includes but not limited to cyclopropyl, cyclopentyl, two ring [3.1.1] heptyl and adamantyls.The cycloalkyl of the application's disclosure can be chosen wantonly by 1,2,3 or 4 and be independently selected from following substituting group replacement: alkenyl, alkoxyl group, alkoxyalkyl, alkyl, arylalkyl, aryl carbonyl, cyano group, cycloalkenyl group, (cycloalkyl) alkyl, halogen, halogenated alkoxy, haloalkyl and (NR jR k) carbonyl; R wherein jAnd R kBe independently selected from hydrogen, alkyl, aryl, arylalkyl and heterocyclic radical; Wherein the aryl moiety of aryl, arylalkyl and heterocyclic radical are optional is replaced by 1 or 2 substituting group that is independently selected from alkoxyl group, alkyl and halogen.
Term " (cycloalkyl) alkyl " means by the alkyl of 1,2 or 3 cycloalkyl substituted as used herein.
Term " naphthene base carbonyl " means the cycloalkyl that is connected in parent molecular moiety by carbonyl as used herein.
Term " cycloalkyl oxy " means the cycloalkyl that is connected in parent molecular moiety by Sauerstoffatom as used herein.
Term " cycloalkyl oxy carbonyl " means the cycloalkyl oxy that is connected in parent molecular moiety by carbonyl as used herein.
Term " halo " and " halogen " mean F, Cl, Br and I as used herein.
Term " halogenated alkoxy " means the haloalkyl that is connected in parent molecular moiety by Sauerstoffatom as used herein.
Term " halogenated alkoxy alkyl " means the alkyl that is replaced by 1,2 or 3 halogenated alkoxy as used herein.
Term " halo alkoxy carbonyl " means the halogenated alkoxy that is connected in parent molecular moiety by carbonyl as used herein.
Term " haloalkyl " means the alkyl that is replaced by 1,2,3 or 4 halogen atom as used herein.
Term " halogenated alkyl carbonyl " means the haloalkyl that is connected in parent molecular moiety by carbonyl as used herein.
Term " heterocyclic radical " means and contains 1,2 or 3 heteroatomic 5,6 or 7 yuan of ring that are independently selected from nitrogen, oxygen and sulphur as used herein.5 yuan of rings have 0-2 two keys, and 6-and 7-unit ring have 0-3 two keys.Term " heterocyclic radical " also comprises bicyclic radicals, and wherein heterocyclic ring and 4-to 6-unit's aromatics or non-aromatic carbocyclic ring or another heterocyclic radical condense.The heterocyclic radical of the application's disclosure can be connected in parent molecular moiety by any carbon atom or the nitrogen-atoms in the group.The example of heterocyclic radical includes but not limited to benzothienyl, furyl, imidazolyl, indolinyl, indyl, isothiazolyl, isoxazolyl, morpholinyl, oxazolyl, piperazinyl, piperidyl, pyrazolyl, pyridyl, pyrrolidyl, pyrrolopyridinyl, pyrryl, thiazolyl, thienyl and thio-morpholinyl.The heterocyclic radical of the application's disclosure can be chosen wantonly and is independently selected from following substituting group by 1,2,3,4 or 5 and replace: alkoxyl group, alkoxy carbonyl, alkyl, aryl, cyano group, halogen, halogenated alkoxy, haloalkyl, second heterocyclic radical, heterocyclic radical alkyl, hydroxyl, hydroxyalkyl, nitro ,-NR xR y, (NR xR y) alkoxyl group, (NR xR y) alkyl, (NR xR y) carbonyl and oxo base; Wherein the heterocyclic radical of aryl, second heterocyclic radical and heterocyclic radical alkyl part can be chosen wantonly and is independently selected from following substituting group by 1,2,3,4 or 5 and replace: alkoxyl group, alkyl, cyano group, halogen, halogenated alkoxy, haloalkyl, hydroxyl and nitro.
Term " heterocyclic radical alkyl " means the alkyl that is replaced by 1,2 or 3 heterocyclic radical as used herein.
Term " hydroxyl " means-OH as used herein.
Term " hydroxyalkyl " means the alkyl that is replaced by 1,2 or 3 hydroxyl as used herein.
Term " NR as used herein aR b", mean two groups that are connected in parent molecular moiety by nitrogen-atoms, R aAnd R bR aAnd R bIndependently be selected from hydrogen, alkoxyl group, alkoxyalkyl, alkyl, aryl, arylalkyl, cycloalkyl, halogenated alkoxy alkyl, haloalkyl, heterocyclic radical and heterocyclic radical alkyl respectively.
Term " (NR as used herein aR b) alkyl ", mean by 1,2 or 3-NR aR bThe alkyl that group replaces.
Term " (NR as used herein aR b) carbonyl ", mean by carbonyl be connected in parent molecular moiety-NR aR bGroup.
Term " (NR as used herein aR b) carbonylic alkyl ", mean by 1,2 or 3-NR aR bThe alkyl of carbonyl substituted.
Term " (NR as used herein aR b) alkylsulfonyl ", mean by alkylsulfonyl be connected in parent molecular moiety-NR aR bGroup.
Term " NR as used herein cR d", mean two groups that are connected in parent molecular moiety by nitrogen-atoms, R cAnd R dR cAnd R dBe independently selected from alkoxyl group, alkyl, aryl, arylalkyl, cycloalkyl, (cycloalkyl) alkyl, heterocyclic radical and heterocyclic radical alkyl; Perhaps R cAnd R dThe nitrogen-atoms that connects with them forms 5 or 6 yuan of monocyclic heterocycles.
Term " NR as used herein eR f", mean two groups that are connected in parent molecular moiety by nitrogen-atoms, R eAnd R fR eAnd R fBe independently selected from hydrogen, alkyl and arylalkyl; Perhaps R eAnd R fThe nitrogen-atoms that connects with them forms 5 or 6 yuan of monocyclic heterocycles, and described heterocycle is optional to be contained one and be selected from O, NR xOther heteroatoms with S; R wherein xBe selected from hydrogen and alkyl; And wherein R ' is selected from hydrogen and alkyl.
Term " NR as used herein gR h", mean two groups that are connected in parent molecular moiety by nitrogen-atoms, R gAnd R hR gAnd R hBe independently selected from hydrogen, alkoxyalkyl, alkoxy carbonyl, alkyl, alkyl-carbonyl, arylalkyl and haloalkyl.
Term " NR as used herein jR k", mean two groups that are connected in parent molecular moiety by nitrogen-atoms, R jAnd R kR jAnd R kBe independently selected from hydrogen, alkyl, aryl, arylalkyl and heterocyclic radical; Wherein the aryl moiety of aryl, arylalkyl and heterocyclic radical are optional is replaced by 1 or 2 substituting group that is independently selected from alkoxyl group, alkyl and halogen.
Term " (NR as used herein jR k) carbonyl ", mean by carbonyl be connected in parent molecular moiety-NR jR kGroup.
Term " (NR as used herein jR k) alkylsulfonyl ", mean by alkylsulfonyl be connected in parent molecular moiety-NR jR kGroup.
Term " NR as used herein xR y", mean two groups that are connected in parent molecular moiety by nitrogen-atoms, R xAnd R yR xAnd R yBe independently selected from hydrogen and alkyl.
Term " (NR as used herein xR y) alkoxyl group ", mean (the NR that is connected in parent molecular moiety by Sauerstoffatom xR y) alkyl.
Term " (NR as used herein xR y) alkyl ", mean by 1,2 or 3-NR xR yThe alkyl that group replaces.
Term " (NR as used herein xR y) carbonyl ", mean by carbonyl be connected in parent molecular moiety-NR xR yGroup.
Term " nitro " means-NO as used herein 2
Term " oxo base " means=O as used herein.
Term " alkylsulfonyl " means-SO as used herein 2-.
The compound of the application's disclosure can be used as prodrug and exists.Term used herein " prodrug " refers to change into rapidly by hydrolysis in blood in vivo the compound of parent compound.The prodrug of the application's disclosure comprises the ester of the hydroxyl on the parent molecule, the ester of the carboxyl on the parent molecule, and the acid amides of the amine on the parent molecule.
The compound of the application's disclosure can be used as pharmacologically acceptable salt and exists.Term " pharmacologically acceptable salt " as used herein, salt or the zwitterionic form of the compound of expression the application disclosure, its in water soluble or the oil or can be scattered in wherein, it is applicable to patient tissue in the category of correct medical judgment and contacts, and there are not excessive toxicity, pungency, anaphylaxis or other problem or complication, this matches with rational interests/risk ratio, and effective to their intended purpose.Can during the final separation of described compound and purifying, prepare salt, perhaps by suitable basic functionality is separated with suitable acid-respons.Representational acid salt comprises acetate, adipate, alginate, Citrate trianion, aspartate, benzoate, benzene sulfonate, hydrosulfate, butyrates, camphorate, camsilate, digluconate, glycerophosphate, Hemisulphate (hemisulfate), enanthate, hexanoate, formate, fumarate, hydrochloride, hydrobromate, hydriodate, the 2-isethionate, lactic acid salt, maleate, mesitylene sulfonate, mesylate, naphthalenesulfonate, nicotinate, the 2-naphthalenesulfonate, oxalate, pamoate, pectate (pectinate), persulphate, 3-phenylpropionic acid salt, picrate, Pivalate, propionic salt, succinate, tartrate, trichloroacetate, trifluoroacetate, phosphoric acid salt, glutaminate, supercarbonate, tosilate and undecane hydrochlorate.The example that can be used to form the acid of pharmaceutically acceptable additive salt comprises mineral acid for example hydrochloric acid, Hydrogen bromide, sulfuric acid and phosphoric acid, and organic acid for example oxalic acid, toxilic acid, succsinic acid and citric acid.
Base addition salt can be during the final separation and purifying of compound, by make acidic-group and suitable alkali for example metallic cation oxyhydroxide, carbonate or supercarbonate or prepare with ammonia or organic primary amine, secondary amine or reactive tertiary amine.The positively charged ion of pharmacologically acceptable salt comprises lithium, sodium, potassium, calcium, magnesium and aluminium, and avirulent quaternary ammonium cation such as ammonium, tetramethyl-ammonium, tetraethyl ammonium, methylamine, dimethylamine, Trimethylamine 99, triethylamine, diethylamine, ethamine, Tributylamine, pyridine, N, accelerine, N-methyl piperidine, N-methylmorpholine, dicyclohexylamine, PROCAINE HCL, PHARMA GRADE, dibenzyl amine, N, N-dibenzyl phenylethylamine and N, N '-dibenzyl-ethylenediamin.Other the representational organic amine that is used to form base addition salt comprises quadrol, thanomin, diethanolamine, piperidines and piperazine.
Term used herein " whose anti-HCV activity " refers to that compound can effectively treat HCV virus.
Term " compound of the application's disclosure " and the expression that is equal to are intended to comprise formula (I) compound, and pharmaceutically acceptable enantiomorph, diastereomer and salt thereof.Similarly, be intended to comprise their salt when mentioning intermediate, need only so in this article salt and allow.
Term " patient " comprises people and other Mammals.
Term " pharmaceutical composition " refers to comprise compound and at least a medication carrier of the application's disclosure, be auxiliary, vehicle or carrier, the composition of thinner, sanitas, filler, flowing regulator, disintegrating agent, wetting agent, emulsifying agent, suspension agent, sweeting agent, seasonings, sweetener, antiseptic-germicide, anti-mycotic agent, lubricant and dispersion agent (depending on mode of administration and formulation) for example.Can use for example Remington ' s PharmaceuticalSciences, the 18th edition, Mack Publishing Company, Easton, the composition of listing among the PA (1999).
Term " pharmaceutically acceptable " be used in this article being illustrated in be fit in the appropriate medical care determination range contact use with patient tissue and do not have excessive toxicity, stimulation, anaphylaxis or other problem or a complication with rational risk/benefit than those compounds, material, composition and/or the formulation that match.
Term " treatment significant quantity " as used herein means the total amount of each activeconstituents that is enough to show patient's benefit, as, reduce viral load.When being applied to single activeconstituents separately, this term means single composition.When being applied to Combined Preparation, this term means the merging amount of the activeconstituents that produces result for the treatment of, and no matter whether be uniting and give in regular turn or simultaneously.
Term " treatment " refers to: (i) prevention may be attacked by a disease easily, obstacle or illness but be not diagnosed as yet in ill patient's body and this disease, obstacle or illness occur; (ii) suppress disease, obstacle or illness, namely stop its development; And/or (iii) alleviate disease, obstacle or illness, cause that namely disease, obstacle or illness disappear.
When being used for the compound of name the application disclosure, label P1 ' used herein, P1, P2, P2 *, P3 and P4 described the binding site with respect to native peptides division substrate, the relative position of the amino-acid residue of proteinase inhibitor combination.Divide between P1 and P1 ' in natural substrate, wherein non-staple position refers to from the terminal beginning of the C-in the natural division of peptide site to the terminal amino acid that extends of N-; And the N-of main position sources spontaneous fission site title is terminal and extend to C-is terminal.For example, P1 ' refers to the first location (being the terminal first location of N-) away from the right-hand side of the C-end that divides the site; And P1 begins counting, P2 from the left-hand side in the terminal division of C-site: from terminal second of C-, etc. (referring to Berger A.﹠amp; Schechter I., Transactions of the Royal Society London series (1970), B257, 249-264).
Figure below has shown the label of the compound of the application's disclosure.
Figure G2007800499569D00141
In the compound of the application's disclosure, there is asymmetric center.For example, this compound can comprise the P1 cyclopropyl unit of following formula
Figure G2007800499569D00151
C wherein 1And C 2Represent the unsymmetrical carbon at 1 and 2 place of cyclopropyl rings separately.
Figure G2007800499569D00152
R 2Be cis R with respect to carbonyl 2Be cis with respect to carbonyl
Figure G2007800499569D00153
R 2Be cis R with respect to acid amides 2Be cis with respect to acid amides
It should be understood that the application's disclosure comprises all stereochemistry heterogeneous forms or its mixture with the ability that suppresses HCV proteolytic enzyme.
Some compound of the application's disclosure also can exist with separable different stable conformation form.By rotating the asymmetry of reversing that causes around the restriction of asymmetric singly-bound, for example because steric hindrance or ring strain can realize the separation of different conformers.The application's disclosure comprises each conformer of these compounds and composition thereof.
Some compound of the application's disclosure can exist with zwitterionic form, and the application's disclosure comprises each zwitterionic form of these compounds and composition thereof.
When being used for therapy, formula (I) compound and the pharmacologically acceptable salt thereof for the treatment of significant quantity might be as the feed chemicals administrations, and it also is possible that activeconstituents occurs as pharmaceutical composition.Therefore, the application's disclosure also provides pharmaceutical composition, and it comprises formula (I) compound or pharmaceutically acceptable salt thereof for the treatment of significant quantity, and one or more pharmaceutically acceptable carrier, thinner or vehicle.Formula (I) compound and pharmacologically acceptable salt thereof are as mentioned above.Carrier, thinner or vehicle are adaptive and the harmless meaning of its recipient said at other composition of itself and preparation, must be acceptable.According to the other side of the application's disclosure, the method for useful in preparing drug formulations also is provided, described method comprises formula (I) compound or pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable carrier, thinner or mixed with excipients.
Pharmaceutical preparation can exist with unit dosage, and its per unit dosage contains the activeconstituents of predetermined amount.At the monotherapy of the disease that is used for prevention and treatment HCV mediation, about 250 milligrams every kilogram (" mg/kg ") body weight of about 0.01-every day, preferably the dosage level of the application's disclosure compound of the about 100mg/kg body weight of about 0.05-every day is typical.Typically, the pharmaceutical composition of the application's disclosure will give for about 5 times with about 1-every day, and perhaps the mode as continuous infusion gives.Such administration can be used as chronic or acute therapy.Can will change according to discharge rate, the course for the treatment of and patient's age, sex, body weight and the physical appearance of the seriousness of the disease of being treated, the state of an illness, administration number of times, route of administration, compound used therefor with the amount of the activeconstituents of manufacture order one formulation with solid support material combination.Preferred unit dose formulations is to contain per daily dose or the sub-doses above-mentioned as this paper, or the activeconstituents of its suitable divided dose.Treatment can begin with the low dose of the dose,optimum that is significantly less than described compound.After this, dosage increases according to little increment, until the best effect that reaches under this situation.In general, the optimal of described compound is that antiviral effective result will be provided usually, and do not cause the concentration level of any harmful or poisonous side effect.
When the composition of the application's disclosure comprises the combination of the compound of the application's disclosure and one or more other therapeutical agents or preventive, compound and other therapeutical agent or preventive exist with the dosage level between about 10 to 150% usually, and more preferably give with the dosage between about 10 to 80% in the monotherapy scheme usually.
Can be suitable for giving pharmaceutical preparation with any suitable approach, for example by oral (comprising cheek or sublingual administration), rectum, intranasal, part (comprising cheek, hypogloeeis or transdermal), vagina or parenteral (comprise in subcutaneous, intracutaneous, intramuscular, intraarticular, the synovial membrane chamber, in the breastbone, in the sheath, intralesional (intralesional), intravenously or intradermal injection or infusion) approach.Can pass through the known any method of pharmacy field, for example by activeconstituents and carrier or mixed with excipients are prepared such preparation.
The pharmaceutical preparation that is applicable to orally give can be used as discrete unit existence, as capsule or tablet; Powder or granule; Solution in moisture or on-aqueous liquid or suspensoid; Edible foaming agent or sweet food agent (whips); Or oil-in-water liquid emulsion or water-in-oil emulsion.
For example, for the oral administration with tablet or capsule form, active pharmaceutical ingredient can mix with oral, atoxic pharmaceutical acceptable inert carriers such as ethanol, glycerine, water etc.Powder prepares by compound powder being broken into suitable fine powder size and mixing with the pharmaceutical carrier of pulverizing similarly (as edible carbohydrate, for example, starch or N.F,USP MANNITOL).Seasonings, sanitas, dispersion agent and tinting material also can exist.
By as above describing the preparation powder mixture and it be filled in the gelatin shell capsule of shaping, the preparation capsule.Also can before stuffing operation, glidant and lubricant such as colloid silica, talcum powder, Magnesium Stearate, calcium stearate or solid polyethylene glycol be added in the powdered mixture.Also can add disintegrating agent or solubilizing agent such as agar-agar, calcium carbonate or yellow soda ash to improve the drug utilization degree after capsule is taken in.
And, when wanting or need, also suitable tackiness agent, lubricant, disintegrating agent and tinting material can be mixed in the mixture.Suitable tackiness agent comprises starch, gelatin, natural sugar such as glucose or beta lactose, corn sweetener, natural and synthetic gum such as Sudan Gum-arabic, tragacanth gum or sodiun alginate, carboxy methyl cellulose, polyoxyethylene glycol etc.The lubricant that is used for these formulations comprises sodium oleate, sodium-chlor etc.Disintegrating agent includes, but are not limited to starch, methylcellulose gum, agar, wilkinite, xanthan gum etc.For example, by the preparation powdered mixture, granulation or make fritter (slugging) is added lubricant and disintegrating agent and is pressed into tablet, the preparation tablet.By the compound that will suitably pulverize and thinner or as described above matrix mix, and optional and tackiness agent such as carboxy methyl cellulose, alginate, jelling agent or polyvinylpyrrolidone, solution retarding agent (retardant) such as paraffin, absorb accelerator such as quaternary ammonium salt and/or absorption agent such as wilkinite, kaolin or Lin Suanergai again and mix the preparation powdered mixture.Can pass through the solution wetted powdered mixture with tackiness agent such as syrup, starch paste, Acadia's mucus (acadia mucilage) or Mierocrystalline cellulose or polymer raw material, and be pressed through the screen cloth granulation.Another kind as granulation is selected, and powdered mixture can be passed through tabletting machine, and with the broken granulating of the piece of imperfect formation.Can adopt the method for adding stearic acid, stearate, talcum powder or vegetables oil that particle is lubricated in case tablet sticks on the tablet shaping punch die.Then lubricated mixture is pressed into tablet.Also the application's disclosure compound can be mixed directly being pressed into tablet with free-pouring inert support, and the step that need not to carry out granulation or make fritter.Dressing and the wax polishing dressing of transparent or opaque protective layer, sugar or the polymer raw be made up of the sealing ply of shellac can be provided.Dyestuff can be added in these dressings to distinguish different unitary doses.
Can be prepared as oral fluid agent such as solution, syrup and the elixir of dosage unit form, like this, the amount of giving contains the compound of predetermined amount.By with compound dissolution in the aqueous solution that suitable process flavoring is handled, can prepare syrup, and by the avirulent solvent of use, can prepare elixir.Isooctadecanol and the polyoxyethylene sorbitol ether that also can add solubilizing agent and emulsifying agent such as ethoxylation, sanitas, flavoring additive such as spearmint oil or natural sweeteners or asccharin or other artificial sweetner etc.
When suitable, the dosage unit preparations of orally give can be by microencapsulation.Preparation also can be by for example with the special material dressing in polymkeric substance, the wax etc. or be embedded in and wherein be prepared into prolonged action preparation or sustained release preparation.
Formula (I) compound and pharmacologically acceptable salt thereof also can give with the form of liposome transfer system, as small unilamellar vesicle (vesicles), big unilamellar liposome and multilamellar liposome.Can form liposome from multiple phosphatide such as cholesterol, octadecylamine or phosphatidylcholine.
Formula (I) compound and pharmacologically acceptable salt thereof also can be by the single carrier transfer of use monoclonal antibody conduct with this compound molecule coupling.But compound also can with as the soluble polymer coupling of target medicine carrier.Such polymkeric substance can comprise polyvinylpyrrolidone, pyran co-polymer, poly-hydroxypropyl Methacrylamide phenol, poly-hydroxyethyl l-asparagine phenol or the polyethylene oxide polylysine that is replaced by palmityl (palitoyl) residue.In addition, compound can be coupled to a class Biodegradable polymeric, be used for realizing that controlled delivery of pharmaceutical agents discharges, for example, the segmented copolymer crosslinked or amphiphilic of poly(lactic acid), poly-epsilon-caprolactone (polepsilon caprolactone), poly-hydroxyl fourth (butyric) acid, poe, polyacetal, poly-dihydropyrane, polybutylcyanoacrylate and hydrogel.
The medicinal preparations that is suitable for transdermal administration can be rendered as the paster agent of dispersion, and the time that it is intended to prolong keeps contacting with recipient's the close of epidermis.For example, can be with activeconstituents from paster by at Pharmaceutical Research 1986,3 (6), the iontophoresis transmission of describing usually in 318.
The medicinal preparations that is suitable for topical can be mixed with ointment, emulsifiable paste, suspension agent, lotion, pulvis, solution, paste, gelifying agent, sprays, aerosol or finish.
For treating eyes or other outside organization for example mouth and skin, preparation preferably applies with the form of topical ointment or emulsion.When being mixed with ointment, activeconstituents can use with paraffinic ointment base or the mixable ointment base of water.Perhaps, activeconstituents can be formulated in the emulsion with oil-in-water emulsion matrix or water-in-oil matrix.
The pharmaceutical preparation that is suitable for the eye topical comprises eye drops, and wherein activeconstituents dissolves or is suspended in the suitable carriers, particularly aqueous solvent.
Be suitable for that the pharmaceutical preparation of topical comprises lozenge, pastille and gargle in the oral cavity.
The pharmaceutical preparation that is suitable for rectal administration can present with the form of suppository or enema.
Be suitable for the pharmaceutical preparation of nose administration, wherein carrier is solid, comprise that particle diameter for example is thick (course) powder of 20-500 micron, it carries out administration in the mode of wherein taking snuffing, namely by sucking by nasal passage fast from the close powder container of nose.The appropriate formulation that is used for wherein carrier with the form administration of nasal spray or nasal drop and is liquid comprises the aqueous solution or the oil solution of activeconstituents.
Be suitable for comprising particle dust or smog by the pharmaceutical preparation of inhalation that it can form by means of aerosol metering, the dosage supercharging, atomizer or the insufflator of various types.
The pharmaceutical preparation that is suitable for vagina administration can present with the form of pesseulum, cotton balls, emulsion, gelifying agent, paste, foam or sprays preparation.
The pharmaceutical preparation that is suitable for administered parenterally comprises moisture and anhydrous aseptic injection liquid, and it can comprise antioxidant, buffer reagent, fungistat and soutes, and it can make the blood isoosmotic pressure of preparation and the acceptor that is intended to; With moisture and anhydrous aseptic suspension, it can comprise suspension agent and thickening material.In the metering of the described preparation unit of may reside in or the many measuring containers, for example in Mi Feng ampoule or the bottle, and can be stored under lyophilize (freeze-drying) condition, only need to add aseptic liquid vehicle, for example water for injection, and then use at once.At once injection solution and suspension can be by sterilized powder, particle and tablet preparation.
Should be understood that except the above-mentioned composition of mentioning especially, described preparation can also comprise other medicament of this area routine, attention be the kind of the preparation discussed, for example be suitable for oral those and can comprise sweetener.
Following table 1 is listed some illustrative examples of the compound that can use with the compound of the application's disclosure.The compound of the application's disclosure can be used in conjoint therapy jointly or separately with other whose anti-HCV active compound, or uses by these compounds are combined into composition.
Table 1
Trade(brand)name The physiology classification The type of inhibitor or target Source company
NIM811 The Cyclophilin inhibitor Novartis
Zadaxin Immunomodulator Sciclone
Suvus Methylenum coeruleum Bioenvision
Actilon(CPG10101) The TLR9 agonist Coley
Batabulin(T67) Anticancer The 'beta '-tubulin inhibitor Tularik Inc.,South San Francisco, CA
ISIS 14803 Antiviral Antisense ISIS Pharmaceuticals Inc,Carlsbad, CA/Elan Phamaceuticals Inc.,New York,NY
Summetrel Antiviral Antiviral Endo Pharmaceuticals Holdings Inc.,Chadds Ford,PA
GS-9132(ACH-806) Antiviral The HCV inhibitor Achillion/Gilead
Pyrazolopyrimidine compound and salt, from WO-2005047288, on May 26th, 2005 Antiviral The HCV inhibitor Arrow Therapeutics Ltd.
Levovirin Antiviral The IMPDH inhibitor Ribapharm Inc.,Costa Mesa,CA
Merimepodib (VX-497) Antiviral The IMPDH inhibitor Vertex Pharmaceuticals Inc., Cambridge,MA
XTL-6865 (XTL-002) Antiviral Monoclonal antibody XTL Biopharmaceuticals Ltd., Rehovot,Isreal
Telaprevir (VX-950,LY-570310) Antiviral The NS3 serpin Vertex Pharmaceuticals Inc., Cambridge, MA/Eli Lilly and Co. Inc., Indianapolis, IN
HCV-796 Antiviral NS5B replicative enzyme inhibitor Wyeth/Viropharma
NM-283 Antiviral NS5B replicative enzyme inhibitor Idenix/Novartis
GL-59728 Antiviral NS5B replicative enzyme inhibitor Gene Labs/Novartis
GL-60667 Antiviral NS5B replicative enzyme inhibitor Gene Labs/Novartis
2’CMeA Antiviral NS5B replicative enzyme inhibitor Gilead
PSI 6130 Antiviral NS5B replicative enzyme inhibitor Roche
R1626 Antiviral NS5B replicative enzyme inhibitor Roche
2 ' C methyladenosine Antiviral NS5B replicative enzyme inhibitor Merck
JTK-003 Antiviral The RdRp inhibitor Japan Tobacco Inc.,Tokyo,Japan
Levovirin Antiviral Ribavirin ICN Pharmaceuticals,Costa Mesa, CA
Ribavirin Antiviral Ribavirin Schering-Plough Corporation, Kenilworth,NJ
Trade(brand)name The physiology classification The type of inhibitor or target Source company
Viramidine Antiviral Prodrugs of ribavirin with Ribapharm Inc.,Costa Mesa,CA
Heptazyme Antiviral Ribozyme Ribozyme Pharmaceuticals Inc., Boulder,CO
BILN-2061 Antiviral Serpin Boehringer Ingelheim Pharma KG, Ingelheim,Germany
SCH 503034 Antiviral Serpin Schering Plough
Zadazim Immunomodulator Immunomodulator SciClone Pharmaceuticals Inc.,San Mateo,CA
Ceplene Immunomodulator Immunomodulator Maxim Pharmaceuticals Inc.,San Diego,CA
CellCept Immunosuppressor HCV IgG immunosuppressor F.Hoffmann-La Roche LTD, Basel,Switzerland
Civacir Immunosuppressor HCV IgG immunosuppressor Nabi Biopharmaceuticals Inc.,Boca Raton,FL
Albuferon-α Interferon, rabbit Albumin IFN-α 2b Human Genome Sciences Inc., Rockville,MD
InfergenA Interferon, rabbit IFN alfacon-1 InterMune Pharmaceuticals Inc., Brisbane,CA
Omega IFN Interferon, rabbit IFN-ω Intarcia Therapeutics
IFN-β and EMZ701 Interferon, rabbit IFN-β and EMZ701 Transition Therapeutics Inc., Ontario,Canada
Rebif Interferon, rabbit IFN-β1a Serono,Geneva,Switzerland
RoferonA Interferon, rabbit IFN-α2a F.Hoffmann-La Roche LTD, Basel,Switzerland
IntronA Interferon, rabbit IFN-α2b Schering-Plough Corporation, Kenilworth,NJ
Intron A and Zadaxin Interferon, rabbit IFN-α 2b/ α 1-thymosin RegeneRx Biopharmiceuticals Inc., Bethesda,MD/ SciClone Pharmaceuticals Inc,San Mateo,CA
Rebetron Interferon, rabbit IFN-α 2b/ ribavirin Schering-Plough Corporation, Kenilworth,NJ
Actimmune Interferon, rabbit INF-γ InterMune Inc.,Brisbane,CA
Interferon-beta Interferon, rabbit Interferon-beta-1a Serono
Multiferon Interferon, rabbit Long-acting IFN Viragen/Valentis
Wellferon Interferon, rabbit Lymphoblastoid IFN-α n1 GlaxoSmithKline plc,Uxbridge, UK
Trade(brand)name The physiology classification The type of inhibitor or target Source company
Omiferon Interferon, rabbit Natural IFN-α Viragen Inc.,Plantation,FL
Pegasys Interferon, rabbit Pegylation IFN-α 2a F.Hoffmann-La Roche LTD, Basel,Switzerland
Pegasys and Ceplene Interferon, rabbit Pegylation IFN-α 2a/ immunomodulator Maxim Pharmaceuticals Inc.,San Diego,CA
Pegasys and ribavirin Interferon, rabbit Pegylation IFN-α 2a/ ribavirin F.Hoffmann-La Roche LTD, Basel,Switzerland
PEG-Intron Interferon, rabbit Pegylation IFN-α 2b Schering-Plough Corporation, Kenilworth,NJ
The PEG-Intron/ ribavirin Interferon, rabbit Pegylation IFN-α 2b/ ribavirin Schering-Plough Corporation, Kenilworth,NJ
IP-501 Protect the liver Anti-fibrosis Indevus Pharmaceuticals Inc., Lexington,MA
IDN-6556 Protect the liver The cysteine proteinase enzyme inhibitors Idun Pharmaceuticals Inc.,San Diego,CA
ITMN-191(R-7227) Antiviral Serpin InterMune Pharmaceuticals Inc., Brisbane,CA
GL-59728 Antiviral NS5B replicative enzyme inhibitor Genelabs
ANA-971 Antiviral The TLR-7 agonist Anadys
TMC-465350 Antiviral Serpin Medivir/ Tibotec
The application's disclosure compound also can be used as laboratory reagent.Compound can be the instrument that the analysis of research and design virus replication, checking animal test system and structure biology research are provided, with the mechanism of the HCV disease of further promoting mutual understanding.Have, the application's disclosure compound for example is used for again, and establishes or measure the binding site of other antiviral compound by competitive inhibition.
The application's disclosure compound also can be used for treating or prevents the virus of material to pollute, and therefore reduce the risk of laboratory or the medical worker who contacts with these materials or patient's virus infection, described material has for example blood, tissue, instruments and outside surface, laboratory apparatus and outside surface and blood collecting or blood transfusion instrument and material.
The application's disclosure is intended to comprise by the synthetic method preparation or by comprising that those occur in human or animal body (in the body) or at the compound with formula (I) of the metabolic approach preparation of external processing.
Used abbreviation in this application, particularly including the abbreviation in following illustrative reaction scheme and embodiment, for those skilled in the art well-known.Some abbreviations are used as follows: CDI represents 1,1 '-carbonyl dimidazoles; THF represents tetrahydrofuran (THF); DBU represents 1,8-diazabicylo [5.4.0], 11 carbon-7-alkene; TFA represents trifluoroacetic acid; HATU represents O-(7-azepine benzo triazol-1-yl)-N, N, N ', N '-tetramethyl-urea phosphoric acid salt; PyBOP represents benzotriazole-1-base-oxygen base-three-pyrrolidino ,-Phosphonium hexafluorophosphates; MeI represents methyl-iodide; Boc or BOC represent tert-butoxycarbonyl; OtBu represents tert.-butoxy; TBME represents t-butyl methyl ether; Et3N represents triethylamine; DMSO represents methyl-sulphoxide; OAc represents acetic ester; DPPA represents the diphenylphosphine acylazide; Me represents methyl; TBAF represents tetrabutylammonium; DMAP represents 4-N, the N-dimethyl aminopyridine; TBuLi represents tert-butyl lithium; LiHMDS represents hexamethyldisilazane base lithium; Tle represents tertiary butyl leucine, also is called tertiary butyl glycine; 4-BiphMgBr represents bromination 4-xenyl magnesium; DCM represents methylene dichloride; MeO represents methoxyl group; EDAC or EDC represent 1-(3-dimethylaminopropyl)-3-ethyl-carbodiimide hydrochloride; And HOBt represents I-hydroxybenzotriazole.
The raw material that can be used for the compound of synthetic the application's disclosure is well known by persons skilled in the art and can easily makes maybe and can buy.
Provide following method to be illustrated as purpose, they are not to limit the application's scope of the disclosure required for protection.Recognize, may preparation wherein use the GPF (General Protection False base to protect the compound of functional group, remove protecting group then so that the compound of the application's disclosure to be provided.Be well known by persons skilled in the art about the details according to the application of the protecting group of the application's disclosure.
Scheme II has shown general method, and its Chinese style (I) compound is by preparing tripeptides carboxylic acid and the coupling of P1` sulphonamide.Described linked reaction need for example be used for example carbonyl dimidazoles processing carboxylic acid (1) of coupling reagent among the THF at solvent, described solvent can be heated to backflow, then at solvent for example in THF or the methylene dichloride, at alkali for example in the presence of the DBU, the derivative of (1) that forms is added in the P1` sulphonamide.
Scheme II
Figure G2007800499569D00241
The other method that has shown preparation formula (I) compound among the scheme II I.Use the method that adopts among the reaction scheme I with P1` sulphonamide composition and the coupling of P1 composition.Then can be with gained P1-P1` part at its N-terminal deprotection.In this general example, adopt the Boc protecting group, but those skilled in the art will recognize that, can adopt multiple suitable amino protecting group in the method.Can solvent for example use in the ethylene dichloride acid for example trifluoroacetic acid remove the Boc protecting group, with the amine that obtains deprotection tfa salt for example.This TFA amine salt can be directly used in linked reaction subsequently, perhaps, can at first this TFA amine salt be changed into the HCl amine salt, and with this HCl amine salt be used for as shown in scheme II I as described in linked reaction.Can be at solvent for example in the methylene dichloride, use coupling reagent for example HATU with this HCl amine salt (3) and the coupling of C-terminal P4-P3-P2 intermediate, with acquisition formula (I) (4) compound.
Scheme II I
Figure G2007800499569D00242
Formula (I) compound
The other method that has shown preparation formula (I) compound among the reaction scheme IV.Wherein be at alkali for example in the presence of the diisopropylamine, and at solvent for example in the methylene dichloride, use coupling reagent for example PyBOP with the free carboxy coupling of hydrochloride and the P2 composition of P1-P1` terminal amine (1).Can be in two step method gained P2-P1-P1` intermediate be transformed an accepted way of doing sth (I) compound, wherein first step be solvent for example use in the methylene dichloride acid for example TFA with the terminal deprotection of P2 amine.Can be at alkali for example in the presence of the diisopropylamine, and use solvent for example in the methylene dichloride, use coupling reagent for example PyBOP with the C-terminal coupling of gained trifluoroacetate and P4-P3 composition, with acquisition formula (I) (4) compound.
Reaction scheme IV
Figure G2007800499569D00251
Formula (I) compound
The P4-P3-P2 intermediate that uses in the superincumbent reaction scheme can be prepared according to the method for former description, has shown further describing of relevant this method among the reaction scheme V in general manner.Wherein, can the free carboxy of P4-P3 intermediate (1) is terminal with the N-terminal coupling of P2 composition to obtain P4-P3-P2 dipeptides (2).By can be with the C-terminal deprotection of this P4-P3-P2 intermediate with the ester group saponification, to obtain the P4-P3-P2 of free carboxy acid (3) form.Can use method described herein with intermediate for example (3) transform an accepted way of doing sth (I) compound.
Reaction scheme V
Figure G2007800499569D00261
Can as described herein formula (I) compound be changed into other formulas (I) compound.An example that has shown such method among the reaction scheme VI, formula (I) (1) compound that wherein will carry the Boc group in the P4 position transforms an accepted way of doing sth (I) (3) compound, and wherein said compound carries urea groups in the P4 position.(1) can in two step method, carry out to the conversion of (3), wherein first step be by solvent for example in the methylene dichloride with acid for example TFA handle (1) (1) changed into amine (2).Can be in the presence of monovalent alkali, with for example tert-butyl isocyanate processing of isocyanic ester, with acquisition formula (I) (3) compound, wherein the P3 part is added cap (capped) by urea with gained amine tfa salt.As mentioned above, those skilled in the art will recognize that intermediate (2) can come preparation formula (I) compound as raw material, wherein the P3 group is added cap by acid amides or sulphonamide or thiocarbamide or SULFAMIDE.The preparation of described formula (I) compound can be used for the standard conditions that formed described P4 functional group by amine and realize.
Reaction scheme VI
Figure G2007800499569D00262
Formula (I) compound
Figure G2007800499569D00271
Formula (I) compound
In the preparation of formula (I) compound, use a kind of above-described general method with the terminal intramolecularly of introducing of P1`, below this is explained in more detail.In some instances, the P1` composition is cycloalkyl-or alkyl-sulphonamide, its can be commercially available or by corresponding alkyl-or cycloalkyl-SULPHURYL CHLORIDE by making with the described SULPHURYL CHLORIDE of ammonia treatment.Perhaps, these sulphonamide can use the general method of describing among the reaction scheme VII to synthesize.Wherein be the sulphonamide that commercially available 3-chloro-sulfonyl propyl chlorine (1) is changed into suitable protection, for example by handling to realize with tert-butylamine.Then by solvent for example among the THF with the alkali of 2 equivalents for example butyllithium handle gained sulphonamide (2) changed into corresponding naphthene sulfamide amine.Can pass through with acid treatment with gained naphthene sulfamide amine deprotection, to obtain the naphthene sulfamide amine of desirable deprotection.
Reaction scheme VII
Figure G2007800499569D00272
Can also use the modification of aforesaid method that the naphthene sulfamide amine that replaces is incorporated in formula (I) compound.For example, can with the alkali of intermediate 2 usefulness 2 equivalents of reaction scheme VIII for example butyllithium handle, and can with the gained reaction mixture with electrophilic reagent for example methyl-iodide handle, with the naphthene sulfamide amine (3) that obtains to replace.Can be with this intermediate (3) at the terminal deprotection of N-, and gained compound (4) is used as intermediate in the preparation of formula (I) compound.
Reaction scheme VIII
Figure G2007800499569D00281
In some cases, the P1` intermediate that adopts in formula (I) compound is derived from sulfamide derivatives.Under these circumstances, the SULFAMIDE intermediate can pass through several route of synthesis, and for example the route of synthesis of describing in reaction scheme IX obtains.
Reaction scheme IX
Figure G2007800499569D00282
Sulfamic acid chloride (2) can make by the following method in position: water (for example 1 equivalent) is added at solvent for example in the chloro sulfonyl isocyanate 1 (for example 1 equivalent) in the THF, remain on low temperature for example-20 ℃ under, and allow gained solution be warmed to 0 ℃.In this solution, add for example anhydrous triethylamine (for example 1 equivalent) of alkali, add amine (for example 1 equivalent) then.Then this reaction mixture is warmed to room temperature, filters and filtrate is concentrated, obtained required SULFAMIDE (3).
Can be according to the route of synthesis of describing among the reaction scheme X with in SULFAMIDE drawing-in system (I) compound.Wherein with carboxylic acid P1 composition (1) with activator for example CDI handle.In independent flask, highly basic is added in the solution of above-mentioned SULFAMIDE, and the gained reaction mixture was stirred several hours, then this reaction mixture is added in the flask of the carboxylic acid that contains activation, to obtain acyl amino sulfone amide derivative (2).Can as described herein intermediate for example 2 be transformed an accepted way of doing sth (I) compound.
Reaction scheme X
Figure G2007800499569D00291
Should be noted that the acyl amino sulfone amide derivative can also be made by the tripeptides carboxylic acid in the one step process described in reaction scheme XI.
Reaction scheme XI
Figure G2007800499569D00292
The P1 composition commercially available acquisition of in formula (I) compound, adopting in some cases, but also can use well known by persons skilled in the art, and the method for describing in this article on unrestricted meaning is synthesized, and uses method described herein to be incorporated in formula (I) compound then.The P1 cyclopropyl amino acid that replaces can synthesize according to the general method of describing among the reaction scheme XII.
In the presence of alkali, with 1,4-dihalo butylene (2) is handled, and obtains the imines (3) that generates with imines (1) commercially available or that synthesize easily.Then (3) are used acid hydrolysis, obtain (4) as principal product, it has the allyl group substituting group that is cis with carboxyl.Can use the Boc group with the amine moiety protection of (4), to obtain the amino acid (5) of protection fully.This intermediate is racemic modification, can it be split by enzyme method, wherein the ester moiety of (5) by protease cracking to obtain corresponding carboxylic acid.Do not want to be entangled in particular theory, but it is believed that this reaction is optionally, an enantiomorph provides the kinetic resolution of this intermediate racemic modification with the speed responsing more faster than its enantiotropy.In an embodiment of the example that this paper mentions, the steric isomer that is used for being incorporated in formula (I) compound is (5a), and it has (1R, 2S) stereochemistry.In the presence of enzyme, this enantiomorph does not experience the ester cracking, therefore this enantiomorph (5a) is reclaimed from reaction mixture.Yet, have (1S, 2R) stereochemical another enantiomorph (5b) can experience the ester cracking, i.e. and hydrolysis is to obtain free acid (6).After this reaction is finished, can for example water extracting process or chromatography be separated ester (5a) from acid product (6) by ordinary method.
Reaction scheme XII
Figure G2007800499569D00301
The method for preparing P2 intermediate and formula (I) compound is shown in the following reaction scheme.Described intermediate, reaction conditions and the method that provides in a particular embodiment is widely used in having the compound of other substitution pattern.For example, the synthetic of the P2 composition that exists in formula (I) compound of reaction scheme XIII can prepare according to defined route of synthesis.Make being easy to or commercially available N-Boc-4-oxo-L-proline(Pro) or N-Cbz-4-oxo-L-proline(Pro) with organometallic reagent Grignard reagent (perhaps alkyl or aryl lithium reagent for example, or alkyl or aryl zincon) handle to obtain intermediate (2), wherein R is carried in the C4 position of proline(Pro) 3Substituting group and free tertiary hydroxyl.The alcohol functional group of intermediate (2) can be carried out then functionalized, to obtain required R 8Functional group.In the method, the alcohol of intermediate (2) can be carried out the good reaction of setting up in a series of this areas.For example, can be with the pure acidylate of (2) to obtain ester, carbamate or carbonic ether; Alkylation is with acquisition ether, and phosphorylation is to obtain phosphoric acid ester.For intermediate (2) being changed into the intermediate (3) of reaction scheme XIII, the hydroxy-acid group protection of (2) shown in may needing at first to incite somebody to action.
The chemical descriptor of relevant carbinol-functionalization is in exercise question is for example following standard textbook: Comprehensive Organic Transformations:A Guide to Functional GroupPreparations.Second Addition, Richard Larock.This textbook is published by Wiley andSons.Wherein those skilled in the art can be easy to adopt the concrete reference and the summary that change into intermediate (3) with the intermediate (2) with reaction scheme XIII to highlight.For example, can be referring to the 883-929 page or leaf of Larock ' s textbook about the condition and the relevant references that are formed ether by alcohol.More specifically, the condition of quoting in the 890-894 page or leaf and reference are maximally related with the preparation of implementing the application's disclosure.Equally, can be referring to the 1952-1955 page or leaf of Laroch ' s textbook about the condition that alcohol is changed into the corresponding esters derivative.In addition, the chemistry of describing in Journal of Organic Chemistry 2001, the 66 volume, the 8926th page and the reference wherein quoted can be used for implementing the preparation of the application's disclosure.
Reaction scheme XIII
Figure G2007800499569D00311
Method 1: with P1-P1 ' coupling, then with the P3-P4 coupling
Method 2: with the P3-P4 coupling, then with P1-P1 ' coupling
Figure G2007800499569D00322
Another the synthetic approach that has shown formula (I) compound as shown in reaction scheme XIII among the reaction scheme XIV.It is last and functionalized that the C4 position of proline(Pro) group adds to later stage intermediate (5) via form, to obtain intermediate 6, can transform an accepted way of doing sth (I) compounds with 6 then.Intermediate (5) can obtain via the 4-step order, and by commercially available intermediate (1) beginning, first step comprises employing coupling reagent known in the art, with (1) and the coupling of P1-P1` intermediate.The acid catalyzed deprotection of the N-Boc group of intermediate (2) obtains unhindered amina intermediate (3), then with itself and the coupling of P3-P4 fragment, with acquisition intermediate (4).The selective oxidation of C4 hydroxyl that be used for to obtain the intermediate (4) of intermediate (5) can use oxidising agent for example Dess-Martin reagent realize.
Reaction scheme XIV
The addition that should be noted that the ketone part of organometallic reagent and proline derivative 1 (reaction scheme XVI) is good foundation in the art.For example, Hruby and co-worker (J.Org.Chem.2001,66,3593) have described the addition of phenyl-magnesium-bromide and general formula 1 intermediate (reaction scheme XVI).These discoveries provide evidence to show, when adopting tert-butyl ester group as the protecting group of C2 carboxy moiety, obtain the optimum yields of required 1,2 adduct (2, reaction scheme XVI).In addition, this research provides the evidence of knowing of X-radiocrystallography form, as the stereochemistry result of this addition reaction.Specifically, as above-mentioned and result ketone 1 form addition, obtained a kind of product, wherein the supposition of C4 hydroxyl and C2 carboxyl is cis with respect to the direction of 5 yuan of rings.Determine to infer from this structure, at R 3Face selectivity in M and 1 the ketone addition is α in the structure 1 of reaction scheme XVI.That is to say that organo-metallic optionally adds to the reverse side (following) of the carbonyl in 1, the stereochemical corresponding tertiary alcohol (2) shown in obtaining to have.
Reaction scheme XVI
Figure G2007800499569D00341
The job description of above-mentioned Hruby the addition (reaction scheme XVI) of specific format reagent and 1 derivative.Yet, in the application's disclosure illustrated the addition of a lot of grignard reagents and proline(Pro) 1.Description comprises that the document of addition of the organometallic reagent of grignard reagent and ketone is quite a lot of, and in this area, be summarized in the general summary, for example: ComprehensiveOrganic Functional Group Transformations.Volume 2:Synthesis:Carbon with one heteroatom attached by a single bond.Editor in ChiefAlan.R.Katritzky, et al.1995.Chapter 2.02, p 37.This class reaction also is described in Comprehensive Organic Synthesis.Editor in Chief Barry M Trost, among Volume 1:Additions to C-X pi-bonds (part 1) .1991.
Nearest research in this area provides the further optimized conditions of the addition reaction of relevant grignard reagent and ketone, and these researchs can be used for the application.For example, Ishihara and co-worker (Org.Lett.2005, Vol.7, No.4,573) have described formation and the purposes of magnesium ate complex recently.Magnesium ate complex R 3MgLi is derived from grignard reagent and lithium alkylide.As described in Ishihara, these title complexs provide in the reaction the good productive rate with 1,2 adduct of ketone.In an independent research, Knochel and co-worker (Angew.Chem Int.Ed.2006,45,497) have described for example LnCl of solubility lanthanon salt 3Combinational use with organomagnesium reagent.Exist these lanthanon salt to make the efficient with 1,2 addition reaction of carbonyl compound improve.These papers and the reference of wherein quoting have been set up this area at the prior art state aspect the optimization of the grignard reaction of the simple addition of carbonyl compound, and as the important source of information among the application.
Shall also be noted that multiple organometallic reagent participates in and the addition reaction of ketone.Comprise reagent for example lithium aryl, lithium alkylide and heteroaryl lithium reagent in this research, they add in the carbonyl moiety in 1,2 mode as everyone knows.For example, in Dondoni and co-worker's nearest paper (J.Org.Chem.2005,70,9257), use BuLi with the benzothiazole lithiumation, and gained C2-lithium material add on the lactone in 1,2 mode.Similarly, estimate that the lithiumation benzothiazole will add on the ketone 1 of reaction scheme XVI in 1,2 mode, to obtain for example 2a of intermediate.
Those skilled in the art will recognize that, also can participate in 1,2 addition with ketone 1 derived from the organometallic reagent of heterocycle Li such as oxazole and thiazole and imidazoles.Have lot of documents to describe to be used for each unique conditions of these heterocyclic ring systems, and this information is that those skilled in the art are easy to obtain.For example, with the addition reaction of ketone in, use the organometallic reagent derived from benzoxazole Huo oxazole need use magnesium acid lithium (lithium magnesate).The particular content of Bayh and co-worker's nearest paper is described in J.Org.Chem., in 2005,70,5190.The addition of the ketone 1 of benzoxazole and reaction scheme XVI provides for example approach of 2b of intermediate that obtains.
For using multiple organometallic reagent and addition ketone derived from heterocycle, an important document precedent is arranged.For example, Behinda and co-worker's paper (Tet.Lett.42,2001,647) described the lithiumation benzoglyoxaline formation and with the addition of simple lactone.Similarly, with the addition reaction of the ketone 1 of reaction scheme XVI in, use this lithiumation benzoglyoxaline to provide and obtain for example approach of 2c of intermediate.In addition, Kawasaki and co-worker's nearest paper (Bioorganic and Medicinal Chem.Lett.13,2003,87) described a series of lithiumation heteroaromaticss formation and with the addition reaction of activating terephthalamide amine.Similarly, with the addition reaction of the ketone 1 of reaction scheme XVI in, use these lithiumation heteroaromatic intermediates that the approach that obtains intermediate 2d-2k will be provided.
With 1,2 addition reaction of ketone 1 in, use also relevant with the application derived from the organometallic reagent of biaryl or heteroaryl-aryl system.The addition of such organometallic reagent and ketone 1 will provide for example approach of 2l and 2m of intermediate that obtains.Should be noted that in the illustrating of the application, may need synthetic biaryl or heteroaryl-aryl organometallic reagent to come to be used for subsequently and the addition of the ketone 1 of reaction scheme XVI.Those skilled in the art will recognize that have important literature to describe preparation this type organometallic reagent and precursor thereof.For example, Chinchilla and co-worker's nearest paper (Chem.Rev.2004,104,2667) has been described preparation of metallization heterogeneous ring compound and uses thereof.Basic chemistry for the preparation of biaryl or heteroaryl-aryl often adopts Suzuki sample linked reaction.The document description of Gregory Fu state and the subclass of following document: the JACS 2004,126,1340 of this area aspect such linked reaction; JACS, 2002,124,13662; Angew.Chem.Int.Ed.2002,41, No.11,1945; Angew.Chem.Int.Ed.2002,41, No.20,3910; JACS 2002,122, and 4020; JACS 2001,123, and 10099; Org.Lett.2001, Vol.3, No.26,4295; Angew.Chem.Int.Ed.1998,37, No.24,3387.In addition, the key of this achievement summary is easy to obtain in this area, for example derives from Rossi in Synthesis 2004, No.15,2419.
Embodiment
Present disclosure is now described in conjunction with some embodiment, and described embodiment is not intended to limit its scope.On the contrary, present disclosure covers all as being included in alternatives, variant and the Equivalent in the claim scope.Thereby, following examples will illustrate a kind of practice of present disclosure, and the purpose that is understood that these embodiment is that to illustrate presenting of some embodiment and they be to be considered to the most useful of its method and conceptual aspect and the content of understandable description in order to provide.
Solution per-cent has been represented weight-volume relationship, and the solution ratio has been represented volume-volume relationship, except as otherwise noted.Nucleus magnetic resonance (NMR) spectrum Bruker300,400 or the 500MHz spectrograph record; Chemical shift (δ) is reported with ppm.According to Still ' s flash chromatography technology (J.Org.Chem.1978,43,2923) at silica gel (SiO 2) on carry out flash chromatography.
The intermediate of describing in following examples can be used for synthesis type 1 compound.
Embodiment 1. preparation P1` intermediates
1. prepare the cyclopropyl sulphonamide
Method 1:
Figure G2007800499569D00371
The cyclopropyl sulphonamide
Reaction scheme 1
Step 1:
(3.0mol 315mL) is dissolved among the THF (2.5L) with tert-butylamine.This solution is cooled to-20 ℃.Add lentamente 3-chloropropane SULPHURYL CHLORIDE (1.5mol, 182mL).This reaction mixture is warmed to room temperature, and stirred 24 hours.This mixture is filtered, and with the filtrate vacuum concentration.Resistates is dissolved in CH 2Cl 2(2.0L).Gained solution with 1.0M HCl (1.0L), water (1.0L) and salt solution (1.0L) washing, is used Na 2SO 4Drying is filtered, and vacuum concentration, has obtained light yellow solid, with its crystallization from hexane, has obtained product, is white solid (316.0g, 99%). 1H NMR(CDCl 3)δ1.38(s,9H),2.30-2.27(m,2H),3.22(t,J=7.35Hz,2H),3.68(t,J=6.2Hz,2H),4.35(b,1H)。
Step 2:
-78 ℃ of products to step 1 (2.14g, 10.0mmol) add in the solution in THF (100mL) n-BuLi (solution of 2.5M in hexane, 8.0mL, 20.0mmol).With this reaction mixture via being warmed to room temperature and vacuum concentration in 1 hour.Resistates is distributed between ethyl acetate and water (each 200mL).With the organic phase salt water washing that separates, use Na 2SO 4Drying is filtered, and vacuum concentration.With resistates recrystallization from hexane, obtained required product, be white solid (1.0g, 56%). 1H NMR(CDCl 3)δ0.98-1.00(m,2H),1.18-1.19(m,2H),1.39(s,9H),2.48-2.51(m,1H),4.19(b,1H)。
Step 3:
(110g, 0.62mmol) solution in TFA (500mL) was stirring at room 16 hours with the product of step 2.Vacuum is removed volatile matter.With resistates recrystallization from ethyl acetate/hexane (60mL/240mL), obtained required product, be white solid (68.5g, 91%). 1H NMR(DMSO-d 6)δ0.84-0.88(m,2H),0.95-0.98(m,2H),2.41-2.58(m,1H),6.56(b,2H)。
Method 2:
Blast gaseous ammonia to being cooled in 0 ℃ the solution of 100mL THF, up to reaching capacity.In this solution, add the solution of 5g (28.45mmol) cyclopropyl SULPHURYL CHLORIDE (available from Array Biopharma) in 50mL THF, this solution is warming up to ambient temperature overnight, and restir 1 day.Enriched mixture is applied to the SiO of 30g up to the solvent residue of 1-2ml 2Filler (with 30%-60% ethyl acetate/hexane wash-out) obtains the cyclopropyl sulphonamide of 3.45g (100%), white solid. 1H NMR (methyl alcohol-d 4) δ 0.94-1.07 (m, 4H), 2.52-2.60 (m, 1H); 13C NMR (methyl alcohol-d 4) δ 5.92,33.01.
2. prepare the cyclopropyl sulphonamide that C1-replaces
2a. the preparation N-tertiary butyl-(1-methyl) cyclopropyl-sulphonamide
Figure G2007800499569D00391
Step 1: the preparation N-tertiary butyl-(3-chlorine) sulfonyl propyl amine
Figure G2007800499569D00392
Prepare as mentioned above.
Step 2: the preparation N-tertiary butyl-(1-methyl) cyclopropyl-sulphonamide
(4.3g, 20mmol) solution is dissolved in and does among the THF (100mL) and be cooled to-78 ℃ with the product of step 1.In this solution, add n-Butyl Lithium (2.5M is in hexane for 17.6mL, 44mmol) at leisure.Remove the dry ice bath, in 1.5 hours, make reaction mixture be warming up to room temperature.This mixture is cooled to-78 ℃ then, adds the solution (2.5M is in hexane for 20mmol, 8mL) of n-Butyl Lithium.Reaction mixture is warming up to room temperature, is cooled to-78 ℃ in 2 hours again, and the pure solution of interpolation methyl-iodide (5.68g, 40mmol).Make reaction mixture be warming up to ambient temperature overnight, at room temperature use saturated NH then 4Cl (100ml) stopped reaction.Extract with ethyl acetate (100ml).Organic phase is washed with salt solution (100ml), dry (MgSO 4), filtering, vacuum concentration obtains yellow oil, and its crystallization and obtain product from hexane is light yellow solid (3.1g, 81%): 1H NMR (CDCl 3) δ 0.79 (m, 2H), 1.36 (s, 9H), 1.52 (m, 2H), 1.62 (s, 3H), 4.10 (br s, 1H).
Step 3: preparation 1-methyl cyclopropyl sulphonamide
Figure G2007800499569D00401
With the product of step 2 (1.91g, 10mmol) solution is dissolved among the TFA (30mL), at room temperature stirred reaction mixture is 16 hours.Desolventizing in a vacuum obtains yellow oil, its from ethyl acetate/hexane (1: 4,40mL) in crystallization, and obtain required product, be white solid (1.25g, 96%): 1H NMR (CDCl 3) δ 0.84 (m, 2H), 1.41 (m, 2H), 1.58 (s, 3H), 4.65 (br s, 2H).C 4H 9NO 2The analytical calculation value of S: C, 35.54; H, 6.71; N, 10.36.Measured value: C, 35.67; H, 6.80; N, 10.40.
2b. preparation 1-propyl group cyclopropyl sulphonamide
Use the method for the described 1-of preparation methyl cyclopropyl sulphonamide to prepare described compound, difference is to use propyl group halogenide for methyl-iodide in second step of this method.
2c. preparation 1-allyl group cyclopropyl sulphonamide
Figure G2007800499569D00403
Step 1: the preparation N-tertiary butyl-(1-allyl group) cyclopropyl sulphonamide
Figure G2007800499569D00404
Obtain described compound according to the method described in the N-tertiary butyl-(1-methyl) cyclopropyl sulphonamide synthetic, yield is 97%, and difference is that the allyl bromide 98 of 1.25 equivalents is used as electrophilic reagent.This compound is directly used in next reaction and need not to purify: 1H NMR (CDCl 3) δ 0.83 (m, 2H), 1.34 (s, 9H), 1.37 (m, 2H), 2.64 (d, J=7.3Hz, 2H), 4.25 (br s, 1H), 5.07-5.10 (m, 2H), 6.70-6.85 (m, 1H).
Step 2: preparation 1-allyl group cyclopropyl sulphonamide
Figure G2007800499569D00411
According to the method described in 1-methyl cyclopropyl sulphonamide synthetic, obtain this compound 1-allyl group cyclopropyl sulphonamide from the product of step 1, yield is 40%.Use 2% ethanol/methylene as eluent at SiO 2Go up by column chromatography purification compound: 1H NMR (CDCl 3) δ 0.88 (m, 2H), 1.37 (m, 2H), 2.66 (d, J=7.0Hz, 2H), 4.80 (s, 2H), 5.16 (m, 2H), 5.82 (m, 1H); 13C NMR (CDCl 3) δ 11.2,35.6,40.7,1190,1336.
2d. preparation 1-benzyl rings sulfonyl propyl amine
Figure G2007800499569D00412
Step 1: the preparation N-tertiary butyl-(1-benzyl) cyclopropyl-sulphonamide.
Figure G2007800499569D00413
Use the method described in the N-tertiary butyl-(1-methyl) cyclopropyl sulphonamide synthetic to obtain described compound, yield is 60%, and difference is to use the bromotoluene of 1.05 equivalents, develops with 10% ethyl acetate/hexane then: 1H NMR (CDCl 3) δ 0.92 (m, 2H), 1.36 (m, 2H), 1.43 (s, 9H), 3.25 (s, 2H), 4.62 (br s, 1H), 7.29-7.36 (m, 5H).
Step 2: preparation 1-benzyl rings sulfonyl propyl amine
Figure G2007800499569D00421
Use the method described in 1-methyl cyclopropyl sulphonamide synthetic, obtain described compound by the N-tertiary butyl (1-benzyl) cyclopropyl sulphonamide, yield is 66%, comes recrystallization by 10% ethyl acetate/hexane of minimum quantity subsequently: 1H NMR (CDCl 3) δ 0.90 (m, 2H), 1.42 (m, 2H), 3.25 (s, 2H), 4.05 (s, 2H), 7.29 (m, 3H), 7.34 (m, 2H); 13C NMR (CDCl 3) δ 11.1,36.8,41.9,127.4,128.8,129.9,136.5.
2e. preparation 1-(1-cyclohexenyl) cyclopropyl-sulphonamide
Figure G2007800499569D00422
Step 1: the preparation N-tertiary butyl-[1-(1-hydroxyl) cyclohexyl]-cyclopropyl sulphonamide
Figure G2007800499569D00423
Use the synthetic described method of the N-tertiary butyl-(1-methyl) cyclopropyl sulphonamide to obtain this compound, yield is 84%, and difference is, uses the pimelinketone of 1.30 equivalents, comes recrystallization by 20% ethyl acetate/hexane of minimum quantity then: 1H NMR (CDCl 3) δ 1.05 (m, 4H), 1.26 (m, 2H), 1.37 (s, 9H), 1.57-1.59 (m, 6H), 1.97 (m, 2H), 2.87 (br s, 1H), 4.55 (br s, 1H).
Step 2: preparation 1-(1-cyclohexenyl) cyclopropyl-sulphonamide
Use the synthetic described method of 1-methyl cyclopropyl sulphonamide, obtain this compound 1-(1-cyclohexenyl)-cyclopropyl sulphonamide by the N-tertiary butyl-[1-(1-hydroxyl) cyclohexyl]-cyclopropyl sulphonamide, yield is 85%, and ethyl acetate and the hexane by minimum quantity comes recrystallization subsequently: 1H NMR (DMSO-d 6) δ 0.82 (m, 2H), 1.28 (m, 2H), 1.51 (m, 2H), 1.55 (m, 2H), 2.01 (s, 2H), 2.16 (s, 2H), 5.89 (s, 1H), 6.46 (s, 2H); 13C NMR (DMSO-d 6) δ 11.6,21.5,22.3,25.0,27.2,46.9,131.6,132.2; LR-MS (ESI): 200 (M +-1).
2f. preparation 1-benzoyl cyclopropyl sulphonamide
Step 1: the preparation N-tertiary butyl-(1-benzoyl) cyclopropyl-sulphonamide
Figure G2007800499569D00432
Use the synthetic described method of the N-tertiary butyl-(1-methyl) cyclopropyl sulphonamide to obtain this compound, yield is 66%, and difference is that the methyl benzoate of 1.2 equivalents is used as electrophilic reagent.Use the 30%-100% dichloromethane/hexane at SiO 2Go up by column chromatography purification compound: 1H NMR (CDCl 3) δ 1.31 (s, 9H), 1.52 (m, 2H), 1.81 (m, 2H), 4.16 (br s, 1H), 7.46 (m, 2H), 7.57 (m, 1H), 8.05 (d, J=8.5Hz, 2H).
Step 2: preparation 1-benzoyl ring-sulfonyl propyl amine
Use the synthetic described method of 1-methyl cyclopropyl sulphonamide, obtain this compound by the N-tertiary butyl (1-benzoyl) cyclopropyl sulphonamide, yield is 87%, comes recrystallization by the ethyl acetate/hexane of minimum quantity subsequently: 1H NMR (DMSO-d 6) δ 1.39 (m, 2H), 1.61 (m, 2H), 7.22 (s, 2H), 7.53 (t, J=7.6Hz, 2H), 7.65 (t, J=7.6Hz, 1H), 8.06 (d, J=8.2Hz, 2H); 13C NMR (DMSO-d 6) δ 12.3,48.4,128.1,130.0,133.4,135.3,192.0.
2g. the preparation N-tertiary butyl-(1-phenyl amino carboxyl)-cyclopropyl sulphonamide
Figure G2007800499569D00441
Use the synthetic described method of the N-tertiary butyl-(1-methyl) cyclopropyl sulphonamide, use the phenyl isocyanate of 1 equivalent, obtain this compound, yield is 42%, comes recrystallization by the ethyl acetate/hexane of minimum quantity subsequently. 1H NMR(CDCl 3)δ1.38(s,9H),1.67-1.71(m,4H),4.30(br s,1H),7.10(t,J=7.5Hz,1H),7.34(t,J=7.5Hz,2H),7.53(t,J=7.5Hz,2H)。
3. prepare the cyclopropane sulphonamide that C1-replaces: the use of N-Boc protecting group
3a. preparation cyclopropyl sulphonamide t-butyl carbamate is at the cyclopropyl sulphonyl of preparation C1-replacement Key intermediate in the amine
Figure G2007800499569D00442
Step 1: preparation 3-chloropropyl sulphonamide
Figure G2007800499569D00443
(55g 310.7mmol) is dissolved in to be added drop-wise among the THF (200ml) and in 30 minutes and is cooled to 0 ℃ NH with 3-chloropropane chloride solution 4In OH (200ml) solution.Reaction mixture is warming up to room temperature, stirred 1 hour, (4 * 500mL) with the water layer distribution for several times with methylene dichloride.The dichloromethane layer that merges is washed with 1N HCl (150ml), water (150ml), use MgSO 4Drying is filtered vacuum concentration.Recrystallization goes out rough solid in the methylene dichloride from the hexane of minimum quantity, obtains 3-chloropropyl sulphonamide, white solid (45.3g, 93%). 1H NMR(CDCl 3)δ2.34(m,2H),3.32(t,J=7.3Hz,2H),3.70(t,J=6.2Hz,2H),4.83(s,2H); 13C NMR(CDCl 3)δ27.10,42.63,52.57。
Step 2: preparation 3-chloropropyl sulfonyl amine t-butyl carbamate
In 30 minutes, to the 3-chloropropyl sulphonamide (30.2g that is cooled in 0 ℃ the methylene dichloride (350mL), 191.5mmol), triethylamine (30.2mL, 217.0mmol) and 4-DMAP (2.40g, 19.6mmol) tert-Butyl dicarbonate (47.2g, solution 216.9mmol) in the drip dichloromethane (250ml) lentamente in the solution.Make reaction mixture be warming up to room temperature, stirred extra 3 hours, with 1N HCl (300mL), water (300mL) and salt solution (300mL) washing, use MgSO 4Drying is filtered, and vacuum concentration obtains crude product.Develop this material with 5% methylene dichloride in the 70mL hexane, obtain 3-chloropropyl sulfonyl amine t-butyl carbamate, be pale solid (47.2g, 96%): 1H NMR (CDCl 3) δ 1.51 (s, 9H), 2.33 (m, 2H), 3.60 (t, J=7.3Hz, 2H), 3.68 (t, J=6.21Hz, 2H); 13C NMR (CDCl 3) δ 26.50,27.95,42.37,50.40,8476,149.53.
Step 3: preparation cyclopropyl sulfonyl amine t-butyl carbamate
Figure G2007800499569D00452
Under argon gas atmosphere, n-butyllithium solution (1.6M is in hexane for 74.7mL, 119.5mmol) is dissolved in does among the THF (105mL) and be cooled to-78 ℃.In 20-30 minute, in this solution, dropwise add 3-chloropropyl sulfonyl amine t-butyl carbamate (14g, 54.3mmol) solution of doing among the THF (105mL).Remove the dry ice bath, in 2 hours, make reaction mixture be warming up to room temperature.With Glacial acetic acid (3.4mL) cold soaking reaction mixture, vacuum concentration, between methylene dichloride (100mL) and water (100mL), distribute.With salt solution (100mL) washing organic phase, dry (MgSO 4), filtering, vacuum concentration obtains cyclopropyl sulfonyl amine t-butyl carbamate, is the pale solid (12.08g, 100%) of wax shape: 1H NMR (CDCl 3) δ 1.10 (m, 2H), 1.34 (m, 2H), 1.50 (s, 9H), 2.88 (m, 1H), 7.43 (s, 1H). 13C NMR(CDCl 3)δ6.21,28.00,31.13,84.07,149.82。
3b. preparation 1-methoxyl group-methyl cyclopropyl-sulphonamide
Step 1: preparation 1-methoxymethyl cyclopropyl sulfonyl amine t-butyl carbamate
Figure G2007800499569D00461
(1.0g 4.5mmol) in the solution, adds n-Butyl Lithium (1.6M is in hexane for 6.4mL, 10.2mmol), and stirred reaction mixture 1 hour to the cyclopropyl sulfonyl amine t-butyl carbamate that is dissolved in THF (30ml) that is cooled to-78 ℃.(0.40mL, pure solution 5.24mmol) make this mixture slowly be warming up to room temperature in a whole night to add chloromethyl methyl ether in this solution.Use the 1N HCl aqueous solution, the pH value of solution value is adjusted to 3, use ethyl acetate (4 * 50mL partly) to extract then.With the extract drying (MgSO that merges 4), filter, concentrate, obtain 1-methoxymethyl cyclopropyl sulfonyl amine t-butyl carbamate, be the solid (1.20g, 100%) of wax shape, it is directly used in next reaction and need not further purification: 1H NMR (CDCl 3) δ 1.03 (m, 2H), 1.52 (s, 9H), 1.66 (m, 2H), 3.38 (s, 3H), 3.68 (s, 2H), 7.54 (s, 1H); 13C NMR (CDCl 3) δ 11.37,28.29,40.38,58.94,73.43,83.61,149.57.
Step 2: preparation 1-methoxymethyl cyclopropyl sulphonamide
Figure G2007800499569D00462
(1.14g, 4.30mmol) solution is dissolved in 50%TFA/ methylene dichloride (30ml) solution, at room temperature stirs 16 hours with 1-methoxymethyl cyclopropyl sulfonyl amine t-butyl carbamate.Desolventizing in a vacuum is at the SiO of 80g 2On, resistates is carried out stratographic analysis (with 0%-60% ethyl acetate/hexane wash-out), obtain 1-methoxymethyl cyclopropyl sulphonamide, white solid (0.55g, the total of 77%, two step): 1H NMR (CDCl 3) δ 0.95 (m, 2H), 1.44 (m, 2H), 3.36 (s, 3H), 3.65 (s, 2H), 4.85 (s, 2H); 13C NMR (CDCl 3) δ 11.17,40.87,59.23,74.80; LRMS m/z 183 (M ++ NH 4).
3c. preparation 1-cyclopropyl methyl cyclopropyl sulphonamide
Figure G2007800499569D00471
Step 1: preparation 1-cyclopropyl methyl cyclopropyl sulfonyl amine t-butyl carbamate
Figure G2007800499569D00472
According to the method that is described in 1-methoxymethyl cyclopropyl sulfonyl amine t-butyl carbamate synthetic; obtain 1-cyclopropyl methyl cyclopropyl sulfonyl amine t-butyl carbamate; yield is 92%, and difference is that the cyclopropyl monobromomethane of 1.10 equivalents is used as electrophilic reagent.This compound is directly used in next reaction and need not to purify: 1H NMR (CDCl 3) δ 0.10 (m, 2H), 0.51 (m, 2H), 0.67 (m, 1H), 1.10 (m, 2H), 1.49 (s, 9H), 1.62 (m, 2H), 1.87 (d, J=7.0Hz, 2H).
Step 2: preparation 1-cyclopropyl methyl-cyclopropyl sulphonamide
Figure G2007800499569D00473
According to the synthetic described method of 1-methoxymethyl cyclopropyl sulphonamide, obtain this compound from 1-cyclopropyl methyl cyclopropyl sulfonyl amine t-butyl carbamate, yield is 65%.Use the 0%-60% ethyl acetate/hexane as elutriant at SiO 2Go up by column chromatography purification compound: 1H NMR (CDCl 3) δ 0.15 (m, 2H), 0.51 (m, 2H), 1.01 (m, 2H), 1.34 (m, 3H), 1.86 (d, J=7.0Hz, 2H), 4.83 (s, 2H); 13C NMR (CDCl 3) δ 4.65,7.74,11.26,35.62,41.21; LRMS m/z 193 (M ++ NH 4).
3d. preparation 1-propyl group formamyl cyclopropane-sulphonamide
Figure G2007800499569D00481
Step 1: preparation 1-propyl group formamyl cyclopropane sulphonamide t-butyl carbamate
Figure G2007800499569D00482
Synthetic described method according to the 1-methoxymethyl cyclopropyl sulfonyl amine tertiary butyl-carbamate has obtained this compound, and the crude product yield is 100%, and difference is that the propyl isocyanate of 1.10 equivalents is used as electrophilic reagent.This compound is directly used in next reaction and need not to purify: 1H NMR (CDCl 3) δ 0.10 (m, 2H), 0.51 (m, 2H), 0.67 (m, 1H), 1.10 (m, 2H), 1.49 (s, 9H), 1.62 (m, 2H), 1.87 (d, J=7.0Hz, 2H).
Step 2: preparation 1-propyl group formamyl cyclopropane-sulphonamide
Figure G2007800499569D00483
Synthetic described method according to 1-methoxymethyl cyclopropyl sulphonamide; obtain this compound from 1-propyl group formamyl cyclopropane sulphonamide t-butyl carbamate; the yield of optimizing is 50%; difference is because recrystallization goes out this material from the dichloromethane/hexane of minimum quantity, so do not use chromatography: 1H NMR (CDCl 3) δ 0.15 (m, 2H), 0.51 (m, 2H), 1.01 (m, 2H), 1.34 (m, 3H), 1.86 (d, J=7.0Hz, 2H), 4.83 (s, 2H); 13C NMR (CDCl 3) δ 4.65,7.74,11.26,35.62,41.21; LRMS m/z 193 (M ++ NH 4).
3e. preparation 1-(3,5-dimethyl isoxazole-4-yl) formamyl cyclopropane sulphonamide
Figure G2007800499569D00491
Step 1: preparation 1-(3,5-dimethyl isoxazole-4-yl) formamyl cyclopropane sulphonamide amino T-butyl formate
Figure G2007800499569D00492
Synthetic described method according to 1-methoxymethyl cyclopropyl sulfonyl amine t-butyl carbamate; obtained this compound; the crude product yield is 100%, and difference is 3 of 1.20 equivalents, and 5-dimethyl isoxazole-4-isocyanic ester is used as electrophilic reagent.This compound is directly used in next reaction and need not to purify.
Step 2: preparation 1-(3,5-dimethyl isoxazole-4-yl) formamyl cyclopropane sulphonamide
Use the 4N HCl/ dioxane of 30mL (120mmol) from the 1-(3 of 1.62g (4.52mmol); 5-dimethyl isoxazole-4-yl) carbamyl basic ring-propane sulphonamide t-butyl carbamate obtains this compound; yield is 50% (580mg); stirring is spent the night; concentrate, compose purify (with 0%-5% ethanol/methylene wash-out) in the enterprising circumstances in which people get things ready for a trip of Biotage 40M post: 1H NMR (methyl alcohol-d 4) δ 1.57 (m, 2H), 1.61 (m 2H), 2.15 (s, 3H), 2.30 (s, 3H), 4.84 (s, 3H); 13C NMR (methyl alcohol-d 4) δ 9.65,10.94,15.01,46.11,114.82,159.45,165.55,168.15; LRMS m/z 260 (M ++ H).
4. prepare naphthene sulfamide amine from the cycloalkyl bromine
4a. prepare the cyclobutyl sulphonamide from the cyclobutyl bromine
Figure G2007800499569D00501
Add 1.7M tert-butyl lithium/pentane of 44mL (74.8mmol) in the cyclobutyl bromine solutions that is cooled to the 5.0g (37.0mmol) in-78 ℃ the anhydrous diethyl ether (ether) of 30mL, in 1.5 hours, solution slowly is warming up to-35 ℃.This mixture is slowly added to through conduit in the chloride solution of up-to-date distillation of the 5.0g (37.0mmol) in the 100ml hexane that is cooled to-40 ℃, in 1 hour, be warming up to 0 ℃ and carry out vacuum concentration carefully.This mixture is dissolved in the ether again, with some ice cold water washings once, dry (MgSO 4), filter, concentrate carefully.This mixture is dissolved among the THF of 20mL again, is added drop-wise to the saturated NH among the THF of 500mL 3, and its stirring is spent the night.In a vacuum this mixture is condensed into rough yellow solid, drips methyl alcohol with its recrystallization from the dichloromethane/hexane of minimum quantity with 1-2, and obtain the cyclobutyl sulphonamide of 1.90g (38%), white solid. 1H NMR(CDCl 3)δ1.95-2.06(m,2H),2.30-2.54(m,4H),3.86(p,J=8Hz,1H),4.75(brs,2H); 13C NMR(CDCl 3)δ16.43,23.93,56.29。C 4H 8NSO 2HRMSm/z (M-H) -Calculated value be 134.0276, measured value is 134.0282.
4b. preparation cyclopentyl sulphonamide
Figure G2007800499569D00502
The 2M chlorination cyclopentyl magnesium solution of 18.5mL in the ether (37.0mmol) is added drop-wise in SULPHURYL CHLORIDE (available from the Aldrich) solution of the up-to-date distillation of 3.0mL (37.0mmol) in the 100ml hexane that is cooled to-78 ℃.In 1 hour, mixture is warming up to 0 ℃, carries out vacuum concentration then carefully.This mixture is dissolved in the ether (200ml) again, with some ice cold waters (200ml) washing once, dry (MgSO 4), filter, concentrate carefully.This mixture is dissolved among the THF of 35mL again, is added drop-wise to the saturated NH among the THF of 500mL 3, and its stirring is spent the night.Concentrate this mixture in a vacuum to rough yellow solid, use 70% ethyl acetate-hexane as elutriant, resistates is filtered the silica gel of 50g, concentrate this solution then.Drip methyl alcohol with resistates recrystallization from the dichloromethane/hexane of minimum quantity with 1-2, and obtain the cyclopentyl sulphonamide of 2.49g (41%), white solid. 1H NMR(CDCl 3)δ1.58-1.72(m,2H),1.74-1.88(m,2H),1.94-2.14(m,4H),3.48-3.59(m,1H),4.80(br s,2H); 13C NMR(CDCl 3)δ25.90,28.33,63.54;MS m/e 148(M-H) -
4c. preparation cyclohexyl sulfonamide
2M chlorination cyclohexyl magnesium (TCI Americas) drips of solution of 18.5mL in the ether (37.0mmol) is added to and is cooled in the chloride solution of the up-to-date distillation of 3.0mL (37.0mmol) in-78 ℃ the 100ml hexane.In 1 hour, mixture is warming up to 0 ℃, carries out vacuum concentration then carefully.This mixture is dissolved in the ether (200ml) again, with some ice cold waters (200ml) washing once, dry (MgSO 4), filter, concentrate carefully.This mixture is dissolved among the THF of 35mL again, is added drop-wise to the saturated NH among the THF of 500mL 3, and its stirring is spent the night.Concentrate this mixture in a vacuum to rough yellow solid, use 70% ethyl acetate-hexane as elutriant, resistates is filtered the silica gel of 50g, concentrate then.Drip methyl alcohol with resistates recrystallization from the dichloromethane/hexane of minimum quantity with 1-2, and obtain the cyclohexyl-sulphonamide of 1.66g (30%), white solid. 1H NMR(CDCl 3)δ1.11-1.37(m,3H),1.43-1.56(m,2H),1.67-1.76(m,1H),1.86-1.96(m,2H),2.18-2.28(m,2H),2.91(tt,J=12,3.5Hz,1H),4.70(br s,2H); 13C NMR(CDCl 3)δ25.04,25.04,26.56,62.74;MS m/e 162(M-1) -
4d. preparation neo-pentyl sulphonamide
Figure G2007800499569D00512
According to the method for preparing cyclohexyl sulfonamide, the 0.75M neo-pentyl magnesium chloride (Alfa) in the ether of 49mL (37mmol) is converted into the neo-pentyl sulphonamide of 1.52g (27%), white solid. 1H NMR(CDCl 3)δ1.17(s,9H),3.12(s,2H),4.74(brs,2H); 13CNMR(CDCl 3)δ29.46,31.51,67.38;MS m/e 150(M-1) -
4e. preparation cyclobutylmethyl sulphonamide
Figure G2007800499569D00521
The acetone soln of sodium iodide/150mL of the cyclobutylmethyl bromine (Aldrich) of 12.3g (83mmol) and 13.7g (91mmol) is refluxed spend the night, be cooled to room temperature then.Leach inoganic solids, under envrionment conditions and in 150 holders, distill out acetone and cyclopropyl methyl-iodide (8.41g, 46%) respectively at 80 ℃.
Anhydrous diethyl ether (ether) solution of cyclobutylmethyl iodine/30mL that is cooled to-78 ℃ 4.0g (21.98mmol) adds in the 1.3M second month in a season-butyllithium/cyclohexane solution of 17mL (21.98mmol) and stirred this solution 5 minutes through conduit.In this mixture, add the hexane solution of the SULPHURYL CHLORIDE/110mL of the up-to-date distillation of 3.0g (21.98mmol) be cooled to-78 ℃ through conduit, mixture is warming up to room temperature in 1 hour, carries out vacuum concentration then carefully.This mixture is dissolved in the ether again, with some ice cold water washings once, dry (MgSO 4), filter, concentrate carefully.This mixture is dissolved among the THF of 30mL again, is added drop-wise to the saturated NH among the THF of 500mL 3, and its stirring is spent the night.In a vacuum this mixture is condensed into rough yellow solid, drips methyl alcohol with its recrystallization from the dichloromethane/hexane of minimum quantity with 1-2, and obtain the cyclobutylmethyl sulphonamide of 1.39g (42%), white solid. 1H NMR(CDCl 3)δ1.81-2.03(m,4H),2.14-2.28(m,2H),2.81-2.92(m,1H),3.22(d,J=7Hz,2H),4.74(brs,2H); 13C NMR(CDCl 3)δ19.10,28.21,30.64,60.93;MS m/e 148(M-1) -
4f. preparation cyclopropyl sulfonyloxy methyl amine
Figure G2007800499569D00522
Use prepares cyclopropyl sulfonyloxy methyl amine (referring to JACS 1981, p.442-445) for the preparation of the method for cyclobutylmethyl sulphonamide by cyclopropyl monobromomethane (Aldrich). 1H NMR(CDCl 3)δ0.39-0.44(m,2H),0.67-0.76(m,2H),1.13-1.27(m,1H),3.03(d,J=7.3Hz,2H),4.74(brs,2H); 13C NMR(CDCl 3)δ4.33,5.61,59.93;MS m/e 134(M-1)。
4g. preparation 2-thiophenesulfonamide
Figure G2007800499569D00531
Use the method for following document to be prepared by 2-thiophene SULPHURYL CHLORIDE (available from Aldrich): JustusLiebigs Ann.Chem., 501,1933, p.174-182.
4h. preparation 4-bromobenzene sulphonamide
Figure G2007800499569D00532
Prepare 4-bromophenyl sulphonamide by handling the commercially available 4-bromo SULPHURYL CHLORIDE that gets with the saturated ammonia among the THF.
5. the general approach for preparing sulphonamide
Figure G2007800499569D00533
Prepare the intermediate sulfamic acid chloride by water (1 equivalent)/THF being added into chloro sulfonyl isocyanate (1 equivalent)/THF solution that cold (20 ℃) stir and making gained solution be warming up to 0 ℃.In this solution, add anhydrous triethylamine (1 equivalent), add essential secondary amine (1 equivalent) subsequently.Reaction mixture is warming up to room temperature, filters then, filtrate is concentrated, obtain desired sulphonamide.
Embodiment 2: preparation P1 intermediate
5.1-tert-butoxycarbonyl amino-cyclopropane formic acid is commercially available getting
Figure G2007800499569D00541
6. prepare the amino cyclobutane formate methyl ester hydrochloride of 1-
(100mg, 0.869mmol) (Tocris) is dissolved in the methyl alcohol of 10mL with the amino cyclobutane formate of 1-.Blasted HCl gas 2 hours.Stirred reaction mixture 18 hours, vacuum concentration then obtains the yellow oil of 144mg.With the ether development of 10mL, obtain the title product of 100mg, white solid. 1H NMR(CDCl 3)δ2.10-2.25(m,1H),2.28-2.42(m,1H),2.64-2.82(m,4H),3.87(s,3H),9.21(br s,3H)。
7a. preparation (1R, 2R)/(1S, 2S) 1-amino-2-ethyl cyclopropane-carboxylic acid tert-butyl ester (mix by racemize Thing)
Figure G2007800499569D00543
Ethyl is cis with respect to carboxyl
Step 1: preparation 2-ethyl cyclopropane-1, the 1-dicarboxylate, as follows
Figure G2007800499569D00544
(92.4g is at 185mL H to the 50%NaOH aqueous solution 2Among the O) in benzyltriethylammonium chloride (21.0g, 92.2mmol) suspension add 1,2-dibromobutane (30.0g, 138.9mmol) and the propanedioic acid di tert butyl carbonate (20.0g, 92.5mmol).At room temperature powerful stirred reaction mixture is 18 hours, adds the mixture of ice and water then.Crude product extracts with methylene dichloride (3 *), water (3 *), salt water washing sequentially, and merge organic extract.Dry (MgSO 4) organic layer, filter and vacuum concentration.The gained residuum carries out fast chromatographic (100g SiO 2, 3% ether/hexane), obtain title product (18.3g, 67.8mmol, 73% yield), it is directly used in next one reaction.
Step 2: prepare racemic 2-ethyl cyclopropane-1, the 1-dioctyl phthalate tert-butyl ester, as follows
Figure G2007800499569D00551
At 0 ℃, (18.3g, (33.55g in suspension 299.0mmol), is H then 67.8mmol) to add potassium tert.-butoxide in the dry diethyl ether (500ml) to the product of step 1 2O (1.35mL, 75.0mmol) and at room temperature spend the night by powerful the stirring.Reaction mixture poured in ice and the mixture of water and with ether (3 *) wash.Extract with 10% Citric acid acidified aqueous solution water layer and with ethyl acetate (3 *) at 0 ℃.The organic layer water (2 *), the salt water washing that merge, dry (MgSO 4), filtering, vacuum concentration obtains title product, is light yellow oil (10g, 46.8mmol, 69% yield).
Step 3: preparation (1R, 2R)/(1S, 2S) 2-ethyl-1-(2-trimethylsilylethoxy) carbonyl ammonia Base) the cyclopropane-carboxylic acid tert-butyl ester is as follows
Figure G2007800499569D00552
To the product of step 2 (10g, 46.8mmol) and the activation recently of 3g
Figure G2007800499569D00553
In the suspension of molecular sieve/dry benzene (160mL), add triethylamine (7.50mL, 53.8mmol) and DPPA (11mL, 10.21mmol).Made reaction mixture refluxed 3.5 hours, (13.5mL 94.2mmol), spends the night reaction mixture refluxed to add 2-trimethyl silyl-ethanol then.Filter reaction mixture is with the ether dilution, with the 10% Citric acid aqueous solution, water, saturated NaHCO 3The aqueous solution, water (2 *), salt solution (2 *) washing, dry (MgSO 4), filter and vacuum concentration.The Aldrich polymeric polyisocyanate scavenger resin of resistates by 10g is suspended in the methylene dichloride of 120mL, and stirring is at room temperature spent the night and is filtered, and obtains title product (8g, 24.3mmol; 52%) light yellow oil: 1H NMR (CDCl 3) δ 0.03 (s, 9H), 0.97 (m, 5H), 1.20 (br m, 1H), 1.45 (s, 9H), 1.40-1.70 (m, 4H), 4.16 (m, 2H), 5.30 (brs, 1H).
Step 4: preparation (1R, 2R)/(1S, 2S) 1-amino-2-ethyl cyclopropane-carboxylic acid tert-butyl ester (mix by racemize Compound), as follows
Ethyl is cis with respect to carboxyl
(3g, (9.3mL, 9.3mmol) the 1.0MTBAF solution in, mixture are heated and refluxed 1.5 hours, are cooled to room temperature, then with the ethyl acetate dilution of 500mL 9mmol) to add THF to the product of step 3.Water (2 * 100mL) and salt solution (2 * 100mL) washing solns, dry (MgSO continuously 4), filter, and vacuum concentration, required product obtained.
7b. it is full accordingly shown in changing into to be used for carrying substituent formula 1 compound of P1 vinyl P1 General method with P 1 analogue
Figure G2007800499569D00562
P1 vinyl substituted base P1 ethyl substituting group
The compd A compd B
With compd A (approximately 100mg) and Pt (S)/C (5%, 10mg) the suspension hydrogenation in about 15mLEtOAc, H 2(30PSI) 0.5 hour.After the filtration of Ceilite plug, filtrate is concentrated and purifying, obtained required product, compd B.
The preparation racemize (1R, 2S)/(1S, 2R)-1-amino-2-vinyl cyclopropane-carboxylic acid ethyl ester:
Figure G2007800499569D00571
Step 1:
(304g 2.16mol) is suspended in the t-butyl methyl ether (1.6L) with glycine ethyl ester hydrochloride.Add phenyl aldehyde (231g, 2.16mol) and anhydrous sodium sulphate (154.6g, 1.09mol) and use ice-water-bath cooling mixture to 0 ℃.(455mL 3.26mol) and at room temperature stirred the mixture 48 hours to drip triethylamine in 30 minutes.By adding ice cold water (1L) stopped reaction and separating organic layer.With t-butyl methyl ether (0.5L) aqueous phase extracted and use saturated NaHCO 3The organic phase that the mixture washing of the aqueous solution (1L) and salt solution (1L) merges.Organic layer MgSO 4Drying is filtered, vacuum concentration, and the expectation product of acquisition 392.4g is goldenrod oil, it is directly used in next step. 1H NMR(CDCl 3,300MHz)δ1.32(t,J=7.1Hz,3H),4.24(q,J=7.1Hz,2H),4.41(d,J=1.1Hz,2H),7.39-7.47(m,3H),7.78-7.81(m,2H),8.31(s,1H)。
Step 2:
In 60 minutes, to trimethyl carbinol lithium (84.1g, 1.05mol)/drip in the suspension of dry toluene (1.2L) glycine ethyl ester N-benzyl imines (100g, 0.526mol) and anti-form-1,4-two bromo-2-butylene (107g, 0.500mol)/mixture of dry toluene (0.6L).After interpolation was finished, (TBME 1L) handled the scarlet mixture by adding water (1L) and t-butyl methyl ether.Water phase separated and extract again with TBME (1L).Merge organic phase, add 1.0M HCl (1L), at room temperature stirred the mixture 2 hours.Separate the extraction of organic phase and water (0.8L).Merge water then, saturated with salt (700g), add TBME (1L), mixture is cooled to 0 ℃.By dripping 10.0M NaOH stirred mixture is alkalized to pH value 14 then, separate organic layer, with TBME (2 * 500mL) aqueous phase extracted.With the organic extract drying (MgSO that merges 4), filter, concentrate, to volume be 1L.(131g 0.600mol) and at room temperature stirred the mixture 4 days to add tert-Butyl dicarbonate in this solution.(50g 0.23mol) adds in the reaction, and mixture was refluxed 3 hours, makes it be cooled to ambient temperature overnight then with extra tert-Butyl dicarbonate.Reaction mixture MgSO 4Drying is filtered, vacuum concentration, the raw material of acquisition 80g.By flash chromatography this resistates (SiO of 2.5Kg that purifies 2, use 1%-2%CH 3OH/CH 2Cl 2Wash-out), acquisition 57g (53%) racemize N-Boc-(1R, 2S)/(1S, 2R)-1-amino-2-vinyl cyclopropane-carboxylic acid ethyl ester, be yellow oil, when being placed in the refrigerator, it solidifies. 1H NMR(CDCl 3,300MHz)δ1.26(t,J=7.1Hz,3H),1.46(s,9H),1.43-1.49(m,1H),1.76-1.82(br m,1H),2.14(q,J=8.6Hz,1H),4.18(q,J=7.2Hz,2H),5.12(dd J=10.3,1.7Hz,1H),5.25(br s,1H),5.29(dd,J=17.6,1.7Hz,1H),5.77(ddd,J=17.6,10.3,8.9Hz,1H);MS m/z 254.16(M-1)。
9. split N-Boc-(1R, 2S)/(1S, 2R)-1-amino-2-vinyl cyclopropane-carboxylic acid ethyl ester
Figure G2007800499569D00581
Split A
To the aqueous solution (0.1M that is arranged on 12 liters of sodium phosphate buffer agents in the band plug reactor, 4.25L, the pH value is 8) in (its be maintained at 39 ℃ and under 300rpm, stir), add the Alcalase 2.4L (about 425mL) (Novozymes North America Inc.) of 511 grams.When the temperature of mixture reached 39 ℃, the pH value was adjusted to 8.0, realized by adding the 50%NaOH aqueous solution.Racemic N-Boc-among the DMSO of 850mL (1R, 2S)/(1S, 2R)-solution of 1-amino-2-vinyl cyclopropane-carboxylic acid ethyl ester was added in 40 minutes then.Temperature of reaction is maintained at 40 ℃ then and reaches 24.5 hours,, uses the 50%NaOH aqueous solution at the time point place of 1.5 hours and 19.5 hours the pH value of mixture to be adjusted to 8.0 therebetween.24.5 after hour, the enantiomeric excess of measuring ester is 97.2%, reaction is cooled to room temperature (26 ℃) and stirs and spends the night (16 hours), the enantiomeric excess of measuring ester thereafter is 100%.With the pH value to 8.5 of 50%NaOH conditioned reaction mixture, the gained mixture extracts (2 * 2L) with MTBE then.Then with the MTBE extract 5%NaHCO that merges 3(3 * 100mL), water (3 * 100mL) washing and vacuum concentration, obtain enantiomer-pure N-Boc-(1R, 2S)/-1-amino-2-vinyl cyclopropane-carboxylic acid ethyl ester, be light yellow solid (42.55g; Purity: 97%210nm does not contain acid; 100% enantiomer excessive (" ee ").
Use 50%H then from the water layer that extraction process obtains 2SO 4Being acidified to the pH value is 2, and (2 * 2L) extract with MTBE.(3 * 100mL) washings and concentrated obtain acid, are light yellow solid (42.74g with MTBE extract water then; Purity: 99%210nm does not contain ester).
Figure G2007800499569D00591
1R, 2S-ester 1S, 2R-acid
Figure G2007800499569D00592
Figure G2007800499569D00601
Split B
Hole (capacity: Savinase 16.0L (from the proteolytic enzyme of Bacillus clausii) (Novozymes North America Inc.) and the racemize N-Boc-(1R in the DMSO of 0.1mL of 0.5mL 100mM HepsNa buffer reagent (pH value 8.5) the interpolation 0.1mL the 10mL/ hole) to 24 orifice plates, 2S)/(1S, 2R)-solution of 1-amino-2-vinyl cyclopropane-carboxylic acid ethyl ester (10mg).This plate is sealed and under 40 ℃, cultivating under the 250rpm.After 18 hours, the enantiomeric excess of following measurement ester is 44.3%: remove the reaction mixture of 0.1mL and fully mix with 1mL ethanol; After centrifugal, analyze the supernatant liquid of 10 microlitres (" μ L ") with chirality HPLC.Add the DMSO of 0.1mL in the remaining reaction mixture, under 250rpm, cultivated this plate again 3 days at 40 ℃, add the ethanol of 4mL then in the hole.After centrifugal, the enantiomeric excess of analyzing the supernatant liquid of 10 μ L and measuring ester with chirality HPLC is 100%.
Split C
Hole (capacity: the Esperase 8.0L (from the proteolytic enzyme of Bacillus halodurans) (Novozymes North America Inc.) and the racemize N-Boc-(1R in the DMSO of 0.1mL that add 0.1mL in the 0.5mL 100mM HepsNa buffer reagent (pH value 8.5) the 10mL/ hole) to 24 orifice plates, 2S)/(1S, 2R)-solution of 1-amino-2-vinyl cyclopropane-carboxylic acid ethyl ester (10mg).This plate is sealed and under 40 ℃, cultivating under the 250rpm.After 18 hours, the enantiomeric excess of following measurement ester is 39.6%: remove the reaction mixture of 0.1mL and fully mix with 1mL ethanol; After centrifugal, analyze the supernatant liquid of 10 μ L with chirality HPLC.Add the DMSO of 0.1mL in the remaining reaction mixture, under 250rpm, cultivated this plate again 3 days at 40 ℃, add the ethanol of 4mL then in the hole.After centrifugal, the enantiomeric excess of analyzing the supernatant liquid of 10 μ L and measuring ester with chirality HPLC is 100%.
Carry out sample analysis as follows:
1) specimen preparation: the reaction mixture of about 0.5mL fully mixes with 10 volume of ethanol.After centrifugal, the supernatant liquid of 10 μ L is expelled on the HPLC post.
2) transformation efficiency is measured:
Post: YMC ODS A, 4.6 * 50mm, S-5 μ m
Solvent: A, the 1mM HCl aqueous solution; B, acetonitrile;
Gradient: 30%B, 1 minute; 30%-45%B, 0.5 minute; 45%B, 1.5 minutes; The B of 45%-30%, 0.5 minute.
Flow velocity: 2mL/ minute
UV detects: 210nm
Retention time: acid, 1.2 minutes; Ester, 2.8 minutes.
3) enantiomeric excess of ester is measured:
Post: CHIRACEL OD-RH, 4.6 * 150mm, S-5 μ m
The HClO of moving phase: acetonitrile/50mM 4The aqueous solution (67/33)
Flow velocity: 0.75mL/ minute.
UV detects: 210nm.
Retention time: (1S, 2R)-1-amino-2-vinyl cyclopropane-carboxylic acid, 5.2 minutes;
Racemic modification (1R, 2S)/(1S, 2R)-1-amino-2-vinyl cyclopropane-carboxylic acid ethyl ester, 18.5 minutes and 20.0 minutes;
(1R, 2S)-1-amino-2-vinyl cyclopropane-carboxylic acid ethyl ester, 18.5 minutes.
Split D
The 0.3M sodium phosphate buffer agent of 5L (the pH value is 8) stirs under 130rpm in the reactor of 38 ℃ of band plugs that are maintained at 20L.4 liters Alcalase 2.4L (Novozymes NorthAmerica Inc.) and 1 liter deionized water are added in the reactor.When the temperature of mixture during close to 38 ℃, use 10N NaOH that the pH value is adjusted to 7.8.In 1 hour via feed hopper, with the racemize N-Boc-among 5 liters of DMSO (1R, 2S)/(1S, 2R)-solution of 1-amino-2-vinyl cyclopropane-carboxylic acid ethyl ester (500 gram) adds in the reactor.Conditioned reaction temperature to 48 ℃ then.After 21 hours, the enantiomeric excess of ester is 99.3%.At 24 hours, stop heating, make sluggish be cooled to room temperature (about 25 ℃) and stir and spend the night.With the pH value to 8.5 of 10N NaOH conditioned reaction mixture, mixture extracts (2 * 4L) with MTBE then.With the MTBE extract 5%NaHCO that merges 3(3 * 400mL), water (3 * 400mL) washings and concentrate, obtain enantiomer-pure N-Boc-(1R, 2S)/-1-amino-2-vinyl cyclopropane-carboxylic acid ethyl ester, be pale yellow crystals (259g; Purity: 96.9%210nm does not contain acid; 100%ee).
Split E
The 0.1M sodium phosphate buffer agent of 10L (the pH value is 8) stirs under 360rpm in 40 ℃ of band plug reactors that are maintained at 20L.1.5L Alcalase 2.4L (Novozymes NorthAmerica Inc.) be added to reactor.When the temperature of mixture during close to 38 ℃, use 10N NaOH that the pH value is adjusted to 8.0.In 1 hour via feed hopper, with the racemic N-Boc-among the 2L DMSO (1R, 2S)/(1S, 2R)-solution of 1-amino-2-vinyl cyclopropane-carboxylic acid ethyl ester (200 gram) adds in the reactor.Conditioned reaction temperature to 40 ℃ then.After 3 hours, regulate pH value to 8.0 with 10N NaOH.After 21 hours, reaction is cooled to 25 ℃.With the pH value to 8.5 of 10N NaOH conditioned reaction mixture, mixture extracts (2 * 5L) with MTBE then.The MTBE extract 5%NaHCO that merges 3(3 * 500mL) and water (3 * 200mL) washings concentrate and obtain the yellow oil of 110g.At room temperature described oil is placed in the clean vacuum (house vacuum) and obtains enantiomer-pure N-Boc-(1R, 2S)/-1-amino-2-vinyl cyclopropane-carboxylic acid ethyl ester, be colourless long rhabdolith (101g; Purity: 97.9%210nm does not contain acid; 100%ee).
The N-Boc-of enantiomer-pure (1R, 2S)/-crystalline structure of 1-amino-2-vinyl cyclopropane-carboxylic acid ethyl ester is by monocrystalline analysis and characterization (X ray NB#:52795-093, identifying code: 634592N1).For want of known chiral centre or bigger atom are not measured absolute configuration.Chain structure along crystalline alpha-axle forms (N...O via intermolecular hydrogen bonding between amide group and carbonylic oxygen atom
Figure G2007800499569D00621
)..
N-Boc-(1R, 2S)-structure of 1-amino-2-vinyl cyclopropane-carboxylic acid ethyl ester:
Figure G2007800499569D00631
The crystal data experimental section:
Molecular formula: C 13H 21N 1O 4 Crystallization
Crystallographic system: orthorhombic crystallization source: MTBE
Spacer: P2 12 12 1Crystal is described: colourless bar-shaped
Figure G2007800499569D00632
α=90 ° crystalline sizes (mm): 0.12 * 0.26 * 0.30
Figure G2007800499569D00633
β=90 ° Data gathering
Figure G2007800499569D00634
γ=90 ° temperature (K): 293
Figure G2007800499569D00635
θ Maximum(°): 65.2 (Cu K α)
Z=4 d x=1.155gcm -3Measured reflection number: 7518
Reflection number for unit cell parameters: 6817 independently reflect number: 2390 (R Int=0.0776)
For the θ scope of unit cell parameters (°): the reflection number (I 〉=2 σ) that 2.2-65.2 is observed: 2284
Uptake factor (mm -1): 0.700 absorbs correction (T Minimum-T Maximum): 0.688-1.000
Split F
The 0.2M sodium borate buffer agent of 5L (the pH value is 9) is in the reactor of 45 ℃ of band plugs that are maintained at 20L and stir under 400rpm.With the deionized water of 3L and the Savinase16L of 4L, EX type (Novozymes North America Inc.) adds in the reactor.When the temperature of mixture during close to 45 ℃, use 10N NaOH that the pH value is adjusted to 8.5.In 40 minutes via feed hopper, with the racemize N-Boc-among the 2L DMSO (1R, 2S)/(1S, 2R)-solution of 1-amino-2-vinyl cyclopropane-carboxylic acid ethyl ester (200 gram) adds in the reactor.Conditioned reaction temperature to 48 ℃ then.After 2 hours, regulate pH value to 9.0 with 10N NaOH.At 18 hours, the enantiomeric excess of ester arrived 72%, pH value and is adjusted to 9.0 with 10N NaOH.At 24 hours, temperature was reduced to 35 ℃.At 42 hours, temperature rose to 48 ℃, and the pH value is adjusted to 9.0 with 10N NaOH.At 48 hours, stop heating, make sluggish be cooled to room temperature (about 25 ℃) and stir and spend the night.At 66 hours, the pH value of reaction mixture was 8.6.With MTBE (2 * 4L) extraction mixtures.With the MTBE extract 5%NaHCO that merges 3(6 * 300mL), water (3 * 300mL) washings and concentrate, obtain enantiomer-pure N-Boc-(1R, 2S)/-1-amino-2-vinyl cyclopropane-carboxylic acid ethyl ester, be pale yellow crystals (101Ag; Purity: 95.9%210nm does not contain acid; 98.6%ee).
The preparation chirality (1R, 2S)-1-amino-2-vinyl cyclopropane-carboxylic acid carbethoxy hydrochloride
At room temperature, at N 2Under the atmosphere, by the 4N HCl/ dioxane (Aldrich) of 200mL, stirring N-Boc-(1R, 2S)-(8.5g 33.3mmol) reaches 3 hours to 1-amino-2-vinyl cyclopropane-carboxylic acid ethyl ester.Desolventizing under reduced pressure keeps temperature to be lower than 40 ℃.This output 6.57g (about 100%) (1R 2S)-1-amino-2-vinyl cyclopropane-carboxylic acid carbethoxy hydrochloride, is faint yellow solid. 1H NMR(300MHz,CD 3OD)δ1.31(t,J=7.0Hz,3H),1.69-1.82(m,2H),2.38(q,J=8.8Hz,1H),4.29(q,J=7.0Hz,2H),5.22(d,J=10.3Hz,1H),5.40(d,J=17.2Hz,1H),5.69-5.81(m,1H)。MS m/z 156(M ++1)。
11. preparation N-Boc-(1R, 2S)-1-amino-2-cyclopropyl rings propane ethyl formate
Figure G2007800499569D00642
With acid chloride (5mg, 0.022mmol) handle N-Boc-in the ether (10ml) (1R, 2S)-1-amino-2-vinyl cyclopropane-carboxylic acid (255mg, solution 1.0mmol).Should be placed on N by orange/red solution 2Under the atmosphere.In 1 hour, drip the diazomethane in the excessive ether.Stirring at room gained solution 18 hours.Use nitrogen gas stream, remove excessive diazomethane.Solution by the concentrated gained of rotary evaporation obtains crude product.Flash chromatography (10% ethyl acetate/hexane) provides 210mg (78%), and (1R 2S)-N-Boc-1-amino-2-cyclopropyl rings propane ethyl formate, is colorless oil.MS m/z 270(M ++H)。
Preparation P1 '-P1 intermediate
12. preparation P1P1 ':
Reaction scheme 1
Figure G2007800499569D00651
The individual isomer that enzyme splits
Figure G2007800499569D00652
Step 1:
To 1 (R)-tert-butoxycarbonyl amino-2 (S)-vinyl cyclopropane-carboxylic acid ethyl ester (3.28g, 13.2mmol)/add in THF (7mL) and methyl alcohol (7mL) solution LiOH (1.27g, 53.0mmol)/suspension of water (14mL).At room temperature stir this mixture overnight, and end with 1NNaOH (15mL) and water (20ml).The gained mixture extracts with 20mL 0.5N NaOH with ethyl acetate (20ml) washing, organic phase.With the water that merges 1N HCl acidifying, be 4 up to the pH value, and (3 * 40ml) extract with ethyl acetate.With the organic extract salt water washing that merges, dry (MgSO 4), filter, concentrate, obtain title compound, white solid (2.62g, 87%). 1H NMR:(DMSO-d 6) δ 1.22-1.26 (m, 1H), 1.37 (s, 9H), 1.50-1.52 (m, 1H), 2.05 (q, J=9Hz, 1H), 5.04 (d, J=10Hz, 1H), 5.22 (d, J=17Hz, 1H), 5.64-5.71 (m, 1H), 7.18,7.53 (s, NH (rotational isomer), 12.4 (brs, 1H)); LC-MS (retention time: 1.67 minutes, method B), MS m/z 228 (M ++ H).
Step 2:
Under nitrogen with the product of step 1 (2.62g, 11.5mmol) and CDI (2.43g, 15.0mmol)/THF (40ml) vlil 50 minutes.Cooling solution to room temperature and by sleeve pipe transfer to the cyclopropyl sulphonamide (1.82g, 15.0mmol)/THF (10ml) solution in.To gained solution add DBU (2.40mL, 16.1mmol) and continue to stir 20 hours.Mixture with 1NHCl end to the pH value be 1 and the THF vacuum concentration.Suspension is with ethyl acetate (2X50mL) extraction and the organic extract that the merges (Na that is dried 2SO 4), filter and concentrate.Recrystallization by hexane-ethyl acetate (1: 1) carries out purifying, obtains title compound (2.4g), white solid.Mother liquor is purified in (wash-out, 9% acetone/methylene dichloride) by Biotage 40S post, obtains second batch of title compound (1.1g).With two batches of merging (total recovery 92%). 1H NMR (DMSO-d 6) δ 0.96-1.10 (m, 4H), 1.22 (dd, J=5.5,9.5Hz, 1H), 1.39 (s, 9H), 1.70 (t, J=5.5Hz, 1H), and 2.19-2.24 (m, 1H), 2.90 (m, 1H), 5.08 (d, J=10Hz, 1H), 5.23 (d, J=17Hz, 1H), 5.45 (m, 1H), 6.85,7.22 (s, NH (rotational isomer); LC-MS (retention time: 1.70 minutes, method B), MS m/z 331 (M ++ H).
Step 3:
The product of stirring at room step 2 (3.5g, 10.6mmol)/methylene dichloride (35mL) and TFA (32mL) solution 1.5 hours.Remove volatile matter in a vacuum and resistates is suspended in the 1NHCl/ ether (20ml) vacuum concentration.Repeat this process once.By pentane development gained mixture and filtration, obtain title compound, be water absorbability pale solid (2.60g, 92%). 1HNMR:(DMSO-d 6) δ 1.01-1.15 (m, 4H), 1.69-1.73 (m, 1H), 1.99-2.02 (m, 1H), 2.38 (q, J=9Hz, 1H), 2.92-2.97 (m, 1H), 5.20 (d, J=11Hz, 1H), 5.33 (d, J=17Hz, 1H), 5.52-5.59 (m, 1H), 9.17 (br s, 3H); LC-MS (retention time: 0.24 minute, method B), MS m/z 231 (M ++ H).
13. preparation P1-P1 ' sulfamide derivatives
Figure G2007800499569D00661
To (1R, 2S) 1-tert-butoxycarbonyl amino-2-vinyl cyclopropane-carboxylic acid (217mg, 1.194mmol)/THF (5ml) solution adds CDI, and (290mg, 1.791mmol), and reaction mixture under refluxad heated 45 minutes.In another round-bottomed flask, LiHMDS (solution of 1.0M in hexane, 2.4mL, 2.4mmol) be added to N-ethylmethylamino sulphonamide (330mg, 2.388mmol)/THF (5ml) solution and stirring at room reaction mixture 1 hour.Two kinds of reaction mixtures were added on together and at room temperature stir 2 hours.Add water with stopped reaction and use the ethyl acetate extraction reaction soln.Separate organic layer and use MgSO 4Dry.Filter and concentrate this solvent, obtain crude product, it purifies to obtain the shielded N-acyl amino of desired N-Boc sulphonamide by preparation property HPLC.Then, compound dissolution in 4N HCl/ dioxane (2mL) solution and when at room temperature stirring 4 hours, is being removed the Boc protecting group.With solution evaporation, obtain hazel oily matter (112mg, 33% yield) with the form of HCl salt. 1H NMR(400Mz,CD 3OD)δ1.16(t,J=7.21Hz,3H),1.68(dd,J=10.03,7.83Hz,1H),2.15(m,1H),2.37(m,1H),2.89(s,3H),3.30(m,2H),5.31(d,J=10.27Hz,1H),5.42(d,J=17.12Hz,3H),5.68(m,1H)。LC-MS (retention time: 0.883 minute), MS m/z 270 (M+Na +).
Embodiment 3: preparation embodiment 3
Figure G2007800499569D00671
Embodiment 3
Figure G2007800499569D00681
Embodiment 3
Step 1:
-50 ℃ to (S)-1-(tert-butoxycarbonyl)-4-oxo-pyrrolidine-2-formic acid (460mg, 2.0mmol) drip in the solution in THF (10mL) bromination 4-xenyl magnesium (0.5M THF, 16.0mL, 8.0mmol).After this temperature stirs 4 hours, with this mixture 5% citric acid treatment, with ethyl acetate (50mL) extraction.With organic layer salt water washing, use MgSO 4Drying is filtered, and concentrates.With residual solid from ethyl acetate: hexane (15mL: recrystallization 15mL), obtained intermediate 1, be white solid (415mg, 54%). 1H NMR(CD 3OD)δ1.49,1.51(d,9H),2.48-2.51(m,1H),2.79-2.82(m,1H),3.75-3.81(m,2H),4.48-4.53(m,1H),7.33-7.36(m,1H),7.43-7.46(m,2H),7.58-7.65(m,6H);MS m/z 384(M ++H)。
Step 2:
To intermediate 1 (383mg, 1.0mmol), cyclopropane sulfonic acid (1-(R)-amino-2-(S)-vinyl cyclopropane carbonyl) amide hydrochloride (293mg, 1.1mmol) and HATU (570mg, 1.5mmol) add in the ice-cold mixture in methylene dichloride (10mL) diisopropylethylamine (560mg, 5.0mmol).Formed solution was warmed to room temperature 4 hours, and vacuum is removed volatile matter.Resistates is developed with ethyl acetate (100mL), and filtered.Filtrate with 5% citric acid (50mL, * 2) and salt water washing, is used MgSO 4Drying is filtered, and concentrates.Resistates with methyl alcohol (4mL) development, has been obtained the required product of 314mg (53%), intermediate 2.MS m/z 596 (M ++ H).
Step 3:
To intermediate 2 (150mg, 0.25mmol) add in the ice-cold suspension in 1, the 4-dioxane (1mL) HCl (the 4M dioxane, 5mL).Stirring at room 2 hours, and vacuum was removed volatile matter, dried overnight under high vacuum with formed solution.Product is directly used in next step.MS m/z 496(M ++H)。
Step 4:
To intermediate 3 (134mg, 0.25mmol) drip in the ice-cold suspension in methylene dichloride (2.5mL) diisopropylethylamine (560mg, 5.0mmol).Add in the formed solution HATU (144mg, 0.38mmol) and (S)-2-(tert-butoxycarbonyl)-3, the 3-acid dimethyl (64mg, 0.28mmol).Final mixture is warmed to ambient temperature overnight, and vacuum is removed volatile matter.Resistates is developed with ethyl acetate (10mL), and filtered.Filtrate with 5% citric acid (10mL, * 2) and salt water washing, is used MgSO 4Drying is filtered, and concentrates.Resistates by preparation HPLC purifying, has been obtained the required product of 16mg (9%), and embodiment 4, are white solid. 1H NMR(CD 3OD)δ1.08-1.13(m,11H),1.27-1.29(m,2H),1.47-1.53(m,10H),1.89-1.93(m,1H),2.26-2.32(m,2H),2.68-2.71(m,1H),2.97-2.99(m,1H),4.05-4.12(m,1H),4.34-4.37(m,2H),4.45-4.47(m,1H),5.15(d,J=12Hz,1H),5.33(d,J=18.5Hz,1H),5.77-5.82(m,1H),7.36-7.37(m,1H),7.44-7.47(m,2H),7.62-7.67(m,6H);MS m/z 709(M ++H)。
Embodiment 4: preparation embodiment 4
Figure G2007800499569D00691
Embodiment 4
Step 1:
This product is the same procedure of describing according in embodiment 3 steps 1, and difference is to use phenyl-magnesium-bromide to make. 1H NMR(DMSO-d 6)δ1.36-1.41(m,9H),2.25-2.28(m,1H),2.60-2.64(m,1H),3.56-3.66(m,2H),4.27-4.29(m,1H),5.50(s,1H),7.25-7.36(m,3H),7.46-7.48(m,2H),12.40(br,1H);MS m/z 308(M ++H)。
Step 2:
This product is the same procedure of describing according in embodiment 3 steps 2, and difference replaces uses the product of embodiment 4 steps 1 to make.MS m/z 520(M ++H)。
Step 3:
Figure G2007800499569D00703
This product is the same procedure of describing according in embodiment 3 steps 3, and difference replaces uses the product of embodiment 4 steps 2 to make.MS m/z 420(M ++H)。
Step 4:
This product is the same procedure of describing according in embodiment 3 steps 4, and difference replaces uses the product of embodiment 4 steps 3 to make. 1H NMR(CD 3OD)δ1.08-1.12(m,11H),1.27-1.28(m,2H),1.48-1.52(m,10H),1.89-1.90(m,1H),2.26-2.32(m,2H),2.68-2.71(m,1H),2.97-2.99(m,1H),4.05-4.09(m,1H),4.34-4.35(m,2H),4.45-4.47(m,1H),5.14(d,J=12Hz,1H),5.34(d,J=18.5Hz,1H),5.77-5.82(m,1H),7.31-7.33(m,1H),7.37-7.40(m,2H),7.57-7.59(m,2H);MS m/z 633(M ++H)。
Embodiment 5: preparation embodiment 5
Figure G2007800499569D00711
Embodiment 5
Step 1:
This product is the same procedure of describing according in embodiment 3 steps 1, and difference replaces use 2-naphthyl magnesium bromide and makes. 1H NMR(CD 3OD)δ1.50,1.52(d,9H),2.54-2.56(m,1H),2.89-2.91(m,1H),3.84-3.87(m,2H),4.51-4.53(m,1H),7.45-7.52(m,2H),7.62-7.64(m,1H),7.85-7.94(m,3H),7.99(s,1H);MS m/z 358(M ++H)。
Step 2:
Figure G2007800499569D00721
This product is the same procedure of describing according in embodiment 3 steps 2, and difference replaces uses the product of embodiment 5 steps 1 to make.MS m/z 570(M ++H)。
Step 3:
Figure G2007800499569D00722
This product is the same procedure of describing according in embodiment 3 steps 3, and difference replaces uses the product of embodiment 5 steps 2 to make.MS m/z 470(M ++H)。
Step 4:
Embodiment 5 is same procedure of describing by in embodiment 3 steps 4, and difference replaces uses the product of embodiment 5 steps 3 to make. 1H NMR(CD 3OD)δ1.05-1.12(m,11H),1.28-1.29(m,2H),1.48-1.53(m,10H),1.91-1.92(m,1H),2.25-2.40(m,2H),2.75-2.79(m,1H),2.97-3.00(m,1H),4.13-4.16(m,1H),4.38-4.47(m,2H),5.14(d,J=12Hz,1H),5.33(d,J=18.5Hz,1H),5.77-5.82(m,1H),7.49-7.51(m,2H),7.70-7.73(m,1H),7.86-7.92(m,3H),8.04(s,1H);MS m/z 683(M ++H)。
Embodiment 6: preparation embodiment 6
Figure G2007800499569D00731
Embodiment 6
Step 1:
Figure G2007800499569D00732
This product is the same procedure of describing according in embodiment 3 steps 1, and difference replaces use 4-fluorophenyl magnesium bromide and makes. 1H NMR(CD 3OD)δ1.48,1.50(d,9H),2.44-2.47(m,1H),2.74-2.95(m,2H),3.69-3.76(m,2H),4.45-4.53(m,1H),7.08-7.12(m,2H),7.52-7.54(m,2H);MS m/z 326(M ++H)。
Step 2:
Figure G2007800499569D00733
This product is the same procedure of describing according in embodiment 3 steps 2, and difference replaces uses the product of embodiment 6 steps 1 to make.MS m/z 538(M ++H)。
Step 3:
Figure G2007800499569D00741
This product is the same procedure of describing according in embodiment 3 steps 3, and difference replaces uses the product of embodiment 6 steps 2 to make.MS m/z 438(M ++H)。
Step 4:
Embodiment 6 is same procedure of describing by in embodiment 3 steps 4, and difference replaces uses the product of embodiment 6 steps 3 to make. 1H NMR(CD 3OD)δ1.02-1.12(m,11H),1.26-1.28(m,2H),1.47-1.52(m,10H),1.89-1.90(m,1H),2.25-2.28(m,2H),2.55-2.65(m,1H),2.92-2.99(m,1H),4.06-4.08(m,1H),4.25-4.33(m,2H),4.41-4.49(m,1H),5.15(d,J=12Hz,1H),5.32(d,J=18.5Hz,1H),5.77-5.82(m,1H),7.09-7.12(m,2H),7.60-7.63(m,2H);MS m/z 651(M ++H)。
Embodiment 7: preparation embodiment 7
Figure G2007800499569D00742
Embodiment 7
Figure G2007800499569D00751
Step 1:
Figure G2007800499569D00752
At 0 ℃ to (2S, 4R)-4-hydroxyl pyrrolidine-2-methyl-formiate HCl salt (5.45g, 30.0mmol), (S)-2-(tert-butoxycarbonyl)-3,3-acid dimethyl (6.93g, 30.0mmol) and HATU (17.1g, 45.0mmol) drip in the slurries in methylene dichloride (100mL) diisopropylethylamine (16.8g, 150mmol).Formed solution in stirred overnight at room temperature, with 5% ice-cold citric acid and 1M NaOH (aq) washed twice, is used the salt water washing then, use MgSO 4Drying, and filter.With the filtrate vacuum concentration, obtained 10.75g (100%), be the light brown foam. 1H NMR(CD 3OD)δ1.04(s,9H),1.46(s,9H),2.01-2.06(m,1H),2.27-2.29(m,1H),3.73(s,3H),3.77-3.87(m,2H),4.31(s,1H),4.49(br,1H),4.56(t,J=8.5Hz,1H)。
Step 2:
Figure G2007800499569D00761
Product (10.75g, 30.0mmol) adding LiOH monohydrate (6.30g, aqueous solution 150mmol) (100mL) in the solution in THF (100mL) and methyl alcohol (100mL) to embodiment 7 steps 1.With final solution in stirred overnight at room temperature.Vacuum is removed volatile matter.Resistates is acidified to pH 2 with 1M HCl (aq).Extract with ethyl acetate (100mL).Organic layer with 5% citric acid and salt water washing, is used MgSO 4Drying, and filter.With the filtrate vacuum concentration, obtained the required product of 8.95g (87%), be the canescence foam. 1H NMR(CD 3OD)δ1.05(s,9H),1.45(s,9H),2.03-2.06(m,1H),2.32-2.36(m,1H),3.79-3.86(m,2H),4.32(br,1H),4.49(br,1H),4.54(t,J=8.5Hz,1H)。
Step 3:
Figure G2007800499569D00762
0 ℃ of product to embodiment 7 steps 2 (1.95g, 5.68mmol) add in the solution in ethyl acetate (150mL) (1R, 2S)-1-amino-N-(cyclopropyl alkylsulfonyl)-2-vinyl cyclopropane carboxamide HCl salt (1.51g, 5.68mmol).This mixture was stirred 5 minutes under this temperature, drip then diisopropylethylamine (1.91g, 17.0mmol).With formed settled solution 0 ℃ of restir 5 minutes, add then EDC (1.41g, 7.38mmol) and HOBt (0.77g, 5.68mmol).With final slurries in stirred overnight at room temperature.Formed settled solution is used 5% ice-cold citric acid, saturated Trisodium Citrate (aq) and salt solution washed twice respectively, use MgSO 4Drying, and filter.With the filtrate vacuum concentration, by quick column purification, with 1: 1 hexane-acetone wash-out, obtained the required product of 2.50g (79%), be white foam shape thing. 1H NMR(CD 3OD)δ1.00-1.10(m,11H),1.24-1.28(m,2H),1.41-1.46(m,10H),1.86-1.91(m,1H),2.00-2.04(m,1H),2.12-2.28(m,1H),2.92-2.99(m,1H),3.80-3.95(m,2H),4.30-4.40(m,2H),4.51(br,1H),5.14(d,J=12Hz,1H),5.35(d,J=18.5Hz,1H),5.77-5.82(m,1H);MS m/z 557(M ++H)。
Step 4:
To Dess Martin reagent (940mg, 2.2mmol) add in the solution in methylene dichloride (20mL) embodiment 7 steps 3 product (556mg, 1.0mmol).With this solution stirring at room 4 hours, and vacuum concentration.Resistates is developed with hot ethyl acetate (10mL), and via diatomite
Figure G2007800499569D00772
Filter.With the filtrate vacuum concentration.Resistates by quick column purification, with 1: 1 hexane-acetone wash-out, is obtained the required product of 550mg (99%), be white foam shape thing. 1H NMR(CD 3OD)δ1.09-1.14(m,11H),1.25-1.28(m,2H),1.43-1.46(m,10H),1.88-1.91(m,1H),2.22-2.28(m,1H),2.51-2.60(m,1H),2.92-2.96(m,1H),4.17-4.34(m,2H),4.77-4.80(m,1H),5.16(d,J=12Hz,1H),5.33(d,J=18.5Hz,1H),5.72-5.82(m,1H);MS m/z555(M ++H)。
Step 5:
-50 ℃ of products to embodiment 7 steps 4 (23mg, 0.05mmol) drip in the solution in THF (0.5mL) 4-p-methoxy-phenyl magnesium bromide (0.5mL, the THF solution of 0.5M, 0.25mmol).Formed solution was stirred 2 hours under this temperature, with ammonium chloride (aq) stopped reaction, use ethyl acetate extraction then.With organic layer salt water washing, use MgSO 4Drying is filtered, and concentrates.Resistates by preparation HPLC purifying, is obtained 4.5mg (14%) embodiment 7, has been white solid. 1H NMR (CD 3OD) δ 1.09-1.12 (m, 11H), 1.26-1.28 (m, 2H), 1.46-1.52 (m, 10H), 1.89-1.91 (m, 1H), 2.25-2.28 (m, 2H), 2.58-2.65 (m, 1H), 2.92-2.99 (m, 1H), 3.81 (s, 3H), 4.04-4.08 (m, 1H), 4.25-4.35 (m, 2H), 4.41-4.49 (m, 1H), 5.15 (d, J=12Hz, 1H), 5.32 (d, J=18.5Hz, 1H), 5.74-5.82 (m, 1H), and 6.92-6.94 (m, 2H), 7.48-7.50 (m, 2H); MS m/z 663 (M ++ H).
Embodiment 8: preparation embodiment 8
Figure G2007800499569D00781
Embodiment 8
Embodiment 8 is same procedure of describing by in embodiment 7 steps 5, and difference replaces use 4 '-methoxyl group-3-xenyl magnesium bromide and makes. 1H NMR(CD 3OD)δ1.02-1.10(m,11H),1.29-1.31(m,2H),1.40-1.50(m,10H),1.90-1.92(m,1H),2.25-2.27(m,2H),2.68-2.69(m,1H),2.92-2.98(m,1H),3.84(s,3H),4.10-4.15(m,1H),4.35-4.52(m,2H),5.15(d,J=12Hz,1H),5.32(d,J=18.5Hz,1H),5.75-5.82(m,1H),7.00-7.03(m,3H),7.40-7.60(m,5H);MS m/z 739(M ++H)。
Embodiment 9: preparation embodiment 9
Figure G2007800499569D00782
Embodiment 9
At-78 ℃ to (S)-1-{ (the S)-2-{[(1R that makes as described in example 7 above; 2S)-and 1-(cyclopropyl sulfuryl amino formyl radical)-2-vinyl cyclopropyl] formamyl }-4-oxo-pyrrolidine-1-yl }-3; 3-dimethyl-1-oxo fourth-2-base-t-butyl carbamate (77mg; 0.14mmol) add the allyl group bromination magnesium (solution of 1.0M in ether in the solution in THF (5mL); 0.7mL, 0.7mmol).This reaction mixture is warmed to room temperature, and stirs and spend the night.Use the saturated aqueous ammonium chloride stopped reaction.With this mixture ethyl acetate extraction, use MgSO 4Drying is filtered, and vacuum concentration.Crude product by quick silica gel chromatography, with 2: 1 hexane/acetone wash-outs, has been obtained this title product (50mg, 60%). 1H NMR(400MHz,CD 3OD)δ0.92-0.96(m,2H),0.93-1.09(m,10H),1.19-1.25(m,2H),1.38-1.48(m,10H),1.83-1.94(m,2H),2.18-2.31(m,2H),2.31-2.46(m,2H),2.88-2.96(m,1H),3.70(d,J=9.82Hz,1H),3.81(d,J=10.32Hz,1H),4.22(d,J=9.32Hz,1H),4.32(dd,J=9.06,4.78Hz,1H),5.09-5.20(m,3H),5.30(dd,J=17.25,1.38Hz,1H),5.69-5.80(m,1H),5.87-5.99(m,1H),6.69(d,J=9.06Hz,1H);MS m/z 619(M+Na) +
The general method of preparation embodiment 10-25
Figure G2007800499569D00791
At-78 ℃ to (S)-1-{ (S)-2-{[(1R; 2S)-and 1-(cyclopropyl sulfuryl amino formyl radical)-2-vinyl cyclopropyl] formamyl }-4-oxo-pyrrolidine-1-yl }-3; (83mg 0.15mmol) adds grignard reagent (0.75-0.90mmol) in the solution in THF (1-5mL) to the 3-dimethyl-1-oxo fourth-2-aminocarbamic acid tert-butyl ester.This reaction mixture is warmed to room temperature, and stirred 1-3 hour.With saturated aqueous ammonium chloride (1mL) stopped reaction.This mixture is neutralized with 1N HCl, and (MgSO is used in 2 * 20mL) extractions with ethyl acetate 4Drying is filtered, and vacuum concentration.Crude product has obtained required product by preparation HPLC purifying.
Embodiment 10: preparation embodiment 10
Figure G2007800499569D00801
Embodiment 10
Embodiment 10 is according to described general method, and (solution of 1.0M in THF, 0.75mL 0.75mmol) makes, and has obtained 5mg (6%) product to use (3-fluorophenyl) magnesium bromide. 1H NMR(400MHz,CD 3OD)δ0.98-1.01(m,2H),1.03-1.11(m,10H),1.21-1.27(m,2H),1.37-1.51(m,10H),1.87(dd,J=8.18,5.41Hz,1H),2.18-2.28(m,2H),2.56-2.64(m,1H),2.90-2.99(m,1H),4.03(d,J=10.83Hz,1H),4.24(d,J=10.58Hz,1H),4.28(s,1H),4.43(dd,J=9.06,4.03Hz,1H),5.12(dd,J=10.45,1.64Hz,1H),5.30(dd,J=17.12,1.51Hz,1H),5.70-5.82(m,1H),6.99-7.07(m,1H),7.30-7.41(m,3H);MS m/z 651(M+H) +
Embodiment 11: preparation embodiment 11
Embodiment 11
Embodiment 11 is according to described general method, and (solution of 1.0M in THF, 0.75mL 0.75mmol) makes, and has obtained 7mg (7%) product to use (3-p-methoxy-phenyl) magnesium bromide. 1H NMR(400MHz,CD 3OD)δ0.98-1.01(m,2H),1.03-1.12(m,10H),1.20-1.27(m,2H),1.42-1.47(m,10H),1.87(dd,J=8.18,5.41Hz,1H),2.17-2.29(m,2H),2.55-2.64(m,1H),2.90-3.00(m,1H),3.79(s,3H),3.98-4.06(m,1H),4.21-4.33(m,2H),4.43(dd,J=9.19,3.90Hz,1H),5.12(dd,J=10.45,1.64Hz,1H),5.29(dd,J=17.12,1.51Hz,1H),5.69-5.82(m,1H),6.85(dd,J=8.06,2.01Hz,1H),7.08(d,J=7.81Hz,1H),7.14(s,1H),7.23-7.30(m,1H);MS m/z 663(M+H) +
Embodiment 12: preparation embodiment 12
Figure G2007800499569D00811
Embodiment 12
Embodiment 12 is according to described general method, and (solution of 0.5M in THF, 1.8mL 0.90mmol) makes, and has obtained 5mg (5%) product to use (3-chloro-phenyl-) magnesium bromide. 1HNMR(400MHz,CD 3OD)δ0.98-1.01(m,2H),1.03-1.11(m,10H),1.22-1.27(m,2H),1.41-1.50(m,10H),1.87(dd,J=8.18,5.41Hz,1H),2.19-2.28(m,2H),2.56-2.64(m,1H),2.90-2.98(m,1H),4.04(d,J=10.58Hz,1H),4.22(d,J=10.83Hz,1H),4.27(s,1H),4.44(dd,J=9.19,3.90Hz,1H),5.12(dd,J=10.32,1.51Hz,1H),5.30(dd,J=17.12,1.51Hz,1H),5.70-5.81(m,1H),7.28-7.37(m,2H),7.47(t,J=7.68Hz,1H),7.61(s,1H));MS m/z 689(M+Na) +
Embodiment 13: preparation embodiment 13
Figure G2007800499569D00812
Embodiment 13
Embodiment 13 is according to described general method, and (solution of 1.0M in THF, 0.90mL 0.90mmol) makes tolyl bromination magnesium between use, has obtained 11mg (11%) product. 1HNMR(400MHz,CD 3OD)δ0.99(s,2H),1.04-1.09(m,10H),1.21-1.27(m,2H),1.42-1.47(m,10H),1.87(dd,J=8.31,5.54Hz,1H),2.18-2.29(m,2H),2.35(s,3H),2.56-2.63(m,1H),2.90-2.98(m,1H),4.03(d,J=11.08Hz,1H),4.24(d,J=10.83Hz,1H),4.28-4.32(m,1H),4.41(dd,J=9.06,4.03Hz,1H),5.12(dd,J=10.32,1.51Hz,1H),5.29(dd,J=17.25,1.38Hz,1H),5.70-5.81(m,1H),7.11(d,J=7.55Hz,1H),7.23(t,J=7.55Hz,1H),7.32(d,J=7.81Hz,1H),7.37(s,1H));MS m/z 669(M+Na) +
Embodiment 14: preparation embodiment 14
Embodiment 14
Embodiment 14 is according to described general method, and (solution of 0.5M in THF, 1.5mL 0.75mmol) makes, and has obtained 12mg (12%) product to use (3-isopropyl phenyl) magnesium bromide. 1H NMR(400MHz,CD 3OD)δ0.98-1.01(m,2H),1.04-1.11(m,10H),1.22-1.28(m,8H),1.41-1.47(m,10H),1.87(dd,J=8.18,5.41Hz,1H),2.18-2.34(m,2H),2.55-2.63(m,1H),2.87-2.98(m,2H),4.00-4.07(m,1H),4.26(d,J=10.83Hz,1H),4.29-4.33(m,1H),4.42(dd,J=9.06,4.03Hz,1H),5.12(dd,J=10.45,1.64Hz,1H),5.29(dd,J=17.12,1.51Hz,1H),5.70-5.81(m,1H),7.17(d,J=7.30Hz,1H),7.24-7.36(m,2H),7.44(s,1H).MS m/z 675(M+H) +
Embodiment 15: preparation embodiment 15
Figure G2007800499569D00831
Embodiment 15
Embodiment 15 is according to described general method, and (solution of 0.5M in THF, 1.5mL 0.75mmol) makes, and has obtained 12mg (12%) product to use (3-isopropyl phenyl) magnesium bromide. 1H NMR(400MHz,CD 3OD)δ0.98-1.01(m,2H),1.04-1.11(m,10H),1.21-1.27(m,2H),1.29(d,J=6.04Hz,6H),1.42-1.47(m,10H),1.87(dd,J=8.18,5.41Hz,1H),2.18-2.29(m,2H),2.55-2.63(m,1H),2.90-2.99(m,1H),3.98-4.06(m,1H),4.23(d,J=10.83Hz,1H),4.29(s,1H),4.45(dd,J=9.32,3.78Hz,1H),4.57-4.66(m,1H),5.12(dd,J=10.32,1.76Hz,1H),5.30(dd,J=17.12,1.51Hz,1H),5.70-5.81(m,1H),6.83(dd,J=8.06,1.76Hz,1H),7.05(d,J=7.81Hz,1H),7.12(s,1H),7.20-7.28(m,1H);MS m/z 691(M+H) +
Embodiment 16: preparation embodiment 16
Figure G2007800499569D00832
Embodiment 16
Embodiment 16 is according to described general method, and (solution of 0.5M in THF, 1.5mL 0.75mmol) makes, and has obtained 7mg (7%) product to use [3-(N, N-dimethyl) aniline] magnesium bromide. 1H NMR(400MHz,CD 3OD)δ1.00(s,2H),1.02-1.13(m,9H),1.20-1.27(m,2H),1.40-1.47(m,10H),1.88(dd,J=8.18,5.41Hz,1H),2.19-2.29(m,2H),2.59-2.70(m,1H),2.90-3.00(m,1H),3.08-3.19(m,6H),4.01-4.14(m,2H),4.24(d,J=10.83Hz,1H),4.28(s,1H),4.50(dd,J=9.44,3.15Hz,1H),5.13(dd,J=10.32,1.76Hz,1H),5.30(dd,J=17.25,1.39Hz,1H),5.70-5.82(m,1H),7.16-7.23(m,1H),7.28-7.36(m,1H),7.42(t,J=7.43Hz,1H),7.52(s,1H);MSm/z 676(M+H) +
Embodiment 17: preparation embodiment 17
Figure G2007800499569D00841
Embodiment 17
Embodiment 17 is according to described general method, and (solution of 0.25M in THF, 3.0mL 0.75mmol) makes, and has obtained 9mg (9%) product to use [3-(1-pyrrolidyl methyl) phenyl] magnesium bromide. 1H NMR(400MHz,CD 3OD)δ0.98-1.02(m,3H),1.04-1.09(m,9H),1.24(d,J=1.51Hz,3H),1.39-1.51(m,11H),1.87(dd,J=8.31,5.54Hz,1H),2.18-2.29(m,2H),2.35(s,3H),2.56-2.63(m,1H),2.90-2.98(m,1H),4.03(d,J=11.08Hz,1H),4.24(d,J=10.83Hz,1H),4.30(s,1H),4.41(dd,J=9.06,4.03Hz,1H),5.12(dd,J=10.32,1.51Hz,1H),5.29(dd,J=17.25,1.38Hz,1H),5.70-5.81(m,1H),7.11(d,J=7.55Hz,1H),7.23(t,J=7.55Hz,1H),7.32(d,J=7.81Hz,1H),7.37(s,1H);MS m/z 716(M+H) +
Embodiment 18: preparation embodiment 18
Figure G2007800499569D00842
Embodiment 18
Embodiment 18 is according to described general method, and (solution of 0.25M in THF, 3.0mL 0.75mmol) makes, and has obtained 15mg (14%) product to use [3-(piperidino methyl) phenyl] magnesium bromide. 1H NMR(400MHz,CD 3OD)δ0.99-1.04(m,5H),1.07(s,9H),1.15-1.26(m,3H),1.41-1.47(m,13H),1.77-1.92(m,5H),2.11-2.40(m,2H),2.65-2.76(m,1H),2.86-2.97(m,1H),4.08-4.19(m,2H),4.28(s,3H),4.52(dd,J=9.82,2.77Hz,1H),5.05-5.15(m,1H),5.22-5.34(m,1H),5.72-5.85(m,1H),7.44(d,J=7.20Hz,1H),7.49(t,J=7.43Hz,1H),7.66(d,J=7.55Hz,1H),7.81(s,1H);MS m/z732(M+H) +
Embodiment 19: preparation embodiment 19
Figure G2007800499569D00851
Compound 19
Embodiment 19 is according to described general method, and (solution of 0.25M in THF, 3.0mL 0.75mmol) makes, and has obtained 16mg (15%) product to use [3-(4-morpholinyl methyl) phenyl] magnesium bromide. 1H NMR(400MHz,CD 3OD)δ0.98-1.02(m,2H),1.07(s,9H),1.19-1.27(m,3H),1.40-1.50(m,10H),1.88(dd,J=7.93,5.67Hz,1H),2.16-2.38(m,2H),2.60-3.01(m,6H),3.71-3.83(m,4H),4.00-4.16(m,3H),4.22(d,J=10.83Hz,1H),4.26-4.32(m,1H),4.48(dd,J=9.32,2.77Hz,1H),5.12(d,J=10.58Hz,1H),5.30(d,J=16.87Hz,1H),5.70-5.83(m,1H),6.79(d,J=8.81Hz,1H),7.33-7.45(m,2H),7.56(d,J=8.06Hz,1H),7.66(s,1H);MS 732m/z(M+H) +
Embodiment 20: preparation embodiment 20
Figure G2007800499569D00861
Embodiment 20
Embodiment 20 is according to described general method, and (solution of 0.25M in THF, 3.6mL 0.90mmol) makes, and has obtained 4mg (4%) product to use [2-(4-morpholinyl methyl) phenyl] magnesium bromide. 1H NMR(400MHz,CD 3OD)δ0.94-0.98(m,2H),1.01-1.11(m,10H),1.16-1.24(m,2H),1.41-1.48(m,10H),1.85(dd,J=8.06,5.29Hz,1H),2.12-2.22(m,1H),2.45(dd,J=12.59,7.55Hz,1H),2.55-2.68(m,4H),2.72-2.85(m,1H),2.87-2.97(m,1H),3.59-3.84(m,6H),4.00-4.07(m,1H),4.13-4.21(m,1H),4.34-4.38(m,1H),4.47(d,J=10.83Hz,1H),5.10(dd,J=10.20,1.64Hz,1H),5.26(dd,J=17.12,1.26Hz,1H),5.68-5.80(m,1H),6.77(d,J=9.57Hz,1H),7.28-7.40(m,3H),7.44-7.49(m,1H);MS m/z(M+H) +
Embodiment 21: preparation embodiment 21
Figure G2007800499569D00862
Embodiment 21
Embodiment 21 is according to described general method, and (solution of 0.5M in THF, 1.8mL 0.90mmol) makes, and has obtained 7mg (7%) product to use (3,5-3,5-dimethylphenyl) magnesium bromide. 1H NMR(400MHz,CD 3OD)δ0.97-1.11(m,12H),1.19-1.27(m,2H),1.37-1.51(m,10H),1.87(dd,J=8.18,5.41Hz,1H),2.15-2.25(m,2H),2.30(s,6H),2.54-2.62(m,1H),2.90-2.98(m,1H),4.02(d,J=10.58Hz,1H),4.21(d,J=10.83Hz,1H),4.27-4.32(m,1H),4.41(dd,J=9.06,3.78Hz,1H),5.12(dd,J=10.32,1.51Hz,1H),5.29(dd,J=17.12,1.51Hz,1H),5.70-5.81(m,1H),6.94(s,1H),7.15(s,2H);MS m/z 683(M+Na) +
Embodiment 22: preparation embodiment 22
Figure G2007800499569D00871
Embodiment 22
Embodiment 22 is according to described general method, and (solution of 1.0M in THF, 0.9mL 0.90mmol) makes, and has obtained 8mg (8%) product to use (4-fluoro-3-aminomethyl phenyl) magnesium bromide. 1H NMR(400MHz,CD 3OD)δ0.97-1.02(m,2H),1.03-1.11(m,10H),1.20-1.27(m,2H),1.41-1.47(m,10H),1.87(dd,J=8.18,5.41Hz,1H),2.20-2.25(m,1H),2.27(d,J=1.51Hz,3H),2.55-2.63(m,1H),2.89-2.98(m,1H),4.03(d,J=10.83Hz,1H),4.21(d,J=10.83Hz,1H),4.26-4.31(m,1H),4.41(dd,J=9.19,3.90Hz,1H),5.12(dd,J=10.32,1.76Hz,1H),5.30(dd,J=17.12,1.26Hz,1H),5.70-5.81(m,1H),6.79(d,J=9.06Hz,1H),6.96-7.03(m,1H),7.33-7.40(m,1H),7.41-7.46(m,1H);MS m/z 687(M+Na) +
Embodiment 23: preparation embodiment 23
Figure G2007800499569D00872
Embodiment 23
Embodiment 23 is according to described general method, and (solution of 0.5M in THF, 1.8mL 0.90mmol) makes, and has obtained 5mg (5%) product to use (3-fluoro-4-aminomethyl phenyl) magnesium bromide. 1H NMR(400MHz,CD 3OD)δ0.99(s,2H),1.03-1.10(m,10H),1.21-1.27(m,2H),1.42-1.47(m,10H),1.87(dd,J=8.31,5.54Hz,1H),2.19-2.22(m,J=8.06Hz,1H),2.24(d,J=1.51Hz,3H),2.54-2.61(m,1H),2.90-2.98(m,1H),4.01(d,J=10.83Hz,1H),4.22(d,J=10.83Hz,1H),4.26-4.30(m,1H),4.41(dd,J=9.06,4.03Hz,1H),5.12(dd,J=10.32,1.76Hz,1H),5.29(dd,J=17.25,1.13Hz,1H),5.70-5.81(m,1H),7.20-7.27(m,3H);MS m/z 665(M+H) +
Embodiment 24: preparation embodiment 24
Figure G2007800499569D00881
Embodiment 24
Embodiment 24 is according to described general method, and (solution of 0.5M in THF, 1.8mL 0.90mmol) makes, and has obtained 5mg (5%) product to use (2,5-3,5-dimethylphenyl) magnesium bromide. 1H NMR(400MHz,CD 3OD)δ0.94-0.97(m,2H),1.02-1.10(m,10H),1.20-1.26(m,2H),1.40-1.49(m,10H),1.86(dd,J=8.18,5.41Hz,1H),2.16(d,J=4.78Hz,1H),2.17-2.25(m,1H),2.28(s,3H),2.48(s,3H),2.72(dd,J=12.46,8.18Hz,1H),2.88-2.97(m,1H),4.09(d,J=10.83Hz,1H),4.13-4.21(m,1H),4.37(d,J=9.32Hz,1H),4.44(d,J=10.83Hz,1H),5.11(dd,J=10.32,1.51Hz,1H),5.28(dd,J=17.12,1.51Hz,1H),5.70-5.80(m,1H),6.82(d,J=9.57Hz,1H),6.99(t,J=7.68Hz,1H),7.11(d,J=7.30Hz,1H),7.17(d,J=7.81Hz,1H);MS m/z 683(M+Na) +
Embodiment 25: preparation embodiment 25
Figure G2007800499569D00891
Embodiment 25
Embodiment 25 is according to described general method, and (solution of 0.5M in THF, 1.8mL 0.90mmol) makes, and has obtained 5mg (5%) product to use (5-fluoro-2-p-methoxy-phenyl) magnesium bromide. 1H NMR(400MHz,CD 3OD)δ1.00-1.10(m,11H),1.20-1.26(m,2H),1.40-1.47(m,10H),1.83-1.89(m,1H),2.20-2.29(m,1H),2.34-2.43(m,1H),2.90-2.98(m,1H),3.80(s,1H),3.85(s,3H),3.93(d,J=10.83Hz,1H),4.18-4.28(m,1H),4.45(d,J=10.83Hz,1H),4.56(dd,J=9.69,2.64Hz,1H),5.12(dd,J=10.58,1.51Hz,1H),5.30(d,J=17.37Hz,1H),5.68-5.83(m,1H),6.75(d,J=8.81Hz,1H),6.98-7.04(m,2H),7.36(d,J=10.32Hz,1H);MS m/z 703(M+Na) +
Embodiment 26: preparation embodiment 26
Figure G2007800499569D00892
Embodiment 26
-40 ℃ of products to embodiment 7 steps 4 (55mg, 0.10mmol) drip to add in the solution in THF (4mL) (4-Phenoxyphenyl) magnesium bromide (0.5M/THF, 1.0mL, 0.50mmol).Stirring under this temperature 2 hours and after 0 ℃ is stirred 1 hour, with this mixture 5% citric acid treatment, and extracting with EtOAc (10mL).With organic layer salt water washing, use MgSO 4Drying is filtered and evaporation.Resistates is developed and filtered with hot hexane.Thus obtained solid by preparation HPLC purifying, has been obtained compound 100, be white solid (2.1mg, 3%).
1H NMR (CD 3OD) δ 1.02-1.09 (m, 11H), 1.26-1.29 (m, 2H), 1.45-1.50 (m, 10H), 1.90-1.92 (m, 1H), 2.25-2.27 (m, 2H), 2.68-2.69 (m, 1H), 2.95-2.99 (m, 1H), 4.05-4.07 (m, 1H), 4.35-4.52 (m, 2H), 5.15 (d, J=12Hz, 1H), 5.32 (d, J=18.5Hz, 1H), 5.75-5.82 (m, 1H), 6.97-7.01 (m, 4H), 7.15-7.16 (m, 1H), and 7.36-7.39 (m, 2H), 7.56-7.58 (m, 2H); LC-MS (retention time: 2.94min, method B), MS m/z 707 (M +-H2O).
Embodiment 27: preparation embodiment 27
Figure G2007800499569D00901
Embodiment 27
Reaction scheme 5
Figure G2007800499569D00902
Step 1.
To (2S, 4R)-4-(biphenyl-4-yl)-4-hydroxyl pyrrolidine-1,2-dioctyl phthalate 1-benzyl ester 2-methyl esters (1.294g, 3mmol) add previously prepared lithium hydroxide monohydrate (0.252g, 6.00mmol) solution in water (10.00ml) in the solution in THF (10ml) and MeOH (10.00ml).With formed turbid solution stirring at room 6 hours.Dilute with water is acidified to pH 3 with 1M HCl, with EtOAc (200ml) extraction. organic layer with 5% citric acid and salt water washing, is used MgSO 4Drying is filtered, evaporation.Residual powder with hexane-EtOAc development in 4: 1, is obtained required product (1.20g, 96% productive rate), be the white powder thing.
1H NMR(CD 3OD)δ2.53-2.57(m,1H),2.79-2.86(m,1H),3.83-3.90(m,2H),4.62-4.66(m,1H),5.16-5.20(m,2H),7.24-7.46(m,8H),7.57-7.66(m,6H);
LC-MS (retention time: 2.64min, method B), MS m/z 400 (M +-H2O).
Step 2.
Will (73.3mg 0.548mmol) stirs in DMF (2.5mL) with the potassium hydride KH of hexane pre-wash.0 ℃ of disposable adding solid form (2S, 4R)-1-(benzyloxycarbonyl)-4-(biphenyl-4-yl)-4-hydroxyl pyrrolidine-2-formic acid (104mg, 0.249mmol).Formed white gels shape thing is stirred 30 minutes (at this moment become almost clarification) under this temperature, add then allyl iodide (0.027mL, 0.299mmol).Final turbid solution was stirred 2 hours under this temperature.5% citric acid stopped reaction with ice-cold extracts with EtOAc.With organic layer salt water washing, use MgSO 4Drying is filtered, evaporation.By the prep-HPLC purifying, obtained 35mg raw material and required product (45mg, 40% productive rate), be white solid.
1H NMR(CD 3OD)δ2.56-2.57(m,1H),2.81-2.84(m,1H),3.82-3.87(m,2H),4.69-4.73(m,3H),5.14-5.23(m,3H),5.30-5.41(m,1H),5.87-6.01(m,1H),7.34-7.46(m,8H),7.57-7.66(m,6H);
LC-MS (retention time: 2.64min, method B), MS m/z 458 (M ++ H).
Embodiment 28: preparation embodiment 28
Figure G2007800499569D00911
Embodiment 28
Reaction scheme 6
Figure G2007800499569D00912
Step 1.
At 0 ℃ to (2S, 4R)-4-(biphenyl-4-yl)-4-hydroxyl pyrrolidine-1,2-dioctyl phthalate 1-benzyl ester 2-methyl esters (540mg, 1.252mmol) at diacetyl oxide (10mL, 106mmol) drip previously prepared trifluoromethanesulfonic acid scandium (61.6mg, 0.125mmol) solution in acetonitrile (0.5ml) in Nei the solution.Formed baby pink solution was stirred 1 hour under this temperature.Use the saturated ammonium chloride stopped reaction, extract with EtOAc.With organic layer salt water washing, use MgSO 4Drying is filtered, evaporation.Resistates by the prep-HPLC purifying, is obtained required product (290mg, 49%), be non-enantiomer mixture.
1H NMR (CDCl 3) δ 1.97 (s, 1/3H), 2.00 (s, 2/3H), 2.42-2.50 (m, 1/3H), 2.68-2.79 (m, 2/3H), 3.03-3.05 (m, 2/3H), 3.24-3.41 (m, 1/3H), 3.54,3.77 (s, 1H, rotational isomers), 3.66,3.79 (s, 2H, rotational isomer), 3.80-3.90 (m, 1/3H), 4.10-4.19 (m, 2/3H), 4.20-4.29 (m, 2/3H), 4.49-4.57 (m, 1/3H), 4.52-4.60 (m, 1H), and 5.09-5.26 (m, 2H), 7.30-7.56 (m, 14H); LC-MS (retention time: 2.78min, method B), MS m/z 496 (M +-CH 3CO 2OH).
Embodiment 101: preparation embodiment 101
Figure G2007800499569D00921
Embodiment 101
Reaction scheme 1
Figure G2007800499569D00922
Step 1.
Add in the two neck flasks magnesium bromide ether compound that grinds (258mg, 1.0mmol).This flask was heated 4 hours under high vacuum at 70 ℃ with oil bath.After this flask cooled off in ice bath, (110mg 0.20mmol) and THF (5mL), and spent the night the gained pale yellow syrup in the room temperature vigorous stirring to add the product of embodiment 7 steps 4.
Step 2.
-78 ℃ to contain 1-(4-bromophenyl)-1H-pyrroles (239mg, 1.0mmol) and drip in another two necks flask of THF (4mL) n-BuLi (2.5M, 0.4mL, 1.0mmol).The gained mixture was stirred 15 minutes under this temperature.These slurries are joined via intubate in the flask of the step 1 that is cooled to-78 ℃ in advance.Final mixture was stirred 2 hours under this temperature, and 0 ℃ of restir 1 hour.With this mixture saturated ammonium chloride stopped reaction, with the EtOAc dilution, and isolate top organic phase.With organic layer salt water washing, use MgSO 4Drying is filtered and evaporation.Resistates is developed and filtered with hot hexane.The gained solid by preparation HPLC purifying, has been obtained embodiment 101, be white solid (16.5mg, 12%).
1H NMR (CD 3OD) δ 1.02-1.09 (m, 11H), 1.26-1.29 (m, 2H), 1.45-1.50 (m, 10H), 1.90-1.92 (m, 1H), 2.25-2.27 (m, 2H), 2.68-2.69 (m, 1H), 2.95-2.99 (m, 1H), 4.05-4.07 (m, 1H), 4.35-4.52 (m, 2H), 5.15 (d, J=12Hz, 1H), 5.32 (d, J=18.5Hz, 1H), 5.75-5.82 (m, 1H), 6.30 (m, 2H), 7.20-7.21 (m, 2H), and 7.48-7.51 (m, 2H), 7.66-7.68 (m, 2H); LC-MS (retention time: 2.81min, method B), MS m/z 698 (M ++ H), 680 (M +-H2O).
Embodiment 102: preparation embodiment 102
Figure G2007800499569D00931
Embodiment 102
Embodiment 102 is by the same procedure of describing among the embodiment 101, difference uses 4 in step 2 '-bromo-2-methoxyl biphenyl replaces 1-(4-bromophenyl)-1H-pyrroles and makes.
1H NMR (CD 3OD) δ 1.02-1.09 (m, 11H), 1.26-1.29 (m, 2H), 1.45-1.50 (m, 10H), 1.90-1.92 (m, 1H), 2.25-2.27 (m, 2H), 2.68-2.69 (m, 1H), 2.95-2.99 (m, 1H), 3.80 (s, 3H), 4.05-4.07 (m, 1H), 4.35-4.52 (m, 2H), 5.15 (d, J=12Hz, 1H), 5.32 (d, J=18.5Hz, 1H), 5.75-5.82 (m, 1H), 7.00-7.09 (m, 2H), and 7.27-7.34 (m, 2H), 7.44-7.51 (m, 4H); LC-MS (retention time: 2.90min, method B), MS m/z 729 (M ++ H), 721 (M +-H2O).
Embodiment 103: preparation embodiment 103
Figure G2007800499569D00941
Embodiment 103
Step 1.
Add in the two neck flasks Cerium II Chloride (III) heptahydrate that grinds (373mg, 1.0mmol).This flask was heated 20 hours under high vacuum at 70 ℃ with oil bath, then 100 ℃ of heating 2 hours, 120 ℃ of heating 2 hours, at last 160 ℃ of heating 16 hours.After this flask cooled off in ice bath, add embodiment 7 steps 4 product (110mg, 0.20mmol) and THF (5mL).With the gained pale yellow syrup room temperature vigorous stirring 4 hours.
Step 2.
-78 ℃ to contain 2-(4-bromophenyl) pyridine (JOC, 2003, p6959,233mg, 1.0mmol) and drip n-BuLi in another two necks flask of THF (4mL) (2.5M, 0.4mL 1.0mmol), and stir this mixture 15 minutes under this temperature.Formed slurries are joined via intubate in the flask of the step 1 that is cooled to-78 ℃ in advance.Final mixture was stirred 2 hours under this temperature, then 0 ℃ of restir 1 hour.With this mixture saturated ammonium chloride stopped reaction, with the EtOAc dilution, and isolate top organic layer.With organic layer salt water washing, use MgSO 4Drying is filtered and evaporation.Resistates is developed and filtered with hot hexane.The gained solid by preparation HPLC purifying, has been obtained embodiment 103, be white solid (17mg, 12%).
1H NMR(CD 3OD)δ1.02-1.09(m,11H),1.26-1.29(m,2H),1.45-1.50(m,10H),1.90-1.92(m,1H),2.25-2.27(m,2H),2.68-2.69(m,1H),2.95-2.99(m,1H),4.05-4.07(m,1H),4.35-4.52(m,2H),5.15(d,J=12Hz,1H),5.32(d,J=18.5Hz,1H),5.75-5.82(m,1H),7.38-7.40(m,1H),7.71-7.74(m,2H),7.87-8.00(m,4H),7.63-7.64(m,1H);
LC-MS (retention time: 2.24min, method B), MS m/z 710 (M ++ H).
Embodiment 104: preparation embodiment 104
Figure G2007800499569D00951
Embodiment 104
Embodiment 104 is by the same procedure of describing in embodiment 101, and difference is used also [d] thiazole replacement 1-(4-bromophenyl)-1H-pyrroles and making of 2-bromobenzene in step 2.
1H NMR(CD 3OD)δ1.02-1.09(m,11H),1.26-1.29(m,2H),1.45-1.50(m,10H),1.90-1.92(m,1H),2.25-2.27(m,2H),2.68-2.69(m,1H),2.95-2.99(m,1H),3.80(s,3H),4.05-4.07(m,1H),4.35-4.52(m,2H),5.15(d,J=12Hz,1H),5.32(d,J=18.5Hz,1H),5.75-5.82(m,1H),7.43-7.52(m,2H),7.93-8.01(m,2H);
LC-MS (retention time: 2.79min, method B), MS m/z 690 (M ++ H).
Embodiment 105: preparation embodiment 105
Figure G2007800499569D00952
Embodiment 105
Embodiment 105 is same procedure of describing by in embodiment 101, and difference is used 2-(4-bromophenyl) thiazole to replace 1-(4-bromophenyl)-1H-pyrroles in step 2 and made.
1H NMR(CD 3OD)δ1.02-1.09(m,11H),1.26-1.29(m,2H),1.45-1.50(m,10H),1.90-1.92(m,1H),2.25-2.27(m,2H),2.68-2.69(m,1H),2.95-2.99(m,1H),3.80(s,3H),4.05-4.07(m,1H),4.35-4.52(m,2H),5.15(d,J=12Hz,1H),5.32(d,J=18.5Hz,1H),5.75-5.82(m,1H),7.64(m,1H),7.72-7.74(m,2H),7.90(m,1H),7.98-7.99(m,2H);
LC-MS (retention time: 2.67min, method B), MS m/z 716 (M ++ H).
The preparation The compounds of this invention
Embodiment 106: preparation compound 106
Figure G2007800499569D00961
Compound 106
Reaction scheme 2
Figure G2007800499569D00971
Step 1.
-78 ℃ to methyl sulfoxide (23.90ml, 337mmol) drip in the solution in DCM (100ml) oxalyl chloride (solution of 2M in DCM, 84ml, 168mmol).Formed solution was stirred 30 minutes under this temperature.-78 ℃ of droppings (2S, 4R)-4-hydroxyl pyrrolidine-1,2-dioctyl phthalate 1-benzyl ester 2-methyl esters (21.38g, 77mmol) solution in DCM (100ml).Formed slurries were stirred 2 hours at-78 ℃, drip N then, and the N-diisopropylethylamine (66.7ml, 383mmol).With final solution stirring at room 3 hours.This mixture is used ice-cold 1M HCl, 5% citric acid and salt water washing successively, use MgSO 4Drying is filtered, and evaporation.Remaining light brown oily thing by silica gel chromatography, is used 4: 1,3: 1 and 2: 1 hexane-EtOAc wash-outs successively, obtained (S)-4-oxo-pyrrolidine-1,2-dioctyl phthalate 1-benzyl ester 2-methyl esters (14.8g, 70% productive rate) is light brown viscous oily matter.
1H NMR (CDCl 3) δ 2.58-2.63 (m, 1H), 2.90-2.99 (m, 1H), 3.62,3.77 (s, 3H, rotational isomers), 3.95-4.02 (m, 2H), 4.82-4.89 (m, 1H), 5.11-5.24 (m, 2H), 7.32-7.39 (m, 5H).
Step 2.
0 ℃ to (S)-4-oxo-pyrrolidine-1,2-dioctyl phthalate 1-benzyl ester 2-methyl esters (14.0g, 50.5mmol) drip in the solution in toluene (500mL) biphenyl-4-base magnesium bromide (152mL, the 0.5M solution in THF, 75.75mmol).Formed pale yellow solution was stirred 1 hour under this temperature.Use the ammonium chloride stopped reaction, isolate organic layer.Water layer is extracted with EtOAc.With the organic layer salt water washing that merges, use MgSO 4Drying is filtered, and evaporation.Resistates is passed through the silica gel plug purifying, use 4: 1 successively, 3: 1,2: 1 and 3: 2 hexane-EtOAc wash-outs, obtained the 11.70g white solid, with its recrystallization from EtOAc-hexane (50ml-150ml), obtained 7.8g (2S, 4R)-and 4-(biphenyl-4-yl)-4-hydroxyl pyrrolidine-1,2-dioctyl phthalate 1-benzyl ester 2-methyl esters is little spicule.Mother liquor is concentrated and by quick column purification, use 4: 1 successively, 3: 1,2: 1 and 3: 2 hexane-EtOAc wash-outs, obtained the required product of 2.41g in addition.
1H NMR (CDCl 3) δ 2.39-2.45 (m, 1H), 2.70-2.75 (m, 1H), 3.66,3.86 (s, 3H, rotational isomers), 3.80-3.90 (m, 1H), 4.00-4.07 (m, 1H), 4.62 (dd, J 1,2=9.5,28Hz, 1H), 5.09-5.15 (m, 1H), 5.21-5.25 (m, 1H), 7.31-7.38 (m, 6H), 7.42-7.45 (m, 2H), 7.54-7.59 (m, 6H);
LC-MS (retention time: 2.77min, method B), MS m/z 414 (M +-H 2O), 370 (M +-H 2O-CO 2).
Step 3.
0 ℃ to (2S, 4R)-4-(biphenyl-4-yl)-4-hydroxyl pyrrolidine-1,2-dioctyl phthalate 1-benzyl ester 2-methyl esters (8.08g, 18.73mmol) add in the solution in DMF (150ml) sodium hydride (0.520g, 20.60mmol).Formed light brown solution was stirred 30 minutes under this temperature.0 ℃ drip methyl-sulfate (1.949ml, 20.60mmol).With final solution stirring at room 2 hours.With 5% citric acid stopped reaction, extract with EtOAc.With salt water washing organic layer, use MgSO 4Drying is filtered, and evaporation.Resistates by quick silica gel chromatography, is used 4: 1,3: 1 and 2: 1 hexane-EtOAc wash-outs successively, obtained the required product of 1.45g, with its recrystallization in MeOH (10ml), obtained 1.20g (14.38% productive rate), be white solid.Also reclaiming the 4.50g raw material during the column purification fast.
1H NMR (CDCl 3) δ 2.51-2.56 (m, 1H), 2.85-2.89 (m, 1H), 2.95,2.97 (s, 3H, rotational isomers), 3.67,3.80 (s, 3H, rotational isomers), 3.69-3.86 (m, 1H), 4.02-4.08 (m, 1H), 4.62 (dd, J 1,2=9.5,28Hz, 1H), 5.09-5.17 (m, 1H), 5.20-5.29 (m, 1H), 7.29-7.46 (m, 10H), 7.57-7.60 (m, 4H); LC-MS (retention time: 2.92min, method B), MS m/z 446 (M ++ H), 414 (M +-MeOH), 370 (M +-MeOH-CO 2).
Step 4.
To containing (2S, 4R)-and 4-(biphenyl-4-yl)-4-methoxyl group tetramethyleneimine-1, (1.29g 2.90mmol) is added in palladium (0.308g on the carbon to 2-dioctyl phthalate 1-benzyl ester 2-methyl esters in the ice-cold Parr shaking flasks of the solution in MeOH (30ml), 0.290mmol) (10%, wet).This container was being placed 5 hours under 25psi pressure hydrogen on the Parr wobbler device.Handle with diatomite, filter, evaporation has obtained the required product of 0.811g (91%), is pale powder shape thing.Products therefrom need not be further purified and be used for next linked reaction.
LC-MS (retention time: 1.92min, method B), MS m/z 312 (M ++ H), 280 (M +-MeOH).
Step 5.
At 0 ℃ to (2S, 4R)-4-(biphenyl-4-yl)-4-methoxyl group tetramethyleneimine-2-methyl-formiate (500mg, 1.606mmol), (S)-2-(tert-butoxycarbonyl amino)-3,3-acid dimethyl (409mg, 1.77mmol) and HATU (867mg, 2.409mmol) add N in the solution in DCM (15ml), and the N-diisopropylethylamine (0.839ml, 4.82mmol).With formed solution stirring at room 4 hours.With the DCM dilution, with 5% citric acid and salt water washing, use MgSO 4Drying is filtered, evaporation.Resistates by the silicagel column purifying, with 2: 1 hexane-acetone wash-outs, is obtained required product (600mg, 71% productive rate), be white foam shape thing.
1H NMR(CD 3OD)δ1.11(s,9H),1.42(s,9H),2.61-2.66(m,1H),2.71-2.75(m,1H),3.00(s,3H),3.75(s,3H),4.20-4.25(m,2H),4.30-4.35(m,1H),4.75-4.79(m,1H),7.35-7.72(m,9H);
LC-MS (retention time: 3.04min, method B), MS m/z 525 (M ++ H), 493 (M +-MeOH).
Step 6.
To (2S; 4R)-4-(biphenyl-4-yl)-1-((S)-2-(tert-butoxycarbonyl amino)-3; 3-dimethyl butyrate acyl group)-4-methoxyl group tetramethyleneimine-2-methyl-formiate (600mg; 1.144mmol) add previously prepared lithium hydroxide monohydrate (96mg, 2.287mmol) solution in water (5.00ml) in the solution in THF (5ml) and MeOH (5.00ml).With formed turbid solution stirring at room 8 hours.Remove volatile matter, with the dilution of 5% citric acid, extract with EtOAc.With organic layer salt water washing, use MgSO 4Drying is filtered, and evaporation.With residual powder (534mg, 91% productive rate) recrystallization from EtOAc-hexane (2ml-4ml), obtained the required product of 450mg, be white crystal.
LC-MS (retention time: 2.93min, method B), MS m/z 511 (M ++ H), 479 (M +-MeOH).
Step 7.
At 0 ℃ to (2S; 4R)-4-(biphenyl-4-yl)-1-((S)-2-(tert-butoxycarbonyl amino)-3; 3-dimethyl butyrate acyl group)-4-methoxyl group tetramethyleneimine-2-formic acid (255mg, 0.499mmol), (1R, 2S)-1-amino-N-(cyclopropyl alkylsulfonyl)-2-vinyl cyclopropane carboxamide; toluenesulphonic acids; hydrate (231mg, 0.549mmol) and HATU (228mg 0.599mmol) adds N in the slurries in CH2Cl2 (5ml); the N-diisopropylethylamine (0.435ml, 2.497mmol).Formed pale yellow solution was stirred 3 hours at 0 ℃.With EtOAc (10mL) dilution, with 5% citric acid and salt water washing, use MgSO 4Drying is filtered, and vacuum-evaporation.Resistates by preparation HPLC purifying (Phenomenex-Luna 30X100mm S10,30%B-100%B, 15 minutes gradient time, 20 minutes stand-by times), is obtained compound 106 (248mg, 68.7% productive rate), be white solid.
1H NMR(CD 3OD)δ1.01-1.12(m,11H),1.26-1.27(m,2H),1.43-1.50(m,10H),1.87-1.90(m,1H),2.20-2.25(m,1H),2.49-2.57(m,2H),2.95-2.99(m,1H),3.11(s,3H),4.07-4.09(m,1H),4.22-4.24(m,1H),4.42(d,J=9Hz,1H),4.58-4.60(m,1H),5.15(d,J=12Hz,1H),5.32(d,J=18.5Hz,1H),5.75-5.82(m,1H),7.38-7.39(m,1H),7.45-7.48(m,2H),7.57-7.67(m,6H);
LC-MS (retention time: 2.96min, method B), MS m/z 723 (M ++ H).
Embodiment 107: preparation compound 107
Figure G2007800499569D01001
Compound 107
Reaction scheme 3
Figure G2007800499569D01011
Step 1.
At 0 ℃ to (2S, 4R)-4-(biphenyl-4-yl)-4-methoxyl group tetramethyleneimine-2-methyl-formiate (150mg, 0.482mmol), (S)-2-(tert-butoxycarbonyl amino) ninth of the ten Heavenly Stems-8-olefin(e) acid (144mg, 0.530mmol) and HATU (260mg, 0.723mmol) add N in the solution in DCM (5ml), the N-diisopropylethylamine (0.252ml, 1.445mmol).With formed solution in stirred overnight at room temperature.With the DCM dilution, with 5% citric acid and salt water washing, use MgSO 4Drying is filtered and evaporation.Resistates by preparation HPLC purifying, is obtained required product (140mg, 51.5% productive rate), be white foam shape thing.
1H NMR(CD 3OD)δ1.31-1.57(m,15H),1.62-1.65(m,1H),1.78-1.82(m,1H),2.11-2.13(m,2H),2.66-2.69(m,1H),2.84-2.89(m,1H),3.00(s,3H),3.76(s,3H),4.16(s,2H),4.30-4.35(m,1H),4.79-4.81(m,1H),4.95(d,J=12Hz,1H),5.03(d,J=18.5Hz,1H),5.83-5.87(m,1H),7.32-7.39(m,1H),7.45-7.56(m,4H),7.64-7.71(m,4H);
LC-MS (retention time: 3.20min, method B), MS m/z 565 (M ++ H).
Step 2.
To (2S; 4R)-4-(biphenyl-4-yl)-1-((S)-2-(tert-butoxycarbonyl amino) ninth of the ten Heavenly Stems-8-enoyl-)-4-methoxyl group tetramethyleneimine-2-methyl-formiate (166mg; 0.294mmol) add previously prepared lithium hydroxide monohydrate (37mg, 0.882mmol) solution in water (2mL) in the solution in THF (2mL) and MeOH (2mL).With formed turbid solution in stirred overnight at room temperature.Vacuum is removed volatile matter.Resistates is placed water, be acidified to pH 2 with 1M HCl.Extract with EtOAc.Organic layer with 5% citric acid and salt water washing, is used MgSO 4Drying is filtered and evaporation.Remaining white solids (148mg, 91% productive rate) need not be further purified and be used for next linked reaction.
LC-MS (retention time: 3.14min, method B), MS m/z 551 (M ++ H).
Step 3.
At 0 ℃ to (2S; 4R)-4-(biphenyl-4-yl)-1-((S)-2-(tert-butoxycarbonyl amino) ninth of the ten Heavenly Stems-8-enoyl-)-4-methoxyl group tetramethyleneimine-2-formic acid (78mg; 0.142mmol), (1R; 2S)-and 1-amino-N-(cyclopropyl alkylsulfonyl)-2-vinyl cyclopropane carboxamide, toluenesulphonic acids, hydrate (65.5mg; 0.156mmol) and HATU (77mg; 0.212mmol) add N in the slurries in DCM (3ml), and the N-diisopropylethylamine (0.074ml, 0.425mmol).With final mixture in stirred overnight at room temperature.With the DCM dilution, with 5% citric acid and salt water washing, use MgSO 4Drying is filtered and evaporation.Resistates by preparation HPLC purifying, is obtained compound 107 (64mg, 59% productive rate), be white solid.
1H NMR(CD 3OD)δ1.11-1.12(m,2H),1.26-1.28(m,2H),1.37-1.51(m,15H),1.64-1.72(m,1H),1.87-1.89(m,2H),2.08-2.11(m,2H),2.28-2.31(m,1H),2.95-2.99(m,1H),3.09(s,3H),4.19(d,J=9Hz,1H),4.38-4.41(m,2H),4.95(d,J=12Hz,1H),5.03(d,J=18.5Hz,1H),5.15(d,J=12Hz,1H),5.32(d,J=18.5Hz,1H),5.75-5.84(m,2H),7.38-7.39(m,1H),7.45-7.48(m,2H),7.53-7.58(m,2H),7.63-7.71(m,4H);
LC-MS (retention time: 3.15min, method B), MS m/z 763 (M ++ H).
Embodiment 108: preparation compound 108
Figure G2007800499569D01031
Compound 108
Reaction scheme 4
Figure G2007800499569D01032
Step 1.
(40.0mg 1.000mmol) uses hexane wash, and stirs in DMF (5mL) with sodium hydride 60% oily dispersion liquid.0 ℃ of disposable adding solid form (2S, 4R)-4-(biphenyl-4-yl)-4-hydroxyl pyrrolidine-1,2-dioctyl phthalate 1-benzyl ester 2-methyl esters (216mg, 0.5mmol).Formed pale yellow solution was stirred 30 minutes under this temperature, add then bromotoluene (0.072mL, 0.600mmol).Final solution was stirred 2 hours at 0 ℃.5% citric acid stopped reaction with ice-cold extracts with EtOAc.With organic layer salt water washing, use MgSO 4Drying is filtered and evaporation.Resistates by the silicagel column purifying, is used 4: 1,3: 1 and 2: 1 hexane-EtOAc wash-outs successively, obtained the required product of 64mg (24%yield), be non-enantiomer mixture and 150mg raw material.
1H NMR(CDCl 3)δ2.28-2.34(m,0.5H),2.51-2.60(m,0.5H),2.90-3.02(m,1H),3.41,3.55,3.56,3.78(s,3H),3.82-3.99(m,1H),4.11-4.23(m,3H),4.41-4.70(m,1H),5.09-5.29(m,2H),7.17-7.63(m,19H);
LC-MS (retention time: 3.18min, method B), MS m/z 522 (M ++ H).
Step 2.
To containing (4R)-4-(benzyloxy)-4-(biphenyl-4-yl) tetramethyleneimine-1,2-dioctyl phthalate 1-benzyl ester 2-methyl esters (58mg, 0.111mmol) be added in the palladium (12mg on the carbon in the ice-cold Parr shaking flasks of solution in ethyl acetate (1mL) and MeOH (1mL), 0.011mmol) (10%, wet).This container was being placed 3 hours under 25psi pressure hydrogen on the Parr wobbler device.Handle with diatomite, filter, evaporation has obtained required product (38mg, 88% productive rate), is pale powder shape thing.Products therefrom need not be further purified and be used for next linked reaction.
LC-MS (retention time: 2.36min, method B), MS m/z 388 (M ++ H).
Step 3.
At 0 ℃ to (4R)-4-(benzyloxy)-4-(biphenyl-4-yl) tetramethyleneimine-2-methyl-formiate (60mg, 0.155mmol), (S)-2-(tert-butoxycarbonyl amino)-3,3-acid dimethyl (39.3mg, 0.17mmol) and HATU (84mg, 0.232mmol) add N in the solution in DCM (2ml), the N-diisopropylethylamine (0.081ml, 0.465mmol).With formed solution in stirred overnight at room temperature.With the DCM dilution, with 5% citric acid and salt water washing, use MgSO 4Drying is filtered and evaporation.Resistates by the silicagel column purifying, with 3: 1 hexane-acetone wash-outs, is obtained required product (77mg, 83% productive rate), be white foam shape thing.
1H NMR(CDCl 3)δ1.04,1.11(s,9H),1.42,1.43(s,9H),2.38-2.40(m,0.5H),2.55-2.60(m,0.5H),2.90-3.02(m,1H),3.52,3.75(s,3H),4.10-4.18(m,2H),4.24-4.26(m,1H),4.34-4.38(m,1H),4.63-4.64(m,0.5H),4.80-4.82(m,0.5H),5.20-5.29(m,1H),7.16-7.61(m,14H);
LC-MS (retention time: 3.28min, method B), MS m/z 601 (M ++ H).
Step 4.
To (4R)-4-(benzyloxy)-4-(biphenyl-4-yl)-1-((S)-2-(tert-butoxycarbonyl amino)-3; 3-dimethyl butyrate acyl group) tetramethyleneimine-2-methyl-formiate (74mg; 0.123mmol) add previously prepared lithium hydroxide monohydrate (15.5mg, 0.370mmol) solution in water (1mL) in the solution in THF (1mL) and MeOH (1mL).With formed turbid solution in stirred overnight at room temperature.Vacuum is removed volatile matter.Resistates is placed water, be acidified to pH 2 with 1M HCl.Extract with EtOAc.Organic layer with 5% citric acid and salt water washing, is used MgSO 4Drying is filtered and evaporation.Remaining white solids (58mg, 80% productive rate) need not be further purified and be used for next linked reaction.
LC-MS (retention time: 3.19min, method B), MS m/z 587 (M ++ H).
Step 5.
At 0 ℃ to (4R)-4-(benzyloxy)-4-(biphenyl-4-yl)-1-((S)-2-(tert-butoxycarbonyl amino)-3; 3-dimethyl butyrate acyl group) tetramethyleneimine-2-formic acid (63mg; 0.107mmol), (1R; 2S)-1-amino-N-(cyclopropyl alkylsulfonyl)-2-vinyl cyclopropane carboxamide; toluenesulphonic acids; hydrate (49.7mg; 0.118mmol) and HATU (58.0mg; 0.161mmol) add N in the slurries in methylene dichloride (2mL); the N-diisopropylethylamine (0.056mL, 0.322mmol).With final mixture in stirred overnight at room temperature.With the DCM dilution, with 5% citric acid and salt water washing, use MgSO 4Drying is filtered and evaporation.Resistates by preparation HPLC purifying (MeOH-water is as eluent), is obtained diastereomer mixture of products (48mg, 55.9% productive rate), be white solid.This solid is experienced another preparation HPLC condition (acetonitrile-water is as eluent) again, obtained homochiral product (11mg).
1H NMR(CD 3OD)δ1.05-1.15(m 11H),1.25-1.31(m,2H),1.47-1.50(m,10H),1.80-1.82(m,1H),2.15-2.20(m,1H),2.57-2.70(m,2H),2.94-2.98(m,1H),4.08-4.19(m,2H),4.30(s,2H),4.45(d,J=9Hz,1H),4.72-4.76(m,1H),5.13(d,J=12Hz,1H),5.28(d,J=18.5Hz,1H),5.75-5.82(m,1H),7.29-7.34(m,6H),7.38-7.40(m,2H),7.63-7.69(m,6H);
LC-MS (retention time: 3.22min, method B), MS m/z 799.5 (M ++ H).
Embodiment 109: preparation compound 109
Figure G2007800499569D01061
About 1: 1 mixture of P1 isomer
Compound 109
Step 1: preparation (E/Z)-2-(benzyloxycarbonyl amino)-4,4-difluoro but-2-ene acid methyl esters
-78 ℃ under nitrogen to potassium tert.-butoxide (3.91g, 34.9mmol) add in the suspension in tetrahydrofuran (THF) (50mL) 2-(benzyloxycarbonyl amino)-2-(dimethoxy phosphoryl) methyl acetate (10.5g, 31.7mmol).Stirring at room 30 minutes, then with 1-oxyethyl group-2, (7.99g 63.4mmol) added the 2-difluoroethanol lentamente with it.The gained mixture is warmed to room temperature, and stirred 18 hours.The water stopped reaction is adjusted to pH=5 by adding several 1N HCl then.This mixture is extracted with EtOAc.With organic layer MgSO 4Drying is filtered and is concentrated, and has obtained the 10g crude product, is yellow oil.By the Biotage purifying, with 30%EtOAc/ hexane wash-out, obtained 7.12g (79%) of (E/Z) methyl 2-(benzyloxycarbonyl amino)-4,4-difluoro but-2-ene acid esters is oily matter.
LC-MS, about 1: 8 isomer mixture.LC-MS,MS m/z 286(M+1)。
Main isomer: 1H NMR (500MHz, CDCl 3) δ ppm 3.84 (s, 3H), 5.18 (s, 2H), 6.21-6.43 (m, 1H), 6.70 (dt, J 1=55.2Hz, J 2=5.19,1H), 6.90-7.05 (brs, 1H), 7.31-7.50 (m, 5H).
Step 2: preparation (E/Z)-2-((benzyloxycarbonyl) (tert-butoxycarbonyl) amino)-4,4-difluoro fourth -2-olefin(e) acid methyl esters, as follows
Figure G2007800499569D01071
With (E/Z)-2-(benzyloxycarbonyl amino)-4,4-difluoro but-2-ene acid methyl esters (19g, 66.6mmol), tert-Butyl dicarbonate (29.1g, 133mmol) and the DMAP of catalytic amount (0.814g, 6.66mmol) solution in tetrahydrofuran (THF) (200mL) was stirring at room 2 hours.With the EtOAc dilution, wash with water then, use MgSO 4Drying is filtered and is concentrated, and has obtained 39.1g of crude product, is colorless oil.By the Biotage purifying, with 25%EtOAc/ hexane wash-out, obtained 22g (86%) of (E/Z)-methyl 2-((benzyloxycarbonyl) (tert-butoxycarbonyl) amino)-4,4-difluoro but-2-ene acid esters is colorless oil.
LC-MS,MS m/z 408(M+1+Na)。Main isomer: 1H NMR (500MHz, CDCl 3) δ ppm 1.45 (s, 9H), 3.73 (s, 3H), 5.21 (s, 2H), 6.12-6.39 (dt, J 1=54.3Hz, J 2=6.10Hz, 1H), 6.72-6.81 (m, 1H), 7.29-7.41 (m, 5H).
Step 3: preparation 1-((benzyloxycarbonyl) (tert-butoxycarbonyl) amino)-2-(difluoromethyl) ring third The alkane methyl-formiate
Figure G2007800499569D01072
To sodium hydride (0.850g, 21.26mmol) add in the mixture in DMSO (50ml) the iodate trimethylsulfonium (4.90g, 22.25mmol), and with this mixture stirring at room 1 hour.Add (E/Z)-2-((benzyloxycarbonyl) (tert-butoxycarbonyl) amino)-4 then, 4-difluoro but-2-ene acid methyl esters (6.4g, 16.61mmol), and with this mixture 80 ℃ of heating 2 hours.Then this reaction is cooled to room temperature, and stirred 18 hours.Add water (10mL), add 1N HCl then to regulate pH to 5.(3 * 250mL) extract with EtOAc with this mixture.With organic layer water (20mL) washing, use MgSO 4Drying is filtered, and concentrates, and has obtained 7g of crude product, is yellow oil.By the Biotage purifying, with 15%EtOAc/ hexane wash-out, obtained 0.8g 1-((benzyloxycarbonyl) (tert-butoxycarbonyl) amino)-2-(difluoromethyl) cyclopropane-carboxylic acid methyl esters, be oily matter.LC-MS,MS m/z 422(M+1+Na)。
Also obtained other 1.35g from the Biotage purifying and taken off-the Cbz product, 1-(tert-butoxycarbonyl amino)-2-(difluoromethyl) cyclopropane-carboxylic acid methyl esters is oily matter.LC-MS,MS m/z 288(M+1+Na)。The productive rate that merges is 42%.
Step 4: preparation 1-(tert-butoxycarbonyl amino)-2-(difluoromethyl) cyclopropane-carboxylic acid
Figure G2007800499569D01081
With 1-((benzyloxycarbonyl) (tert-butoxycarbonyl) amino)-2-(difluoromethyl) cyclopropane-carboxylic acid methyl esters (1.05g, 2.63mmol), 1-(tert-butoxycarbonyl amino)-2-(difluoromethyl) cyclopropane-carboxylic acid methyl esters (1.43g, 5.39mmol) (10mL, 40.0mmol) mixture in was stirring at room 18 hours at MeOH (20mL) and sodium hydroxide.This reaction mixture is concentrated, and use 1N HCl to be adjusted to pH=4.From this solution, be settled out white solid, collected by filtering.Filter cake is washed with water, obtained the 1.2g crude product, be white solid.Crystallization from hexane/EtOAc has obtained 1.0g (50%) 1-(tert-butoxycarbonyl amino)-2-(difluoromethyl) cyclopropane-carboxylic acid, is white solid.Isolate this intermediate, be independent diastereomer, wherein CF 2H substituting group and carboxyl are cis.
LC-MS,MS m/z 274(M+1+Na)。
1H NMR(300MHz,d4-MeOH)δppm 1.31-1.54(m,10H)1.66-1.86(m,1H)1.85-2.10(m,1H)5.92(t,J=55.81Hz,1H)。
Step 5: the amino first of preparation 1-(cyclopropyl sulfuryl amino formyl radical)-2-(difluoromethyl) cyclopropyl Tert-butyl acrylate
With 1-(tert-butoxycarbonyl amino)-2-(difluoromethyl) cyclopropane-carboxylic acid (975mg, 3.88mmol) and CDI (755mg, 4.66mmol) mixture heating up in tetrahydrofuran (THF) (10mL) refluxed 1 hour.Then it is cooled to room temperature, add afterwards the cyclopropane sulphonamide (564mg, 4.66mmol), add then DBU (0.702mL, 4.66mmol).With this mixture stirring at room 18 hours.Then with its vacuum concentration, dilute with water, and use 1N HCl to be adjusted to pH=4.This acid solution is extracted water (5 * 15mL) washings, dry (MgSO with EtOAc 4) and vacuum concentration, obtained the 1.6g crude product, be white solid.Recrystallization from hexane/EtOAc has obtained 1.15g (84%) 1-(cyclopropyl sulfuryl amino formyl radical)-2-(difluoromethyl) cyclopropyl t-butyl carbamate, is white solid.LC-MS,MS m/z377(M+1+Na)。
1H NMR(500MHz,d4-MeOH)δppm 1.03-1.17(m,2H),1.18-1.25(m,1H),1.25-1.35(m,1H),1.39-1.46(m,J=5.49Hz,1H),1.49(s,9H),1.90-2.03(m,1H),2.04-2.20(m,1H),2.99(s,1H),5.50-5.89(m,1H)。
Step 6: preparation 1-amino-N-(cyclopropyl alkylsulfonyl)-2-(difluoromethyl) cyclopropane carboxamide, HCl salt
Figure G2007800499569D01092
HCl salt
With 1-(cyclopropyl sulfuryl amino formyl radical)-2-(difluoromethyl) cyclopropyl t-butyl carbamate (1.1g, 3.10mmol) and the mixture of 4N HCl/ dioxane (10mL) stirring at room 2 hours.Then this reaction mixture is concentrated, obtained 0.93g (103%) 1-amino-N-(cyclopropyl alkylsulfonyl)-2-(difluoromethyl) cyclopropane carboxamide, HCl salt is white solid.LC-MS,MS m/z 277(M+1+Na)。 1H NMR(500MHz,d4-MeOH)δppm1.07-1.21(m,2H)1.20-1.37(m,2H)1.70-1.85(m,1H)2.21-2.36(m,2H)2.96-3.11(m,1H)5.86-6.13(dt,J 1=55.24Hz,J 2=5.19Hz,1H)。
Step 7: preparation compound 109:
With (2S; 4R)-4-(biphenyl-4-yl)-1-((S)-2-(tert-butoxycarbonyl amino)-3; 3-dimethyl butyrate acyl group)-4-methoxyl group tetramethyleneimine-2-formic acid (30mg; 0.059mmol), 1-amino-N-(cyclopropyl alkylsulfonyl)-2-(difluoromethyl) cyclopropane carboxamide; HCl salt (17.08mg; 0.059mmol), HATU (26.8mg, 0.071mmol) and the 1-methyl piperidine (29.1mg, 0.294mmol) mixture in DCM (3mL) was stirring at room 18 hours.Then with its vacuum concentration.Resistates is distributed between EtOAc (50mL) and 5mL water.Use 1N HCl to be adjusted to pH=4.Then with organic phase water (3 * 5mL) washings, dry (MgSO 4), filter and concentrate, obtained the 50mg crude product, be white solid.By preparation HPLC purifying, obtained 38mg compound 109, be in white solid (mixtures of two kinds of P1 diastereomers of~1: 1).LC-MS,MS m/z 747(M+1)。
1H NMR(500MHz,d4-MeOH)δppm 0.82-1.68(m,23H),1.89-1.91(m,2H),2.41-2.74(m,2H),2.92-3.05(m,1H),3.05-3.21(m,3H),3.98-4.17(m,1H),4.13-4.29(m,1H),4.27-4.49(m,1H),4.49-4.73(m,1H),5.67-6.14(m,1H),7.38(t,J=7.32Hz,1H),7.47(t,J=7.32Hz,2H),7.52-7.81(m,6H)。
Embodiment 110: preparation compound 110
Figure G2007800499569D01111
Compound 110
Intermediate 111: preparation compound 111
Figure G2007800499569D01112
Compound 111
Figure G2007800499569D01113
Compound 111
To containing (S)-1-((2S; 4R)-4-(biphenyl-4-yl)-2-((1R; 2S)-1-(cyclopropyl sulfuryl amino formyl radical)-2-vinyl cyclopropyl formamyl)-4-methoxyl group tetramethyleneimine-1-yl)-3; be added in the palladium (2.061mg on the carbon in the ice-cold Parr shaking flasks of the 3-dimethyl-1-oxo fourth-solution of 2-aminocarbamic acid tert-butyl ester ester in EtOAc (1mL); μ mol) 1.937 (10%, wet).This container was being placed 2 hours under 10psi pressure hydrogen on the Parr wobbler device.Handle with diatomite, filter, evaporation.Resistates by the prep-HPLC purifying, is obtained required product (6mg, 43% productive rate), be white solid.
1H NMR(CD 3OD)δ0.98-1.02(m,4H),1.10-1.14(m,10H),1.26-1.35(m,4H),1.29-1.62(m,12H),2.48-2.51(m,1H),2.55-2.61(m,1H),2.95-2.99(m,1H),3.11(s,3H),4.07-4.09(m,1H),4.22-4.24(m,1H),4.42(s,1H),4.58-4.60(m,1H),7.38-7.39(m,1H),7.45-7.48(m,2H),7.57-7.67(m,6H);
LC-MS (retention time: 3.00min, method B), MS m/z 725 (M ++ H), 693 (M +-MeOH).
Embodiment 200: preparation compound 200
Figure G2007800499569D01121
Compound 200
Step 1: preparation (S)-3,3-dimethyl-2-(2,2,2-, three fluoro-1,1-dimethyl-ethoxy carbonyl amino)-butyric acid
Step 1a: preparation carbonic acid pyridine-2-base ester 2,2,2-three fluoro-1,1-dimethyl-ethyl ester
Figure G2007800499569D01123
To KH (1.03g, 25.8mmol) add in the slurries in THF (70mL) 1,1,1-, three fluoro-2-methyl propan-2-ols (3g, 23.42mmol).This is reflected at 0 ℃ stirred 20 minutes.Then with carbonic acid two pyridines-(5.06g, 23.42mmol) solution in THF (30mL) is added in this mixture 2-base ester.With this mixture in stirred overnight at room temperature.From this solution, obtained a collection of solid.With this solid filtering, and wash with EtOAc (30mL).With the THF/EtOAc solution with water washing that merges.With organic layer Na 2SO 4Drying is filtered and is concentrated.Gained white solid (1.24g, 21%) is directly used in next step.
Step 1b: preparation (S)-3,3-dimethyl-2-(2,2,2-, three fluoro-1,1-dimethyl-ethoxy carbonyl amino)-methyl-butyrate
Figure G2007800499569D01131
To (S)-2-amino-3,3-acid dimethyl methyl esters (0.411g 2.262mmol) adds N in the ice-cold solution in DCM (20mL), and the N-diisopropylethylamine (1.182mL, 6.79mmol).With formed solution stirring 5 minutes, add carbonic acid pyridine-2-base ester 2,2 then, 2-three fluoro-1, the solution of 1-dimethyl-ethyl ester (1.24g, 4.98mmol derive from step 1a) in DCM (10mL).With final pale yellow solution in stirred overnight at room temperature.This reaction mixture with 5% aqueous citric acid solution, the 0.1M NaOH aqueous solution and salt water washing, is used Na then 2SO 4Drying is filtered, and is evaporated to dried.Resistates by the silicagel column purifying, with (4: 1) hexane-EtOAc wash-out, has been obtained colorless oil, be product (673mg, 99%).
1H-NMR(CDCl 3-d)δ0.96(s,9H),1.65(s,6H),3.73(s,3H),.5.31(m,1H);LCMS RT=2.72min,[M+Na] +=322.3。
Step 1c: preparation (S)-3,3-dimethyl-2-(2,2,2-, three fluoro-1,1-dimethyl-ethoxy carbonyl amino)-butyric acid
Figure G2007800499569D01132
To (S)-3,3-dimethyl-2-(2,2,2-three fluoro-1,1-dimethyl-ethoxy carbonyl amino)-methyl-butyrate (673mg, 2.249mmol, derive from step 1b) and add previously prepared lithium hydroxide monohydrate (189mg, 4.50mmol) solution in water (4.00mL) in the solution in THF (4mL).Should the muddiness white solution stirring at room 5 hours.Again the solution of 100mg LiOH in 1mL water is added in this mixture, and with this solution in stirred overnight at room temperature.Remove volatile matter, and this reaction mixture is diluted with 5% aqueous citric acid solution, with EtOAc (3 * 10mL) extractions.With organic layer salt water washing, use Na 2SO 4Drying is filtered and evaporation.Need not be further purified and be used for the next step reaction as the crude product (325mg, 51%) of white solid.
LCMS RT=2.55min,[M+Na] +=308.2。
Step 2: preparation (2S, 4R)-4-biphenyl-4-base-1-[(S)-3,3-dimethyl-2-(2,2,2-, three fluoro-1,1-dimethyl-ethoxy carbonyl amino)-butyryl radicals]-4-methoxyl group-tetramethyleneimine-2-methyl-formiate
Figure G2007800499569D01141
This compound is the same procedure of describing according among the embodiment 106, difference is to use (S)-3 in step 5,3-dimethyl-2-(2,2,2-three fluoro-1,1-dimethyl-ethoxy carbonyl amino)-and butyric acid replacement (S)-2-(tert-butoxycarbonyl amino)-3, the 3-acid dimethyl makes as raw material.
1H NMR(CD 3OD)δ1.11(s,9H),1.57(s,3H),1.62(s,3H),2.59(m,1H),2.76(m,1H),2.94(s,3H),3.71(s,3H),4.12(m,1H),4.21(m,1H),4.29(m,1H),4.72(m,1H),7.31-7.68(m,9H);LCMS RT=3.33min,[M+Na] +=601.2。
Step 3: preparation (2S, 4R)-4-biphenyl-4-base-1-[(S)-3,3-dimethyl-2-(2,2,2-, three fluoro-1,1-dimethyl-ethoxy carbonyl amino)-butyryl radicals]-4-methoxyl group-tetramethyleneimine-2-formic acid
Figure G2007800499569D01151
This compound is the same procedure of describing according in embodiment 106 steps 6; difference is to use (2S; 4R)-4-biphenyl-4-base-1-[(S)-3; 3-dimethyl-2-(2; 2,2-, three fluoro-1,1-dimethyl-ethoxy carbonyl amino)-butyryl radicals]-4-methoxyl group-tetramethyleneimine-2-methyl-formiate replacement (2S; 4R)-4-(biphenyl-4-yl)-1-((S)-2-(tert-butoxycarbonyl amino)-3,3-dimethyl butyrate acyl group)-4-methoxyl group tetramethyleneimine-2-methyl-formiate makes.
LCMS RT=3.26min,[M+Na] +=587.2。
Step 4: preparation (S)-1-[(2S; 4R)-4-biphenyl-4-base-2-((S)-(1R; 2S)-1-cyclopropane sulfonyl amino carbonyl-2-vinyl-cyclopropyl formamyl)-4-methoxyl group-tetramethyleneimine-1-carbonyl]-2; 2-dimethyl-propyl group }-carboxylamine 2; 2; 2-three fluoro-1,1-dimethyl-ethyl ester
Figure G2007800499569D01152
Compound 200
Compound 200 is same procedure of describing according in embodiment 106 steps 7; difference is to use (2S; 4R)-4-biphenyl-4-base-1-[(S)-3; 3-dimethyl-2-(2; 2,2-, three fluoro-1,1-dimethyl-ethoxy carbonyl amino)-butyryl radicals]-4-methoxyl group-tetramethyleneimine-2-formic acid replacement (2S; 4R)-4-(biphenyl-4-yl)-1-((S)-2-(tert-butoxycarbonyl amino)-3,3-dimethyl butyrate acyl group)-4-methoxyl group tetramethyleneimine-2-formic acid makes.
1H NMR(CD 3OD)δ1.12(s,9H),1.30-1.26(m,4H),1.43(m,1H),1.64(s,3H),1.69(s,3H),1.88(m,1H),2.22(m,1H),2.49(m.1H),2.62(m,1H),2.96(m,1H),3.11(s,3H),4.07(m,1H),4.23(m,1H),4.42(d,J=9Hz,1H),4.56(m,1H),5.15(d,J=12Hz,1H),5.32(d,J=18.5Hz,1H),5.74(m,1H),7.36-7.70(m,9H);LCMS RT=3.24min,[M+Na] +=799.3。
Embodiment 201: preparation compound 201
Figure G2007800499569D01161
Compound 201
Step 1: preparation (2S, 4R)-4-biphenyl-4-base-1-((S)-2-tert-butoxycarbonyl amino-3-methyl-butyryl radicals)-4-methoxyl group-tetramethyleneimine-2-methyl-formiate
This compound is the same procedure of describing according among the embodiment 106, difference is to use (S)-2-(tert-butoxycarbonyl amino)-3 Methylbutanoic acid (commercially available from Aldrich) to replace (S)-2-(tert-butoxycarbonyl amino)-3 in step 5, and the 3-acid dimethyl makes as raw material.
LCMS RT=3.22min,[M+Na] +=533.4。
Step 2: preparation (2S, 4R)-4-biphenyl-4-base-1-((S)-2-tert-butoxycarbonyl amino-3-methyl-butyryl radicals)-4-methoxyl group-tetramethyleneimine-2-formic acid
Figure G2007800499569D01171
This compound is the same procedure of describing according in embodiment 106 steps 6; difference is to use (2S; 4R)-4-biphenyl-4-base-1-((S)-2-tert-butoxycarbonyl amino-3-methyl-butyryl radicals)-4-methoxyl group-tetramethyleneimine-2-methyl-formiate replacement (2S; 4R)-4-(biphenyl-4-yl)-1-((S)-2-(tert-butoxycarbonyl amino)-3,3-dimethyl butyrate acyl group)-4-methoxyl group tetramethyleneimine-2-methyl-formiate makes.
LCMS RT=3.07min,[M+H] +=497.4。
Step 3: preparation (S)-1-[(2S; 4R)-4-biphenyl-4-base-2-((S)-(1R, 2S)-1-cyclopropane sulfonyl amino carbonyl-2-vinyl-cyclopropyl formamyl)-4-methoxyl group-tetramethyleneimine-1-carbonyl]-2-methyl-propyl group }-t-butyl carbamate
Figure G2007800499569D01172
Compound 201
Compound 201 is same procedure of describing according in embodiment 106 steps 7; difference is to use (2S; 4R)-4-biphenyl-4-base-1-((S)-2-tert-butoxycarbonyl amino-3-methyl-butyryl radicals)-4-methoxyl group-tetramethyleneimine-2-formic acid replacement (2S; 4R)-4-(biphenyl-4-yl)-1-((S)-2-(tert-butoxycarbonyl amino)-3,3-dimethyl butyrate acyl group)-4-methoxyl group tetramethyleneimine-2-formic acid makes.
1H NMR(CD 3OD)δ0.87(m,1H),0.93(m,1H),1.00(m,4H),1.07(m,2H),1.18-1.30(m,4H),1.38(m,2H),1.43(m,6H),1.85(m,1H),2.08(m,1H),2.22(m,1H),2.49(m.1H),2.53(m,1H),2.96(m,1H),3.07(s,3H),4.06(m,1H),4.12(m,1H),4.22(t,1H),4.54(d,J=9Hz,1H),5.10(d,J=12Hz,1H),5.27(d,J=18.5Hz,1H),5.74(m,1H),7.36-7.70(m,9H);LCMS RT=3.11min,[M+Na] +=731.2。
Embodiment 202: preparation compound 202
Figure G2007800499569D01181
Compound 102
Step 1: preparation (2S, 4R)-4-biphenyl-4-base-1-((S)-2-tert-butoxycarbonyl amino-3-methyl-pentanoyl)-4-methoxyl group-tetramethyleneimine-2-methyl-formiate
Figure G2007800499569D01182
This compound is the same procedure of describing according among the embodiment 106, difference is to use (2S)-2-(tert-butoxycarbonyl amino)-3 methylvaleric acid (commercially available from Aldrich) to replace (S)-2-(tert-butoxycarbonyl amino)-3 in step 5, and the 3-acid dimethyl makes as raw material.
LCMS RT=3.27min,[M+Na] +=547.4。
Step 2: preparation (2S, 4R)-4-biphenyl-4-base-1-((S)-2-tert-butoxycarbonyl amino-3-methyl-pentanoyl)-4-methoxyl group-tetramethyleneimine-2-formic acid
Figure G2007800499569D01191
This compound is the same procedure of describing according in embodiment 106 steps 6; difference is to use (2S; 4R)-4-biphenyl-4-base-1-((S)-2-tert-butoxycarbonyl amino-3-methyl-butyryl radicals)-4-methoxyl group-tetramethyleneimine-2-methyl-formiate replacement (2S; 4R)-4-(biphenyl-4-yl)-1-((S)-2-(tert-butoxycarbonyl amino)-3,3-dimethyl butyrate acyl group)-4-methoxyl group tetramethyleneimine-2-methyl-formiate makes.
LCMS RT=3.15min,[M+H] +=511.5。
Step 3: preparation (S)-1-[(2S; 4R)-4-biphenyl-4-base-2-((S)-(1R, 2S)-1-cyclopropane sulfonyl amino carbonyl-2-vinyl-cyclopropyl formamyl)-4-methoxyl group-tetramethyleneimine-1-carbonyl]-2-methyl-butyl }-t-butyl carbamate
Figure G2007800499569D01192
Compound 202
Compound 202 is same procedure of describing according in embodiment 106 steps 7; difference is to use (2S; 4R)-4-biphenyl-4-base-1-((S)-2-tert-butoxycarbonyl amino-3-methyl-pentanoyl)-4-methoxyl group-tetramethyleneimine-2-formic acid replacement (2S; 4R)-4-(biphenyl-4-yl)-1-((S)-2-(tert-butoxycarbonyl amino)-3,3-dimethyl butyrate acyl group)-4-methoxyl group tetramethyleneimine-2-formic acid makes.
1H NMR(CD 3OD)δ0.91-0.97(m,4H),1.03(m,2H),1.10(m,2H),1.20-1.34(m,5H),1.40-1.50(m,9H),1.68(m,1H),1.88(m,1H),2.25(m.1H),2.56(m,2H),2.98(m,1H),3.11(s,3H),4.10(m,1H),4.22(m,2H),4.63(d,J=9Hz,1H),5.15(d,J=12Hz,1H),5.32(d,J=18.5Hz,1H),5.74(m,1H),7.36-7.70(m,9H);LCMS RT=3.24min,[M+Na] +=745.3。
Embodiment 203: preparation compound 203
Figure G2007800499569D01201
Compound 203
Step 1: preparation (2S, 4R)-4-biphenyl-4-base-1-((S)-2-tert-butoxycarbonyl amino-4-methyl-pentanoyl)-4-methoxyl group-tetramethyleneimine-2-methyl-formiate
This compound is the same procedure of describing according among the embodiment 106, difference is to use (S)-2-(tert-butoxycarbonyl amino)-4-methylvaleric acid (commercially available from Aldrich) to replace (S)-2-(tert-butoxycarbonyl amino)-3 in step 5, and the 3-acid dimethyl makes as raw material.
LCMS RT=3.26min,[M+Na] +=547.4。
Step 2: preparation (2S, 4R)-4-biphenyl-4-base-1-((S)-2-tert-butoxycarbonyl amino-4-methyl-pentanoyl)-4-methoxyl group-tetramethyleneimine-2-formic acid
This compound is the same procedure of describing according in embodiment 106 steps 6; difference is to use (2S; 4R)-4-biphenyl-4-base-1-((S)-2-tert-butoxycarbonyl amino-3-methyl-butyryl radicals)-4-methoxyl group-tetramethyleneimine-2-methyl-formiate replacement (2S; 4R)-4-(biphenyl-4-yl)-1-((S)-2-(tert-butoxycarbonyl amino)-3,3-dimethyl butyrate acyl group)-4-methoxyl group tetramethyleneimine-2-methyl-formiate makes.
LCMS RT=3.16min,[M+Na] +=533.5。
Step 3: preparation (S)-1-[(2S; 4R)-4-biphenyl-4-base-2-((S)-(1R, 2S)-1-cyclopropane sulfonyl amino carbonyl-2-vinyl-cyclopropyl formamyl)-4-methoxyl group-tetramethyleneimine-1-carbonyl]-3-methyl-butyl }-t-butyl carbamate
Figure G2007800499569D01212
Compound 203
Compound 203 is same procedure of describing according in embodiment 106 steps 7; difference is to use (2S; 4R)-4-biphenyl-4-base-1-((S)-2-tert-butoxycarbonyl amino-4-methyl-pentanoyl)-4-methoxyl group-tetramethyleneimine-2-formic acid replacement (2S; 4R)-4-(biphenyl-4-yl)-1-((S)-2-(tert-butoxycarbonyl amino)-3,3-dimethyl butyrate acyl group)-4-methoxyl group tetramethyleneimine-2-formic acid makes.
1H NMR(CD 3OD)δ0.91-0.98(m,2H),1.02(m,4H),1.24-1.32(m,6H),1.39-1.48(m,9H),1.64(m,1H),1.76(m,1H),1.88(m,1H),2.30(m.1H),2.62(m,2H),2.98(m,1H),3.11(s,3H),4.11(m,1H),4.18(d,J=9Hz,1H),4.37(m,1H),4.49(m,1H),5.15(d,J=12Hz,1H),5.32(d,J=18.5Hz,1H),5.74(m,1H),7.36-7.70(m,9H);LCMS RT=3.24min,[M+Na] +=745.3。
Embodiment 204: preparation compound 204
Figure G2007800499569D01221
Compound 204
Step 1a: preparation (S)-2-(diamantane-1-base oxygen base carbonylamino)-3,3-dimethyl-methyl-butyrate
To (S)-2-amino-3,3-acid dimethyl methyl esters (200mg, 1.377mmol) and DIEA (0.481mL, 2.75mmol) add in the mixture in DCM (2mL) solution fluorine formic acid 1-adamantane esters (334mg, 1.515mmol).With this reaction mixture in stirred overnight at room temperature.Compound has obtained colorless oil (358mg, 68%) as product by silica gel chromatography (20%EtOAc/80% hexane).
1H-NMR(CDCl 3-d)δ0.94(s,9H),1.62(s,6H),2.07(s,6H),2.13(s,3H),3.70(s,3H),.4.07(d,J=9Hz,1H),5.09(broad s,1H,NH);LCMS RT=3.05min,[M+H] +=324.4。
Step 1b: preparation (S)-2-(diamantane-1-base oxygen base carbonylamino)-3,3-dimethyl-butyric acid
Figure G2007800499569D01231
To (S)-2-(diamantane-1-base oxygen base carbonylamino)-3,3-dimethyl-methyl-butyrate (358mg, 1.107mmol) add previously prepared lithium hydroxide monohydrate (93mg, 2.214mmol) solution in water (4.00mL) in the solution in THF (4mL) and MeOH (4mL).Should the muddiness white solution stirring at room 5 hours.Remove volatile matter, and this reaction mixture is diluted with rare citric acid solution, with EtOAc (3 * 10mL) extractions.With organic layer salt water washing, use Na 2SO 4Drying is filtered and evaporation, has obtained white solid (291.4mg, 85%) as product.This crude product need not be further purified direct use.
LCMS RT=2.91min,[M+H] +=310.4。
Step 2: preparation (2S, 4R)-1-[(S)-2-(diamantane-1-base oxygen base carbonylamino)-3,3-dimethyl-butyryl radicals]-4-biphenyl-4-base-4-methoxyl group-tetramethyleneimine-2-methyl-formiate
Figure G2007800499569D01232
This compound is the same procedure of describing according in embodiment 106 steps 5, difference is to use (S)-2-(diamantane-1-base oxygen base carbonylamino)-3,3-dimethyl-butyric acid replaces (S)-2-(tert-butoxycarbonyl amino)-3, and the 3-acid dimethyl makes.
LCMS RT=3.63min,[M+Na] +=625.4。
Step 3: preparation (2S, 4R)-1-[(S)-2-(diamantane-1-base oxygen base carbonylamino)-3,3-dimethyl-butyryl radicals]-4-biphenyl-4-base-4-methoxyl group-tetramethyleneimine-2-formic acid
Figure G2007800499569D01241
This compound is the same procedure of describing according in embodiment 106 steps 6; difference is to use (2S; 4R)-1-[(S)-2-(diamantane-1-base oxygen base carbonylamino)-3; 3-dimethyl-butyryl radicals]-4-biphenyl-4-base-4-methoxyl group-tetramethyleneimine-2-methyl-formiate replacement (2S; 4R)-4-(biphenyl-4-yl)-1-((S)-2-(tert-butoxycarbonyl amino)-3,3-dimethyl butyrate acyl group)-4-methoxyl group tetramethyleneimine-2-methyl-formiate makes.
LCMS RT=3.16min,[M+Na] +=611.3。
Step 4: preparation (S)-1-[(2S; 4R)-4-biphenyl-4-base-2-((S)-(1R; 2S)-1-cyclopropane sulfonyl amino carbonyl-2-vinyl-cyclopropyl formamyl)-4-methoxyl group-tetramethyleneimine-1-carbonyl]-2,2-dimethyl-propyl group }-carboxylamine diamantane-1-base ester
Figure G2007800499569D01242
Compound 204
Compound 204 is same procedure of describing according in embodiment 106 steps 7; difference is to use (2S; 4R)-1-[(S)-2-(diamantane-1-base oxygen base carbonylamino)-3; 3-dimethyl-butyryl radicals]-4-biphenyl-4-base-4-methoxyl group-tetramethyleneimine-2-formic acid replacement (2S; 4R)-4-(biphenyl-4-yl)-1-((S)-2-(tert-butoxycarbonyl amino)-3,3-dimethyl butyrate acyl group)-4-methoxyl group tetramethyleneimine-2-formic acid makes.
1H NMR(CD 3OD)δ1.00-1.10(m,10H),1.22-1.31(m,3H),1.43(m,1H),1.64-1.71(m,6H),1.88(m,1H),1.97-2.22(m,10H),2.50(m.1H),2.63(m,1H),2.98(m,1H),3.11(s,3H),4.04(d,J=9Hz,1H),4.23(m,1H),4.38(m,1H),4.57(m,1H),5.12(d,J=12Hz,1H),5.32(d,J=18.5Hz,1H),5.77(m,1H),7.33-7.67(m,9H);LCMS RT=3.43min,[M+H] +=801.4。
Embodiment 205: preparation compound 205
Figure G2007800499569D01251
Compound 205
Step 1a: preparation (S)-2-(3-diamantane-1-base-urea groups)-3,3-dimethyl-methyl-butyrate
Figure G2007800499569D01252
To (S)-2-amino-3,3-acid dimethyl methyl esters (200mg, 1.377mmol) and DIEA (0.481mL, 2.75mmol) add in the mixture in DCM (2mL) solution isocyanic acid 1-adamantane esters (334mg, 1.515mmol).With this reaction mixture in stirred overnight at room temperature.Compound has obtained white solid (265mg, 59%) as product by silica gel chromatography (20%EtOAc/80% hexane).
1H-NMR(CDCl 3-d)δ0.94(s,9H),1.64(s,6H),1.93(s,6H),2.04(s,3H),3.70(s,3H),.4.24(d,J=9Hz,1H),4.34(broad s,1H,NH),4.88(broad s,1H,NH);LCMS RT=2.87min,[M+H] +=323.4。
Step 1b: preparation (S)-2-(3-diamantane-1-base-urea groups)-3,3-dimethyl-butyric acid
Figure G2007800499569D01261
To (S)-2-(3-diamantane-1-base-urea groups)-3,3-dimethyl-methyl-butyrate (265mg, 0.822mmol) add previously prepared lithium hydroxide monohydrate (69mg, 1.644mmol) solution in water (4.00mL) in the solution in THF (4mL) and MeOH (4mL).Should the muddiness white solution stirring at room 5 hours, remove volatile matter, and this reaction mixture diluted with rare citric acid solution, with EtOAc (3 * 10mL) extractions.With organic layer salt water washing, use Na 2SO 4Drying is filtered, and evaporation has obtained white solid (232mg, 92%) as product.This crude product need not be further purified direct use.
LCMS RT=3.09min,[M+H] +=309.4。
Step 2: preparation (2S, 4R)-1-[(S)-2-(3-diamantane-1-base-urea groups)-3,3-dimethyl-butyryl radicals]-4-biphenyl-4-base-4-methoxyl group-tetramethyleneimine-2-methyl-formiate
Figure G2007800499569D01262
This compound is the same procedure of describing according in embodiment 106 steps 5, and difference is to use (S)-2-(3-diamantane-1-base-urea groups)-3, and 3-dimethyl-butyric acid replaces (S)-2-(tert-butoxycarbonyl amino)-3, and the 3-acid dimethyl makes.
LCMS RT=3.51min,[M+Na] +=624.4。
Step 3: preparation (2S, 4R)-1-[(S)-2-(3-diamantane-1-base-urea groups)-3,3-dimethyl-butyryl radicals]-4-biphenyl-4-base-4-methoxyl group-tetramethyleneimine-2-formic acid
Figure G2007800499569D01271
This compound is the same procedure of describing according in embodiment 106 steps 6; difference is to use (2S; 4R)-1-[(S)-2-(3-diamantane-1-base-urea groups)-3; 3-dimethyl-butyryl radicals]-4-biphenyl-4-base-4-methoxyl group-tetramethyleneimine-2-methyl-formiate replacement (2S; 4R)-4-(biphenyl-4-yl)-1-((S)-2-(tert-butoxycarbonyl amino)-3,3-dimethyl butyrate acyl group)-4-methoxyl group tetramethyleneimine-2-methyl-formiate makes.
LCMS RT=3.42min,[M+H] +=588.3。
Step 4: preparation (2S; 4R)-1-[(S)-2-(3-diamantane-1-base-urea groups)-3; 3-dimethyl-butyryl radicals]-4-biphenyl-4-base-4-methoxyl group-tetramethyleneimine-2-formic acid ((1R, 2S)-1-cyclopropane sulfonyl amino carbonyl-2-vinyl-cyclopropyl)-acid amides
Figure G2007800499569D01272
Compound 205
Compound 205 is same procedure of describing according in embodiment 106 steps 7; difference is to use (2S; 4R)-1-[(S)-2-(3-diamantane-1-base-urea groups)-3; 3-dimethyl-butyryl radicals]-4-biphenyl-4-base-4-methoxyl group-tetramethyleneimine-2-formic acid replacement (2S; 4R)-4-(biphenyl-4-yl)-1-((S)-2-(tert-butoxycarbonyl amino)-3,3-dimethyl butyrate acyl group)-4-methoxyl group tetramethyleneimine-2-formic acid makes.
1H NMR(CD 3OD)δ1.05-1.11(m,9H),1.23-1.33(m,5H),1.41(m,1H),1.72(s,6H),1.87(m,1H),1.97-2.05(m,8H),2.21(m,1H),2.56(m.1H),2.63(m,1H),2.95(m,1H),3.11(s,3H),4.06(d,J=9Hz,1H),4.21(m,1H),4.46(m,1H),4.67(m,1H),5.12(d,J=12Hz,1H),5.28(d,J=18.5Hz,1H),5.77(m,1H),7.33-7.67(m,9H);LCMS RT=3.40min,[M+Na] +=800.5。
Embodiment 206: preparation compound 206
Figure G2007800499569D01281
Compound 206
Step 1a: preparation (S)-2-(the 3-tertiary butyl-urea groups)-3,3-dimethyl-methyl-butyrate
Figure G2007800499569D01282
This compound is the same procedure of describing according among the embodiment 205 step 1a, and difference is to use tert-butyl isocyanate to replace isocyanic acid 1-adamantane esters to make.
LCMS RT=2.24min,[M+H] +=245.2。
Step 1b: preparation (S)-2-(the 3-tertiary butyl-urea groups)-3,3-dimethyl-butyric acid
This compound is the same procedure of describing according among the embodiment 205 step 1b, difference is to use (S)-2-(the 3-tertiary butyl-urea groups)-3,3-dimethyl-methyl-butyrate replaces (S)-2-(3-diamantane-1-base-urea groups)-3, and 3-dimethyl-methyl-butyrate makes.
1H-NMR(CDCl 3-d)δ1.00(s,9H),1.56(s,9H),3.49(s,1H);LCMSRT=0.96min,[M+H] +=231.3。
Step 2: preparation (2S, 4R)-4-biphenyl-4-base-1-[(S)-2-(the 3-tertiary butyl-urea groups)-3,3-dimethyl-butyryl radicals]-4-methoxyl group-tetramethyleneimine-2-methyl-formiate
Figure G2007800499569D01291
This compound is the same procedure of describing according among the embodiment 106, and difference is to use (S)-2-(the 3-tertiary butyl-urea groups)-3 in step 5, and 3-dimethyl-butyric acid makes as raw material.
LCMS RT=3.26min,[M+Na] +=546.3。
Step 2: preparation (2S, 4R)-4-biphenyl-4-base-1-[(S)-2-(the 3-tertiary butyl-urea groups)-3,3-dimethyl-butyryl radicals]-4-methoxyl group-tetramethyleneimine-2-formic acid
Figure G2007800499569D01292
This compound is the same procedure of describing according in embodiment 106 steps 6; difference is to use (2S; 4R)-4-biphenyl-4-base-1-[(S)-2-(the 3-tertiary butyl-urea groups)-3; 3-dimethyl-butyryl radicals]-4-methoxyl group-tetramethyleneimine-2-methyl-formiate replacement (2S; 4R)-4-(biphenyl-4-yl)-1-((S)-2-(tert-butoxycarbonyl amino)-3,3-dimethyl butyrate acyl group)-4-methoxyl group tetramethyleneimine-2-methyl-formiate makes.
LCMS RT=3.14min,[M+Na] +=532.3。
Step 3: preparation (2S, 4R)-4-biphenyl-4-base-1-[(S)-2-(the 3-tertiary butyl-urea groups)-3,3-dimethyl-butyryl radicals]-4-methoxyl group-tetramethyleneimine-2-formic acid ((1R, 2S)-1-cyclopropane sulfonyl amino carbonyl-2-vinyl-cyclopropyl)-acid amides
Compound 138
Compound 206 is same procedure of describing according in embodiment 106 steps 7; difference is to use (2S; 4R)-4-biphenyl-4-base-1-[(S)-2-(the 3-tertiary butyl-urea groups)-3; 3-dimethyl-butyryl radicals]-4-methoxyl group-tetramethyleneimine-2-formic acid replacement (2S; 4R)-4-(biphenyl-4-yl)-1-((S)-2-(tert-butoxycarbonyl amino)-3,3-dimethyl butyrate acyl group)-4-methoxyl group tetramethyleneimine-2-formic acid makes.
1H NMR(CDCl 3-d)δ1.03-1.08(m,12H),1.23-1.38(m,11H),1.75(m,1H),1.92(m,1H),2.08(m.1H),2.47(m,1H),2.96(m,1H),3.11(s,3H),4.10(m,1H),4.49(m,1H),4.53(d,J=9Hz,1H),4.80(m,1H),5.11(d,J=12Hz,1H),5.28(d,J=18.5Hz,1H),5.74(m,1H),7.57-7.61(m,4H),7.40-7.46(m,5H);LCMS RT=3.26min,[M+H] +=722.4。
Embodiment 207: preparation compound 207 and compound 208
Figure G2007800499569D01311
Compound 207
Figure G2007800499569D01312
Compound 208
Step 1: preparation 2-(3-methoxyl group-phenyl amino)-3,3-dimethyl-butyric acid
Figure G2007800499569D01313
Room temperature to the 3-anisidine (100mg, 0.812mmol) and the trimethylammonium pyruvic acid (211mg, 1.624mmol) add in the mixture in methyl alcohol (3mL) acetic acid (0.930mL, 16.24mmol).With this mixture stirring at room 30 minutes, then 70 ℃ of heating 2 hours.This mixture is cooled to room temperature, add then cyano group three sodium borohydrides (1.624mL, 1.624mmol).With this mixture in stirred overnight at room temperature.Add water (10mL), and (3 * 10mL) extract with ethyl acetate with this mixture.With the saturated NaHCO of organic layer 3With the salt brine solution washing, use Na then 2SO 4Drying is filtered, and is evaporated to dried.Resistates by preparation HPLC purifying, has been obtained white solid (30.5mg, 16%) as product.
LCMS RT=3.46min,[M+H] +=238.2。
Step 2: preparation (2S, 4R)-4-biphenyl-4-base-1-[(S)-2-(the 3-tertiary butyl-urea groups)-3,3-dimethyl-butyryl radicals]-4-methoxyl group-tetramethyleneimine-2-methyl-formiate
This compound is the same procedure of describing according in embodiment 106 steps 5, and difference is to use 2-(3-methoxyl group-phenyl amino)-3, and 3-dimethyl-butyric acid replaces (S)-2-(tert-butoxycarbonyl amino)-3, and the 3-acid dimethyl makes.
LCMS RT=3.31min,[M+H] +=531.5。
Step 2: preparation (2S, 4R)-4-biphenyl-4-base-4-methoxyl group-1-[2-(3-methoxyl group-phenyl amino)-3,3-dimethyl-butyryl radicals]-tetramethyleneimine-2-formic acid
Figure G2007800499569D01322
This compound is the same procedure of describing according in embodiment 106 steps 6; difference is to use (2S; 4R)-4-biphenyl-4-base-1-[(S)-2-(the 3-tertiary butyl-urea groups)-3; 3-dimethyl-butyryl radicals]-4-methoxyl group-tetramethyleneimine-2-methyl-formiate replacement (2S; 4R)-4-(biphenyl-4-yl)-1-((S)-2-(tert-butoxycarbonyl amino)-3,3-dimethyl butyrate acyl group)-4-methoxyl group tetramethyleneimine-2-methyl-formiate makes.
LCMS RT=3.20min,[M+H] +=517.3。
Embodiment 207: preparation compound 207 and compound 208
Figure G2007800499569D01331
Compound 207 compounds 208
Compound 207 and compound 208 are same procedure of describing according in embodiment 106 steps 7; difference is to use (2S; 4R)-4-biphenyl-4-base-4-methoxyl group-1-[2-(3-methoxyl group-phenyl amino)-3; 3-dimethyl-butyryl radicals]-tetramethyleneimine-2-formic acid replacement (2S; 4R)-4-(biphenyl-4-yl)-1-((S)-2-(tert-butoxycarbonyl amino)-3,3-dimethyl butyrate acyl group)-4-methoxyl group tetramethyleneimine-2-formic acid makes.This reaction mixture is separated by preparation HPLC; compound 207 (2S have been obtained; 4R)-4-biphenyl-4-base-4-methoxyl group-1-[(S)-2-(3-methoxyl group-phenyl amino)-3; 3-dimethyl-butyryl radicals]-tetramethyleneimine-2-formic acid ((1R; 2S)-1-cyclopropane sulfonyl amino carbonyl-2-vinyl-cyclopropyl)-acid amides and compound 208 (2S; 4R)-4-biphenyl-4-base-4-methoxyl group-1-[(R)-2-(3-methoxyl group-phenyl amino)-3; 3-dimethyl-butyryl radicals]-tetramethyleneimine-2-formic acid ((1R, 2S)-1-cyclopropane sulfonyl amino carbonyl-2-vinyl-cyclopropyl)-acid amides.
Compound 207:
1H NMR(CDCl 3-d)δ1.00-1.10(m,2H),1.14(s,9H),1.31-1.43(m,3H),1.92(m,1H),2.03(m,1H),2.44(m,1H),2.91-2.98(m,2H),3.00(s,3H),3.69(s,3H),3.75(m,1H),3.95(d,J=10Hz,1H),4.36(d,J=9Hz,1H),4.57(d,J=9Hz,1H),5.15(d,J=12Hz,1H),5.28(d,J=18.5Hz,1H),5.72(m,1H),6.17(s,1H),6.21(d,J=9.0Hz,1H),6.25(d,J=9.0Hz,1H),7.00(t,1H),7.34(m,3H),7.45(m,2H),7.58(m,4H);LCMS RT=3.29min,[M+H] +=729.4。
Compound 208:
1H NMR(CDCl 3-d)δ1.09(s,9H),1.30(m,2H),1.44(m,1H),1.56(m,2H),1.95(m,1H),2.64(m,1H),2.72(m,1H),2.86(m,2H),2.90(s,3H),3.77(m,1H),3.80(s,3H),4.01(d,J=10Hz,1H),4.60(d,J=9Hz,1H),4.66(d,J=9Hz,1H),5.11(d,J=12Hz,1H),5.28(d,J=18.5Hz,1H),5.86(m,1H),6.36(s,1H),6.37(d,J=9.0Hz,1H),6.43(d,J=9.0Hz,1H),7.12(t,1H),7.39(t,1H),7.44-7.48(m,4H),7.59(d,J=9Hz,2H),7.66(d,J=9Hz,2H),;LCMS RT=3.37min,[M+H] +=729.4
Embodiment 208: preparation compound 209 and compound 210
Compound 209
Figure G2007800499569D01342
Compound 210
Step 1: preparation 2-(3,4-, two fluoro-phenyl aminos)-3,3-dimethyl-butyric acid
Figure G2007800499569D01343
This compound is the same procedure of describing according in embodiment 207 steps 1, and difference is to use 3,4-difluoroaniline to replace the 3-anisidine to make.
LCMS RT=2.64min,[M+H] +=244.2。
Step 2: preparation (2S, 4R)-4-biphenyl-4-base-1-[2-(3,4-, two fluoro-phenyl aminos)-3,3-dimethyl-butyryl radicals]-4-methoxyl group-tetramethyleneimine-2-methyl-formiate
Figure G2007800499569D01351
This compound is the same procedure of describing according in embodiment 207 steps 2, and difference is to use 2-(3,4-, two fluoro-phenyl aminos)-3, and 3-dimethyl-butyric acid replaces 2-(3-methoxyl group-phenyl amino)-3, and 3-dimethyl-butyric acid makes.
LCMS RT=3.37min,[M+H] +=537.5。
Step 2: preparation (2S, 4R)-4-biphenyl-4-base-1-[2-(3,4-, two fluoro-phenyl aminos)-3,3-dimethyl-butyryl radicals]-4-methoxyl group-tetramethyleneimine-2-formic acid
Figure G2007800499569D01352
This compound is the same procedure of describing according in embodiment 106 steps 6; difference is to use (2S; 4R)-4-biphenyl-4-base-1-[2-(3; 4-two fluoro-phenyl aminos)-3; 3-dimethyl-butyryl radicals]-4-methoxyl group-tetramethyleneimine-2-methyl-formiate replacement (2S; 4R)-4-(biphenyl-4-yl)-1-((S)-2-(tert-butoxycarbonyl amino)-3,3-dimethyl butyrate acyl group)-4-methoxyl group tetramethyleneimine-2-methyl-formiate makes.
LCMS RT=3.26min,[M+H] +=523.3。
Embodiment 208: preparation compound 209 and compound 210
Figure G2007800499569D01361
Compound 209 compounds 210
Compound 209 and compound 210 are same procedure of describing according in embodiment 106 steps 7; difference is to use (2S; 4R)-4-biphenyl-4-base-1-[2-(3; 4-two fluoro-phenyl aminos)-3; 3-dimethyl-butyryl radicals]-4-methoxyl group-tetramethyleneimine-2-formic acid replacement (2S; 4R)-4-(biphenyl-4-yl)-1-((S)-2-(tert-butoxycarbonyl amino)-3,3-dimethyl butyrate acyl group)-4-methoxyl group tetramethyleneimine-2-formic acid makes.This reaction mixture is passed through preparation HPLC purifying; compound 209 (2S have been obtained; 4R)-4-biphenyl-4-base-1-[(S)-2-(3; 4-two fluoro-phenyl aminos)-3; 3-dimethyl-butyryl radicals]-4-methoxyl group-tetramethyleneimine-2-formic acid ((1R; 2S)-1-cyclopropane sulfonyl amino carbonyl-2-vinyl-cyclopropyl)-acid amides and compound 210 (2S; 4R)-4-biphenyl-4-base-1-[(R)-2-(3; 4-two fluoro-phenyl aminos)-3; 3-dimethyl-butyryl radicals]-4-methoxyl group-tetramethyleneimine-2-formic acid ((1R, 2S)-1-cyclopropane sulfonyl amino carbonyl-2-vinyl-cyclopropyl)-acid amides.
Compound 209:
1H NMR(CDCl 3-d)δ1.00-1.10(m,2H),1.14(s,9H),1.31-1.43(m,3H),1.92(m,1H),2.03(m,1H),2.44(m,1H),2.91-2.98(m,2H),3.00(s,3H),3.71(d,J=10Hz,1H),3.80(d,J=10Hz,1H),4.36(d,J=9Hz,1H),4.57(d,J=9Hz,1H),5.15(d,J=12Hz,1H),5.28(d,J=18.5Hz,1H),5.74(m,1H),6.26(s,1H),6.41(m,1H),6.86(q,1H),7.26(m,1H),7.29-7.39(m,2H),7.45(t,2H),7.58(t,4H);LCMS RT=3.05min,[M+Na] +=757.3。
Compound 210:
1H NMR(CDCl 3-d)δ1.00-1.10(m,3H),1.07(s,9H),1.48(m,2H),1.65(m,1H),2.03(m,1H),2.69(m,1H),2.87(m,1H),2.90(s,3H),3.71(d,J=10Hz,1H),3.90(d,J=10Hz,1H),4.34(d,J=9Hz,1H),4.61(d,J=9Hz,1H),4.66(d,J=9Hz,1H),5.15(d,J=12Hz,1H),5.28(d,J=18.5Hz,1H),5.89(m,1H),6.28(s,1H),6.60(m,1H),7.03(q,1H),7.39(t,1H),7.44-7.48(m,4H),7.59(d,J=9Hz,2H),7.66(d,J=9Hz,2H);LCMS RT=3.13min,[M+H] +=757.3。
Embodiment 209: preparation compound 211
Figure G2007800499569D01371
Compound 211
Biological test
In the application's disclosure, adopt HCV NS3/4A proteolytic enzyme prozyme method of inspection and cell based HCV to copy method of inspection, and be prepared as follows, carry out and verify:
The generation of recombinant HCV NS3/4A proteolytic enzyme mixture
Generation as described below is derived from the HCV NS3 proteolytic enzyme mixture of BMS strain, H77 strain or J4L6S strain.Produce the recombinant protein of these purifications to be used in the homogeneous phase method of inspection (seeing below) how to suppress HCV NS3 proteolytic activity effectively with the compound of indicating the application's disclosure.
From the serum of HCV infected patient available from Dr.T.Wright, San FranciscoHospital.By the dna fragmentation of reverse transcription-PCR (RT-PCR) acquisition of passing through serum RNA (Yeast Nucleic Acid) and the primer that use is selected based on the homology between other genotype 1a strain, the genomic engineering full-length cDNA of structure HCV (complementation (compliment) thymus nucleic acid) template.By measuring the whole genome sequence, according to people's such as Simmonds classification, with genotype 1a owing to the HCV isolate (referring to P Simmonds, KA Rose, S Graham, SW Chan, F McOmish, BC Dow, EA Follett, PL Yap and H Marsden, J.Clin.Microbiol., 31 (6), 1493-1503 (1993)).The aminoacid sequence of non-structural region NS2-5B equals HCV genotype 1a (H77) and 87% according to the show>97% and equals genotype 1b (J4L6S).Infecting the clone is that H77 (1a genotype) and J4L6S (1b genotype) are disclosed in (AAB67036 among the Genbank available from R.Purcell (NIH) and sequence, referring to Yanagi, M., Purcell, R.H., Emerson, S.U. and Bukh, J.Proc.Natl.Acad.Sci.U.S.A.94 (16), 8738-8743 (1997); AF054247, referring to Yanagi, M., St Claire, M., Shapiro, M., Emerson, S.U., Purcell, R.H. and Bukh, J, Virology 244 (1), 161-172. (1998)).
H77 and J4L6S strain are for the manufacture of reorganization NS3/4A proteolytic enzyme mixture.As people such as P.Gallinari (referring to Gallinari P, Paolini C, Brennan D, Nardi C, Steinkuhler C, De Francesco R.Biochemistry.38 (17): 5620-32, (1999)) described use is recombinant HCV NS3/4A proteolytic enzyme mixture (amino acid/11 027 to the 1711) coded DNA of these strains.In brief, in 3 ' of NS4A coding region-end adds three Methionin solubilising tails (solubilizing tail).The halfcystine that NS4A-NS4B is divided in the P1 position of site (amino acid/11 711) changes glycine into to avoid the proteolysis of tyrosine label.In addition, introducing halfcystine at amino acid position 1454 places by PCR divides to prevent the self-dissolving in the NS3 helicase territory to mutant serine.Clonal vaviation dna fragmentation in pET21b bacterial expression vector (Novagen), and the rules of describing according to the people such as P.Gallinari that revise are (referring to Gallinari P, Brennan D, Nardi C, Brunetti M, Tomei L, Steinkuhler C, De Francesco R., J Virol.72 (8): 6758-69 (1998)) expresses the NS3/4A mixture at e. coli strains BL21 (DE3) in (Invitrogen).In brief, cause NS3/4A proteolytic enzyme mixture with 0.5 mmole (mM) isopropyl ss-D-1-thio-galactose pyran-glucoside (IPTG) down at 20 ℃ and express 22 hours (h).Typical fermentation (1 liter (L)) produces about 10 gram (g) wet cells and sticks with paste.Described cell is resuspended in by 25mM N-(2-hydroxyethyl) piperazine-N '-(2-ethanesulfonic acid) (HEPES), pH 7.5, in molten born of the same parents' damping fluid that 20% glycerine, 500mM sodium-chlor (NaCl), 0.5%TritonX-100,1 mcg/ml (" μ g/mL ") N,O-Diacetylmuramidase, 5mM magnesium chloride (MgCl2), 1 μ g/mlDnaseI, 5mM beta-mercaptoethanol (β ME), proteinase inhibitor-ethylenediamine tetraacetic acid (EDTA) (EDTA) free (Roche) constitute (10 milliliters/gram), homogenate is also cultivated 20 minutes (min) under 4 ℃.With this homogenate sonication and by under 235000g 4 ℃ of following super centrifugal 1 hour clarifications.Add imidazoles in the supernatant liquor and be adjusted to 8.0 to the ultimate density of 15mM and with the pH value.Should be carried on nickel-nitrilotriacetic acid(NTA) (Ni-NTA) post of using buffer B (25mMHEPES, pH 8.0,20% glycerine, 500mM NaCl, 0.5%Triton X-100,15mM imidazoles, 5mM β ME) pre-equilibration by rough protein extract.Flow velocity load sample with 1 ml/min.This post is with the damping fluid C of 15 times of column volumes (identical with buffer B, but contain 0.2%Triton X-100) washing.Damping fluid D (C is identical with damping fluid, but contains the 200mM imidazoles) this protein of wash-out with 5 times of column volumes.
Compile the fraction that contains NS3/4A proteolytic enzyme mixture and be carried on the desalting column Superdex-S200 with damping fluid D (25mMHEPES, pH 7.5,20% glycerine, 300mM NaCl, 0.2%Triton X-100,10mM β ME) pre-equilibration.Flow velocity load sample with 1 ml/min.Compile the fraction that contains NS3/4A proteolytic enzyme mixture and be condensed into about 0.5 mg/ml.Judge that by SDS-PAGE and mass spectroscopy purity derived from the NS3/4A proteolytic enzyme mixture of BMS, H77 and J4L6S post is greater than 90%.This enzyme is stored under-80 ℃, thaws on ice and before using, dilute in damping fluid in check.
FRET peptide method of inspection is with monitoring HCV NS3/4A proteolytic activity
The purpose of this external method of inspection is the compound of measuring the application's disclosure to as mentioned above derived from the inhibition of the HCV NS3 proteolytic enzyme mixture of BMS strain, H77 strain or J4L6S strain.How the compound of this method of inspection indication the application disclosure suppresses HCV NS3 proteolytic activity effectively.
In order to monitor HCV NS3/4A protease activity, use the NS3/4A peptide substrates.This substrate is RET S1 (the Resonance Energy Transfer Depsipeptide Substrate that people such as Taliani describe in Anal.Biochem.240 (2): 60-67 (1996); AnaSpec, Inc.cat#22991) (FRET peptide).The sequence of this peptide roughly divides the site naturally based on the NS4A/NS4B of HCV NS3 proteolytic enzyme, just has ester bond but not amido linkage in this division site.This peptide also contains fluorescence near an end of peptide gives body EDANS and contain acceptor DABCYL near another end.By the fluorescence to intermolecular resonance energy transfer (RET) quencher peptide between body and the acceptor, but along with NS3 proteolytic enzyme makes this peptide division, the fluorescence apparition that discharges product and give body from the RET quencher.
Under the situation of the compound that has or do not exist the application's disclosure, cultivate peptide substrates with one of three kinds of reorganization NS3/4A proteolytic enzyme mixtures.By using the formation of Cytofluor Series 4000 monitoring fluorescent reaction in real time product, measure the inhibition of compound.
Reagent is as follows: HEPES and Glycerol (Ultrapure) are available from GIBCO-BRL.Methyl-sulphoxide (DMSO) is available from Sigma.Beta-mercaptoethanol is available from Bio Rad.
The check damping fluid: 50mM HEPES, pH 7.5; 0.15M NaCl; 0.1%Triton; 15% glycerine; 10mM β ME.Substrate: 2 μ M ultimate densities (from the 2mM liquid storage in DMSO that is stored under-20 ℃).HCV NS3/4A proteolytic enzyme 1a type (1b), 2-3nM ultimate density (coming 5 μ M liquid storages among the comfortable 25mM HEPES, pH 7.5,20% glycerine, 300mM NaCl, 0.2%Triton-X100,10mM β ME).For rendeing a service the compound that approaches the check limit, by making this check more responsive to check with adding 50 mcg/ml fetal bovine serum albumin (Sigma) in the damping fluid and final protease concentration being down to 300pM.
In 96 hole polystyrene blackboards from Falcon, test.Each hole contain check with 25 μ l NS3/4A proteolytic enzyme mixtures in the damping fluid, 50 microlitres the 10%DMSO/ check with the compound of the application's disclosure in the damping fluid and 25 microlitres in check with the substrate in the damping fluid.Also prepare contrast (no compound) at identical inspection panel.This enzyme complex was mixed 1 minute with compound or contrast solution, cause enzymatic reaction by adding substrate then.Use Cytofluor Series 4000 (Perspective Biosystems) to read inspection panel immediately.This instrument is set under 25 ℃, reads 340 nanometers emissions and 490 nanometers excite.Reaction was carried out about 15 minutes usually.
Calculate inhibition per-cent with following formula:
100-[(δF inh/δF con)×100]
Wherein δ F is the interior change in fluorescence of linearity range of this curve.Inhibition-concentration data is used non-linear curve fitting, and use formula y=A+ ((B-A)/(1+ ((C/x) ^D))) Excel XLfit computed in software 50% effective concentration (IC 50).
All test-compounds all are found to suppress with 1.6 μ M or lower IC50 ' s the activity of NS3/4A proteolytic enzyme mixture.In addition, it is found that to have similar inhibition activity at the application's disclosure compound more than a kind of NS3/4A mixture test, although this compound shows the effectiveness to the 1b strain of comparison 1a plant height without exception.
The specificity check
Carry out specificity and check to prove with other Serine or L-Cysteine HCL Anhydrous and compare, the compound of the application's disclosure is in the external selectivity that suppresses aspect the HCV NS3/4A proteolytic enzyme mixture.
Contrast the specificity of the compound of various serine stretch protein enzymatic determination the application disclosures: human neutrophil elastoser (HNE), porcine pancreatic elastase (PPE) and people's Quimotrase and a kind of L-Cysteine HCL Anhydrous: human liver tissue proteolytic enzyme B.In all cases, use the 96 orifice plate form rules (it uses colorimetric p-Nitroaniline (pNA) substrate or fluorescence amino-methyl-tonka bean camphor (AMC) substrate to each enzyme spcificity) (PCT patent application No.WO 00/09543) of some modifications of process as mentioned above.All enzymes are all available from Sigma, EMDbiosciences, and substrate is from Bachem, Sigma and EMDbiosciences.
Compound concentration is renderd a service from 100 to 0.4 μ M with it and is not waited.Come the primase check in enzyme-inhibitor of at room temperature cultivating 10 minutes in advance by substrate is added to, and be hydrolyzed into 15% transformation efficiency of measuring at cytofluor.
The final condition of each check is as follows:
50mM three (methylol) aminomethane hydrochloride (Tris-HCl) pH 8,0.5M sodium sulfate (Na2SO4), 50mM NaCl, 0.1mM EDTA, 3%DMSO, 0.01%Tween-20 contains: 5 μ M LLVY-AMC and 1nM Quimotrase.
50mM Tris-HCl, pH 8.0,50mM NaCl, 0.1mM EDTA, 3%DMSO, 0.02%Tween-20,5 μ M succ-AAPV-AMC and 20nM HNE or 8nM PPE;
100mM NaOAC (sodium acetate) pH 5.5,3%DMSO, 1mM TCEP (three (2-carboxy ethyl) phosphonium salt hydrochlorate), 5nM cathepsin B (before using, containing the enzyme liquid storage that activates in the damping fluid of 20mMTCEP) and at H 2The 2 μ M Z-FR-AMC that dilute among the O.
Use following formula to calculate and suppress per-cent:
[1-((UV Inh-UV Blank)/(UV Ctl-UV Blank))] * 100
The inhibition concentration data are used non-linear curve fitting, and use Excel XLfit computed in software 50% effective concentration (IC 50).
The generation of HCV replicon
As Lohmann V, Korner F, Koch J, Herian U, Theilmann L, Bartenschlager R., Science 285 (5424): the described full cell system of HCV replicon of setting up of 110-3 (1999).This system makes us can evaluate and test our HCV proteolytic enzyme compound to the influence of HCV rna replicon.In brief, use the HCV strain 1b sequence described in the Lohmann paper (Assession number: AJ238799), Operon Technologies, Inc. (Alameda, CA) synthesized HCV cDNA, then at plasmid pGem9zf (+) (Promega, Madison, WI) the middle standard molecule biotechnology assembling total length replicon that uses.This replicon is made of following ingredients: (i) be fused to the HCV 5 ' UTR on preceding 12 amino acid of housing albumen, (ii) neomycin phosphotransferase gene (neo), (iii) from the IRES of encephalomyocarditis virus (EMCV) and (iv) HCV NS3 to NS5B gene and HCV 3 ' UTR.(Ambion, Austin is TX) according to the synthetic rna transcription product of the indication of manufacturers with the ScaI linearizing and at external use T7MegaScript transcript reagent box for plasmid DNA.Be among the HUH-7 with the in-vitro transcription product transfection of cDNA to the human liver cell oncocyte.In the presence of selectable marker Xin Meisu (G418), be implemented in the selection that constitutes the cell of going up the expression of HCV replicon.In time at normal chain and strand RNA generation and protein generation sign gained clone.
HCV replicon FRET method of inspection
Exploitation HCV replicon FRET method of inspection is with the inhibition of the compound described in monitoring the application disclosure to the HCV virus replication.Contain 10% foetal calf serum (FCS) (Sigma) and the Dulbecco ' s Modified EagleMedia (DMEM) of 1mg/ml G418 (Gibco-BRL) make in (Gibco-BRL) constituting on the HUH-7 cell growth of expression of HCV replicon.Eve with cell inoculation (1.5 * 10 4Individual cells/well) in 96 hole tissue culture sterile plate.Dilution containing in the plate 4%FCS, 1: 100 penicillin/streptomycin (Streptomysin) (Gibco-BRL), 1: the contrast (0.5%DMSO ultimate density in this check) of preparation compound and no compound among the DMEM of 100L-glutamine and 5%DMSO.In cell, add compound/DMSO mixture and descend cultivation 4 days at 37 ℃.After 4 days, at first use the cytotoxicity of alamar Blue (Trek Diagnotstic Systems) assessment cell at the CC50 reading.Add in the substratum of culturing cell by the alamar Blue with 1/10 volume, measure the toxicity (CC of compound 50).After 4 hours, use Cytofluor Series 4000 (PerspectiveBiosystems) read the fluorescent signal from each hole under 530 nanometer excitation wavelengths and 580 nanometer emission wavelengths.Use fully each plate of rinsing of phosphate buffered saline (PBS) (PBS) (150 microlitres 3 times) then.Molten born of the same parents' testing reagent (the plain enzyme molten born of the same parents' reagent of cell cultures (Promega#E153A) of 5 * cell fluorescence that contains the HCV protease substrate with 25 microlitres, with distilled water diluting to 1 *, add NaCl to 150mM ultimate density, FRET peptide substrates (described to the enzyme method of inspection as mentioned) is diluted to 10 μ M ultimate densities by the 2mM liquid storage in 100%DMSO) dissolved cell.The HCV protease substrate.Then this plate is put into and be set to 340 nanometers and excite/Cytofluor 4000 instruments of 490 nanometers emissions, automatic mode moved for 21 cycles, and read this plate with dynamic mode.As to IC 50Measure the described EC that carries out 50Measure.
HCV replicon luciferase reporter gene method of inspection
As the secondary check method, checking is from the IC50 measurement result of replicon FRET method of inspection in replicon luciferase reporter gene method of inspection.People such as Krieger (Krieger N, Lohmann V and Bartenschlager R, J.Virol.75 (10): 4614-4624 (2001)) have described the application of replicon luciferase reporter gene method of inspection at first.The cDNA of the peopleization form by inserting coding Renilla luciferase gene and directly be fused to connexon sequence on 3 ' of luciferase gene-end, modification is constructed the described replicon of our FRET method of inspection.Use just is arranged at the Asc1 restriction site of the nuclear of neomycin marker upstream region of gene introduces the replicon structure with this inset.The adaptive mutation that position 1179 (Serine is to Isoleucine) is located (Blight KJ, Kolykhalov, AA, Rice, CM, Science290 (5498): 1972-1974) have also been introduced.Be created in the stable cell lines of this HCV replicon structure of textural expression as mentioned above.As described in to HCV replicon FRET method of inspection, with following modification the luciferase reporter gene method of inspection is set.At 37 ℃/5%CO 2After 4 days, use the Renilla uciferase activity of Promega Dual-Glo luciferase checking system analysis of cells in the incubator.From celliferous each hole, take out substratum (100 μ l).In all the other 50 microlitre substratum, add 50 μ l Dual-Glo luciferase reagent, and plate was at room temperature shaken 10 minutes to 2 hours.In each hole, add Dual-Glo Stop ﹠amp then; Glo Reagent (50 microlitre), and plate at room temperature shaken 10 minutes to 2 hours again.Use the fluorescence program to read plate at Packard TopCount NXT.
Use following formula to calculate and suppress per-cent:
Figure G2007800499569D01431
Use the XLfit drafting and analyze numerical value to obtain EC 50Value.
The representational the application's disclosure compound of assessment in HCV enzyme detection method, HCV replicon cell detection method and/or the specificity method of inspection in some general introductions.For example, find that compound 3 has the IC to 4.4 nmoles (nM) of NS3/4A BMS strain in the enzyme method of inspection 50With the disclosed H77 (IC of 1.6nM 50) and the J4L6S (IC of 1.1nM 50) strain obtains similar effectiveness value.EC in the replicon FRET method of inspection 50Value is 11nM and is 1.4nM in replicon luciferase method of inspection.
In the specificity method of inspection, find that same compound has following activity: HLE=82 μ M; PPE>100 μ M; Quimotrase=75 μ M; Cathepsin B>100 μ M.These results show, this compounds suppresses the HCV replicon to NS3 proteolytic enzyme high degree of specificity and many these members to be copied.
The compound of test the application disclosure and discovery have the activity in following scope:
IC 50Field of activity (NS3/4A BMS strain): A is 1nM to 50nM; B is>50nM to 100nM is to 500nM; C is>500nM.
EC 50Field of activity (for test-compound): A is 1nM to 50nM; B is>50nM to 100nM is to 500nM; C is>500nM.
Come record by the patent compound sequence number that shows in the use table 2, the structure of compound can be referring to this paper.
Table 2
Compound HCV proteolytic enzyme (IC50) HCV protease replicon (EC50)
Compound 106 1.00 11.00
Compound 107 B C
Compound 108 A A
Compound 109 A B
Compound 110 A B
Compound 200 A A
Compound 201 A B
Compound 202 A B
Compound 203 A B
Compound 204 A A
Compound 205 A A
Compound 206 A A
Compound 207 A A
Compound 208 315.00 1,303
Compound 209 A A
Compound 210 C C
Compound 211 1600.00
It will be apparent to one skilled in the art that present disclosure is not limited to above-mentioned illustrative examples, under the situation that does not deviate from its essential nature, it can be with other specific forms performance.Therefore expectation is to consider each embodiment all sidedly, as to claims but not describing property of above-described embodiment and nonrestrictive reference, and therefore the connotation and the whole variations in the scope that fall into the claim Equivalent are intended to be contained in wherein.

Claims (9)

1. formula (I) compound
Or its pharmacologically acceptable salt, wherein
N is 1;
R 1Be NHSO 2R 7
R 2Be selected from alkenyl, alkyl or haloalkyl;
R 3It is aryl;
R 4Be-OR 8
R 5Be selected from alkenyl, alkyl or arylalkyl;
R 6Be selected from alkoxy carbonyl, cycloalkyl oxy carbonyl, halo alkoxy carbonyl, (NR aR b) carbonyl; Perhaps
R 6Be phenyl, wherein said phenyl is optional to be independently selected from following substituting group replacement by 1 or 2: alkoxyl group and halogen;
R 7It is cycloalkyl; With
R aAnd R bBe independently selected from hydrogen or cycloalkyl,
R 8Be selected from alkoxyalkyl, alkyl, alkyl-carbonyl, arylalkyl, cycloalkyl, (cycloalkyl) alkyl, naphthene base carbonyl, halogenated alkoxy alkyl, haloalkyl, (NR eR f) carbonyl and-P (O) (OR ') 2R wherein eAnd R fBe independently selected from hydrogen, alkyl and arylalkyl; Perhaps R eAnd R fThe nitrogen-atoms that connects with them forms 5 or 6 yuan of monocyclic heterocycles, and described heterocycle is optional to be contained one and be selected from O, NR xOther heteroatoms with S; R wherein xBe selected from hydrogen and alkyl; And wherein R ' is selected from hydrogen and alkyl.
2. compound, it is selected from following compound:
Figure FSB00001068905400021
Figure FSB00001068905400031
Or its pharmacologically acceptable salt.
3. composition, described composition comprises compound or pharmaceutically acceptable salt thereof and pharmaceutically acceptable carrier of claim 1.
4. the composition of claim 3, described composition also comprises at least a additional compounds with whose anti-HCV activity.
5. the composition of claim 4, wherein said at least a additional compounds is Interferon, rabbit or ribavirin.
6. the composition of claim 5, wherein said Interferon, rabbit is selected from interferon alpha 2B, glycol interferon alpha, Interferon alfacon-1, interferon alpha 2A and lymphoblastoid interferon-tau.
7. the composition of claim 4, wherein said at least a additional compounds be selected from interleukin-22, interleukin 6, interleukin 12, promotion 1 type helper cell reply the compound of development, RNA interfering, sense-rna, Imiquimod, ribavirin, inosine 5 '-monophosphate dehydrogenase inhibitor, Symmetrel and Rimantadine.
8. the composition of claim 4, wherein said at least a additional compounds effectively suppress to be selected from the function of following target: HCV metalloprotease, HCV serine protease, HCV polysaccharase, HCV helicase, HCV NS4B albumen, HCV enter, HCV assemble, HCV deviates from, HCV NS5A albumen and the IMPDH that is used for the treatment of the HCV infection.
9. the purposes in the compound or pharmaceutically acceptable salt thereof of claim 1 medicine that HCV infects in for the preparation of the treatment patient.
CN200780049956.9A 2006-11-16 2007-11-15 Hepatitis c virus inhibitors Expired - Fee Related CN101652383B (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US86613006P 2006-11-16 2006-11-16
US60/866,130 2006-11-16
US11/939,780 US7888464B2 (en) 2006-11-16 2007-11-14 Hepatitis C virus inhibitors
US11/939,780 2007-11-14
PCT/US2007/084799 WO2008064066A1 (en) 2006-11-16 2007-11-15 Hepatitis c virus inhibitors

Publications (2)

Publication Number Publication Date
CN101652383A CN101652383A (en) 2010-02-17
CN101652383B true CN101652383B (en) 2013-09-18

Family

ID=41674126

Family Applications (1)

Application Number Title Priority Date Filing Date
CN200780049956.9A Expired - Fee Related CN101652383B (en) 2006-11-16 2007-11-15 Hepatitis c virus inhibitors

Country Status (2)

Country Link
CN (1) CN101652383B (en)
ZA (1) ZA200903379B (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012039132A1 (en) * 2010-09-22 2012-03-29 三井化学アグロ株式会社 Method for producing amino acid amide derivative having fluorine-containing carbamate group, production intermediate thereof, and method for producing ethylene diamine derivative

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006122188A2 (en) * 2005-05-10 2006-11-16 Bristol-Myers Squibb Company Tripeptides as hepatitis c virus inhibitors

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006122188A2 (en) * 2005-05-10 2006-11-16 Bristol-Myers Squibb Company Tripeptides as hepatitis c virus inhibitors

Also Published As

Publication number Publication date
CN101652383A (en) 2010-02-17
ZA200903379B (en) 2010-07-28

Similar Documents

Publication Publication Date Title
CN101228180B (en) Tripeptides as hepatitis C virus inhibitors
US7888464B2 (en) Hepatitis C virus inhibitors
US7964560B2 (en) Hepatitis C virus inhibitors
US7935670B2 (en) Hepatitis C virus inhibitors
CN103936819B (en) Hepatitis c virus inhibitors
CN101541784B (en) Hepatitis c virus inhibitors
CN102164928B (en) Hepatitis c virus inhibitors
CN101024671B (en) Hepatitis c virus inhibitors
US7763584B2 (en) Hepatitis C virus inhibitors
US8003604B2 (en) Hepatitis C virus inhibitors
CN101646667A (en) Inhibitors of hepatitis C virus
CN101583611A (en) Inhibitors of hepatitis C virus
CN101578294A (en) Inhibitors of hepatitis C virus
CN101189223A (en) Hepatitis C virus inhibitors
CN101652383B (en) Hepatitis c virus inhibitors
CN101568543B (en) Macrocyclic peptides as hepatitis c virus inhibitors
CN101568336B (en) Macrocyclic peptides as hepatitis c virus inhibitors

Legal Events

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

Granted publication date: 20130918

Termination date: 20161115