CN1942467A - 2, 4, 6-substituted pyridyl derivative compounds useful as beta-secretase inhibitors for the treatment of Alzheimer's disease - Google Patents

2, 4, 6-substituted pyridyl derivative compounds useful as beta-secretase inhibitors for the treatment of Alzheimer's disease Download PDF

Info

Publication number
CN1942467A
CN1942467A CNA2005800118430A CN200580011843A CN1942467A CN 1942467 A CN1942467 A CN 1942467A CN A2005800118430 A CNA2005800118430 A CN A2005800118430A CN 200580011843 A CN200580011843 A CN 200580011843A CN 1942467 A CN1942467 A CN 1942467A
Authority
CN
China
Prior art keywords
methyl
alkyl
compound
cycloalkyl
amino
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.)
Pending
Application number
CNA2005800118430A
Other languages
Chinese (zh)
Inventor
J·C·巴罗
G·B·麦戈伊
P·G·南特梅特
H·A·拉亚帕克斯
H·G·塞尔尼克
S·R·斯陶费尔
J·P·瓦卡
S·J·斯塔彻尔
C·A·科伯恩
M·G·斯坦顿
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.)
Merck and Co Inc
Original Assignee
Merck and Co Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Merck and Co Inc filed Critical Merck and Co Inc
Publication of CN1942467A publication Critical patent/CN1942467A/en
Pending legal-status Critical Current

Links

Classifications

    • BPERFORMING OPERATIONS; TRANSPORTING
    • B23MACHINE TOOLS; METAL-WORKING NOT OTHERWISE PROVIDED FOR
    • B23KSOLDERING OR UNSOLDERING; WELDING; CLADDING OR PLATING BY SOLDERING OR WELDING; CUTTING BY APPLYING HEAT LOCALLY, e.g. FLAME CUTTING; WORKING BY LASER BEAM
    • B23K35/00Rods, electrodes, materials, or media, for use in soldering, welding, or cutting
    • B23K35/02Rods, electrodes, materials, or media, for use in soldering, welding, or cutting characterised by mechanical features, e.g. shape
    • B23K35/0255Rods, electrodes, materials, or media, for use in soldering, welding, or cutting characterised by mechanical features, e.g. shape for use in welding
    • B23K35/0261Rods, electrodes, wires
    • CCHEMISTRY; METALLURGY
    • C23COATING METALLIC MATERIAL; COATING MATERIAL WITH METALLIC MATERIAL; CHEMICAL SURFACE TREATMENT; DIFFUSION TREATMENT OF METALLIC MATERIAL; COATING BY VACUUM EVAPORATION, BY SPUTTERING, BY ION IMPLANTATION OR BY CHEMICAL VAPOUR DEPOSITION, IN GENERAL; INHIBITING CORROSION OF METALLIC MATERIAL OR INCRUSTATION IN GENERAL
    • C23CCOATING METALLIC MATERIAL; COATING MATERIAL WITH METALLIC MATERIAL; SURFACE TREATMENT OF METALLIC MATERIAL BY DIFFUSION INTO THE SURFACE, BY CHEMICAL CONVERSION OR SUBSTITUTION; COATING BY VACUUM EVAPORATION, BY SPUTTERING, BY ION IMPLANTATION OR BY CHEMICAL VAPOUR DEPOSITION, IN GENERAL
    • C23C28/00Coating for obtaining at least two superposed coatings either by methods not provided for in a single one of groups C23C2/00 - C23C26/00 or by combinations of methods provided for in subclasses C23C and C25C or C25D
    • C23C28/02Coating for obtaining at least two superposed coatings either by methods not provided for in a single one of groups C23C2/00 - C23C26/00 or by combinations of methods provided for in subclasses C23C and C25C or C25D only coatings only including layers of metallic material
    • C23C28/021Coating for obtaining at least two superposed coatings either by methods not provided for in a single one of groups C23C2/00 - C23C26/00 or by combinations of methods provided for in subclasses C23C and C25C or C25D only coatings only including layers of metallic material including at least one metal alloy layer
    • CCHEMISTRY; METALLURGY
    • C23COATING METALLIC MATERIAL; COATING MATERIAL WITH METALLIC MATERIAL; CHEMICAL SURFACE TREATMENT; DIFFUSION TREATMENT OF METALLIC MATERIAL; COATING BY VACUUM EVAPORATION, BY SPUTTERING, BY ION IMPLANTATION OR BY CHEMICAL VAPOUR DEPOSITION, IN GENERAL; INHIBITING CORROSION OF METALLIC MATERIAL OR INCRUSTATION IN GENERAL
    • C23CCOATING METALLIC MATERIAL; COATING MATERIAL WITH METALLIC MATERIAL; SURFACE TREATMENT OF METALLIC MATERIAL BY DIFFUSION INTO THE SURFACE, BY CHEMICAL CONVERSION OR SUBSTITUTION; COATING BY VACUUM EVAPORATION, BY SPUTTERING, BY ION IMPLANTATION OR BY CHEMICAL VAPOUR DEPOSITION, IN GENERAL
    • C23C28/00Coating for obtaining at least two superposed coatings either by methods not provided for in a single one of groups C23C2/00 - C23C26/00 or by combinations of methods provided for in subclasses C23C and C25C or C25D
    • C23C28/02Coating for obtaining at least two superposed coatings either by methods not provided for in a single one of groups C23C2/00 - C23C26/00 or by combinations of methods provided for in subclasses C23C and C25C or C25D only coatings only including layers of metallic material
    • C23C28/023Coating for obtaining at least two superposed coatings either by methods not provided for in a single one of groups C23C2/00 - C23C26/00 or by combinations of methods provided for in subclasses C23C and C25C or C25D only coatings only including layers of metallic material only coatings of metal elements only
    • CCHEMISTRY; METALLURGY
    • C23COATING METALLIC MATERIAL; COATING MATERIAL WITH METALLIC MATERIAL; CHEMICAL SURFACE TREATMENT; DIFFUSION TREATMENT OF METALLIC MATERIAL; COATING BY VACUUM EVAPORATION, BY SPUTTERING, BY ION IMPLANTATION OR BY CHEMICAL VAPOUR DEPOSITION, IN GENERAL; INHIBITING CORROSION OF METALLIC MATERIAL OR INCRUSTATION IN GENERAL
    • C23CCOATING METALLIC MATERIAL; COATING MATERIAL WITH METALLIC MATERIAL; SURFACE TREATMENT OF METALLIC MATERIAL BY DIFFUSION INTO THE SURFACE, BY CHEMICAL CONVERSION OR SUBSTITUTION; COATING BY VACUUM EVAPORATION, BY SPUTTERING, BY ION IMPLANTATION OR BY CHEMICAL VAPOUR DEPOSITION, IN GENERAL
    • C23C4/00Coating by spraying the coating material in the molten state, e.g. by flame, plasma or electric discharge
    • C23C4/12Coating by spraying the coating material in the molten state, e.g. by flame, plasma or electric discharge characterised by the method of spraying
    • C23C4/131Wire arc spraying
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B23MACHINE TOOLS; METAL-WORKING NOT OTHERWISE PROVIDED FOR
    • B23KSOLDERING OR UNSOLDERING; WELDING; CLADDING OR PLATING BY SOLDERING OR WELDING; CUTTING BY APPLYING HEAT LOCALLY, e.g. FLAME CUTTING; WORKING BY LASER BEAM
    • B23K35/00Rods, electrodes, materials, or media, for use in soldering, welding, or cutting
    • B23K35/02Rods, electrodes, materials, or media, for use in soldering, welding, or cutting characterised by mechanical features, e.g. shape
    • B23K35/0255Rods, electrodes, materials, or media, for use in soldering, welding, or cutting characterised by mechanical features, e.g. shape for use in welding
    • B23K35/0261Rods, electrodes, wires
    • B23K35/0272Rods, electrodes, wires with more than one layer of coating or sheathing material
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B23MACHINE TOOLS; METAL-WORKING NOT OTHERWISE PROVIDED FOR
    • B23KSOLDERING OR UNSOLDERING; WELDING; CLADDING OR PLATING BY SOLDERING OR WELDING; CUTTING BY APPLYING HEAT LOCALLY, e.g. FLAME CUTTING; WORKING BY LASER BEAM
    • B23K35/00Rods, electrodes, materials, or media, for use in soldering, welding, or cutting
    • B23K35/22Rods, electrodes, materials, or media, for use in soldering, welding, or cutting characterised by the composition or nature of the material
    • B23K35/24Selection of soldering or welding materials proper
    • B23K35/30Selection of soldering or welding materials proper with the principal constituent melting at less than 1550 degrees C
    • B23K35/3033Ni as the principal constituent
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B23MACHINE TOOLS; METAL-WORKING NOT OTHERWISE PROVIDED FOR
    • B23KSOLDERING OR UNSOLDERING; WELDING; CLADDING OR PLATING BY SOLDERING OR WELDING; CUTTING BY APPLYING HEAT LOCALLY, e.g. FLAME CUTTING; WORKING BY LASER BEAM
    • B23K35/00Rods, electrodes, materials, or media, for use in soldering, welding, or cutting
    • B23K35/22Rods, electrodes, materials, or media, for use in soldering, welding, or cutting characterised by the composition or nature of the material
    • B23K35/24Selection of soldering or welding materials proper
    • B23K35/30Selection of soldering or welding materials proper with the principal constituent melting at less than 1550 degrees C
    • B23K35/3053Fe as the principal constituent

Abstract

The present invention is directed to 2, 4, 6-substituted pyridyl derivative compounds which are inhibitors of the beta-secretase enzyme and that are useful in the treatment of diseases in which the beta-secretase enzyme is involved, such as Alzheimer's disease. The invention is also directed to pharmaceutical compositions comprising these compounds and the use of these compounds and compositions in the treatment of such diseases in which the beta-secretase enzyme is involved.

Description

Be used for as β-short Secretase inhibitors Alzheimer treatment 2,4, the pyridine derivative compound that 6-replaces
The related application of cross reference
60/653,037 the right of priority that the application enjoys according to 35 U.S.C.119 (e) request that the U.S. Provisional Application series number of submitting on April 20th, 2,004 60/563,612 and 15,60/630,539 and 2005 on the February of submitting on November 23rd, 2004 submit to.
Technical field
The present invention relates to a class new 2,4, the pyridine derivative compound that 6-replaces, they are effective β-short Secretase inhibitors and are applicable to and wherein involve the β-disease of short Secretases (secretase) such as the treatment of Alzheimer.
Background technology
Alzheimer is characterised in that the abnormal deposition of amyloid in brain forms neurofibrillary tangles in extracellular plaque and the cell.The speed that amyloid is accumulated is the function that forms, assembles and fail speed outside brain.People are generally accepted to be that the main composition of amyloid plaque is that (β A4 is also referred to as A β to the 4kD amyloid protein; Beta-protein matter and β AP), it is a kind of very large protein hydrolysate of size of precursor protein.Amyloid precursor protein (APP or A β; PP) has acceptor spline structure and have big ectodomain, stride diaphragm area and short cytoplasmic tail.A beta structure territory comprises a plurality of parts of the outer and membrane spaning domain of the born of the same parents of APP, thereby so its release mean that the existence of two kinds of different proteolysis incidents produces its NH 2-and the COOH-end.At least exist two kinds to discharge APP and produce the APP (APP of solubility, COOH-butt form from film s) secretion matrix.Proteolytic enzyme and the film forming fragment of structure thereof that discharges APP is called " short Secretases ".Most APP sBe to be discharged by the α that infers-short Secretases, it is separated in a matter implosion and discharges α-APP sAnd stoped the release of complete A β.The APP of small part sBe to be discharged by β-short Secretases (" β-short Secretases "), it is at the NH of APP 2Near-terminal cracking also produces the COOH-terminal fragment (CTFs) that contains whole A beta structures territory.
Therefore, the activity of β-short Secretases or β-site amyloid precursor protein-lyase (" BACE ") causes the unusual cracking of APP, produce A β, with amyloid-beta plaque accumulating in brain, it is characterized in that Alzheimer (referring to R.N.Rosenberg, Arch.Neurol., vol.59, Sep 2002, pp.1367-1368; H.Fukumoto etc., Arch.Neurol., vol.59, Sep 2002, pp.1381-1389; J.T.Huse etc., J.Biol.Chem., vol 277, No.18, May 3,2002 publishes pp.16278-16284; K.C.Chen and W.J.Howe, Biochem.Biophys.Res.Comm, vol.292, pp 702-708,2002).So, can suppress the therapeutical agent of β-short Secretases or BACE and can effectively treat Alzheimer.
Compound of the present invention prevents the formation of insoluble A β thus by suppressing the active effectively treatment Alzheimer of β-short Secretases or BACE; With the generation that stops A β.
Summary of the invention
The present invention relates to as 2,4 of β-short Secretase inhibitors, the pyridyl derivative compound that 6-replaces, and be used for the treatment of the disease that wherein relates to β-short Secretases, for example Alzheimer.The invention still further relates to the pharmaceutical composition that contains these compounds and these compounds and composition and relate to application in the disease of β-short Secretases in treatment.
Detailed Description Of The Invention
The present invention relates to the compound of formula (I):
Figure A20058001184300221
Wherein: X is selected from group
Figure A20058001184300231
R wherein 11And R 12Be independently selected from
(a) hydrogen,
(b)-C 1-10Alkyl,
(c) C 2-10Alkenyl,
(d) C 2-10Alkynyl group,
(e)-C 3-12Cycloalkyl and
(f) be selected from the aryl of phenyl and naphthyl;
This alkyl wherein, cycloalkyl, alkenyl, alkynyl group or aryl are not substituted or are replaced by one or more following groups
(i) halogen,
(ii)-OH,
(iii)-CN,
(iv)-C 1-10Alkyl,
(v)-C 3-12Cycloalkyl and
(vi)-O-C 1-10Alkyl,
Y 1Be N, and Y 2And Y 3Be respectively CH, or
Y 2Be N, and Y 1And Y 3Be respectively CH, or
Y 3Be N, and Y 1And Y 2Be respectively CH;
A is selected from group
(1) hydrogen,
(2)-C 1-10Alkyl and
(3)-C 2-10Alkenyl,
Wherein this alkyl or alkenyl are not replace or replaced by one or more following groups
(a) halogen,
(b)-C 3-12Cycloalkyl,
(c)-OH,
(d)-CN,
(e)-O-C 1-10Alkyl,
(f) phenyl, or
(g) heteroaryl is selected from pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidyl, pyrryl, tetrazyl, furyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl,  azoles base, different  azoles base, thiazolyl, the  di azoly, indyl, quinolyl, isoquinolyl, benzimidazolyl-, benzofuryl, indoles alkynyl (indynyl) and benzoxazol base
With this phenyl and heteroaryl be not replace or replaced by one or more following groups
(i) halogen,
(ii)-OH,
(iii)-CN,
(iv)-O-C 1-10Alkyl,
(v)-C 1-10Alkyl, or
(vi)-C 3-12Cycloalkyl;
Q is-C 0-3Alkyl, wherein this alkyl is not replace or replaced by one or more following groups
(1) halogen,
(2)-C 3-12Cycloalkyl,
(3)-OH,
(4)-CN,
(5)-O-C 1-10Alkyl and
(6)-C 1-10Alkyl;
R 1Be the aryl that (1) is selected from phenyl and naphthyl,
(2) heteroaryl is selected from pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidyl, pyrryl, tetrazyl, furyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl,  azoles base, different  azoles base, thiazolyl, the  di azoly, indyl, quinolyl, isoquinolyl, benzimidazolyl-, benzofuryl, indoles alkynyl and benzoxazol base
(3)-C 1-10Alkyl and
(4) C 3-8Cycloalkyl, this cycloalkyl randomly with C 6-10It is aryl-condensed,
This alkyl wherein, cycloalkyl, aryl or heteroaryl are not replace or replaced by one or more following groups
(a) halogen,
(b)-C 1-10Alkyl, wherein this alkyl is not replace or replaced by halogen,
(c)-OH,
(d)-CN,
(e)-O-C 1-10Alkyl,
(f)-C 3-12Cycloalkyl and
(g)-NR 8R 9
R 2Be selected from
(1)-OH and
(2)-NR 8R 9, R wherein 8And R 9Be selected from
(a) hydrogen,
(b) C 1-10Alkyl and
(c) C0-6 alkyl-C 6-10Aryl,
Or work as R 2Be NR 8R 9, and R 8When being hydrogen with A, Q then, R 1And R 9Formation-CH can link together 2CH 2CH 2-, work as R 2Be NR 8R 9The time, then Q, R 1Can link together with A constitutes 4 or 5 carbon alkyl chains, and wherein the one or more carbon atoms in the alkyl chain can be used N, O or S atom, or SO 2Group substitutes;
R 4Be selected from group
(1)-C 1-10Alkyl, or
(2)-C 3-12Cycloalkyl, wherein this alkyl and cycloalkyl are not substituted or are replaced by one or more following groups
(a) halogen,
(b)-OH,
(c)-CN,
(d)-O-C 1-10Alkyl,
(e)-C 1-10Alkyl,
(f)-C 3-12Cycloalkyl,
(g) be selected from the aryl of phenyl and naphthyl, or
(h) heteroaryl is selected from pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidyl, pyrryl, tetrazyl, furyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl,  azoles base, different  azoles base, thiazolyl, the  di azoly, indyl, quinolyl, isoquinolyl, benzimidazolyl-, benzofuryl, indoles alkynyl and benzoxazol base
With this aryl and heteroaryl be not replace or replaced by one or more following groups
(i) halogen,
(ii)-OH,
(iii)-CN,
(iv)-O-C 1-10Alkyl,
(v)-C 3-12Cycloalkyl, or
(vi)-C 1-10Alkyl;
R 7Be selected from group
(1) hydrogen,
(2)-C 1-10Alkyl and
(3) be selected from the aryl of phenyl and naphthyl;
Wherein this alkyl and aryl are not replace or replaced by one or more following groups
(a) halogen,
(b)-OH,
(c)-CN,
(d)-O-C 1-10Alkyl,
(e)-C 3-12Cycloalkyl,
(f) be selected from the aryl of phenyl and naphthyl, or
(g) heteroaryl is selected from pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidyl, pyrryl, tetrazyl, furyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl,  azoles base, different  azoles base, thiazolyl,  di azoly, indyl, quinolyl, isoquinolyl, benzimidazolyl-, benzofuryl, indoles alkynyl and benzoxazol base;
This cycloalkyl wherein, aryl or heteroaryl are not replace or replaced by one or more following groups
(i) halogen,
(ii)-OH,
(iii)-CN,
(iv)-O-C 1-10Alkyl,
(v)-C 3-12Cycloalkyl, or
(vi) be selected from the aryl of phenyl and naphthyl;
Or R 4And R 7Formation-CH can link to each other 2CH 2CH 2-;
R 5And R 6Be independently selected from
(1) hydrogen,
(2)-C 1-10Alkyl,
(3)-C 2-10Alkenyl,
(4)-C 2-10Alkynyl group and
(5)-C 1-10Alkyl-C 3-12Cycloalkyl;
This alkyl wherein, cycloalkyl, alkenyl or alkynyl group are not replace or replaced by one or more following groups
(a) halogen,
(b)-OH,
(c)-CN,
(d)-C 1-10Alkyl
(e)-C 3-12Cycloalkyl,
(f)-O-C 1-10Alkyl,
(g) heteroaryl is selected from pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidyl, pyrryl, tetrazyl, furyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl,  azoles base, different  azoles base, thiazolyl, the  di azoly, indyl, quinolyl, isoquinolyl, benzimidazolyl-, benzofuryl, indoles alkynyl and benzoxazol base
Wherein this heteroaryl can not replace or be replaced by halogen;
(h) phenyl, or
(i)-NR 8R 9
Or R 5And R 6The continuous 4-6 person of formation encircles with the nitrogen-atoms that they connected, and it does not replace or is replaced by one or more following groups
(a)-C 1-10Alkyl,
(b)-C 3-12Cycloalkyl,
(c)-(CH 2) positive phenyl,
(d)-C 2-10Alkenyl, or
(e)-C 2-10Alkynyl group, this alkyl wherein, alkenyl and alkynyl group are not replace or replaced by one or more following groups
(i) halogen,
(ii)-OH,
(iii)-CN,
(iv)-O-C 1-10Alkyl, or
(v)-C 3-12Cycloalkyl;
With this cycloalkyl and phenyl be not replace or replaced by one or more following groups
(i) halogen,
(ii)-C 1-10Alkyl,
(iii)-OH,
(iv)-CN,
(v)-C 3-12Cycloalkyl, or
(vi)-O-C 1-10Alkyl;
N is 0,1,2,3 or 4;
With its pharmaceutically acceptable salt, with and each enantiomer and diastereomer.
In a preferred embodiment, Y 1Be N and Y 2And Y 3Be respectively CH.
In one embodiment, the compound that the present invention relates to formula (I) R wherein 1Be preferably CH of the phenyl that do not replace or replace and Q 2Preferably, R 1It is unsubstituted phenyl.
In other embodiments, R 1It is heteroaryl.Preferred R 1Heteroaryl comprises pyridyl (2-pyridyl, 3-pyridyl or 4-pyridyl), thienyl (preferred 2-thienyl or 3-thienyl), thiazole and indoles alkynyl.
In other embodiments, R 1Be C 1-12Alkyl or C 3-8Cycloalkyl.Preferred C 1-12Alkyl R 1Group comprises C 1-6Alkyl (preferred unsubstituted C 1-6Alkyl comprises methyl and sec.-propyl).Preferred C 3-8Cycloalkyl comprises cyclopropyl, and cyclopentyl and cyclohexyl are preferably unsubstituted.The ring carbon atom of two cycloalkyl can link to each other and constitute C 6-12Aryl.The isomery example of this embodiment condenses group and is:
Figure A20058001184300291
In another embodiment, the compound that the present invention relates to formula (I) R wherein 2Be NR 8R 9, and preferred R 8And R 9Be hydrogen.
In another embodiment of formula (I) compound, work as R 2Be NR 8R 9And R 8When being hydrogen respectively with A, Q then, R 1And R 9Formation-CH can link together 2CH 2CH 2-, constitute the pyrrolidyl pyrrolidyl thus.
In another embodiment of formula (I) compound, work as R 2Be NR 8R 9, Q then, R 1Constitute 4 or 5 carbon alkyl chains together with the A connexon, wherein the one or more carbon atoms in this alkyl chain can be by N, O or S atom, or by SO 2Group substitutes.
In another embodiment of formula (I) compound, R 2Be OH.
In another embodiment of formula (I) compound, A is the C that does not replace or replace (preferably not replacing) 1-10Alkyl, the C of preferred (preferably not the replacing) that does not replace or replace 1-6Alkyl and especially more preferably methyl.
In another embodiment, A can be a hydrogen.
In another embodiment of formula (I) compound, R 5And R 6With the continuous formation of its nitrogen-atoms that connects simultaneously pyrrolidine ring.
In another embodiment of formula (I) compound, R 4And R 7Be C 1-10Alkyl, preferred C 1-6Alkyl.More preferably, R 4Be methyl or sec.-propyl and R 7It is methyl.
In an embodiment of formula (I) compound, X is that the  diazole is selected from
Figure A20058001184300292
In another embodiment of formula (I) compound, X is that the  azoles is selected from
Figure A20058001184300301
In another embodiment of formula (I) compound, X is that thiazole is selected from
Figure A20058001184300302
At another embodiment of formula (I) compound, X is 1,2, the 4-triazine
Furans
Figure A20058001184300304
Pyrazoles
Pyrimidine
Figure A20058001184300306
In another embodiment, the present invention relates to the compound of formula (II)
A wherein, X, Y 1, Y 2, Y 3, Q, R 1, R 2, R 4, R 5And R 7Define as above, and R 10Be hydrogen or C 1-6Alkyl and its pharmaceutically acceptable salt and its each enantiomer and diastereomer.
At a preferred implementation of the compound of formula (II), Y 1Be N and Y 2And Y 3Be respectively CH.In an embodiment of formula (II) compound, R 1Be that phenyl and Q are CH 2Preferably, R 1It is unsubstituted phenyl.
In another embodiment, the compound that the present invention relates to formula (II) R wherein 2Be NR 8R 9And preferred R 8And R 9Be hydrogen simultaneously.
In another embodiment of formula (II) compound, R 2Be OH.
In another embodiment of formula (II) compound, R 10Be hydrogen.In another embodiment, R 10Be C 1-6Alkyl, preferable methyl, ethyl or sec.-propyl.
In another embodiment of formula (II) compound, A is C 1-10Alkyl, preferred C 1-6Alkyl, more preferably methyl.
In another embodiment of formula (II) compound, R 4And R 7Be C 1-10Alkyl, preferred C 1-6Alkyl.More preferably, R 4Be methyl or sec.-propyl and R 7It is methyl.
In another embodiment of formula (II) compound, R 5Be hydrogen or, C 1-10Alkyl, wherein C 1-10Alkyl is replaced by one or more following groups or does not replace:
(1) halogen (preferred fluorine),
(2)-OH,
(3)-CN,
(4) phenyl,
(5)-OC 1-10Alkyl, or
(6)-NR 8R 9(preferred R 8And R 9Be respectively C 1-10Alkyl).
In the compound of formula (II), preferred R 5Group comprises hydrogen, methyl, benzyl and-C 1-10Alkyl-NR 8R 9, R wherein 8And R 9Be respectively C 1-10Alkyl.
In an embodiment of formula (II) compound, X is that the  diazole is selected from
Figure A20058001184300321
In another embodiment of formula (II) compound, X is that the  azoles is selected from
In another embodiment of formula (II) compound, X is that thiazole is selected from
Figure A20058001184300323
In another embodiment of formula (II) compound, X is 1,2, the 4-triazine
Furans
Pyrazoles
Figure A20058001184300331
Pyrimidine
Work as R 10When being hydrogen, formula (II) compound of a preferred enantiomorph configuration is to have trans-S on methyl-cyclopropyl-methyl moiety, the compound of S configuration, shown in (IIA):
In another embodiment, the present invention relates to the compound of formula (III)
A wherein, X, Q, R1, R 2, R 4, R 5, R 6And R 7Define as above, and R 10Be hydrogen or C 1-6Alkyl and its pharmaceutically acceptable salt and its each enantiomer and diastereomer.
Another embodiment of the present invention comprises the compound of the title compound that is selected from the following example and its pharmaceutically acceptable salt.
So used, term " alkyl " itself or as another substituent integral part is meant saturated straight chain or branched hydrocarbyl (for example, C with defined amount carbon atom 1-10Alkyl is meant the alkyl with 1-10 carbon atom).The preferred alkyl that the present invention uses is C 1-6Alkyl has 1-6 carbon atom.The example alkyl comprises methyl, ethyl, n-propyl, sec.-propyl, normal-butyl, isobutyl-, the tertiary butyl, amyl group, hexyl etc.
So used, term " alkenyl " itself or as another substituent integral part is meant straight or branched alkyl (for example, the C that has a carbon-to-carbon double bond and specify carbonatoms 2-10Alkenyl is meant the alkenyl with 2-10 carbon atom).The preference chain thiazolinyl that the present invention uses is C 2-6Alkenyl has 2-6 carbon atom.The example chain thiazolinyl comprises vinyl and propenyl.
So used, term " cycloalkyl " itself or as another substituent integral part is meant to have the saturated monocycle of specifying carbonatoms, encircle or bridged cyclic hydrocarbon group (for example, C more 3-12Cycloalkyl is meant the cycloalkyl with 3-12 carbon atom).Preferred cycloalkyl comprises C 3-8Cycloalkyl, especially C 3-8Monocyclic cycloalkyl.The example monocyclic cycloalkyl comprises cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl etc.Example bridged ring alkyl comprises adamantyl and norcamphyl.
So used, term " aryl " itself or as another substituent integral part is meant to have aromatics or cyclic group (for example, the C that specifies carbonatoms 6-10Aryl is meant the aryl with 6-10 carbon atom).The preferred aryl groups that the present invention uses comprises phenyl and naphthyl.
Term " halo " or " halogen " comprise fluorine, chlorine, bromine and iodine.
So used, term " heteroaryl " itself or as another substituent integral part is meant have at least one ring hetero atom aromatics cyclic group of (O, N or S).The example heteroaryl that the present invention uses comprises furyl, pyranyl, benzofuryl, isobenzofuran-base, chromenyl, thienyl, the benzo thiophenyl, pyrryl, pyrazolyl, imidazolyl, pyridyl, pyrazinyl, pyrimidyl, pyridazinyl, indyl, indazolyl, benzimidazolyl-, quinolyl and isoquinolyl.
When the heteroaryl of this paper definition is substituted, this substituting group can with the ring carbon atom bonding of heteroaryl, or be positioned on the ring hetero atom (that is, nitrogen, oxygen or sulphur), they have and allow the valency that replaces.Preferably, this substituting group bonding ring carbon atom.Similarly, when heteroaryl was defined herein as substituting group, tie point can be on the ring carbon atom of heteroaryl, or on ring hetero atom (that is, nitrogen, oxygen or sulphur), and they have and allow the valency that replaces.Preferably, connection can be positioned at ring carbon atom.
Compounds more of the present invention have at least one asymmetric center.May there be additional asymmetric neutrality in character according to different substituents on the molecule.Compound with asymmetric center produces enantiomer (optical isomer); Diastereomer (conformer) or both and scope of the present invention comprise the possible enantiomer and the diastereomer of all miscellanys and purifying and partial purification compound.The present invention includes all these type of isomeric form of these compounds.
The independence of enantiomerism or diastereo-isomerism enrichment compound is synthetic, or its chromatographic separation, can obtain by suitable improvement method disclosed herein according to methods known in the art.Its absolute stereo chemistry can be measured by the X-radiocrystallgraphy of crystalline product and deutero-crystal intermediate, if necessary, adopts the reagent of the asymmetric center that contains known absolute configuration.
If wish that the racemize miscellany of described compound can separate, thereby isolates single enantiomer.This separation can be undertaken by method well known in the art, for example the coupling of the compound of the racemize miscellany of described compound and enantiomer-pure forms the diastereomer miscellany, subsequently by standard method for example fractional crystallization and the single diastereomer of chromatographic separation.Linked reaction utilizes the acid of enantiomer-pure or alkali to form salt usually.The diastereomer derivative can be converted into pure enantiomer by the additional chirality residue of cracking.The racemize miscellany of described compound can also directly utilize chiral solid phase to separate by chromatographic process, and this method is well known in the art.
In addition, any enantiomer of compound can utilize the optical purity starting raw material of configuration known or reagent to obtain by method well known in the art by stereospecific synthesis.
In the compound of formula (I)-(III), with R 2, A and Q bonding carbon atom remove and be chiral carbon.So can there be racemic modification in the compound of formula (I)-(III), perhaps with pure (R) of stereochemistry or (S) form.The isomer of formula (I) compound is as follows:
Figure A20058001184300351
(be generally (R) configuration, for example work as A is CH to preferred above-mentioned first configuration 3, R 2Be NH 2, Q is-CH 2-and R 1When being phenyl).
Compound of the present invention can be according to following universal method and specific embodiment 1-94 preparation (route plan 1-25).
At route plan, the amino acid derivative of 1 type is converted into corresponding diazanyl acid amides 3 through two step programs.For the amino acid derivative that obtains to be purchased, can adopt two step alkylated reactions of glycine Schiff's base 4.The Schiff's base deprotection, Boc protection and ester hydrolysis provide another approach that obtains compound 2.Be used for synthetic 5 with 4 alkylated reaction can with the enantioselectivity mode according to the described method of document carry out (referring to K.Maruoka etc., J.Am.Chem.Soc.2000,122,5228-5229 and M.North etc., Tetrahedron Lett.2003,44,2045-2048).
Route plan 1
Figure A20058001184300361
In route plan 2, the amino acid 2 of N-protected is converted into carboxylic acid amides 6a (with thiocarboxamide 6b), and subsequent dewatering obtains nitrile 7.7 use NH 2OH-HCl handles under weak basic condition and obtains oxyamide thing 8.
Route plan 2
Figure A20058001184300371
In route plan 3, the amino acid/11 BH that generates on the spot 3Reduction obtains corresponding amino alcohol, and this compound can be obtained compound 9 by N-protected subsequently.9 oxidation generates aldehyde 10, and it can be converted into imines 11 subsequently under mild conditions.
Route plan 3
Figure A20058001184300372
The alcohol of amino alcohol 12 is shown in route plan 4.10 epoxidation reaction is opened with ammonia subsequently and is obtained amino alcohol 12.
Route plan 4
Figure A20058001184300381
Route plan 5 illustrates the preparation of regional isomer amino alcohol 13: the Ellman derivative of aldehyde 10 synthetic, and the Ge Shi addition of vinyl, ozone decomposes and reduction is handled, and removes the Ellman assistant agent subsequently.
Route plan 5
Figure A20058001184300382
Route plan 6 has been described the preparation of bromoketone 14 processes derived from two azo ketone of acid 2.
Route plan 6
Route plan 7 has been described the preparation of intermediate 15-18, is used to modify different heterocycles.In addition, after heterocycle X has formed (intermediate 15 '-18 '), can stay next complete cl radical so that R is introduced in the back 5R 6N.Note, if desired, also can form back quilt-(R at heterocycle X by the residual second complete down cl radical 7) N-SO 2R 4Substitute.
Route plan 7
Route plan 8-21 has described the preparation of different heterocyclic X.1,3 of 19 types, the synthetic of 4- diazole can dewater with Burgess reagent ring subsequently, and the Boc deprotection be finished (route plan 8) by with acid 15 and 3 couplings of diazanyl acid amides.
Route plan 8
In addition, acyl group-hydrazides can be directly derived from the acid of 15 types so that can be incorporated into subsequently in the  diazole ring by the different aminoacids derivative.Last R 1Introducing subsequently of group can be carried out (route plan 8A) by the Schiff's base of 45 types.
Route plan 8A
Figure A20058001184300402
Two kinds possible 1,2, a kind of synthetic shown in route plan 9 in the 4- diazole.Acid 15 and 11 couplings of oxyamide thing, ring dehydration and Boc deprotection obtain  diazole 20 under alkaline condition subsequently.
Route plan 9
The synthetic of 1,2, the 4  diazole that other are possible can be obtained by nitrile 17.Reaction sequence is similar to aforesaid method and obtains  diazole 21 (route plan 10).
Route plan 10
Figure A20058001184300412
1,2,4-triazoles 22 is described in route plan 11.Nitrile 17 transforms to corresponding inferior amine salt under alkaline condition, obtains required heterocycle with 3 backflows of diazanyl acid amides and Boc deprotection subsequently.
Route plan 11
Figure A20058001184300413
Imidazoles 24 is according to route plan 12 described preparations.12 with formic acid coupling and oxidation, closed loop obtains imidazoles 23 subsequently.Bromide 16 is converted into corresponding boric acid through the lithium halogen exchange, has palladium mediatedly finished this reaction process with coupling imidazoles 23 and Boc deprotection, obtains compound 24.
Route plan 12
Figure A20058001184300421
1,2,3-triazoles 26 synthetic as described in the route plan 13.The two azo  intermediates that obtain via aniline 8 form aromatic yl azide, obtain 25 with the cycloaddition of suitable short of electricity alkynes subsequently.The formation of Ellman sulfinyl imines, nucleophilic addition, synthesizing subsequently by chirality assistant agent cracking realization 26.
Route plan 13
Synthesizing shown in route plan 14 of  azole 27, thiazoles 28 and imidazoles 29.Acid 15 reduction is oxidized to aldehyde, epoxidation reaction, and epoxide is opened with ammonia, subsequently the amino alcohol of gained and sour 2 couplings and oxidation is obtained general intermediate ketone group acid amides.The ring dehydration generates 27,28 or 29 under the described conditions.
Route plan 14
Figure A20058001184300441
The regional isomerism  azoles of second series and thiazoles can be synthetic according to route plan 15 described methods by aromatic bromide 16.Crosslinked, the asymmetric amination of Sharpless and obtain general midbody acid amide-aldehyde with sour 2 couplings.Adopt above-mentioned condition to obtain  azoles 30, thiazole 31, and another obtains the approach of imidazoles 29.
Route plan 15
Figure A20058001184300451
The another way of route plan 16 expression preparation heterocycle 29-31.Acid 15 is converted into corresponding bromoketone through intermediate two azo ketone.Substitute and cyclisation with carboxylic acid amides 6a, remove Boc subsequently and obtain  azoles 30.Substitute and cyclisation with thiocarboxamide 6b, remove Boc subsequently and obtain thiazole 31.Substitute and cyclisation in the presence of the ammonia source with acid 2, remove Boc subsequently and obtain imidazoles 29.
Route plan 16
Following route plan 17 is described regional isomerism  azole 32, thiazoles 33 and imidazoles 34 synthetic of the 3rd series.Acid 15 and amino alcohol 12 couplings, ring dehydration under proper condition, the Boc deprotection obtains required compound subsequently.
Route plan 17
Figure A20058001184300462
Synthesizing shown in route plan 18 of the  azole 35 in the 3rd possibility XiLin, thiazoles 36.Amino alcohol 13 obtains general intermediate aldehydes-acid amides with the EDC coupling of acid 15.The ring dehydration obtains required 5-membered heterocycles under proper condition.
Route plan 18
Figure A20058001184300471
The mode of the another kind of preparation of route plan 19 expressions heterocycle 34-36.Bromoketone 14 usefulness acid 15 substitutes, and cyclisation in the presence of the ammonia source is removed Boc subsequently and obtained imidazoles 34.Substitute and cyclisation with carboxylic acid amides 37, remove Boc subsequently and obtain  azoles 35.Substitute and cyclisation with thiocarboxamide 38, remove Boc subsequently and obtain thiazole 36.
Route plan 19
Figure A20058001184300481
The different  azole of 40 types can be according to route plan 20 described method preparations.Aromatic bromide 16 obtains 39 with the cross-coupling and the deprotection of TMS acetylene.The cycloaddition of the nitrile oxide derivative made from oxidation 11 on the spot, the Boc deprotection obtains 40 subsequently.
Route plan 20
Figure A20058001184300482
The second different  azoles regional isomer synthetic can utilize and be similar to route plan 20 described flow processs and finish.Aldehyde 10 can utilize the Corey-Fuchs method to be processed as alkynes 41.The acid reduction, the formation of oxidation and hydroxyl imide salt obtains 42.Generate nitrile oxide and cycloaddition on the spot by 41, remove Boc subsequently and obtain different  azoles 43.
Route plan 21
Figure A20058001184300491
Route plan 22 has been described the preparation of intermediate 49, is used to prepare the  diazoles compound of 50 types.Carboxylic acid 48 also can be used for preparing the multiple heterocyclic compound of above-mentioned route plan.
Route plan 22
Route plan 23 expressions have the preparation method of the The compounds of this invention of pyrazoles or pyrimidine X group.
Route plan 23
Figure A20058001184300502
Route plan 24 has been described the preparation of intermediate 54, is used to prepare the  diazoles compound of 55 types.Carboxylic acid 54 also can be used to modify the multiple heterocyclic compound described in the above-mentioned multiple route plan.
Route plan 24
Figure A20058001184300511
Route plan 25 has been described compound synthetic of 58 types.Be purchased the diazotization and the iodate of 2-chloro-4-amino-6-chloropyridine, obtain Furan Aldehydes 56 by the Suzuki cross-coupling subsequently.The Ge Shi addition obtains intermediate ethanol 57 with rear oxidation and organo-metallic addition subsequently.Above-mentioned R 4And R 7Introducing, Ritte reaction subsequently, R 5And R 6Assembling and trinitride reduction obtain 58 compounds of final structure.
Route plan 25
Term " pure substantially " is meant by analytical technology known in the art and measures, and purity is at least 90% and preferred purity 95% and the special more preferably separate substance of purity 99%.
Term " pharmaceutically acceptable salt " is meant the salt that can be accepted nontoxic alkali or acid preparation by pharmacy, comprises inorganic or organic bases and inorganic or organic acid.Compound Ke Yishi one, two of the present invention or three salt depend on the acid functionality's who exists in this compound free alkali number.Free alkali and comprise aluminium by mineral alkali deutero-salt, ammonium, calcium, copper, iron, ferrous, lithium, magnesium, manganese salt, inferior manganese, potassium, sodium, zinc etc.Preferred especially ammonium, calcium, magnesium, potassium and sodium salt.May there be more than a kind of crystalline structure in the salt of solid form and can also be the form of hydrate.The salt that can accept organic nontoxic alkali derived from pharmacy comprises primary, the salt of the second month in a season and tertiary amine comprises natural substitutional amine-group, cyclic amine and deacidite, arginine for example, trimethyl-glycine, caffeine, choline, N, N '-dibenzyl ethylidene-diamines, diethylamine, 2-DEAE diethylaminoethanol, 2-dimethylaminoethanol, thanomin, ethylene diamine, N-ethyl-morpholine, N-ethylpiperidine, glycosamine, glucosamine, Histidine, hydrab amine, Isopropylamine, Methionin, methylglucosamine, morpholine, piperazine, piperidines, versamid 900, PROCAINE HCL, PHARMA GRADE, the purine class, Theobromine, triethylamine, Trimethylamine 99, tripropyl amine, Trometamol etc.When compound of the present invention was alkalescence, salt can be accepted the non-toxic acid preparation by pharmacy, comprises inorganic and organic acid.This type of acid comprises acetate, trifluoroacetic acid, Phenylsulfonic acid, phenylformic acid, camphorsulfonic acid, citric acid, ethyl sulfonic acid, fumaric acid, glyconic acid, L-glutamic acid, Hydrogen bromide, hydrochloric acid, isethionic acid, lactic acid, toxilic acid, oxysuccinic acid, amygdalic acid, methylsulfonic acid, glactaric acid, nitric acid is pounced on acid, pantothenic acid, phosphoric acid, succsinic acid, sulfuric acid, tartrate, tosic acid etc.Special optimization citric acid, Hydrogen bromide, hydrochloric acid, trifluoroacetic acid, toxilic acid, phosphoric acid, sulfuric acid, fumaric acid and tartrate.
The present invention relates to compound disclosed herein as β-short secretase activity or the active inhibitor of β-site amyloid precursor protein-lyase (" BACE ") in for example application in the Mammals of this inhibiting patient of needs or object, comprise the described compound of using significant quantity.Term " β-short Secretases ", " β-site amyloid precursor protein-lyase " and " BACE " can exchange use in this manual.Except the mankind, many other Mammalss can be handled according to method of the present invention.
The invention further relates to and prepare the medicine or the method for compositions that contain The compounds of this invention and pharmaceutical carriers or thinner that is used for suppressing human body and animal β-short secretase activity.
Compound of the present invention has utilizability in the danger of treatment, improvement, control or reduction Alzheimer.For example, described compound can be used to prevent the dementia of Alzheimer type, and be used for the Alzheimer type early stage, mid-term or late period dementia.Described compound can also be used for the treatment of, improves, controls or reduce danger and other illnesss that can pass through inhibition β-short Secretases treatment or divide of the disease that unusual cracking mediated (being also referred to as APP) of amyloid precursor protein.This type of illness comprises mild cognitive damage, Trisomy 21 (mongolism), brain amyloid blood vessel disease, sex change dementia, hereditary cerebral hemorrhage are followed Dutch type amyloidosis (HCHWA-D), Creutzfeld-JakobShi disease, prion disease, amyotrophic lateral sclerosis is benumbed on the carrying out property nuclear, head trauma, apoplexy, mongolism, pancreatitis, embedding gonosome myositis, other periphery amyloid, diabetes and atherosclerosiss.
Object that The compounds of this invention is used or patient generally are the mankind that wherein wish to suppress β-short secretase activity, sex, but also can comprise other Mammalss, dog for example, cat, mouse, rat, ox, horse, sheep, rabbit, monkey, chimpanzee or other apes or primates wherein need to suppress β-short secretase activity or treat above-mentioned disease.
Compound of the present invention can be united with one or more other drugs and is used for disease or the treatment of conditions that The compounds of this invention is suitable for, and the coupling of its Chinese traditional medicine keeps away that it is separately safe in utilization or more effective.In addition, compound of the present invention can or reduce the side effect or the toxic medication combined use of The compounds of this invention with one or more other treatments, prevention, control, improvement.This type of other drug can be by certain approach and with its usual amounts and The compounds of this invention administration simultaneously or sequentially.So pharmaceutical composition of the present invention comprises those that contain one or more other activeconstituentss except that The compounds of this invention.The calcium cooperative programs can be used as the integral part administration of a unit dosage joint product, and perhaps as cover box or treatment plan administration, wherein one or more medications are to separate a part of administration of formulation as treatment plan.
The cooperative programs example of the unit dosage of The compounds of this invention and other drug or cover box-like formula comprises the cooperative programs that contain the Kang Aercihaimoshi medicine, for example other β-short Secretase inhibitors or γ-short Secretase inhibitors; The tau phosphorylation inhibitor; The retarding agent that the A beta oligomers forms; The p25/CDK5 inhibitor; The HMG-CoA reductase inhibitor; The NK1/NK3 receptor antagonist; NSAID class medicine comprises Ibuprofen BP/EP; Vitamin-E; Anti-amyloid antibody; Anti-inflammatory compound, for example (R)-flurbiprofen; CB-1 receptor antagonist or CB-1 receptor inverse agonists; Antibody is doxycycline and Rifampin for example; N-methyl-D-aspartate (NMDA) receptor antagonist, for example Memantine hydrochloride; Anticholinesterase is lycoremine for example, and thunder department replaces bright, solely Buddhist nun's inferior and tacrine; The tethelin succagoga is ibutamoren for example, methylsulfonic acid ibutamoren, and capromorelin; Histamine H 3Antagonist; The AMPA agonist; PDE IV inhibitor; The GABAA inverse agonist; Neuronal nicotinic sample agonist; P-450 inhibitor, for example ritonavir; Or other raising The compounds of this invention that influence acceptor or enzyme are tired, security, accessibility or reduce adverse side effect or toxic medicine.The above-mentioned catalogue of cooperative programs only is to illustrate and qualification never in any form.
Term used herein " composition " comprises the product of the specific components that contains predetermined amount or ratio, and any product that is directly or indirectly obtained by the cooperative programs of the specific components of specified quantitative.This term relevant with pharmaceutical composition comprised the product of the carrier that contains one or more activeconstituentss and choose wantonly, described carrier comprises inert fraction, and it is any directly or indirectly by the associating of two or more components arbitrarily, compound or assemble the product that obtains, perhaps by other reactions of one or more components or the product that reacts to each other and obtain.Usually, pharmaceutical composition closely makes activeconstituents and liquid vehicle or absorption solid financial group or two kinds combine to prepare by all even, and subsequently, if necessary, is required preparation with this product shaping.Active target compound is enough to exist the process of disease or the amount of situation generation predictive role in pharmaceutical composition.So pharmaceutical composition of the present invention comprises any composition by The compounds of this invention and pharmaceutical acceptable carrier cyclisation are made.
The pharmaceutical composition that orally uses can contain one or more reagent that are selected from sweeting agent, correctives, tinting material and sanitas according to the preparation of the method for pharmaceutical compositions known in the art and this based composition provides pharmaceutically attractive in appearance and good to eat preparation.Tablet contains the activeconstituents mixed mutually with the pharmaceutical acceptable excipient that is fit to the preparation tablet.These vehicle can be for example, inert diluent, lime carbonate for example, yellow soda ash, lactose, calcium phosphate or sodium phosphate; Granulating agent and disintegrating agent, for example W-Gum or alginic acid; Tackiness agent, starch for example, gelatin, polyvinylpyrrolidone or gum arabic, and lubricant, for example, Magnesium Stearate, stearic acid or talcum powder.Tablet is dressing not, and perhaps they can also obtain slow releasing function for more time with its dressing with the disintegration and the absorption of delay in gi tract thus by known technology.
The preparation that orally uses also can be used as hard gelatin capsule and exists, and wherein activeconstituents and inert solid diluent are mixed, and this inert solid diluent for example is lime carbonate, calcium phosphate or kaolin; Or as the soft gelatin capsule existence, wherein this activeconstituents and water or oil medium such as peanut oil, whiteruss or sweet oil are mixed.
The other drug composition comprises aqueous suspension, and it contains and the mixed active substance of vehicle that is fit to the preparation aqueous suspension.In addition, oil-based suspension can for example be prepared in the whiteruss by activeconstituents being suspended in vegetables oil such as peanut oil, sweet oil, sesame oil or Oleum Cocois or being suspended in mineral oil.Oil-based suspension can contain different vehicle.Pharmaceutical composition of the present invention can also be an O/w emulsion, and it also can contain vehicle for example sweeting agent and correctives.
This pharmaceutical composition can be the form of sterilization injectable water or oil-based suspension, and it can be prepared according to methods known in the art, perhaps can use with the form of rectal administration suppository.
Compound of the present invention also can be by sucking, utilize suction apparatus administration well known by persons skilled in the art, perhaps by the transdermal patch administration.
So-called " pharmacy can be accepted " be meant carrier, thinner or vehicle must with other component compatibility of preparation and harmless to the recipient.
Term " is used " and " administration " should be understood to be meant compound of the present invention is offered in the individuality that needs treatment being incorporated into the intravital form of individual machine with treatment useful form and treatment significant quantity, includes, but are not limited to oral dosage form, tablet for example, capsule, syrup, suspensoid etc.; Injectable dosage formulations, IV for example, IM, or IP etc.; The transdermal formulation comprises creme, frozen glue, powder or patch; Through buccal dosage form; Suck powder, sprays, suspensoid etc.; And rectal suppository.
Term " significant quantity " or " treatment significant quantity " are meant the active compound that is searched out by researchist, animal doctor, clinician and other clinical positions person of described compound or medicament produces biology or medical response in tissue, system, animal or human's body amount.So used, term " treatment " is meant above-mentioned treatment of conditions, particularly in the patient of the symptom that has been proved disease or illness.
So used, term " treatment " or " processing " is meant and uses compound of the present invention and comprise that (1) suppresses this disease (promptly in experience or performance remove the animal of the pathology of disease or symptom, stop further developing of pathology and/or symptom), or (2) improve disease (that is, reversing this pathology and/or symptom) in experience or performance remove the animal of the pathology of disease or symptom.
Term " control " comprises preventative processing, elimination, improvement or reduces the seriousness of controlled illness.
The composition that contains The compounds of this invention generally exists with unit dosage and can prepare by the method that pharmaceutical field is known.Term " unit dosage " is meant that wherein all active and non-active ingredients are combined into the single formulation of a suitable system, the patient can open a container or packing maybe for this medicament administration patient's people and wherein contains all dosage thus, and does not need any composition in two or more containers or the packing mixed.The representative instance of unit dosage is the tablet or the capsule of oral administration, the single dose bottle of injection, or the suppository of rectal administration.The above-mentioned catalogue of unit dosage limits never in any form, and only is the existing representative instance of unit dosage.
The composition that contains The compounds of this invention can be used as the cover box usually and exists, two or more components thus, it can be activity or inactive component, carrier, thinner etc., offers the patient or gives the people of patient's drug administration with the specification sheets of reagent preparation formulation.This type of cover box can contain and material and the component that is necessary be provided, or they can comprise and using or preparation patient or need the material that can independently obtain or the specification sheets of composition for the people of patient's drug administration.
When pointing out with compounds for treating of the present invention, improvement, control or reducing Alzheimer or other diseases dangerous, can obtain gratifying result when usually The compounds of this invention is with the per daily dose administration of the about 100mg/kg per weight of about 0.1mg-, preferably as single per daily dose administration or with separate doses administration every day 2-6 time, or take the slowly-releasing form.Total per daily dose is the about 2000mg of about 1.0mg-, the about 20mg/kg body weight of preferably about 0.1mg-.In the adult situation of 70kg, total per daily dose generally is that about 7mg-is about 1,400mg.This dosage can be adjusted to produce nearest therapeutic response.Described compound can adopt 1-4 time scheme administration every day, preferred every day 1 or 2 times.
Concrete dosage The compounds of this invention or its pharmaceutically acceptable salt comprises 1mg, 5mg, 10mg, 30mg, 80mg, 100mg, 150mg, 300mg and 500mg.Pharmaceutical composition of the present invention can provide to contain the 0.5mg-1000mg formulations of active ingredients of having an appointment; More preferably contain the 0.5mg-500mg activeconstituents of having an appointment; Or 0.5mg-250mg activeconstituents; Or 1mg-100mg activeconstituents.The certain drug composition that is used for the treatment of can contain the 1mg that has an appointment, 5mg, 10mg, 30mg, 80mg, 100mg, 150mg, the activeconstituents of 300mg and 500mg.
Yet, should understand the concrete dosage level of any particular patient and administration and can change this moment and should depend on multiple factor, comprise the activity of used specific compound, the time length of metabolic stability and compound effects, age, body weight, whole body health, sex, diet, mood and administration time, excretion rate, the therapy medication combined, that the seriousness of particular disorder and host carry out.
The compounds of this invention can confirm by methods known in the art as the practicality of β-short secretase activity inhibitor.Enzyme inhibition is measured as follows.
The ECL analytical method: evenly terminal point electrochemiluminescence (ECL) analysis is adopted biotinylation BACE substrate.The Km of substrate is greater than 100 μ M and owing to the restriction of this unit solubleness can't be measured.Type reaction contains the 0.1nM enzyme of having an appointment, the substrate of 0.25M, and damping fluid (50mM NaOAc, pH4.5,0.1mg/ml BSA, 0.2% CHAPS, 15mM EDTA and 1mM Deferoxamine) and total reaction volume are 100 μ l.Reaction process is 30 minutes, adds the 1M Tris-HCL of 25 μ L subsequently, and pH8.0 ends.Gained enzyme product is analyzed by adding rutheniumization (ruthenylated) antibody, and this antibodies specific is discerned the C-end of this product.Join the magnetic bead of streptavidin coating in this solution and sample carries out M-384 (Igen Inc., Gaithersburg MD) analyze.Under these conditions, handle with BACE 1 less than 10% substrate.Used enzyme dissolves in the human protein in (do not comprise membrane spaning domain and kytoplasm stretch) baculovirus expression system in these researchs.For the inhibition of measuring compound is tired, contain the solution (inhibitor of 12 kind concentration be by 100M begun preparation and carried out three times serial dilutions) of inhibitor in DMSO in the reaction mixture (the DMSO final concentration is 10%).All tests at room temperature utilize above-mentioned standard reaction condition to carry out.For measuring the IC of compound 50, four parameter formula are used for fitting of a curve.The error of reproducing dissociation constant is generally less than 2 times.
The HPLC test: evenly (tonka bean camphor-CO-REVNFEVEFR), its cracking under BACE 1 effect discharges the N-terminal fragment that is connected with tonka bean camphor to terminal point HPLC test employing substrate.The Km of this substrate is greater than 100 μ M and owing to the restriction of this unit solubleness can't be measured.Type reaction contains the 2nM enzyme of having an appointment, and the substrate of 1.0 μ M and damping fluid (50mM NaOAc, pH4.5,0.1mg/ml BSA, 0.2% CHAPS, 15mM EDTA and 1mM deferoxamine) and cumulative volume are 100 μ l.This reaction was carried out 30 minutes and by adding the 1M Tris-HCL of 25 μ L, pH8.0 ends.The gained reaction mixture is carried on the HPLC and product separates with substrate with 5 development linear gradients.Under these conditions, carry out BACE 1 less than 10% substrate.Used enzyme dissolves in the human protein in (do not comprise membrane spaning domain and kytoplasm stretch) baculovirus expression system in these researchs.For the inhibition of measuring compound is tired, contain the solution (inhibitor of 12 kind concentration be by 100M begun preparation and carried out three times serial dilutions) of inhibitor in DMSO in the reaction mixture (the DMSO final concentration is 10%).All tests at room temperature utilize above-mentioned standard reaction condition to carry out.For measuring the IC of compound 50, four parameter formula are used for fitting of a curve.The error of reproducing dissociation constant is generally less than 2 times.
Particularly, the compound of the following example has the activity of inhibition β-short Secretases, the IC that generally has in one or both above-mentioned tests 50Be about 1nM-100 μ M.This type of result is the index of used inhibitor as the intrinsic activity of β-short secretase activity inhibitor.
The Several Methods of preparation The compounds of this invention is in route plan and the embodiment illustrated of this paper.Starting raw material is according to methods known in the art or in this method that exemplifies preparation.Provide the following example to understand the present invention more comprehensively.These embodiment should not limit the present invention for example by any way.
Intermediate compound I: R-N-(tert-butoxycarbonyl)-Alpha-Methyl phenylalanine hydrazi acid amides (route plan 1)
Figure A20058001184300581
Steps A: Boc protection
To D-Alpha-Methyl phenylalanine (1.00g, 5.58mmol) add in the slurries in 20mL two  alkane 3N NaOH (7.4mL, 22.32mmol) and Boc2O (1.28g, 5.86mmol).Make this reaction carry out 14 hours.Reduce pH by dripping 1N HCl, dilute with water, and water extracts with EtOAc (3x).The organism Na that merges 2SO 4Drying is filtered and is concentrated and obtains expecting product, and it is a white foam.This material just need not to be further purified and can use. 1H?NMR(d 4-MeOH)δ7.25-7.17(m,3H),7.12(d,J=6.6Hz,2H),3.27(d,J=13.4Hz,1H),3.15(d,J=13.4Hz,1H),1.45(s,9H),1.39(s,3H)。LCMS[(M-Boc)+H] +=180
Step B: the formation of diazanyl acid amides
(1.50g is 5.37mmol) at 25mL CH to the R-N-Boc-of steps A Alpha-Methyl phenylalanine 3Add in the solution among the CN EDC (1.75g, 9.13mmol), add subsequently hydrazine (0.421mL, 13.43mmol).Generate white precipitate at once, this solution became clarification gradually in 1 hour.Make this reaction at room temperature spend the night, add saturated NaHCO 3The aqueous solution is ended it, dilutes with EtOAc.Separate each layer, water layer washs with fresh EtOAc (3x).The organic layer Na that merges 2SO 4Drying is filtered and is concentrated and obtains white foam, and it need not to be further purified and can use. 1H?NMR(d 4-MeOH)δ7.27-7.20(m,3H),7.11(d,J=7.7Hz,2H),3.30(d,J=13.5Hz,1H),3.02(d,J=13.5Hz,1H),1.46(s,9H),1.31(s,3H)。LCMS[[(M-Boc)+H] +=194
Intermediate II: R-N-(tert-butoxycarbonyl)-Alpha-Methyl phenylalanine amides (route plan 2)
(2.04g is 7.30mmol) at 26mL CH to N-Boc-D-Alpha-Methyl phenylalanine 2Cl 2In solution in add EDC (1.54g, 8.03mmol), add subsequently HOAt (0.845g, 6.21mmol).After following 30 minutes of the room temperature, adding 15mL DMF, this reaction is cooled to~and 10 ℃, in this reaction, be blown into ammonia 50 minutes.This reaction is by adding saturated NaHCO 3The aqueous solution is ended and is diluted with EtOAc.Separate each layer, water layer washs with new EtOAc (2x).The organism that merges is used Na with the 3M LiCl aqueous solution and salt water washing 2SO 4Drying is filtered and is concentrated.This resistates is by normal-phase chromatography purifying (0->6% MeOH/CH 2Cl 2) obtain the white solid of required carboxylic acid amides. 1H?NMR(d 4-MeOH)δ7.96(br?s,1H),7.28-7.13(m,3H),7.12(d,J=7.4Hz,2H),6.39(br?s,1H),5.81(br?s,1H),3.32(d,J=13.7Hz,1H),3.10(d,J=13.7Hz,1H),1.44(s,9H),1.40(s,3H)。LCMS[(M-Boc)+H] +=179。
Intermediate III: (3R-amino-1-benzyl-2RS-hydroxyl-1-methyl-propyl) t-butyl carbamate (route plan 3,4)
Steps A: reduction
To D-Alpha-Methyl-phenylalanine (1.74g, 9.71mmol) in the solution in 30mL THF in room temperature property adding next time NaBH4 (0.92g 24.27mmol).This solution is cooled to 0 ℃.Drip in 30 minutes the iodine be present among the 5mL THF (2.46g, 9.71mmol).Behind reinforced the finishing, should react reflux 2 days.This reaction is cooled to 0 ℃ and end until stopping bubbling by adding methyl alcohol subsequently.This reaction mixture is acidified to pH1 by adding 6N HCl, stirs 30 minutes down and vacuum concentration at 50 ℃.Utilize ion-exchange chromatogram purification (SCX box) to obtain the white solid of 2R-amino-2-methyl-3-phenyl third-1-alcohol I. 1H NMR (400MHz, CDCl 3) δ 7.35-7.18 (m, 5H), 3.36 (A of AB, d, J=10.4Hz, 1H), 3.31 (B of AB, d, J=10.4Hz, 1H), 2.70 (s, 2H), 1.04 (s, 3H).
Step B:Boc protection
With 2R-amino-2-methyl-3-phenyl third-1-alcohol (4.14g, 25mmol) and tert-Butyl dicarbonate (7.1g, 32.5mmol) solution at room temperature stirred 16 hours, concentrate and to obtain (1-benzyl-2R-hydroxyl-1-methylethyl) t-butyl carbamate), it is a white solid. 1HNMR (400MHz, CDCl 3) δ 7.35-7.15 (m, 5H), 4.48 (br s, 1H), 4.17 (br s, 1H), 3.76-3.62 (m, 2H), 3.19 (A of AB, d, J=13.6Hz, 1H), 2.81 (B of AB, d, J=13.6Hz, 1H), 1.47 (s, 9H), 1.07 (s, 3H).
Step C: oxidation
To (1-benzyl-2R-hydroxyl-1-methylethyl) t-butyl carbamate (6.7g, 25.2mmol) add in the solution in DCM (100mL) and DMSO (25mL) triethylamine (10.5mL, 75.7mmol) and sulphur trioxide-pyridine (10g, 63.1mmol).This reaction mixture at room temperature stirred 3.5 hours, with the EtOAc dilution, used 10% KHSO 4, saturated NaHCO 3, water, salt solution and LiCl solution washing, use dried over sodium sulfate, vacuum concentration and obtain the white solid of (1-benzyl-1-methyl-2R-oxoethyl) t-butyl carbamate by purification by flash chromatography (silica gel, 0-20% EtOAc/ hexane). 1H NMR (400MHz, CDCl 3) δ 9.53 (s, 1H), 7.35-7.22 (m, 3H), 7.12-7.00 (m, 2H), 4.84 (br s, 1H), 3.17 (A of AB, d, J=13.6Hz, 1H), 3.08 (B of AB, d, J=13.6Hz, 1H), 1.51 (s, 9H), 1.27 (s, 3H).
Step D: epoxidation reaction
To N-((1-benzyl-1-methyl-2R-oxoethyl) t-butyl carbamate (and 1g 3.80mmol) adds 6 and drips in the solution in acetonitrile (15mL), trimethyl sulfonium iodide (775mg, 3.80mmol) and potassium hydroxide (511mg, 9.11mmol).This is reflected at 60 ℃ of lower seals stirred 1.5 hours, add additional trimethyl sulfonium iodide (775mg, 3.80mmol) and potassium hydroxide (511mg 9.11mmol) and stirring under should react 60 ℃, seals and reaches 3 hours.Reaction mixture is diluted with EtOAc, use saturated NaHCO 3The aqueous solution, salt water washing obtain the oil of (1-methyl isophthalic acid-oxyethane-2R-base-2-phenylethyl) t-butyl carbamate with dried over sodium sulfate and vacuum concentration.MS(ES,M+H)278。
Step e: the open loop of epoxide
(986mg is 3.56mmol) at EtOH (35mL) and NH with (1-methyl isophthalic acid-oxyethane-2R-base-2-phenylethyl) t-butyl carbamate 4Solution among the OH (35mL) stirred 16 hours at 60 ℃ of lower seals, vacuum concentration and by purification by flash chromatography (silica gel, 0-30% (10%NH 4OH/MeOH)/CH 2Cl 2) obtain the viscous crude of (3R-amino-1-benzyl-2RS-hydroxyl-1-methyl-propyl) t-butyl carbamate. 1H NMR (400MHz, CD 3OD, 1: 1 non-enantiomer mixture) δ 7.30-7.14 (m, 5H), 4.01 (br d, J=9.2Hz, 0.5H), 3.54 (dd, J=10.0,2Hz, 0.5H), 3.39 (br s, 0.5H), 3.36 (br s, 0.5H), 2.94-2.56 (m, 4H), 1.47 (s, 9H), 1.03 (s, 1.5H), 0.99 (s, 1.5H).
Intermediate compound IV: amino N 2-[(tert-butoxycarbonyl)]-2-methyl-3-pyridin-4-yl propionic acid (route plan 1)
Figure A20058001184300611
Steps A: the formation of Schiff's base
(10.0g is 71.6mmol) at 100mL CH to the alanine methyl ester hydrochloride 2Cl 2In solution in add benzophenone imines (12.0mL, 71.6mmol).Make this reaction at room temperature carry out 15 hours, this solution engenders white precipitate.This reaction H 2O and CH 2Cl 2Dilution separates each layer, and Na is used in the salt water washing of this organic layer 2SO 4Drying is filtered and the concentrated viscous oil that obtains N-(phenylbenzene methylene radical) alanine methyl ester, and it need not to be further purified and can use. 1HNMR(CDCl 3,400MHz)δ7.62(m,2H),7.47-7.29(m,7.19-7.16(m,2H),4.16(q,J=6.8Hz,1H),3.7(s,3H,1.40(d,J=6.8Hz,3H)。
Step B: alkylation
(9.78g 36.6mmol) adds two (trimethyl silyl) ammonification sodium at THF (45.72mL, 45.72mmol) the 1M solution in down and in 20 minutes in 0 ℃ in the solution in 60mL DMF to the N-of steps A (phenylbenzene methylene radical) alanine methyl ester.In 30 minutes, in 20 minutes, add 4-picoline villaumite hydrochlorate (3.00g, 18.29mmol) solution in 40mL DMF through sleeve pipe to this reaction.Make this reaction rise to room temperature and stirred 5 hours.The saturated NH of this reaction 4The Cl aqueous solution is ended and is extracted with EtOAc (3x).The organic layer that merges is new with 3MLiCl (2x) and salt solution, uses Na 2SO 4Drying is filtered vacuum concentration.Obtain 2-[(phenylbenzene methylene radical by silica gel chromatography purifying (0-40% EtOAc/ hexane)) amino]-white solid (5.28g, 81%) of 2-methyl-3-pyridin-4-yl methyl propionate.LCMS[M+H]=359.2。 1HNMR (d 4-MeOH) δ 8.51 (dd, J=4.5,1.5Hz, 2H), 7.56 (dd, J=8.4,1.4Hz, 2H), 7.40-7.37 (m, 4H), 7.34-7.30 (m, 2H), 7.26-7.23 (m, 2H), 7.10-7.07 (m, 2H), 3.33 (A of AB, d, J=13.0Hz, 1H), 3.27 (s, 3H), 3.18 (B of AB, d, J=12.9Hz, 1H), 1.32 (s, 3H).
Step C: the removing of Schiff's base
To the 2-[(of step B phenylbenzene methylene radical) amino]-2-methyl-3-pyridin-4-yl methyl propionate (5.28g, 14.73mmol) at 75mL 1: adding 6NHCl in the suspension among the 1MeOH/THF (3.68mL, 22.10mmol).At room temperature stir after 1.5 hours and should react vacuum concentration.Utilize ion-exchange chromatogram purification (SCX box) to obtain 2-amino-2-methyl-3-pyridin-4-yl methyl propionate, it is yellow oil (2.76g, 97%).LCMS[M+H]=195.3。 1H NMR (d 4-MeOH) δ 8.43 (dd, J=4.6,1.6Hz, 2H), 7.24 (dd, J=4.6,1.5Hz, 2H), 3.70 (s, 3H), 3.09 (A of AB, d, J=12.9Hz, 1H), 2.90 (B of AB, d, J=13.0Hz, 1H), 1.39 (s, 3H).
Step D:Boc protection
To the 2-of step C amino-2-methyl-3-pyridin-4-yl methyl propionate (2.76g, 14.21mmol) in the suspension in 70mL THF in 0 ℃ add down tert-Butyl dicarbonate (4.03g, 18.47mmol).After 30 minutes, this reaction is risen to room temperature and makes it spend the night.This reaction is with the EtOAc dilution and use saturated NH 4The Cl aqueous solution, water and salt water washing.Organic layer Na 2SO 4Drying is filtered and vacuum concentration.At silica gel chromatography (0-60%EtOAc/CH 2Cl 2) purifying obtains the 2-[(tert-butoxycarbonyl) amino]-yellow solid (3.22g, 77%) of 2-methyl-3-pyridin-4-yl methyl propionate.LCMS[M+H]=295.2。 1H NMR (400MHz, d4-MeOH) δ 8.43 (d, J=5.1Hz, 2H), 7.21 (d, J=5.9Hz, 2H), 3.73 (s, 3H), 3.44 (A of AB, d, J=13.2Hz, 1H), 3.12 (B of AB, d, J=13.2Hz, 1H), 1.46 (s, 9H), 1.30 (s, 3H).
Step e: saponification
To the 2-[(of step D tert-butoxycarbonyl) amino]-2-methyl-3-pyridin-4-yl methyl propionate (0.25g, 0.85mmol) at 4.25mL 1: adding 3NNaOH in the solution among the 1MeOH/THF (0.43mL, 1.27mmol).This is reflected at carried out under 50 ℃ 1 hour, this moment with its be cooled to room temperature and with 6N HCl (0.21mL, 1.27mmol).To react vacuum concentration and generate the 2-[(tert-butoxycarbonyl) amino]-white solid of 2-methyl-3-pyridin-4-yl propionic acid NaCl.LCMS[M+H]=281.3。 1H NMR (400MHz, d4-MeOH) δ 8.44 (d, J=5.3Hz, 2H), 7.28 (d, J=5.9Hz, 2H), 3.43 (A of AB, d, J=12.6Hz, 1H), 3.33 (B of AB, d, J=12.3Hz, 1H), 1.47 (s, 9H), 1.41 (s, 3H).
Intermediate V:N-(tert-butoxycarbonyl)-2,5-dimethyl remove first leucine (route plan 1)
It is to be prepared according to the described method of preparation intermediate compound IV by iodate Trimethylmethane and L-Ala Schiff's base that N-(tert-butoxycarbonyl)-2,5-dimethyl remove the first leucine.
Intermediate A: N-benzyl-1-(2-trans-methyl cyclopropyl) methylamine
Figure A20058001184300632
Steps A: coupling
In the 2L flask with trans-nicotinic acid (15.0g, 174mmol), benzylamine (20.5g, 192mmol) and DIPEA (36.7g 192mmol) is dissolved in the 700mL methylene dichloride.Under the room temperature in this solution gradation add solid EDC-HCl (36.7g, 192mmol) and stir and spend the night.Reaction mixture is inclined to 10%KHSO 4(250mL) aqueous solution.Separate each layer and use 10%KHSO once more 4The washing 1 this.Organic layer is with H successively 2Na is used in O (200mL), salt solution (150mL) washing 2SO 4Dry and be concentrated into the dried crystallization that obtains white (2E)-N-benzyl but-2-enamides: 1HNMR (400MHz, CDCl 3) δ 7.28 (m, 5H), 6.85 (sext, J=6.8Hz, 1H), 5.78 (dd, J=15.2,1.6Hz, 2H), 4.47 (d, J=5.6Hz, 2H), 1.82 (dd, J=7.2,1.6Hz, 3H).
Step B: cyclisation
Containing Et under the room temperature 2Under vigorous stirring, in 5 minutes, drip in the Erlenmeyer flask of the O (300mL) and the 40% KOH aqueous solution (111mL) 1-methyl-3-nitro-1-nitrosoguanidine (11.1g, 67mmol).Stop stirring and water layer is freezing under-78 ℃ of baths behind reinforced the finishing.The ether layer is decanted among the Erlenmeyer that contains the KOH sheet.Content was placed 5 minutes., curve to the three respectively contain in the flask of KOH sheet and with hypsokinesis to the Et that contains (2E)-N-benzyl but-2-enamides (3.0g, 17.1mmol derive from steps A) 2On the O/THF solution (200mL/50mL).Add subsequently Pd (OAc) 2 (180mg, 0.9mmol) and make this reaction rise to room temperature and stirred 1 hour.Be blown into nitrogen 10 minutes to this reaction.This miscellany H 2O (150mL) washing.Separate organic layer and use Na subsequently 2SO 4Dry.Remove desolvate and by flash chromatography at SiO 2(EtOAc/ hexane) purifying obtains N-benzyl-trans-2-methyl cyclopropane carboxylic acid amides (83%): 1H NMR (400MHz, CDCl 3) δ 7.28 (m, 5H), 5.81 (br s, 1H), 4.43 (dd, J=5.6,2.4Hz, 2H), 1.37 (m, 1H), 1.17 (m, 1H), 1.07 (d, J=6.0Hz, 3H), 1.04 (overlapping m, 1H), 0.56 (m, 1H).
Randomly be prepared chirality HPLC obtain preferred enantiomer trans-S, S.In following intermediate and embodiment, no matter be preferred enantiomer trans-S, S or trans-S, S and trans-R, the racemic mixture of R need not to identify and can use.In order to simplify, this methyl-cyclopropyl-methyl moiety is plotted as trans-racemize.
Step C: reduction
In the 500mL flask, add the N-benzyl-trans-2-methyl cyclopropane carboxylic acid amides be contained among the THF (80mL) (derive from step B, 3.9g, 20.6mmol).Drip BH through dropping funnel 3-THF (1.0M, 105mL, 105mmol).Reinforced finishing back (10 minutes.), this miscellany was refluxed 5 hours.Make this miscellany be cooled to room temperature and carefully use MeOH (15mL) to end.This miscellany is concentrated into dried, is dissolved in methylene dichloride and washs with 3M KOH.Separate organic layer, use the salt water washing, use Na subsequently 2SO 4Dry and be concentrated into dried.Thick material was handled 1 hour under 50 ℃ in two  alkane with 1N HCl.This miscellany is concentrated the white solid that obtains hydrochloride.This solid is dissolved in saturated NaHCO 3The aqueous solution (80mL) is also used CHCl 3(2 * 150mL) extractions.Na is used in the organic layer salt water washing that merges 2SO 4Dry and remove through rotary evaporation and to desolvate, obtain the cream-coloured semisolid (quant.) of N-benzyl-1-(2-trans-methyl cyclopropyl) methylamine after the vacuum-drying: 1H NMR (400MHz, CDCl 3) δ 7.28 (m, 5H), 3.80 (s, 2H), 2.50 (d, J=6.8Hz, 2H), 2.4 (br s, 1H), 1.02 (d, J=6.0Hz, 3H), 0.69 (m, 1H), 0.52 (m, 1H), 0.23 (m, 2H).
Intermediate B: N-methyl isophthalic acid-(2-trans-methyl cyclopropyl) methylamine
Figure A20058001184300651
In N-benzyl-1-(2-trans-methyl cyclopropyl) solution of methylamine (8g, 45.6mmol, intermediate A) in DCE (240mL) and MeOH (120mL), add formaldehyde (34mL, 456mmol, 37% aqueous) and NaBH (OAc) 3(19.3g, 91mmol).Reaction mixture was at room temperature stirred 1 hour, use saturated NaHCO 3The aqueous solution is handled, and concentrates near doing, and dilute with water is also with EtOAc (3x) extraction.Dried over sodium sulfate is used in the organic layer salt water washing that merges, with HCl (50mL, 50mmol, 1M Et 2O) handle, and vacuum concentration obtains the hydrochloride of N-benzyl-methyl isophthalic acid-(2-trans-methyl cyclopropyl) methylamine, it is at 20%Pd (OH) 2Hydrogenation 2 hours under 60 ℃ and the 1atm H2 in EtOH (400mL) under the existence of/C (616mg).Filter and vacuum concentration obtains the hydrochloride of N-methyl isophthalic acid-(2-trans-methyl cyclopropyl) methylamine. 1HNMR(400MHz,CD 3OD)δ2.88(d,J=7.3Hz,2H),2.69(s,3H),1.09(d,J=5.7Hz,3H),0.78-0.70(m,2H),0.52-0.50(m,1H),0.50-0.40(m,1H)。
Intermediate C:2-{ benzyl [2-methyl cyclopropyl) methyl] amino }-6-[methyl (methyl sulphonyl) amino] Yi Yansuan (route plan 7)
Steps A: the introducing of sulphonamide
With 2; 6-dichloro-isonicotinic acid methyl esters (5.0g; 24.3mmol), methyl (methyl sulphonyl) amine (3.18g; 29.12mmol), potassiumphosphate (7.22g; 34.0mmol), Xantphos (0.87g; 1.50mmol) and three (dibenzylidene ketone), two palladiums (0.68g, 0.51mmol) join drying, be blown in the flask of argon gas.Add two  alkane (195mL), this solution is reflected at 100 ℃ of following heating 16 hours with argon-degassed and this.This reaction is cooled to room temperature, through diatomite filtration and vacuum-evaporation.Flash chromatography (silica gel, 0-50%EtOAc/CH 2Cl 2) obtain 2-chloro-6-[methyl (methyl sulphonyl) amino] iso methyl nicotinate, it is a yellow oil: 1H NMR (400MHz, CDCl 3) δ 7.88 (s, 1H), 7.68 (s, 1H), 3.96 (s, 3H), 3.44 (s, 3H), 3.11 (s, 3H).
Step B: hydrolysis
To 2-chloro-6-[methyl (methyl sulphonyl) amino] and iso methyl nicotinate (2g, 7.18mmol) 1: 1THF: adding NaOH in the solution among the MeOH (60mL) (9.3mL, 9.3mmol, 1N).Reaction mixture was at room temperature stirred 0.5 hour, is acidified to pH3-4,, obtain 2-chloro-6-[methyl (methyl sulphonyl) amino with dried over sodium sulfate and vacuum concentration with methylene dichloride (x2) extraction with 1N HCl] white solid of Yi Yansuan. 1H?NMR(400MHz,CD 3OD)δ7.88(s,1H),7.63(s,1H),3.39(s,3H),3.13(s,3H)。
Step C: amination
With 2-chloro-6-[methyl (methyl sulphonyl) amino] Yi Yansuan (1.2g; 4.53mmol), intermediate A (1.85g; 10.55mmol), potassiumphosphate (3.18g; 15.1mmol) and two (tri-butyl phosphine) palladium (0.13g, 0.25mmol) suspension in degassing DMA (10mL) is sealed in the Glass tubing and is heated to 130 ℃ and reaches 4 hours.Reaction mixture water and salt solution dilution are acidified to pH3-4 with 1N HCl, with dichloromethane extraction and vacuum concentration.This resistates is dissolved in EtOAc, with LiCl (x3) solution washing, at Na 2SO 4Last dry, vacuum concentration and by purification by flash chromatography (silica gel, 2-7% (1% AcOH is in MeOH)/CH 2Cl 2) obtain 2-{ benzyl [(2-methyl cyclopropyl) methyl] amino-6-[methyl (methyl sulphonyl) amino] oil of Yi Yansuan. 1HNMR (400MHz, CDCl 3) δ 7.35-7.15 (m, 5H), 7.06 (s, 1H), 7.03 (s, 1H), 4.59-4.28 (m, 2H), 3.55 (A of ABX, dd, J=14.8,5.6Hz, 1H), 3.29 (B of ABX, dd, J=14.8,7.2Hz, 1H), 3.28 (s, 3H), 2.87 (s, 3H), 0.97 (d, J=6.4Hz, 3H), 0.84-0.74 (m, 1H), 0.66-0.56 (m, 1H), and 0.40-0.31 (m, 1H), 0.29-0.21 (m, 1H).
Intermediate D:2-{ methyl [(2-methyl cyclopropyl) methyl] amino }-6-[methyl (methyl sulphonyl) amino] Yi Yansuan (route plan 7)
Figure A20058001184300671
2-{ methyl [(2-methyl cyclopropyl) methyl] amino }-6-[methyl (methyl sulphonyl) amino] Yi Yansuan is by 2-chloro-6-[methyl (methyl sulphonyl) amino] Yi Yansuan and N-methyl isophthalic acid-(2-trans-methyl cyclopropyl) methylamine utilization is similar to the method preparation described in the step C of intermediate C preparation. 1HNMR (400MHz, CD 3OD) δ 7.02 (s, 1H), 6.96 (s, 1H), the 3.55 (A of ABX, dd, J=14.4,6.0Hz, 1H), the 3.29 (B of ABX, dd, J=14.4,7.2Hz, 1H), 3.35 (s, 3H), 3.15 (s, 3H), 3.12 (s, 3H), 1.03 (d, J=6.0Hz, 3H), 0.82-0.66 (m, 2H), 0.48-0.41 (m, 1H), 0.28-0.22 (m, 1H).
Intermediate E: N-(4-(acetyl bromide)-6-{ methyl (2-methyl cyclopropyl) methyl } amino) pyridine-2-yl)-N-methyl Toluidrin (route plan 16)
Figure A20058001184300672
To intermediate D (250mg, 0.76mmol) being cooled in THF (3mL) drip in-78 ℃ the solution N-methylmorpholine (0.088mL, 0.8mmol) and tetryl formate (0.104mL, 0.8mmol), with ℃ down stirring 0.5 hour of this reaction mixture-78.Reaction mixture filters on diatomite, uses ether rinse.Filtrate and washings are cooled to-20 ℃ and with two azomethanes (3.5mL, Et 2O solution is by 150mL Et 2O, the 40mL 40% KOH aqueous solution and 13.2g 1-methyl-3-nitro-1-nitrosoguanidine are made) handle.Stirring is after 1.5 hours down at-20 ℃, and this reaction mixture rises to room temperature, adds 5mL two azomethane solution and this reaction mixture is at room temperature stirred 0.5 hour vacuum concentration (room temperature bath and HOAc are in trap).This resistates is dissolved in Et 2O and EtOAc, water, salt solution are new, with dried over sodium sulfate and vacuum concentration.This resistates is dissolved in Et 2O (5mL) is cooled to-20 ℃, handles with 62% HBr (0.25mL), stirs 0.3 hour down at-20 ℃, uses Et 2The O dilution, water, saturated NaHCO 3The aqueous solution, salt solution are new, obtain thick N-(4-(acetyl bromide)-6-(methyl [(2-methyl cyclopropyl) methyl] amino) pyridine-2-yl)-N-methyl Toluidrin with dried over sodium sulfate and vacuum concentration, and it is a yellow oil. 1H NMR (400MHz, CDCl 3) δ 6.96 (s, 1H), 6.80 (s, 1H), 4.40 (s, 2H), 3.52 (A of ABX, dd, J=14.8,6.4Hz, 1H), 3.36 (B of ABX, dd, J=14.8,6.8Hz, 1H), 3.39 (s, 3H), 3.12 (s, 3H), 3.11 (s, 3H), 1.03 (d, J=6.0Hz, 3H), 0.78-0.62 (m, 2H), and 0.44-0.38 (m, 1H), 0.31-0.26 (m, 1H).
Intermediate F:N-(4-(2-amino-1-hydroxyethyl)-6-{ methyl [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl Toluidrin (route plan 14)
Figure A20058001184300681
Steps A: reduction
(815mg, 2.49mmol) being cooled in THF (10mL) adds BH in 0 ℃ the solution to intermediate D 3-THF (7.5mL, 7.5mmol, 1M THF) and this reaction mixture at room temperature stirred 2.5 hours carefully ends with MeOH, ends with 1N HCl subsequently, with the EtOAc dilution, with 1N HCl, saturated NaHCO 3The aqueous solution, salt water washing, use dried over sodium sulfate, vacuum concentration and obtain the viscous crude of N-(4-(hydroxymethyl)-6-{ methyl [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl Toluidrin by purification by flash chromatography (silica gel, 25-70% EtOAc/ hexane). 1H NMR (400MHz, CDCl 3) δ 6.51 (s, 1H), 6.38 (s, 1H), 4.63 (d, J=6.0Hz, 2H), 3.50 (A of ABX, dd, J=14.4,6.4Hz, 1H), 3.33 (s, 3H), 3.31 (B of ABX, dd, J=14.4,6.8Hz, 1H), 3.10 (s, 3H), 3.07 (s, 3H), 1.84 (t, J=6.0Hz, 1H), 1.02 (d, J=5.6Hz, 3H), 0.77-0.60 (m, 2H), and 0.42-0.35 (m, 1H), 0.27-0.21 (m, 1H).
Step B: oxidation
To N-(4-(hydroxymethyl)-6-{ methyl [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl Toluidrin (480mg, 1.53mmol) add in the solution in methylene dichloride (10mL) and DMSO (2.5mL) triethylamine (1.07mL, 7.66mmol) and SO 3-pyridine (975mg, 6.13mmol).This reaction mixture was at room temperature stirred 0.5 hour, with the EtOAc dilution, with 1N HCl, saturated NaHCO 3Dried over sodium sulfate is used in the aqueous solution, salt water washing, vacuum concentration and obtain N-(4-formyl radical-6-{ methyl [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl Toluidrin by purification by flash chromatography (silica gel, 25-50% EtOAc/ hexane). 1HNMR (400MHz, CDCl 3) δ 9.94 (s, 1H), 6.92 (s, 1H), 6.72 (s, 1H), 3.53 (A of ABX, dd, J=14.8,6.4Hz, 1H), 3.40 (s, 3H), 3.36 (B of ABX, dd, J=14.8,6.8Hz, 1H), 3.15 (s, 3H), 3.13 (s, 3H), 1.04 (d, J=6.0Hz, 3H), 0.78-0.62 (m, 2H), and 0.45-0.37 (m, 1H), 0.31-0.24 (m, 1H).
Step C: the open loop of epoxidation reaction and epoxide
To N-(4-formyl radical-6-(methyl [(2-methyl cyclopropyl) methyl] amino) pyridine-2-yl)-N-methyl Toluidrin (435mg; 1.40mmol) add 5 in the solution in acetonitrile (5mL) and drip; trimethyl sulfonium iodide (342mg; 1.68mmol) and potassium hydroxide (188mg, 3.35mmol).This is reflected at 65 ℃ of lower seals stirred 3 hours,, use saturated NaHCO with the EtOAc dilution 3The aqueous solution, salt water washing use dried over sodium sulfate and vacuum concentration to obtain N-methyl-N-(6-{ methyl [(2-methyl cyclopropyl) methyl] amino }-4-oxyethane-2-yl pyridines-2-yl) Toluidrin, are used for the step down.(440mg is 1.35mmol) at EtOH (10mL) and NH for Toluidrin with N-methyl-N (6-{ methyl [(2-methyl cyclopropyl) methyl] amino }-4-oxyethane-2-yl pyridines-2-yl) 4Solution among the OH (15mL) stirred 16 hours at 60 ℃ of lower seals, vacuum concentration and by purification by flash chromatography (silica gel, 5-15% (10%NH 4OH/MeOH)/CH 2Cl 2) obtain the viscous crude of N-(4-(2-amino-1-hydroxyethyl)-6-{ methyl [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl Toluidrin. 1H NMR (400MHz, CD 3OD) δ 6.52 (s, 1H), 6.47 (s, 1H), the 4.58 (X ' of A ' B ' X ', dd, J=7.6,4.0Hz, 2H), the 3.54 (A of ABX, dd, J=14.4,6.4Hz, 1H), the 3.35 (B of ABX, dd, J=14.4,6.8Hz, 1H), 3.30 (s, 3H), 3.13 (s, 3H), 3.08 (s, 3H), 3.86 (A ' of A ' B ' X ', dd, J=13.2,4.0Hz, 1H), 2.76 (B ' of A ' B ' X ', dd, J=13.2,7.6Hz, 1H), 1.02 (d, J=6.0Hz, 3H), 0.80-0.65 (m, 2H), 0.46-0.39 (m, 1H), 0.25-0.18 (m, 1H).
Intermediate G:N-(tert-butoxycarbonyl)-N-(2-oxo-2-{2-{ methyl [(2-methyl cyclopropyl) methyl] amino }-6-{ methyl (methyl sulphonyl) amino] pyridin-4-yl }-α-D-aminomethyl phenyl alanimamides (route plan 14)
Figure A20058001184300701
Steps A: coupling
With intermediate F (330mg, 096mmol), N-Boc-D-Alpha-Methyl phenylalanine (323mg, 1.16mmol), EDC (240mg, 1.25mmol), HOAt (157mg, 1.16mmol) and diisopropylethylamine (0.34mL, 1.93mmol) solution in DMF (9mL) at room temperature stirred 16 hours.Reaction mixture dilutes with EtOAc; water and salt water washing; use dried over sodium sulfate; vacuum concentration and by purification by flash chromatography (silica gel; 30-65% EtOAc/ hexane) obtain N-(tert-butoxycarbonyl)-N-(2-hydroxyl-2-{2-{ methyl [(2-methyl cyclopropyl) methyl] amino }-6-[methyl (methyl sulphonyl) amino] pyridin-4-yl) ethyl)-the light yellow foam of D-Alpha-Methyl phenyl alanimamides, it directly carries out oxidation step.
Step B: oxidation
To N-(tert-butoxycarbonyl)-N-(2-hydroxyl-2-{2-{ methyl [(2-methyl cyclopropyl) methyl] amino }-6-[methyl (methyl sulphonyl) amino] the pyridin-4-yl ethyl)-D-Alpha-Methyl phenyl alanimamides (458mg; 0.76mmol) add triethylamine (0.53mL in the solution in DCM (6mL) and DMSO (1.5mL); 3.79mmol) and sulphur trioxide-pyridine (483mg, 3.03mmol).Reaction mixture at room temperature stirred 1.5 hours, with the EtOAc dilution, used 10% KHSO 4, saturated NaHCO 3With the salt water washing, use dried over sodium sulfate, vacuum concentration and obtain N-(tert-butoxycarbonyl)-N-(2-oxo-2-{2-{ methyl [(2-5 by purification by flash chromatography (silica gel, 0-50% EtOAc/ hexane)
The methyl cyclopropyl) methyl] amino-6-[methyl (methyl sulphonyl) amino] pyridin-4-yl } ethyl)-orange oil of D-Alpha-Methyl phenyl alanimamides. 1H NMR (400MHz, CDCl 3) δ 7.38-7.10 (m, 5H), 6.91 (s, 1H), 6.75 (s, 1H), 4.82 (br s, 1H), 4.78-4.62 (m, 2H), 3.53 (A of ABX, dd, J=14.7,6.7Hz, 1H), 3.43 (A of AB, d, J=14.4Hz, 1H), 3.38 (s, 3H), 3.36 (B of ABX, dd, J=14.7,6.6Hz, 1H), 3.16 (s, 3H), 3.15 (B of AB, d, J=14.4Hz, 1H), 3.13 (s, 3H), 1.49 (s, 9H), 1.46 (s, 3H), 1.04 (d, J=6.0Hz, 3H), 0.80-0.62 (m, 2H), 0.46-0.38 (m, 1H), 0.31-0.24 (m, 1H).
Intermediate H:[1R-benzyl-1-methyl-3-(the 2-{ methyl [(2-methyl cyclopropyl) methylamino }-6-[methyl (methyl sulphonyl) amino] different nicotinoyl amino)-the 2-oxopropyl] t-butyl carbamate (route plan 17)
[1R-benzyl-1-methyl-3-; ({ 2-; (methyl [; (2-methyl cyclopropyl) methyl] amino }-the 6-[methyl; (methyl sulphonyl) amino] different nicotinoyl amino)-the 2-oxopropyl] t-butyl carbamate is by intermediate III and intermediate D) coupling and utilize tert-butoxycarbonyl subsequently with preparation N-)-N-; (2-oxo-2-{2-{ methyl [; (2-methyl cyclopropyl) methyl] amino }-the 6-[methyl; (methyl sulphonyl) amino] pyridin-4-yl) ethyl)-D-Alpha-Methyl phenyl alanimamides; the hydroxyl oxidize method that (intermediate G) is identical. 1H NMR (400MHz, CDCl 3) δ 7.36-7.22 (m, 3H), 7.13-7.08 (m, 2H), 6.88 (br s, 1H), 6.79 (s, 1H), 6.72 (s, 1H), 4.74 (br s, 1H), 4.68-4.58 (m, 1H), 4.44-4.34 (m, 1H), 3.53 (A of ABX, dd, J=14.0,6.0Hz, 1H), 3.38 (s, 3H), 3.40-3.30 (A of AB and the B of ABX, m, 2H), 3.14 (s, 3H), 3.11 (s, 3H), 3.05 (B of AB, d, J=13.6Hz, 1H), 1.49 (s, 9H), 1.32 (s, 3H), 1.04 (d, J=6.0Hz, 3H), 0.78-0.62 (m, 2H), 0.44-0.36 (m, 1H), 0.29-0.22 (m, 1H).
Intermediate J:2-{ methyl [(2-methyl cyclopropyl) methyl l amino]-6-[methyl (methyl sulphonyl) amino] pyridine-4-carboxylic imidic acid ethyl ester (route plan 7,11)
Steps A: the preparation of nitrile (route plan 7)
With intermediate D (250mg, 0.76mmol), ammonium chloride (204mg, 3.82mmol), EDC (176mg, 0.92mmol), HOAt (104mg, 0.76mmol) and diisopropylethylamine (0.67mL, 3.82mmol) suspension in DMF (10mL) at room temperature stirred 16 hours.Reaction mixture dilutes with EtOAc, water, 10%KHSO 4, saturated NaHCO 3The aqueous solution, LiCl solution washing are with dried over sodium sulfate and vacuum concentration.This resistates is dissolved in THF (5mL), with Burgess reagent (219mg, 0.92mmol) handle and 80 ℃ of following microwaves (Smith synthesizer) irradiation 5 minutes, vacuum concentration and obtain N-(4-cyano group-6-{ methyl [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl Toluidrin by purification by flash chromatography (silica gel, 0-30% EtOAc/ hexane). 1H NMR (400MHz, CDCl 3) δ 6.73 (s, 1H), 6.48 (s, 1H), the 3.47 (A of ABX, dd, J=14.8,6.4Hz, 1H), 3.37 (s, 3H), 3.33 (B of ABX, dd, J=14.8,6.8Hz, 1H), 3.13 (s, 3H), 3.09 (s, 3H), 1.04 (d, J=5.6Hz, 3H), 0.78-0.62 (m, 2H), 0.45-0.37 (m, 1H), 0.32-0.25 (m, 1H).
Step B: the preparation of inferior amine salt
(180mg, 0.58mmol) being cooled in EtOH (10mL) is blown into HCL (g) in 0 ℃ the solution and reaches 10 minutes to N-(4-cyano group-6-{ methyl [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl Toluidrin.With the reaction mixture sealing, rise to room temperature and stirring 3 hours.Nitrogen is blown into this reaction mixture 10 minutes and with this reaction mixture vacuum concentration.Obtain 2-{ methyl [(2-methyl cyclopropyl) methyl] amino with ether development }-6-[methyl (methyl sulphonyl) amino] hydrochloride and the glassy yellow solid of pyridine-4-azomethine acetoacetic ester. 1H NMR (400MHz, CD 3OD) δ 6.96 (s, 1H), 6.88 (s, 1H), 4.63 (q, J=7.2Hz, 2H), 3.60 (A of ABX, dd, J=14.8,6.4Hz, 1H), 3.44 (B of ABX, dd, J=14.8,6.8Hz, 1H), 3.38 (s, 3H), 3.18 (s, 3H), 3.16 (s, 3H), 1.61 (t, J=7.2Hz, 3H), 1.03 (d, J=6.0Hz, 3H), 0.84-0.68 (m, 2H), and 0.50-0.43 (m, 1H), 0.29-0.23 (m, 1H).
Intermediate K:(2,2-two fluoro ethyls) [(2-methyl cyclopropyl) methyl] amine
Figure A20058001184300731
Steps A: coupling
In the 500-mL flask with difluoroacetic acid (0.78g, 8.2mmol), N-benzyl-1-(2-trans-methyl cyclopropyl) methylamine (1.3g, 7.42mmol) and DIPEA (4.2g 32.6mmol) is dissolved in the methylene dichloride of 100mL.Gradation at room temperature adds BOP (3.3g, solid 7.42mmol) and stirring 1 hour in this solution.Should react vacuum concentration and pass through the acid amides that flash column chromatography purifying (silica gel, 0-10% ethyl acetate/hexane) obtains 1.45g (77%), it is a clarified oil: 1HNMR (400MHz, CDCl 3) δ 7.31 (m, 5H), 6.19 (td, J=51,20Hz, 1H), 4.75 (m, 2H), 3.24 (m, 2H), 0.99 (t, J=5.5Hz, 3H), 0.62 (m, 2H), 0.31 (m, 2H); LCMS[M+H] +=254.1.
Step B: reduction
In round-bottomed flask with N-benzyl-2,2-two fluoro-N-{[-2-methyl cyclopropyl methyl]-methyl (1.45g 5.7mmol) is dissolved in the anhydrous THF of 75mL to ethanamide.In this solution, add BH 3-THF (18.9mmol, the 1M THF solution of 18.9mL).Should react according to reflux exchanger and reflux 16 hours.Make this reaction be cooled to 0 ℃ and by adding methyl alcohol, adding dense HCl (5mL) subsequently and end.The gained mixture at room temperature stirred 16 hours.With crude mixture vacuum concentration and between 10% NaOH/ ethyl acetate, distributing subsequently.The organism dried over sodium sulfate is filtered and vacuum-evaporation.Flash column chromatography purifying (silica gel, 0-10% ethyl acetate/hexane) obtains the amine of 1.25g (91%), and it is a clarified oil: 1H NMR (400MHz, CDCl 3) δ 7.31 (m, 5H), 5.75 (tt, 56.5,4.5Hz, 1H), 3.78 (d, J=13.7Hz, 1H), 3.73 (d, J=13.7Hz, 1H), 2.92 (td, J=15,3.8Hz, 2H), 2.56 (dd, J=13.2,6.0Hz, 1H), 2.37 (dd, J=13.2Hz, 7.0Hz, 1H), 1.03 (d, J=6.0Hz, 3H), 0.54 (m, 2H), 0.25 (t, J=6.2Hz, 2H); LCMS[M+H] +=240.2.
Step C: hydrogenation
With N-benzyl-2,2-two fluoro-N-[(2-methyl cyclopropyl) methyl] ethamine (1.25g, 5.2mmol) and the solution of hydrochloric acid (5.7mmol, the 4M two  alkane solution of 1.44mL) in ethanol (50mL) handle with the nitrogen degassing and with palladium hydroxide (125mg).This reaction placed under the nitrogen atmosphere and vigorous stirring 1 hour.This reaction obtains the light yellow oil of (2,2-two fluoro ethyls) [(2-methyl cyclopropyl) methyl] amine through diatomite filtration with methanol wash and vacuum concentration: 1H NMR (400MHz, CD 3OD) δ 6.35 (tt, 48,3Hz, 1H), 3.56 (td, J=15.6,3.1Hz, 2H), 3.03 (m, 2H), 1.11 (d, J=5.9Hz, 3H), 0.83 (m, 2H), 0.60 (m, 1H), 0.50 (m, 1H).
Intermediate L:N, N-dimethyl-N-[(2-methyl cyclopropyl) methyl] the third-1, the 3-diamines
Figure A20058001184300741
Steps A: coupling
The 100-mL flask with vinylformic acid (0.17g, 2.4mmol), N-benzyl-1-(2-trans-methyl cyclopropyl) methylamine (0.5g, 2.4mmol) and DIPEA (0.64g 4.9mmol) is dissolved in the 20mL methylene dichloride.(0.68g, solid 3.5mmol) also stirred 15 hours at room temperature to drip EDC in this solution.Making this be reflected between 1M HCl and the methylene dichloride distributes.The organism dried over sodium sulfate is filtered, vacuum concentration and the crude product that carries out the next step.
Step B:Michael addition
In the 50-mL flask, acrylamide (crude product of steps A) is handled with dimethyl amine (4.5mmol, the 2M methanol solution of 2.2mL) at methyl alcohol (10mL).Should react and at room temperature stir 1 hour, subsequently vacuum concentration.This resistates obtains the 0.47g N in (75% liang of step) by flash column chromatography purifying (2.5-15%MeOH/ methylene dichloride) 1-benzyl-N 3, N 3-dimethyl-N 1The clarified oil of-[(2-methyl cyclopropyl)-methyl]-β-alanimamides: 1H NMR (400MHz, CDCl 3) δ 7.24 (m, 5H), 4.68 (m, 2H), 3.21 (m, 2H), 2.67 (m, 4H), 2.34 (s, 3H), 2.24 (s, 3H), 0.97 (m, 3H), 0.58 (m, 2H), 0.29m, 2H).LCMS[M+H] +=275.4。
Step C: reduction
At round-bottomed flask with N 1-benzyl-N 3, N 3-dimethyl-N 1-(0.47mg 1.7mmol) is dissolved among the anhydrous THF of 10mL [(2-methyl cyclopropyl)-methyl]-β-alanimamides.Add BH to this solution 3-THF (5.1mmol, the 1M THF solution of 5.1mL).This reaction was installed back flow condenser and reflux 16 hours.Make this reaction be cooled to 0 ℃ and with methyl alcohol, use dense HCl (5mL) to end subsequently.The gained mixture heating up was refluxed 16 hours.With crude mixture vacuum concentration and between 10% NaOH/ ethyl acetate, distributing after this.This organism dried over sodium sulfate is filtered and vacuum-evaporation.Obtain the N of 0.42g (49%) by the reverse-phase chromatography purifying 1-benzyl-N 3, N 3-dimethyl-N 1-[(2-methyl cyclopropyl)-methyl] the third-1, the clarified oil of 3-two ammoniums two (trifluoroacetate): LCMS[M+H] +=261.5.
Step D: hydrogenation
With N 1-benzyl-N 3, N 3-dimethyl-N 1-[(2-methyl cyclopropyl)-methyl] the third-1,3-two ammoniums two (trifluoroacetate) (0.42g, 0.86mmol) handle with the nitrogen degassing with palladium hydroxide (75mg) by the solution in ethanol (50mL).This reaction placed under the nitrogen atmosphere and vigorous stirring 1 hour.This reaction obtains N with methanol wash and vacuum concentration, N-dimethyl-N-[(2-methyl cyclopropyl through diatomite filtration) methyl] the third-1, the light yellow oil of 3-two ammoniums two (trifluoroacetate).
Intermediate M:(2-fluoro ethyl) [(2-methyl cyclopropyl) methyl] amine
Figure A20058001184300751
Be similar to the described method preparation of intermediate K by gifblaar poison, utilization. 1HNMR(400MHz,CDCl 3)δ9.79(bs,1H),4.98(bd,J=46.3,2H),3.42(m,2H),3.03(bs,2H),1.12(bs,3H),1.01(bs,1H),0.87(bs,1H),0.66(bs,1H),0.50(bs,1H)。
Intermediate N:(2-methoxy ethyl) [(2-methyl cyclopropyl) methyl] amine
Figure A20058001184300752
Be similar to the described method preparation of intermediate K by the methoxyacetic acid utilization. 1HNMR(400MHz,CDCl 3)δ7.98(bs,1H),3.64(bs,2H),3.38(bs,3H),3.27(bs,2H),3.00(bs,2H),1.06(d,J=5.3Hz,3H),0.79(bs,2H),0.51(m,2H)。
Intermediate O:N, N-dimethyl-N-[(2-methyl cyclopropyl) methyl] second-1, the 2-diamines
By N, the N-N-methylsarcosine adopts and is similar to the described method preparation of intermediate K. 1HNMR(400MHz,CD 3OD)δ3.49(m,4H),2.98(m,2H),2.96(s,6H),1.09(d,J=5.8Hz,3H),0.83(m,2H),0.61(m,1H),0.49(m,1H)。
Intermediate P:{ (1S)-[(1S, 2S)-2-methyl cyclopropyl] ethyl } amine
Steps A. (2E)-1,1-diethoxy but-2-ene
With crotonaldehyde (23.64mL, 285.35mmol), triethyl orthoformate (57.02mL, 342.42mmol) and ammonium nitrate (2.28g 28.54mmol) mixes in 60mL EtOH.After following 22 hours of the room temperature, should react with EtOAc dilution (60mL) and with saturated sodium bicarbonate solution (40mL) and wash.Water layer is used EtOAc (20mL) extraction once more.The organism that merges is used Na with salt water washing (40mL) 2SO 4Drying is filtered and vacuum concentration obtains 1 of 36.5g (89%), 1-diethoxy but-2-ene. 1H?NMR(CDCl 3,400MHz)5.84(m,1H);5.54(m,1H);4.82(d,J=5.7Hz,1H);3.64(m,2H);3.49(m,2H);1.73(m,3H);1.21(m,6H)。
Step B. (4S, 5S)-2-[(lE)-third-1-thiazolinyl]-1,3-dioxolane-4,5-dicarboxylic acid diisopropyl ester
With (2E)-1,1-diethoxy but-2-ene (32.20g, 223.27mmol), D-tartrate (-)-diisopropyl ester (64.64mL, 245.60mmol) and toluenesulphonic acids pyridine  (2.24g, 8.93mmol) solution in the 100mL phenyl ester is heated to 95 ℃, distillation removes desolvates and generates EtOH.95 ℃ after following 7 hours, this reaction is cooled to room temperature and vacuum concentration.By normal-phase chromatography purifying (10->30% EtOAc/ hexane) obtain 35.37g (55%) (4S, 5S)-2-[(1E)-third-1-thiazolinyl]-1,3-dioxolane-4, the orange oil of 5-dicarboxylic acid diisopropyl ester. 1H?NMR(CDCl 3,400MHz)6.03(m,1H);5.86(m,2H);5.12(m,2H);4.71(d,J=3.84Hz,1H);4.63(d,J=3.84Hz,1H);1.78(m,3H);1.30(d,J=6.23Hz,12H);LC/MS[M+H] +=287。
Step C. (4S, 5S)-2-[(1S, 2S)-2-methyl cyclopropyl]-1,3-dioxolane-4,5-dicarboxylic acid diisopropyl ester
To intermediate (4S, 5S)-2-[(1E)-third-1-thiazolinyl]-1,3-dioxolane-4, the 5-diisopropyl ester (4.10g, 14.32mmol) add in 20 ℃ of solution in the 60mL hexane 1M be present in zinc ethyl in the hexane (42.96mL, 42.96mmol).Drip methylene iodide (6.92mL, 85.92mmol) and vigorous stirring.-20 ℃ after following 1 hour, this is reflected at-5 ℃ freezing down.-5 ℃ after following 17 hours, this is reflected at 0 ℃ continues down to stir 5 hours and used cold saturated ammonium chloride solution (100mL) to end and use Et subsequently 2O (100mL * 3) extraction.The organism that merges filters with sodium thiosulfate solution (100mL) and salt solution (100mL) washing, uses Na 2SO 4Drying is filtered and vacuum concentration once more.By normal-phase chromatography purifying (10->30%EtOAc/ hexane) obtain 3.85g (89%) (4S, 5S)-2-[(1S, 2S)-2-methyl cyclopropyl]-1,3-dioxolane-4,5-dicarboxylic acid diisopropyl ester, it is a yellow oil. 1H?NMR(CDCl 3,400MHz)5.12(m,2H);4.78(d,J=6.41Hz,1H);4.66(d,J=4.21Hz,1H);4.57(d,J=4.22Hz,1H);1.30(m,12H);1.09(d,J=5.68Hz,3H);0.94(m,2H);0.67(m,1H);0.39(m,1H);LC/MS[M+H] +=301。
Step D.2-methyl-N-{ (1E)-[(1S, 2S)-2-methyl cyclopropyl] methylene radical) third-2-sulfenimide
To (4S, 5S)-2-[(1S, 2S)-2-methyl cyclopropyl l-1,3-dioxolane-4, (0.450g is 1.50mmol) at 5mL CH for 5-dicarboxylic acid diisopropyl ester 2Cl 2/ 200 μ L H 2Adding tosic acid in the solution among the O (0.071g, 0.38mmol).This is reflected at 50 ℃ of following reflux., 50 ℃ after following 16 hours, this reaction is cooled to room temperature.Remove the water droplet that generates at this reaction top.Add cuprous sulfate (II) (0.507g, 2.85mmol) and R-(+)-tertiary butyl thionyl amines (0.173g, 1.43mmol).After following 5.5 hours of the room temperature, this reaction is filtered through Celite pad.This diatomite CH 2Cl 2(200mL) washing and this filtrate vacuum concentration.By normal-phase chromatography purifying (0->50%EtOAc/ hexane) generate 2-methyl-N-{ (1E) of 0.245g (92%)-[(1S, 2S)-2-methyl cyclopropyl] methylene radical) the colourless resistates of clarification of third-2-thionyl amines. 1H?NMR(CDCl 3,400MHz)7.46(d,J=7.69Hz,1H);1.62(m,1H);1.25(m,2H);1.10(m,12H);0.82(m,1H);LC/MS[M+H] +=188。
Step e .2-methyl-N-{ (1S)-1-[(1S, 2S)-2-methyl cyclopropyl] ethyl } third-2-thionyl amines
To 2-methyl-N-{ (1E)-[(1S, 2S)-2-methyl cyclopropyl] methylene radical (0.300g is 1.60mmol) at 5mL CH for third-2-thionyl amines 2Cl 2In-78 ℃ of solution in add 3M and be present in Et 2Methyl-magnesium-bromide among the O (1.07mL, 3.20mmol).-78 ℃ after 2 hours, this reaction rises to room temperature.After following 1 hour of the room temperature, this reaction is ended with saturated ammonium chloride solution (15mL) and is extracted with EtOAc (30mL * 2).The organism that merges is used Na with salt water washing (15mL) 2SO 4Drying is filtered and vacuum concentration.Generate methyl-N-{ (1S)-1-[(1S of 0.224g (69%) by normal-phase chromatography purifying (0->80% EtOAc/ hexane), 2S)-2-methyl cyclopropyl] ethyl } the colourless resistates of clarification of third-2-thionyl amines. 1H?NMR(CDCl 3,400MHz)2.77(m,1H);1.31(d,J=6.50Hz,3H);1.21(s,9H);1.03(d,J=5.77Hz,3H);0.54(m,3H);0.30(m,1H);LC/MS[M+H] +=204。
Step F. (1S)-1-[(1S, 2S)-2-methyl cyclopropyl] the ethane chlorination ammonium
To 2-methyl-N-{ (1S)-1-[(1S, 2S)-2-methyl cyclopropyl] ethyl } (0.210g 1.03mmol) adds 2M and is contained in Et third-2-thionyl amines in 0 ℃ of solution in 4mL MeOH 2HCl among the O (0.52mL, 1.03mmol).This is reflected at 0 ℃ stirred 18 hours down and vacuum concentration subsequently.The gained material is dissolved in Et 2O (4mL) and vacuum concentration obtain (1S)-1-[(1S 2 times, 2S)-2-methyl cyclopropyl] white solid of ethane chlorination ammonium. 1H?NMR(CDCl 3,400MHz)2.60(m,1H);1.37(d,J=6.59Hz,3H);1.08(d,J=6.04Hz,3H);0.77(m,1H);0.64(m,2H);0.42(m,1H);LC/MS[M+H] +=100。
Intermediate Q:{ (1R)-1-[(1S, 2S)-2-methyl cyclopropyl] ethyl } amine
Figure A20058001184300781
S-(+)-uncle's butane thionyl imide that adopts the described method of intermediate P and improve among the step D prepares.LC/MS[M+H] +=100。
Intermediate R:{ (1S)-1-[(1S, 2S)-2-methyl cyclopropyl] propyl group } amine
Adopt the described scheme of intermediate P, and improve the used ethylmagnesium bromide of step e.LC/MS[M+H] +=114。
Intermediate S:(1S)-2-methyl isophthalic acid-((1S, 25)-2-methyl cyclopropyl) third-1-amine
By the described scheme of intermediate P, and the used sec.-propyl bromination magnesium of improvement step e prepares.LC/MS[M+H] +=128。
The intermediate T:(2-tertiary butyl (dimethyl) silyl) oxygen base } (2-methyl cyclopropyl)-methyl] amine
Figure A20058001184300793
Be similar to the described method preparation of intermediate B by (tertiary butyl (dimethyl) siloxy-) ethamine, utilization. 1H?NMR(400MHz,CD 3OD)δ3.74(t,J=5.3Hz,2H),2.76(m,2H),2.52(m,2H),1.04(d,J=6.0Hz,3H),0.90(s,9H),0.68(m,1H),0.55(m,1H),0.29(m,1H),0.24(m,1H),0.07(s,6H)。
Intermediate a:2-{ (2,2-two fluoro ethyls) [(2-methyl cyclopropyl) methyl] amino)-and 6-[methyl (methyl sulphonyl) amino] Yi Yansuan
Figure A20058001184300801
Be similar to the described method preparation of intermediate C by intermediate K utilization.LCMS[M+H] +=378.3。
Intermediate b:2-{ (2-methoxy ethyl) [(2-methyl cyclopropyl) methyl] amino }-6-[methyl (methyl sulphonyl) amino] Yi Yansuan
Figure A20058001184300802
Be similar to the described method preparation of intermediate C by intermediate N utilization.LCMS[M+H] +=372.3。
Intermediate c:2-{ (2,2-two fluoro ethyls) [(2-methyl cyclopropyl) methyl] amino }-6-[(sec.-propyl alkylsulfonyl) (methyl) amino] Yi Yansuan
Be similar to the described method preparation of intermediate C by intermediate K and (sec.-propyl alkylsulfonyl) methylamine utilization. 1H?NMR(400MHz,CDCl 3)δ7.09(s,1H),7.01(s,1H),6.08(td,J=56.4,4.2Hz,1H),3.93(m,2H),3.46(m,1H),3.44(s,3H),3.32(dd,J=15.0,6.6Hz,1H),1.41(d,J=6.8Hz,6H),1.06(d,J=5.7Hz,3H),0.74(m,2H),0.45(m,1H),0.35(m,1H)。LCMS[M+H] +=406.5。
Intermediate d:2-{ (2-fluoro ethyl) [(2-methyl cyclopropyl) methyl] amino }-6-[(sec.-propyl alkylsulfonyl) (methyl) amino] Yi Yansuan
Figure A20058001184300811
Be similar to the method preparation of intermediate C by intermediate M and (sec.-propyl alkylsulfonyl) methylamine utilization.LCMS[M+H] +=388.0。
Intermediate: (methyl) amino 2-[(sec.-propyl alkylsulfonyl)]-6-{ (2-methoxy ethyl) [(2-methyl cyclopropyl) methyl] amino) Yi Yansuan
Figure A20058001184300812
Be similar to the described method preparation of intermediate C by intermediate N and (sec.-propyl alkylsulfonyl) methylamine utilization. 1H?NMR(400MHz,CDCl 3)δ7.01(s,1H),6.98(s,1H),3.99(m,1H),3.75(m,1H),3.59(m,1H),3.47(m,1H),3.44(s,3H),3.36(s,3H),3.34(m,1H),1.40(d,J=3.3Hz,6H),1.05(d,J=5.8Hz,3H),0.74(m,2H),0.43(m,1H),0.29(m,1H)。LCMS[M+H] +=400.5。
Intermediate f:2-{[2-(dimethylamino) ethyl] [(2-methyl cyclopropyl) methyl] amino }-6-[(sec.-propyl alkylsulfonyl) (methyl) amino] Yi Yansuan
Being similar to the described mode of intermediate C by intermediate O and (sec.-propyl alkylsulfonyl) methylamine utilization prepares. 1H?NMR(400MHz,CDCl 3)δ6.95(s,1H),6.85(s,1H),4.00(m,2H),3.88(m,1H),3.44(m,2H),3.43(s,3H),3.34(d,J=5.9Hz,2H),2.95(bs,6H),1.38(d,J=7.0Hz,6H),1.03(d,J=5.7Hz,3H),0.70(m,2H),0.45(m,1H),0.31(m,1H)。LCMS[M+H] +=413.3。
Intermediate g:2-{[3-(dimethylamino) propyl group] [(2-methyl cyclopropyl) methyl] amino }-6-[(sec.-propyl alkylsulfonyl) (methyl) amino] Yi Yansuan
Figure A20058001184300822
Be similar to the described method preparation of preparation intermediate C by intermediate L and (sec.-propyl alkylsulfonyl) methylamine utilization.LCMS[M+H] +=427.4。
Intermediate h:[1-(5-{2-chloro-6-[methyl (methyl sulphonyl) amino] pyridin-4-yl }-1,3,4- diazole-2-yl)-1R-methyl-2-phenylethyl] t-butyl carbamate (route plan 8)
Figure A20058001184300831
Steps A. coupling
To 2-chloro-6-[methyl (methyl sulphonyl) amino) Yi Yansuan (0.200g, 0.76mmol) add in the solution in 3mL DMF triethylamine (0.13mL, 0.91mmol); intermediate compound I (0.222g, 0.76mmol), HOAt (0.123g; 0.91mmol) and EDC (0.174g, 0.91mmol).After following 17 hours of the room temperature, this reaction with saturated sodium bicarbonate solution (30mL) and salt solution (30mL) washing, is used Na with EtOAc dilution (60mL) 2SO 4Drying is filtered and vacuum concentration.Obtain the colourless resistates of clarification of the acyl group hydrazides of 0.544g (99%) by normal-phase chromatography purifying (20->80% EtOAc/ hexane).NMR(CDCl 3,400MHz)7.71(s,1H);7.53(s,1H);7.29(m,3H);7.15(d,J=6.59Hz,2H);3.50(d,J=13.55Hz,1H);3.42(s,3H);3.11(s,3H);3.05(d,J=13.73Hz,1H);1.47(s,9H);1.43(s,3H);LC/MS[M-C4H7]+=484。
Step B. cyclodehydration
To the acyl group hydrazides of steps A (0.550g, 1.02mmol) add in the solution in 5mL DCE Burgess reagent (0.971g, 4.07mmol).This be reflected at 120 ℃ of microwave treatment 10 minutes and directly by normal-phase chromatography purifying (0->50% EtOAc/ hexane) obtain (1R)-1-(5-{2-chloro-6-[methyl (methyl sulphonyl) amino] pyridin-4-yl-1; 3; 4- diazole-2-yl)-and 1-methyl-2-phenylethyl t-butyl carbamate, it is a white foam.NMR(CDCl 3,400MHz)7.89(s,1H);7.72(s,1H);7.29(m,3H);7.08(m,2H);3.60(d,J=13.55Hz,1H);3.46(s,3H);3.38(d,J=13.55Hz,1H);3.15(s,3H);3.15(s,3H);1.70(s,3H);1.42(s,9H);[M+H] +=522。
Intermediate i.{1-[5-(2-chloro-6-{ methyl (2-methyl cyclopropyl) methyl } base of aminopyridine-4-))-1,3,4- diazole-2-yl]-1R-methyl-2-phenylethyl } t-butyl carbamate
Figure A20058001184300841
Steps A: nucleophilicity amination
With 2, the 6-dichloro-isonicotinic acid (0.25g, 1.3mmol), methyl-[(methyl cyclopropyl) methyl] salt acid amide (0.53g, 3.9mmol) and cesium carbonate (1.25g, 6.5mmol) in the solution in 5mL DMF in 120 ℃ of lower seal pipes the heating 72 hours.This is reflected between 1M HCl and the ethyl acetate and distributes.Dried over sodium sulfate is used in this organism water (4x), salt water washing, filters and vacuum-evaporation obtains thick 2-chloro-6-{ methyl [(the 2-methyl cyclopropyl) methyl] amino of 0.3g (90%) } the nucleic acid oil of Yi Yansuan: LCMS (M+H)=255.0.
Step B: coupling
2-chloro-6-{ methyl [(2-methyl cyclopropyl) methyl] amino } Yi Yansuan (0.33g, 1.3mmol), N-(tert-butoxycarbonyl)-D-Alpha-Methyl phenylalanine hydrazi acid amides (alaninhydrazamid) (0.38g, 1.3mmol) and diisopropylethylamine (0.17g, 1.3mmol) (0.57g 1.3mmol) handles with BOP for solution in the 10mL methylene dichloride.Stir under the room temperature should react after 1 hour with additional hydrazi acid amides (0.19g, 0.65mmol) and BOP (0.29g, 0.65mmol).After 1 hour, this reaction vacuum-evaporation and obtain { 1-benzyl-2-oxo-2-[2-(2-chloro-6-{ methyl [(2-methyl cyclopropyl) methyl] amino } different nicotinoyl of 0.224g (33%) by anti-phase LC purifying) diazanyl]-the 1R-methylethyl orange solids of the amino acid tert-butyl ester: LCMS (M+H)=530.3.
Step C: cyclodehydration
With the different nicotinoyl of 1-benzyl-2-oxo-2-[2-(2-chloro-6-{ methyl [(2-methyl cyclopropyl) methyl] amino)) diazanyl]-the 1R-methylethyl) t-butyl carbamate (0.22g; 0.42mmol) (0.22g 0.93mmol) handles with Burgess reagent for solution in 2mLTHF.Should react sealing and ℃ reach 5 minutes at microwave reactor internal heating to 130.Add additional 2 normal Burgess reagent and this container sealed once more and reheat 5 minutes.Should react vacuum-evaporation and by flash column chromatography purifying (25-40% ethyl acetate: hexane) obtain { 1-[5-(2-chloro-6-{ methyl [(2-methyl cyclopropyl) methyl] amino } pyridin-4-yl)-1,3,4- diazole-2-yl]-1R-methyl-2-phenylethyl } the aminobutyric acid tert-butyl ester, it is a yellow oil: 1H NMR (400MHz, CDCl 3) δ 7.28 (m, 3H), 7.06 (m, 2H), 7.00 (s, 2H), 3.57 (m, 2H), 3.39 (m, 2H), 3.15 (s, 3H), 1.73 (s, 3H), 1.56 (s, 9H), 1.05 (d, J=5.7Hz, 3H), 0.72 (m, 2H), 0.45 (m, 1H), 0.29 (m, 1H).LCMS(M+H)=512.2。
Intermediate k:1-(2-{2-chloro-6-[methyl (methyl sulphonyl) amino] pyridin-4-yl }-1,3- azoles-5-yl)-1R-methyl-2-phenylethyl] t-butyl carbamate
Figure A20058001184300851
Steps A. coupling
To 2-chloro-6-[methyl (methyl sulphonyl) amino] Yi Yansuan (1.80g, 6.79mmol) add in the solution in 10mL DMF triethylamine (1.14mL, 8.15mmol); intermediate compound IV (2.00g, 8,15mmol); HOAt (1.11g, 8.15mmol) and EDC (1.56g, 8.15mmol).After following 3.5 hours of the room temperature, this reaction with saturated sodium bicarbonate solution (50mL) and salt solution (50mL) washing, is used Na with w/EtOAc (100mL) dilution 2SO 4Drying is filtered and vacuum concentration.By normal-phase chromatography purifying (0->70% EtOAc/ hexane and 0-7%MeOH/CH 2Cl 2) generate the white solid of (1R)-1-benzyl-3-({ 2-chloro-6-[methyl (methyl sulphonyl) amino] different nicotinoyl } amino)-2-hydroxyl-1-methyl-propyl t-butyl carbamate of 3.18g (87%).NMR(CDCl 3,400MHz)7.66(d,J=2.58Hz,1H);7.50(d,J=4.58Hz,1H);7.31(m,3H);7.18(m,2H);4.05(m,1H);3.84(m,1H);3.59(m,1H);3.43(s,3H);3.23(d,J=13.92Hz,1H);3.09(s,3H);2.68(d,J=13.55Hz,1H);1.45(s,9H);1.21(s,3H);LC/MS[M+H] +=541。
Step B. oxidation
({ 2-chloro-6-[methyl (methyl sulphonyl) amino] different nicotinoyl } amino)-(0.400g is 0.74mmol) at 8mLCH for 2-hydroxyl-1-methyl-propyl t-butyl carbamate to (1R)-1-benzyl-3- 2Cl 2Adding triethylamine in-20 ℃ of solution the among/4mL DMSO (0.41mL, 2.96mmol).After 5 minutes, (0.471g, 2.96mmol) solution in 3mL DMSO adds through conduit with the pyridine sulphur trioxide.After following 16 hours of the room temperature, Et is ended and used to this reaction with salt solution (10mL) 2O (15mL) extraction.Water layer is used Et once more 2O (10mL * 2) extraction.The organism that merges is used Na with 10% sodium bisulfite (10mL) and salt solution (10mL) washing 2SO 4Drying is filtered and vacuum concentration.Obtain the white solid of (1R)-1-benzyl-3-({ 2-chloro-6-[methyl (methyl sulphonyl) amino] different nicotinoyl } amino)-1-methyl-2-oxopropyl t-butyl carbamate of 0.335g (84%) by normal-phase chromatography purifying (20->70% EtOAc/ hexane).NMR(CDCl 3,400MHz)7.69(d,J=1.01Hz,1H);7.51(d,J=0.92Hz,1H);7.31(m,3H);7.12(m,2H);4.62(m,1H);4.41(m,1H);3.44(s,3H);3.11(s,3H);1.48(s,9H);1.31(s,3H);LC/MS[M-C4H7]+=483。
Step C. cyclodehydration
To (1R)-1-benzyl-3-({ 2-chloro-6-[methyl (methyl sulphonyl) amino] different nicotinoyl } amino)-1-methyl-2-oxopropyl t-butyl carbamate (0.190g; 0.35mmol) add in the solution in 2mL toluene Burgess reagent (0.504g, 2.12mmol).This is reflected at 130 ℃ of following microwave treatment 30 minutes.With the clarification of this reaction mixture yellow upper strata vacuum concentration and by normal-phase chromatography purifying (0->50% EtOAc/ hexane) obtain (1R)-1-(2-{2-chloro-6-[methyl (methyl sulphonyl) amino] pyridin-4-yl-1,3- azoles-5-yl)-1-methyl-2-phenylethyl t-butyl carbamate.NMR(CDCl 3,400MHz)7.86(s,1H);7.69(s,1H);7.28(m,3H);7.04(m,3H);3.45(s,3H);3.13(m,4H);1.60(s,3H);1.42(s,9H);LC/MS[M+H] +=521。
Intermediate l:N-(4-(diazanyl carbonyl)-6-{ methyl [(2-methyl cyclopropyl) methyl I amino pyridine-2-yl)-N-methyl Toluidrin
Figure A20058001184300861
Steps A; Coupling
To intermediate D (1.95g, 5.96mmol) and the Boc-hydrazine (0.866g, 6.55mmol) add in the solution in 25mLDMF EDC (1.37g, 7.15mmol) and HOAt (0.080g, 0.596mmol).Make this reaction carry out 15 hours, add subsequently that 3M LiCl ends and dilute with EtOAc.Separate each layer, water layer washs with EtOAc (2x).The organism that merges is used Na with 3MLiCI (2x) and salt water washing 2SO 4Drying is filtered and is concentrated.This resistates obtains required coupling adducts by normal-phase chromatography purifying (15->65% EtOAc/ hexane), and it is a white foam. 1H?NMR(CDCl 3,400MHz)δ8.17(br?s,1H),6.74(s,1H),6.70(s,1H),6.23(br?s,1H),3.50(dd,J=14.5,6.4Hz,1H),3.33(s,3H),3.30(dd,J=14.5,8.3Hz,1H),3.08(s,3H),3.06(s,3H),1.48(s,9H),1.00(d,J=5.9Hz,3H),0.74-0.60(m,2H),0.37(m,1H),0.23(m,1H);LCMS[M+H] +=442。
Step B; Deprotection
(2.50g is 5.662mmol) at 100mLCH under 0 ℃ HCl to be blown into the product of steps A 2Cl 2In solution in reach 10 minutes.This solution changes glassy yellow.This reaction rises to room temperature and reaches 30 minutes, concentrates subsequently and obtains yellow solid, and it need not to be further purified and can use. 1HNMR(CDCl 3,400MHz)δ7.55(s,1H),7.35(s,1H),3.44(m,2H),3.34(s,3H),3.22(s,3H),3.15(s,3H),0.92(d,J=5.8Hz,3H),0.72(m,1H),0.63(m,1H),0.40(m,1H),0.23(m,1H);LCMS[M+H] +=342。
Intermediate m:N-{4-(5-{ (1-((phenylbenzene methylene radical) amino) ethyl)-1,3,4- diazole-2-yl)-6-{ methyl [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl Toluidrin
Figure A20058001184300871
To N-(4-[5-(1-amino-ethyl)-1,3,4- diazole-2-yl]-6-I methyl [(2-methyl cyclopropyl) methyl] amino pyridine-2-yl)-N-methyl Toluidrin (embodiment 22 for 0.544g, 1.26mmol) is at 10mL CH 2Cl 2In solution in add benzophenone imines (0.25mL, 1.51mmol).In reaction process, generate white precipitate gradually.(0.160mL in the time of 0.894mmol), makes this reaction carry out 15 hours when the benzophenone imines that adds another equal portions.After 24 hours, this reaction H 2O and EtOAc dilution separate each layer.Water layer washs with EtOAc (2x), and Na is used in the salt water washing of the organism of merging 2SO 4Drying is filtered and is concentrated.This resistates by normal-phase chromatography purifying (2->35% EtOAc/ hexane) obtain N-(4-(5-{1-[(phenylbenzene methylene radical) amino] ethyl-1,3,4- diazole-2-yl)-and 6-{ methyl [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-the yellow foam of N-methyl Toluidrin. 1H?NMR(CDCl 3,400MHz)δ7.63(m,4H),7.54-7.50(m,4H),7.48(m,2H),7.05(s,1H),6.99(s,1H),4.95(q,J=6.5Hz,1H),3.52(dd,J=14.3,6.2Hz,1H),3.38(s,3H),3.34(dd,J=14.3,6.3Hz,1H),3.14(s,3H),3.12(s,3H),1.67(d,J=6.6Hz,3H),1.01(d,J=6.0Hz,3H),0.74(m,1H),0.65(m,1H),0.40(m,1H),0.25(m,1H);LCMS[M+H] +=559。
Intermediate n:((2E)-and 1-benzyl-{ benzyl [(2-methyl cyclopropyl) methyl] amino }-6-[methyl (methyl sulphonyl) amino] pyridin-4-yl }-1-methyl-4-oxo but-2-ene-1-yl) t-butyl carbamate (route plan 24)
Figure A20058001184300881
Steps A: the preparation of Weinreb acid amides
To intermediate C (1.0g 2.23mmol) adds N in the solution in the 10mL methylene dichloride, the O dimethyl hydroxylamine hydrochloride (226mg, 2.32mmol), diisopropylethylamine (574L, 3.48mmol), EDC (577mg, 3.01mmol), and HOAt (347mg, 2.55mmol).Gained solution at room temperature stirred 16 hours, with hypsokinesis to 0.3N HCl (20mL) with ethyl acetate extraction (3 * 50mL).The organism that merges is with sodium bicarbonate aqueous solution (75mL) washing, uses salt solution (75mL) washing subsequently and with dried over sodium sulfate and concentrated.By automatic purification by flash chromatography (silica gel box; about 20 minutes of 0-100% ethyl acetate/hexane) obtain 2-{ benzyl [(2 methyl cyclopropyl) methyl] amino-6-[(sec.-propyl alkylsulfonyl) (methyl) amino]-N-methoxyl group-N methyl Isonicotinamide, it is a yellow oil.LCMS[M+H]=475.1。
Step B: the preparation of methyl ketone
To the 2-{ of steps A benzyl [(2-methyl cyclopropyl) methyl] amino }-6-[(sec.-propyl alkylsulfonyl) (methyl) amino]-N-methoxyl group-N-methyl Isonicotinamide (860mg; 1.81mmol) add methyl-magnesium-bromide (3.62mL in-78 ℃ of solution in the 6mL dry THF; 10.8mmol 3M solution is in Anaesthetie Ether).Make reaction mixture slowly rise to 0 ℃ in 1 hour, incline it to the saturated ammonium chloride of 20mL and use ethyl acetate extraction (3 * 30mL) this moment.The organism that merges is used dried over sodium sulfate and concentrated with salt water washing (30mL).Obtain the glassy yellow solid of N-(4-ethanoyl-6-{ benzyl [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl-prop-2-sulphonamide by automatic purification by flash chromatography (silica gel box, about 35 minutes of 0-100% ethyl acetate/hexane).LCMS[M+H]=430.1。
Step C: aldol withdrawal
To the N-of step B (4-ethanoyl-6-{ benzyl [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl-prop-2-sulphonamide (100mg; 0.23mmol) add two (trimethyl silyl) lithamide (244L in-78 ℃ of solution in the 1.5mL dry THF; 0.49mmol; the THF solution of 2M), and with the gained mixture stirred 15 minutes.Add (1R)-1-benzyl-1-methyl-2-oxoethyl t-butyl carbamate (61mg, 0.23mmol is according to preparing the method preparation of intermediate III to step C) to this mixture and be dissolved in the 1mL dry THF.This solution slowly rises to room temperature and continue to stir subsequently 16 hours, ends with the saturated solution (15mL) of ammonium chloride this moment and with ethyl acetate extraction (2 * 25mL).The organism that merges is used dried over sodium sulfate and is concentrated.Obtain (1R by the reverse-phase chromatography purifying; 2E)-1-benzyl-4-{2-{ benzyl [(2-methyl cyclopropyl) methyl] amino }-6-[(sec.-propyl alkylsulfonyl) (methyl) amino] pyridin-4-yl)-1-methyl-4-oxo but-2-ene aminocarbamic acid tert-butyl ester; intermediate n, it is an orange oil.LCMS[M+H]=675.3。
Embodiment 1
N-(4-[5-(1R-amino-1-methyl-2-phenylethyl)-1,3,4- diazole-2-yl]-6-(benzyl [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl Toluidrin (route plan 8)
Figure A20058001184300901
Steps A: coupling
To intermediate C (100mg, 0.25mmol) and intermediate compound I (87mg, 0.30mmol) add in the solution in DMF (5mL) HOAt (37mg, 0.27mmol) and EDC (57mg, 0.30mmol).Reaction mixture at room temperature stirred 2 hours, handled with N-(tert-butoxycarbonyl)-D-Alpha-Methyl phenylalanine hydrazi acid amides (20mg, 0.07mmol, intermediate compound I), stirred 1 hour under the room temperature, and with the EtOAc dilution, Na is used in water, LiCl (x3) washing 2SO 4Dry; vacuum concentration and by purification by flash chromatography (silica gel, 10-50% EtOAc/ hexane) obtain [1R-benzyl-2-oxo-1-methyl-2-(2-{2-{ methyl [(2-methyl cyclopropyl) methyl] amino }-6-[methyl (methyl sulphonyl) amino] different nicotinoyl diazanyl) ethyl] white foam of t-butyl carbamate. 1HNMR (400MHz, CDCl 3) δ 9.35 (br s, 1H), 8.64 (br s, 1H), 7.38-7.14 (m, 10H), 6.79 (s, 2H), 4.88 (A of AB, d, J=17.2Hz, 1H), 4.83 (B of AB, d, J=17.2Hz, 1H), 4.66 (s, 1H), 3.60-3.48 (m, 2H), 3.36-3.24 (m, 1H), 3.28 (s, 3H), the 3.07 (B of AB, d, J=14.0Hz, 1H), 2.84 (s, 3H), 1.50 (s, 9H), 1.45 (s, 3H), 0.98 (d, J=6.0Hz, 3H), 0.86-0.76 (m, 1H), 0.65-0.55 (m, 1H), 0.39-0.31 (m, 1H), 0.30-0.23 (m, 1H).
Step B: dehydrocyclization and Boc remove
Will [1R-benzyl-2-oxo-1-methyl-2-(2-{2-{ methyl [(2-methyl cyclopropyl) methyl] amino }-6-[methyl (methyl sulphonyl) amino] different nicotinoyl } diazanyl) ethyl] t-butyl carbamate (140mg; 0.21mmol) and methoxycarbonyl amino-sulfonyl-triethyl ammonium hydroxide (197mg; 0.83mmol; Burgess reagent) solution in THF (5mL) is microwave (Smith synthesizer) and 120 ℃ of following irradiations 10 minutes; vacuum concentration and by purification by flash chromatography (silica gel; the 0-30%EtOAc/ hexane) obtain [1-(5-{2-{ benzyl [(2-methyl cyclopropyl) methyl] amino } }-6-[methyl (methyl sulphonyl) amino] pyridin-4-yl }-1; 3,4- diazole-2-yl)-1-methyl-2-phenylethyl] t-butyl carbamate.The EtOAc solution saturated with HCl (g) contacts 0.5 hour to remove Boc, concentrate and by the dry N-(4-[5-(1-amino-1-methyl-2-phenylethyl)-1 that obtains of two  alkane/water-cooled freeze-drying, 3,4- diazole-2-yl]-6-{ benzyl [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-hydrochloride of N-methyl Toluidrin. 1H?NMR(400MHz,CD 3OD)δ7.33-7.18(m,8H),7.08-7.02(m,3H),6.94(d,J=2.8Hz,1H),4.98-4.82(m,2H),3.74-3.66(m,1H),3.48-3.36(m,1H),3.41(s,3H),3.34-3.26(m,2H),2.96(s,3H),1.82(s,3H),0.98(d,J=6.0Hz,3H),0.91-0.81(m,1H),0.73-0.62(m,1H),0.48-0.39(m,1H),0.29-0.19(m,1H)。HRMS (ES, M+H) calculated value: C 30H 36N 6O 3S:561.2643, measured value: 561.2655.
Use preferred enantiomer among the intermediate A step B trans-S, thereby S prepares the pure S of preferred mapping, S, R example.
Embodiment 2
N-(4-[5-(1R-amino-1-methyl-2-phenylethyl)-1,3,4- diazole-2-yl]-6-([(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl Toluidrin
Figure A20058001184300911
Will [1-(5-{2-{ benzyl [(2-methyl cyclopropyl) methyl] amino }-6-[methyl (methyl sulphonyl) amino] pyridin-4-yl }-1; 3; 4- diazole-2-yl)-and 1R-methyl-2-phenylethyl] t-butyl carbamate (94mg; 0.14mmol; derive from embodiment 1) EtOH (10mL) and TFA (0.011mL, 0.14mmol) in solution with 20% Pd (OH) 2/ C (20mg) hydrogenation 3 hours under 40 ℃ and 1atm H2.Filter and concentrate, Boc removes (the saturated EtOAc of HCl (g)) subsequently, concentrates and at Et 2Development obtains N-(4-[5-(1R-amino-1-methyl-2-phenylethyl)-1,3,4- diazole-2-yl]-6-{[(2-methyl cyclopropyl) methyl among the O] amino } pyridine-2-yl)-light yellow solid of N-methyl Toluidrin hydrochloride. 1H?NMR(400MHz,CD 3OD)δ7.35-7.30(m,3H),7.11-7.05(m,2H),6.99(d,J=1.2Hz,1H),6.93(d,J=1.2Hz,1H),3.45(br?s,2H),3.37(s,3H),3.28-3.20(m,2H),3.19(s,3H),1.82(s,3H),1.06(d,J=6.0Hz,3H),0.90-0.80(m,1H),0.73-0.65(m,1H),0.47-0.41(m,1H),0.31-0.24(m,1H)。HRMS (ES, M+H) calculated value C 23H 30N 6O 3S:471.2173, measured value: 471.2175.
Use preferred enantiomer among the intermediate A step B trans-S, thereby S prepares the pure S of preferred mapping, S, R example.
Embodiment 3
N-(4-[5-(1R-amino-1-methyl-2-phenylethyl)-1,3,4- diazole-2-yl]-6-(methyl [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl Toluidrin
Figure A20058001184300921
By 2-{ benzyl [(2-methyl cyclopropyl) methyl] amino }-6-[methyl (methyl sulphonyl) amino] Yi Yansuan and N-methyl isophthalic acid-(2-trans-methyl cyclopropyl) methylamine adopts and is similar to embodiment 1 described method preparation.HRMS (ES, M+H) calculated value C 24H 32N 6O 3S:485.2330, measured value: 485.2355.
Use preferred enantiomer among the intermediate A step B trans-S, thereby S prepares the pure S of preferred mapping, S, R example.
Embodiment 4
N-(4-[5-(1R-amino-1-methyl-2-phenylethyl)-1,3,4- diazole-2-yl]-6-{ benzyl [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl-prop-2-sulphonamide
Figure A20058001184300922
By 2-{ benzyl [(2-methyl cyclopropyl) methyl] amino }-6-[methyl (sec.-propyl alkylsulfonyl) amino] Yi Yansuan (being similar to the method for preparing intermediate C) and N-(tert-butoxycarbonyl)-D-Alpha-Methyl phenylalanine hydrazi imines, adopt and be similar to embodiment 1 described method preparation by methyl (sec.-propyl alkylsulfonyl) amine.HRMS (ES, M+H) calculated value C 32H 40N 6O 3S:589.2955, measured value: 589.2920.
Use preferred enantiomer among the intermediate A step B trans-S, thereby S prepares the pure S of preferred mapping, S, R example.
Embodiment 5
N-(4-[5-(1R-amino-1-methyl-2-phenylethyl)-1,3,4- diazole-2-yl]-6-{[(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl-prop-2-sulphonamide
Be similar to embodiment 2 described methods preparations by embodiment 4 employings.HRMS (ES, M+H) calculated value C 25H 34N 6O 3S:499.2486, measured value: 499.2499.
Use preferred enantiomer among the intermediate A step B trans-S, thereby S prepares the pure S of preferred mapping, S, R example.
Embodiment 6
N-(4-[5-(1R-amino-1-methyl-2-phenylethyl)-1,3,4- diazole-2-yl]-6-((2,2-two fluoro ethyls) [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl Toluidrin (route plan 8)
Figure A20058001184300932
Be similar to embodiment 1 described method preparation by intermediate P utilization.HRMS[M+1]+calculated value=535.2298, measured value=535.2288.
Use preferred enantiomer among the intermediate A step B trans-S, thereby S prepares the pure S of preferred mapping, S, R example.
Embodiment 7
N-(4-[5-(1R-amino-1-methyl-2-phenylethyl)-1,3,4- diazole-2-yl]-6-{ (2-methoxy ethyl) [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl Toluidrin (route plan 8)
Figure A20058001184300941
Be similar to embodiment 1 described method preparation by intermediate Q utilization.HRMS[M+1]+calculated value=529.2592, measured value=529.2591.
Use preferred enantiomer among the intermediate A step B trans-S, thereby S prepares the pure S of preferred mapping, S, R example.
Embodiment 8
N-(4-[5-(1R-amino-1-methyl-2-phenylethyl)-1,3,4- diazole-2-yl]-6-{ (2,2-two fluoro ethyls) [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl-prop-2-sulphonamide (route plan 8)
Figure A20058001184300942
Be similar to embodiment 1 described method preparation by intermediate R utilization.HRMS[M+1]+calculated value=563.2611, measured value=562.2603.
Use preferred enantiomer among the intermediate A step B trans-S, thereby S prepares the pure S of preferred mapping, S, R example.
Embodiment 9
N-(4-[5-(1R-amino-1-methyl-2-phenylethyl)-1,3,4- diazole-2-yl]-6-{ (2-fluoro ethyl) [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl-prop-2-sulphonamide (route plan 8)
Be similar to embodiment 1 described method preparation by intermediate S utilization.HRMS[M+1]+calculated value=545.2705, measured value=545.2727.
Use preferred enantiomer among the intermediate A step B trans-S, thereby S prepares the pure S of preferred mapping, S, R example.
Embodiment 10
N-(4-[5-(1R-amino-1-methyl-2-phenylethyl)-1,3,4- diazole-2-yl]-6-{ (2-methoxy ethyl) [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl-prop-2-sulphonamide (route plan 8)
Utilize the mode that is similar to embodiment 1 to prepare by intermediate T.HRMS[M+1]+calculated value=557.2905, measured value=557.2900.
Use preferred enantiomer among the intermediate A step B trans-S, thereby S prepares the pure S of preferred mapping, S, R example.
Embodiment 11
N-(4-[5-(1R-amino-1-methyl-2-phenylethyl)-1,3,4- diazole-2-yl]-6-{[2-(dimethylamino) ethyl] [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl-prop-2-sulphonamide (route plan 8)
Be similar to embodiment 1 described method preparation by intermediate U utilization.LCMS[M+H] +=570.4。
Use preferred enantiomer among the intermediate A step B trans-S, thereby S prepares the pure S of preferred mapping, S, R example.
Embodiment 12
N-(4-[5-(1R-amino-1-methyl-2-phenylethyl)-1,3,4- diazole-2-yl]-6-([3-(dimethylamino) propyl group] [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl-prop-2-sulphonamide (route plan 8)
Figure A20058001184300962
Be similar to embodiment 1 described method preparation by intermediate V utilization.HRMS[M+1]+calculated value=584.3378, measured value=584.3382.
Use preferred enantiomer among the intermediate A step B trans-S, thereby S prepares the pure S of preferred mapping, S, R example.
Embodiment 13
N-[4-[5-(1R-amino-1-methyl-2-phenylethyl)-1,3,4- diazole-2-yl]-6-(1-[(1S, 2S)-2-methyl cyclopropyl] ethyl } amino) pyridine-2-yl]-N-methyl Toluidrin (route plan 8, the 2 hurdles)
Steps A .Pd coupling
To (1S)-1-[(1S; 25)-and 2-methyl cyclopropyl] ethane chlorination ammonium (ethanaminiumchloride) (intermediate P; 0.045g; 0.33mmol) and (1R)-1-(5-{2-chloro-6-[methyl (methyl sulphonyl) amino] pyridin-4-yl }-1; 3; 4- diazole-2-yl)-(intermediate h, 0.173g 0.33mmol) add K in the solution in 1mL DMF to 1R-methyl-2-phenylethyl t-butyl carbamate 3PO 4(0.352g, 1.66mmol) and Pd (PtBu 3) 2(0.025g, 0.05mmol).The 110 ℃ of down heating 12 hours and filtering with EtOAc dilution (30mL) and through Celite pad subsequently that are reflected at sealing.This diatomite washs with EtOAc (100mL).With the filtrate vacuum concentration with by normal-phase chromatography purifying (0->50% EtOAc/ hexane), anti-phase subsequently LC purifying.The gained material washs with EtOAc dilution (40mL) and with saturated sodium bicarbonate solution (20mL).Water layer is used EtOAc (20mL) extraction once more.The organism that merges is used Na with salt water washing (40mL) 2SO 4Dry; filter and vacuum concentration obtain (1R)-1R-methyl isophthalic acid of 0.030g (16%)-(5-{2-((1S)-1-[(1S; 2S)-and 2-methyl cyclopropyl] ethyl } amino)-6-[methyl (methyl sulphonyl) amino] pyridin-4-yl }-1; 3; 4- diazole-2-yl)-and 2-phenylethyl t-butyl carbamate, it is a yellow residue.NMR(CDCl 3,400MHz)7.27(m,3H);7.03(m,3H);6.79(s,1H);3.55(d,J=13.46Hz,1H);3.43(m,2H);3.37(s,3H);3.15(s,3H);1.71(s,3H);1.42(s,9H);1.29(d,J=6.41Hz,3H);1.07(d,J=5.77Hz,3H);0.67(m,2H);0.50(m,1H);0.26(m,1H);LC/MS[M+H] +=585。
Step B.Boc removes
With HCl (g) be blown into (1R)-1-methyl isophthalic acid-(5-{2-((1S)-1-[(1S; 2S)-and 2-methyl cyclopropyl] ethyl } amino)-6-[methyl (methyl sulphonyl) amino] pyridin-4-yl }-1; 3; 4- diazole-2-yl)-(0.030g is 0.05mmol) at 1mL CH for 2-phenylethyl t-butyl carbamate 2Cl 2In 0 ℃ of solution reach 5 minutes.0 ℃ after following 10 minutes, should react vacuum concentration and be dissolved in DMF.By anti-phase LC purifying; with postlyophilization generate 0.008g (27%) (2R)-2-(5-{2-((1S)-1-[(1S; 2S)-and 2-methyl cyclopropyl] ethyl } amino)-6-[methyl (methyl sulphonyl) amino] pyridin-4-yl-1; 3,4- diazole-2-yl)-white solid of 1-phenyl third-2-trifluoroacetic acid ammonium.NMR(CDCl 3,400MHz)7.31(m,3H);7.05(m,2H);6.91(d,J=1.1Hz,1H);6.84(d,J=1.1Hz,1H);3.55(m,1H);3.40(s,2H);3.16(s,3H);1.83(s,3H);1.28(d,J=6.4Hz,3H);1.06(d,J=5.7Hz,3H);0.67(m,2H);0.53(m,1H);0.21(m,1H);LC/MS[M+H] +=485。
Wherein X is that the other compound of  diazole is according to the described preparation of table 1.Method A and B are respectively with reference to the method A of route plan 8 and the method B of route plan 8.
Table I- diazole embodiment
Figure A20058001184301001
Embodiment 39
N-(4-[2-(1R-amino-I-methyl-2-phenylethyl)-1,3- azoles-5-yl]-6-(methyl [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl Toluidrin (route plan 16)
With intermediate E (100mg, 0.25mmol) and intermediate II (103mg, 0.37mmol) solution in DMF (0.5mL) is 120 ℃ of down heating 1 hour.Crude product is by preparation HPLC purifying (5->95%CH 3CN/H 2O; add 0.1% TFA; C18 PRO YMC 20 * 150mm) obtain [the 1R-methyl isophthalic acid-(4-{2-{ methyl [(2-methyl cyclopropyl) methyl] amino }-6-[methyl (methyl sulphonyl) amino] pyridin-4-yl-1,3- azoles-2-yl)-the 2-phenylethyl] t-butyl carbamate.Boc removes (the saturated EtOAc of HCl (g)), concentrates and by preparation HPLC purifying (5->95% CH 3CN/H 2O, add 0.1% TFA, C18 PRO YMC 20 * 150mm) obtain N-(4-[2-(1R-amino-1-methyl-2-phenylethyl)-1,3- azoles-5-yl]-tfa salt of 6-(methyl [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl Toluidrin. 1HNMR(400MHz,CD 3OD)δ8.58(s,1H),7.32-7.26(m,3H),7.02-6.96(m,2H),6.92(d,J=0.8Hz,1H),6.84(d,J=0.8Hz,1H),3.62-3.53(m,1H),3.43-3.26(m,3H),3.33(s,3H),3.14(s,3H),3.11(s,3H),1.81(s,3H),1.03(d,J=6.0Hz,3H),0.81-0.66(m,2H),0.48-0.41(m,1H),0.27-0.21(m,1H)。HRMS (ES, M+H) calculated value C 25H 33N 5O 3S:484.2377, measured value: 484.2400.
Use preferred enantiomer among the intermediate A step B trans-S, thereby S prepares the pure S of preferred mapping, S, R example.
Embodiment 40
N-(4-[2-(1R-amino-1-methyl-2-phenylethyl)-1,3- azoles-5-yl]-6-(methyl [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl Toluidrin (route plan 14)
Figure A20058001184301021
With intermediate G (25mg, 0.04mmol) and Burgess reagent (40mg, 0.17mmol) solution in THF (0.5mL) is in microwave (Smith synthesizer) and 80 ℃ irradiation 25 minutes down, under N2, concentrate and by purification by flash chromatography (silica gel, 0-40% EtOAc/ hexane).Remove Boc (1mL with HCl, 4mmol, 4M is in two  alkane), with postlyophilization obtain N-(4-[2-(1R-amino-1-methyl-2-phenylethyl)-1,3- azoles-5-yl]-6-{ methyl [(2-methyl cyclopropyl) methyl] amino pyridine-2-yl)-tfa salt of N-methyl Toluidrin. 1H?NMR(400MHz,CD 3OD)δ7.84(s,1H),7.32-7.26(m,3H),7.04-6.97(m,2H),6.92(s,1H),6.81(s,1H),3.70-3.57(m,1H),3.50-3.36(m,1H),3.42(s,2H),3.38(s,3H),3.18(s,6H),1.84(s,3H),1.04(d,J=6.0Hz,3H),0.84-0.706(m,2H),0.54-0.44(m,1H),0.34-0.26(m,1H)。HRMS (ES, M+H) calculated value C 25H 33N 5O 3S:484.2377, measured value: 484.2390.
Use preferred enantiomer among the intermediate A step B trans-S, thereby S prepares the pure S of preferred mapping, S, R example.
Embodiment 41
N-(4-[2-(1R-amino-1-methyl-2-phenylethyl)-1,3-thiazoles-5-yl]-6-(methyl [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl Toluidrin (route plan 14)
Figure A20058001184301031
With intermediate G (50mg, 0.08mmol) and Lawson ' s reagent (134mg, 0.33mmol) irradiation 15 minutes and 90 ℃ of 15 minutes (Boc also can utilize same reaction to remove) of irradiation down down of 85 ℃ in the inherent microwave of solution in acetonitrile (0.8mL) (Smith synthesizer), flow down at N2 and to concentrate and by preparation HPLC purifying (5->95% CH 3CN/H 2O, add 0.1% TFA, C18PRO YMC 20 * 150mm) obtain N-(4-[2-(1R-amino-1-methyl-2-phenylethyl)-1,3-thiazoles-5-yl]-tfa salt of 6-(methyl [(2 methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl Toluidrin.MS(ES,M+H)500。
Use preferred enantiomer among the intermediate A step B trans-S, thereby S prepares the pure S of preferred mapping, S, R example.
Embodiment 42
N-(4-[2-(1R-amino-1-methyl-2-phenylethyl)-1,3-imidazoles-5-yl]-6-{ methyl [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl Toluidrin (route plan 14)
With intermediate G (50mg, 0.08mmol) and ammonium acetate (64mg, mixture 0.83mmol) is 150 ℃ of down fusings 15 minutes (loss part Boc).Make reaction mixture be cooled to room temperature, use saturated NaHCO 3Dilution is with EtOAc (x3) extraction.Na is used in the organic layer salt water washing that merges 2SO 4Drying and vacuum concentration.The TFA (5mL, 10% TFA is in DCM) that is used on the DCM removes Boc, and vacuum concentration is by preparation HPLC purifying (5->95% CH 3CN/H 2O, add 0.1%TFA, C18 PRO YMC 20 * 150mm) and lyophilize obtain the tfa salt of N-(4-[2-(1R-amino-1-methyl-2-phenylethyl)-1,3-imidazoles-5-yl]-6-{ methyl [(2-methyl cyclopropyl) methyl] aminopyridine-2-yl)-N-methyl Toluidrin. 1H NMR (400MHz, CD 3OD) δ 7.76 (s, 1H), 7.29-7.21 (m, 3H), 7.06 (s, 2H), 6.92-6.86 (m, 2H), and 3.66-3.56 (m, 1H), 3.48-3.35 (m, 1H), the 3.41 (A of AB, d, J=13.6Hz, 1H), 3.34 (s, 3H), the 3.26 (B of AB, d, J=13.6Hz, 1H), 3.18 (s, 3H), 3.16 (s, 3H), 1.74 (s, 3H), 1.04 (d, J=5.7Hz, 3H), 0.85-0.70 (m, 2H), 0.51-0.45 (m, 1H), 0.31-0.25 (m, 1H).HRMS (ES, M+H) calculated value C 25H34N 6O2S:483.2537, measured value: 483.2548.
Use preferred enantiomer among the intermediate A step B trans-S, thereby S prepares the pure S of preferred mapping, S, R example.
Embodiment 43
N-(4-[5-(1R-amino-I-methyl-2-phenylethyl)-1,3- azoles-2-yl]-6-(methyl [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl) N-methyl Toluidrin (route plan 17)
Figure A20058001184301041
Embodiment 43 adopts the described method of preparation that is similar to embodiment 7 to make by intermediate H. 1H NMR (400MHz, CD 3OD) δ 7.32-7.26 (m, 3H), 7.22 (s, 1H), 7.14 (s, 1H), and 7.06-6.99 (m, 3H), 3.62 (A of ABX, dd, J=14.4,6.2Hz, 1H), 3.45 (A of AB, d, J=13.7Hz, 1H), 3.43 (B of ABX, dd, J=14.4,2.3Hz, 1H), 3.39 (s, 3H), 3.26 (B of AB, d, J=13.7Hz, 1H), 3.18 (s, 3H), 3.16 (s, 3H), 1.73 (s, 3H), 1.04 (d, J=6.2Hz, 3H), 0.86-0.70 (m, 2H), 0.52-0.44 (m, 1H), 0.30-0.24 (m, 1H).HRMS (ES, M+H) calculated value C 25H 33N 5O 3S:484.2377, measured value: 484.2374.
Use preferred enantiomer among the intermediate A step B trans-S, thereby S prepares the pure S of preferred mapping, S, R example.
Embodiment 44
N[4-[5-(1R-amino-1-methyl-2-phenylethyl)-1,3- azoles-2-yl]-6-(1-[(1S, 2S)-2-methyl cyclopropyl] ethyl) amino) pyridine-2-yl]-N-methyl Toluidrin
Steps A .Pd coupling
To (1S)-1-[(1S; 2S)-and 2-methyl cyclopropyl] ethane chlorination ammonium (intermediate P; 0.039g; 0.29mmol) and (1R)-1-(2-(2-chloro-6-[methyl (methyl sulphonyl) amino] pyridin-4-yl)-1; 3- azoles-5-yl)-1-methyl-2-phenylethyl t-butyl carbamate (intermediate k; 0.050g, 0.10mmol) add K in the solution in 1mL DMF 3PO 4(0.122g, 0.58mmol) and Pd (PtBu 3) 2(0.007g, 0.01mmol).After following 16 hours of the room temperature, with sealed reaction 110 ℃ down heating 8.5 hours and with after Celite pad filter.This diatomite is washed with EtOAc (60mL).Be dissolved in DMF and pass through anti-phase LC purifying with the filtrate vacuum concentration and with the gained resistates.The gained material washs with EtOAc dilution (40mL) and with saturated sodium bicarbonate solution (20mL).Water layer is used EtOAc (20mL) extraction once more.The organism that merges is used Na with salt water washing (20mL) 2SO 4Dry; filter and vacuum concentration obtain (1R)-1-methyl isophthalic acid of 0.028g (50%)-(2-{2-((1S)-1-[(1S; 2S)-and 2-methyl cyclopropyl] ethyl } amino)-6-[methyl (methyl sulphonyl) amino] pyridin-4-yl }-1; 3- azoles-5-yl)-and 2-phenylethyl t-butyl carbamate, it is a yellow residue.(CDCl 3,400MHz)7.28(m?3H);7.06(d,J=0.91Hz,1H);7.03(m,2H);6.93(s,1H);6.76(s,1H);3.50(m,1H);3.36(m,4H);14(m,4H);1.60(s,3H);1.42(s,9H);LC/MS[M+H] +=584。
Step B.Boc removes
With HCl (g) be blown into (1R)-1-methyl isophthalic acid-(2-{2-((1S)-1-[(1S; 2S)-and 2-methyl cyclopropyl] ethyl } amino)-6-[methyl (methyl sulphonyl) amino] pyridin-4-yl }-1; 3- azoles-5-yl)-(0.028g is 0.05mmol) at 1mL CH for 2-phenylethyl t-butyl carbamate 2Cl 2In 0 ℃ of solution in reach 5 minutes.0 ℃ following 10 minutes, should react vacuum concentration and be dissolved in DMF.By anti-phase LC purifying; obtain (2R)-2-(2-(2-(((1S)-1-[(1S with postlyophilization; 25)-and 2-methyl cyclopropyl] ethyl) amino)-6-[methyl (methyl sulphonyl) amino] pyridin-4-yl }-1,3- azoles-5-yl)-white solid of 1-phenyl-propane-2-ammonium.(CDCl 3,400MHz)7.27(m,3H);7.19(s,1H);7.01(m,3H);6.89(d,J=0.92Hz,1H);3.53(m,1H);3.45(d,J=13.37Hz,1H);3.33(s,3H);3.24d,J=13.55Hz,1H);3.14(s,3H);1.71(s,3H);1.27(d,J=6.41Hz,3H);1.94(d,J=5.67Hz,3H);0.66(m,2H);0.53(m,1H);0.20(m,1H);LC/MS[M+H] +=484。
Other  oxazole derivatives are according to following table 3 described preparations.
Table 3- oxazole derivatives
Figure A20058001184301061
Embodiment 49
N-(4-[5-(1R-amino-1-methyl-2-phenylethyl)-1,3-thiazoles-2-yl]-6-{ methyl [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl Toluidrin (route plan 17)
Embodiment 49 is by intermediate H, utilizes the described method preparation of the preparation that is similar to embodiment 8. 1H NMR (400MHz, CD 3OD) δ 7.55 (s, 1H), 7.26-7.20 (m, 3H), 7.05-7.00 (m, 2H), 6.99 (d, J=1.2Hz, 1H), 6.90 (d, J=1.2Hz, 1H), 3.59 (A of ABX, dd, J=14.4,6.4Hz, 1H), 3.42 (B of ABX, dd, J=14.4,6.8Hz, 1H), 3.37 (s, 3H), 3.17 (s, 3H), 3.15 (s, 3H), 3.09 (s, 2H), 1.60 (s, 3H), 1.04 (d, J=6.0Hz, 3H), 0.85-0.68 (m, 2H), 0.50-0.42 (m, 1H), 0.29-0.21 (m, 1H).HRMS (ES, M+H) calculated value C 25H 33N 5O 2S 2: 500.2149, measured value: 500.2149.
Use preferred enantiomer among the intermediate A step B trans-S, thereby S prepares the pure S of preferred mapping, S, R example.
Embodiment 50
N-(4-[5-(1R-amino-1-methyl-2-phenylethyl)-4H-1,2,4-triazole-3-yl]-6-(methyl [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl Toluidrin (route plan 11)
With intermediate compound I (17mg, 0.06mmol), intermediate J (20mg, 0.05mmol) and diisopropylethylamine (0.013mL, 0.08mmol) solution in EtOH (1mL) sealing and 150 ℃ of heating 2 days down.With the reaction mixture vacuum concentration and by preparation HPLC purifying (5->95%CH 3CN/H2O, the 0.1%TFA that adds, C18 PRO YMC 20 * 150mm) obtain N-(4-[5-(1R-amino-1-methyl-2-phenylethyl)-4H-1,2,4-triazole-3-yl]-6-{ methyl [(2-methyl cyclopropyl) methyl] amino pyridine-2-yl)-tfa salt of N-methyl Toluidrin. 1HNMR(400MHz,CD 3OD)δ7.30-7.22(m,3H),7.13(s,1H),7.04(s,1H),7.02-6.96(m,2H),3.65-3.56(m,1H),3.48-3.42(m,1H),3.38(s,3H),3.36(s,2H),3.18(s,3H),3.17(s,3H),1.78(s,3H),1.04(d,J=6.0Hz,3H),0.86-0.70(m,2H),0.52-0.44(m,1H),0.30-0.23(m,1H)。
Use preferred enantiomer among the intermediate A step B trans-S, thereby S prepares the pure S of preferred mapping, S, R example.
Embodiment 51
N-(4-[5-(1-amino-ethyl)-1,3,4- diazole-2-yl]-6-{ methyl [(2-methyl cyclopropyl) methyl] amino) pyridine-2-yl)-N-methyl Toluidrin (another route plan 8)
Steps A: coupling
(0.610g is 1.614mmol) with N-Boc-L-Ala (0.336g, 1.78mmol) adding HunigShi alkali (0.564mL in the solution in 10mL DMF to intermediate 1,3.23mmol), EDC (0.371g, 1.94mmol) and HOAt (0.025g, 0.161mmol).After 15 hours, this reaction separates each layer with 3M LiCl and EtOAc dilution.Water layer washs with EtOAc (2x), and the organism of merging is used Na with 3M LiCl (2x) and salt water washing 2SO 4Drying is filtered and is concentrated.This resistates obtains required coupling adducts by normal-phase chromatography purifying (35->90% EtOAc/ hexane), and it is yellow foam. 1H?NMR(CDCl 3,400MHz)δ9.11(br?s,1H),8.97(br?s,1H),8.00(s,1H),6.76(s,1H),6.69(s,1H),5.09(d,J=7.5Hz,1H),3.47(dd,J=14.5,6.2,1H),3.32(s,3H),3.29(dd,J=14.5,7.0Hz,1H),3.07(s,3H),2.93(s,3H),1.43(s,9H),1.41(d,J=7.5Hz,3H),1.00(d,J=5.7Hz,3H),0.68(m,1H),0.62(m,1H),0.36(m,1H),0.23(m,1H);LCMS[M+H] +=513。
Step B: dehydration
To the product of steps A (0.763g, 1.49mmol) at 7mL 1, add in the solution in the 2-ethylene dichloride Burgess reagent (1.42g, 5.95mmol).These slurries are 120 ℃ of following microwave treatment 10 minutes, directly incline subsequently to being used for positive purifying (10->55% EtOAc/ hexane) to the top of silicagel column.Obtain expecting product, it is yellow foam. 1H?NMR(CDCl 3,400MHz)δ7.01(s,1H),6.93(s,1H),5.12(m,1H),3.52(dd,J=14.5,6.3Hz,1H),3.38(s,3H),3.33(dd,J=14.5,7.0Hz,1H),3.15(s,3H),3.11(s,3H),1.63(d,J=6.8Hz,3H),1.44(s,9H),1.01(d,J=4.9Hz,3H),0.73(m,1H),0.67(m,1H),0.41(m,1H),0.26(m,1H);LCMS[M+H] +=495。
Step C: deprotection
(0.532g, 1.08mmol) solution in 15mLEtOAc reaches 5 minutes HCl to be blown into the product of step B under 0 ℃.Bright yellow solution was risen to room temperature 1 hour, concentrate subsequently and obtain N-(4-[5-(1-amino-ethyl)-1,3,4- diazole-2-yl]-6-{ methyl [(2-methyl cyclopropyl) methyl] aminopyridine-2-yl)-N-methyl Toluidrin, it is yellow foam, and it need not to be further purified and can use. 1H?NMR(d 4-MeOH,400MHz)87.15(s,1H),7.05(s,1H),4.95(q,J=7.0Hz,1H),3.61(dd,J=14.5,8.1Hz,1H),3.43(dd,J=14.5,6.9Hz,IH),3.38(s,3H),3.16(s,3H),3.15(s,3H),1.79(d,J=7.0Hz,3H),1.01(d,J=5.8Hz,3H),0.82-0.75(m,2H),0.47(m,1H),0.25(m,1H);LCMS[M+H] +=395。
Use preferred enantiomer among the intermediate A step B trans-S, thereby S prepares the pure S of preferred mapping, S, R example.
Embodiment 52
N (4-[5-(1-amino-1-methyl-2-pyridin-4-yl ethyl)-1,3,4- diazole-2-yl]-6-{[(2 methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl Toluidrin (another route plan 8)
N-(4-[5-(1-amino-1-methyl-2-pyridin-4-yl ethyl)-1,3,4- diazole-2-yl]-6-([(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl Toluidrin is to be similar to embodiment 70 described method preparations by intermediate III and intermediate compound I utilization. 1H?NMR(d 4-MeOH)δ8.77(d,J=6.0Hz,2H),7.78(d,J=6:4Hz,2H),6.96(d,J=l?1.7Hz,2H),3.78(app?ABX,JAB,JAX,JBx=13.5Hz,2H),3.37(s,3H),3.24(d,J=6.8Hz,2H),3.16(s,3H),1.94(s,3H),1.06(d,J=6.0,3H),0.87-0.81(m,1H),0.72-0.66(m,1H),0.43(dt,J=8.7,4.6Hz,1H),0.26(dt,J=8.6,4.8Hz,1H)。HRMS (ES, M+H) calculated value C 22H 29N 7O 3S:472.2126, measured value: 472.2163.
Use preferred enantiomer among the intermediate A step B trans-S, thereby S prepares the pure S of preferred mapping, S, R example.
Table IV- oxadiazole derivative
Figure A20058001184301102
Figure A20058001184301111
Figure A20058001184301121
Figure A20058001184301131
Figure A20058001184301141
Figure A20058001184301151
Embodiment 87
N-(4-[5-(1-amino-1-methyl-2-(4-fluorophenyl) ethyl)-1,3,4- diazole-2-yl]-6-(methyl [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl Toluidrin (another route plan 8, the 2 hurdles)
Figure A20058001184301152
(0.051g 0.091mmol) adds down 1M in 0 ℃ in the solution in 0.80mL DMF and is contained in NaHMDS among the THF (0.13mL, 1.30mmol), this reaction becomes blueness to intermediate m.After 5 minutes, (0.180mL, 0.180mmol), behind reinforced finishing, this reaction becomes yellow to add the 1M solution of 4-luorobenzyl bromination thing in DMF with syringe.After 30 minutes, add 0.125mL H 2O and 0.025mL 1H HCl.1.5 after hour, add 0.125mL 1N HCl, this reaction was carried out 15 hours, be carried in the SCX ion exchange column subsequently, use the MeOH wash-out, subsequently with the 2M NH3 wash-out that is contained among the MeOH.Separate impure required product by alkaline cut, it is further by preparation HPLC purifying (5->95% CH 3CN/H 2O, about 30min, 0.1% TFA that adds, obtain after C18 PRO YMC 20 * 150mm) lyophilizes N-(4-{5-[1-amino-2-(4-fluorophenyl)-1-methylethyl]-1,3,4- diazole-2-yl }-6-{ methyl [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-the cotton-shaped yellow solid of N-methyl Toluidrin. 1HNMR (d 4-MeOH, 400MHz) δ 7.12-7.03 (m, 5H), 6.91 (s, 1H), 3.59 (dd, J=14.8,6.2Hz, 1H), 3.43-3.39 (m, 3H), 3.31 (s, 3H), 3.16 (s, 3H), 3.15 (s, 3H), 1.84 (s, 3H), 1.02 (d, J=6.0Hz, 3H), 0.80-0.70 (m, 2H), 0.46 (m, 1H), 0.26 (m, 1H); High resolution mass spectrometry (FT/ICR) calculated value M+H=503.2235 measured value 503.2279.
Use preferred enantiomer among the intermediate A step B trans-S, thereby S prepares the pure S of preferred mapping, S, R example.
Other  oxadiazole derivatives are according to following table 5 described method preparations.
Table 5- oxadiazole derivative
Embodiment 91
Figure A20058001184301162
Steps A: with 4-amino 2, the 6-dichloropyridine (2.0g 12.2mmol) is suspended in the toluene (50mL), to wherein add phenyl aldehyde (6.24mL, 30.7mmol).This reacts reflux 12 hours and collects water with Dean-Stark equipment.Solvent removed in vacuo subsequently.This resistates is dissolved in ethanol (25mL) and is heated to 50 ℃.Dropping NaBH4 (1.86g, 61mmol).This reaction postheating reaches 2 hours for 75 ℃.Solvent removed in vacuo adds entry (100mL) and (3 * 100mL) extract with EtOAc with this solution.The organic layer that merges salt water washing (1 * 100mL), use MgSO 4Drying is filtered and is concentrated.Obtain this benzylaniline (2.93g, 94%) by silica gel chromatography purifying (20%EtOAc/Hex).LCMS[M+H]=253。 1H?NMR(CDCl 3)δ7.40-7.14(m,5H),6.46(s,2H),4.35(d,J=5.5Hz,2H)
Step B: to the aniline of steps A (0.079g, 0.31mmol) add in the solution in the brooethyl cyclopropane (0.05g, 0.37mmol), add subsequently NaH (60% dispersion, 0.014g, 0.37mmol).This solution at room temperature stirred 30 minutes, used H subsequently 2O ends.(3 * 30mL) extract this solution, after this organic layer MgSO of He Binging with EtOAc 4Drying is filtered and is concentrated.Obtain cyclopropyl monomethylaniline (0.078g, 81%) by silica gel chromatography purifying (15% EtOAc/Hex).
LCMS[M+H]=307.1
1H?NMR(CDCl 3)δ7.36-7.13(m,5H),6.51(s,2H),4.64(s,2H),3.29(d,J=6.6Hz,2H),1.08-1.6(m,1H),0.60-0.55(m,2H),0.24-0.21(m,2H)
Step C: with the dichloropyridine of step B (0.078g, 0.25mmol), N-methyl sulphonamide (0.033g, 0.30mmol), K 3PO 4(0.075g, 0.35mmol), and Xanphos (0.01g, two  alkane solution (2mL) 0.17mmol) outgased with being blown into argon gas in 15 minutes.Add Pd2 (dba) 3G, 0.05mmol) and this solution is heated to 100 ℃ reaches 12 hours.Cool off this solution and use H 2O (30mL) dilution.This solution is with EtOAc (3 * 30mL) extractions and the organic layer MgSO that merge 4Drying is filtered and is concentrated.Obtain required sulphonamide (0.034g, 35%) by silica gel chromatography purifying (20% EtOAc/Hex).
LCMS[M+H]=380.3
1H?NMR(CDCl 3)δ7.35-7.19(m,3H),7.16(d,J=7.1Hz,2H),6.68(s,1H),6.46(s,1H),4.62(s,2H),3.31(d,J=6.5Hz,2H),3.29(s,3H),2.93(s,3H),1.12-1.08(m,1H),0.58-0.55(m,2H),0.24-.21(m,2H)。
Step D: (0.17g 1.45mmol), adds Pd (PPh subsequently to add zinc cyanide in the solution of sulphonamide in DMF (4mL) of step C 3) 4(0.017g, 0.15mmol).This solution is heated to 85 ℃ reaches 48 hours.With reaction mixture H 2O (30mL) dilution and with EtOAc (3, * 30mL) extract.The organic layer that merges salt water washing (1 * 50mL), use MgSO 4Drying is filtered, and concentrates.Obtain this cyanopyridine (0.27g, 81%) by silica gel chromatography purifying (30% EtOAc/Hex).
LCMS[M+H]=371.1
1H?NMR(CDCl 3)δ7.36-7.26(m,3H),7.15(d,J=7.2Hz,2H),6.87(s,1H),6.82(s,1H),4.68(s,2H),3.36(d,J=6.5Hz,2H),3.30(s,3H),2.92(s,3H),1.12-1.09(m,1H),0.61(d,J=7.6Hz,2H),0.27(d,J=4.9Hz,2H)。
Step e: (0.15g is 0.41mmol) 9: 1EtOH/H to the cyanopyridine of step D 2Adding solid KOH in the solution among the O (10mL) (0.26g, 4.1mmol).This solution is heated to 85 ℃ reaches 16 hours.Cool off this solution, use 1N HCl acidifying, and (3 * 30mL) extract with EtOAc.The organic layer that merges salt water washing (1 * 50mL), use MgSO 4Drying is filtered and is concentrated.This resistates need not to be further purified and can use.In the solution of this carboxylic acid in DCM (5mL), add the diazanyl phenylalanine (0.16g, 0.59mmol), DIPEA (0.023g, 1.78mmol) and bop reagent (0.25g, 0.59mmol).This reaction was at room temperature stirred 1 hour, obtained diketo hydrazine (0.30g, 77%) by silica gel chromatography purifying (5%MeOH/ chloroform) subsequently
LCMS[M+H]=665.1
1H?NMR(CDCl 3)δ7.41-7.16(m,11H),6.83(s,1H),4.72(s,2H),3.57(d,J=14Hz,1H),3.39(d,J=6.6Hz,2H),3.35(s,3H),3.10(d,J=14Hz,1H),2.91(s,3H),1.50(s,9H),1.45(s,3H),1.13-1.10(m,1H),0.59-0.55(m,2H),0.25(d,J=5.0Hz,2H)。
Step F: to the diketo hydrazine of step e (0.30g, 0.46mmol) 1, add in the 2-dichloroethane solution (3mL) burgess reagent (0.33g, 1.4mmol).With this solution 120 ℃ of following microwave treatment 8 minutes.Obtain required  diazole (0.29g, 62%) by the silica gel chromatography purifying.
LCMS[M+H]=647.1
1H?NMR(CDCl 3)δ7.39-7.04(m,11H),6.75(s,1H),4.73(s,2H),3.54-3.48(m,2H),3.41(d,J=6.6Hz,2H),3.34(s,3H),3.03(s,3H),1.72(br?s,3H),1.41(br?s,9H),1.24-1.13(m,1H),0.60(q,J=7.9Hz,2H),0.26(q,J=5.3Hz,2H)。
Step G: (add TFA (1.5mL) in the 0.027g, methyl alcohol 0.05mmol) (5mL) solution, add the Pd (OH) of catalytic amount subsequently to the  of step F diazole 2Place H2 atmosphere to assign 20 minutes through balloon this solution.This solution obtains the aminopyridine of complete deprotection through diatomite filtration and vacuum concentration.
LCMS[M+H]=457.1
1H?NMR(CD 3OD)δ7.31-7.21(m,3H),7.21(s,1H)。7.08(m,2H),6.69(s,1H),3.46(d,J=4.8Hz,2H),3.36(s,3H),3.17(s,3H),3.08(d,J=7.0Hz,2H),1.87(s,3H),1.14-1.08(m,1H),0.59(d,J=7.0Hz,2H),0.30(d,J=5.1Hz,2H)。
Embodiment 92
N-(4-(5-[(1R)-1-amino-1-methyl-2-phenylethyl]-the 1H-pyrazole-3-yl)-6-{ benzyl [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl-prop-2-sulphonamide
Figure A20058001184301191
To intermediate n (62mg, 0.09mmol) add in the solution in 0.5mL DMF anhydrous hydrazine (14L, 0.46mmol).The gained mixture was 90 ℃ of following reflux 18 hours and be cooled to room temperature subsequently.To wherein adding 2mL water and 3mL ethyl acetate.Separate organic layer, use the salt water washing, use dried over sodium sulfate and be concentrated into dried.By the reverse-phase chromatography purifying obtain (1R)-1-(3-(2-(benzyl [(2-methyl cyclopropyl) methyl] amino)-6-[(sec.-propyl alkylsulfonyl) (methyl) amino] pyridin-4-yl }-1H-pyrazoles-5-yl)-1-methyl-2-phenylethyl t-butyl carbamate, it is dissolved in the methylene dichloride that 1mL contains 20% TFA.After 1 hour, this solution concentration is obtained title compound, N-(4-(5-[(1R)-1-amino-1-methyl-2-phenylethyl]-the 1H-pyrazole-3-yl)-6-(benzyl [(2-methyl cyclopropyl) methyl] amino) pyridine-2-yl)-N-methyl-prop-2-sulphonamide.
1H?NMR(400MHz,d4-MeOH)δ7.31-7.19(m,9H),7.01-6.99(m,2H),6.76(s,1H),6.71(s,1H),6.61(s,1H),4.96-4.76(m,2H),3.81-3.74(m,1H),3.69-3.63(m,1H)3.39(s,3H),3.35-3.22(m,4H),1.70(s,3H),1.14(d,J=6.8Hz,6H),0.94(d,J=6Hz,3H),0.87-0.84(m,1H),0.66-0.60(m,1H),0.42-0.38(m,1H),0.26-0.21(m,1H);LCMS[M+H]=587.2。
Use preferred enantiomer among the intermediate A step B trans-S, thereby S prepares the pure S of preferred mapping, S, R example.
Embodiment 93
N-(4-(6-[(1R)-1-amino-1-methyl-2-phenylethyl] pyrimidine-4-yl }-6-{ benzyl [(2-methyl cyclopropyl) methyl] aminopyridine-2-yl)-N-methyl-prop-2-sulphonamide
Steps A: condensation
The suspension of 69mg (1.01mmol) sodium ethylate in 1mL DMA is added drop-wise in the solution of 81mg (1.01mmol) amitraz hydrochloride in 1mL DMA, and with this miscellany vigorous stirring 5 minutes.(68mg, 0.10mmol) drips of solution in 1mL DMA is added in the single mixture, and gained mixture heating up to 110 ℃ is reached 30 minutes with intermediate n.Make it be cooled to room temperature subsequently.Add entry (10mL) and this miscellany ethyl acetate extraction (3 * 15mL).The organism were washed with 3M aq.LiCI that merges (3 * 25mL), with salt water washing (25mL), with dried over sodium sulfate and concentrated.Obtain (1R)-1-(6-{2-{ benzyl [(2-methyl cyclopropyl) methyl] amino }-6-[(sec.-propyl alkylsulfonyl) (methyl) amino by the reverse-phase chromatography purifying] pyridin-4-yl }-3; 4-dihydro-pyrimidin-4-yl)-and 1-methyl-2-phenylethyl t-butyl carbamate, it is a yellow oil.LCMS[M+H]=701.3
Step B: oxidation and deprotection
To (the 1R)-1-of above-mentioned steps A (6-(2-(benzyl [(2-methyl cyclopropyl) methyl] amino)-6-[(sec.-propyl alkylsulfonyl) (methyl) amino] pyridin-4-yl-3; 4-dihydro-pyrimidin-4-yl)-1-methyl-2-phenylethyl t-butyl carbamate (49mg; 0.07mmol) add in the solution in 2mL toluene dichloro dicyano quinone (24mg, 0.11mmol).Gained solution was heated 30 minutes down at 120 ℃, be cooled to room temperature subsequently and be concentrated into dried.Obtain (1R)-1-(6-(2-(benzyl [(2-methyl cyclopropyl) methyl] amino)-6-[(sec.-propyl alkylsulfonyl) (methyl) amino] pyridin-4-yl } pyrimidine-4-yl)-1-methyl-2-phenylethyl t-butyl carbamate by the reverse-phase chromatography purifying, make it be dissolved in the methylene dichloride that 1mL contains 20%TFA.Make this miscellany place 1 hour, concentrate and obtain this title compound N-(4-{6-[(1R)-1-amino-1-methyl-2-phenylethyl] pyrimidine-4-yl }-6-{ benzyl [(2-methyl cyclopropyl) methyl] amino } pyridine-2-yl)-N-methyl-prop-2-sulphonamide. 1H?NMR(400MHz,d4-MeOH)δ9.29(s,1H),7.66-7.65(m,1H),7.31-7.21(m,8H),7.02-7.01(m,2H),6.96-6.93(m,2H),4.98-4.60(m,2H),3.83-3.66(m,2H),3.38(s,3H),3.35-3.25(m,4H),1.81(s,3H),1.16(d,J=6.8Hz),0.96(dd,J=6Hz,J=1.8Hz,3H)0.90-0.82(m,1H),0.70-0.62(m,1H),0.45-0.40(m,1H),0.28-0.24(m,1H);LCMS[M+H]=599.1。
Use preferred enantiomer among the intermediate A step B trans-S, thereby S prepares the pure S of preferred mapping, S, R example.
Embodiment 94
N (2-(5-[(1R)-1-amino-1-methyl-2-phenylethyl]-1,3,4- diazole-2-yl }-6-[(cyclopropyl methyl) (methyl) amino] pyridin-4-yl }-N-methyl Toluidrin
Figure A20058001184301211
Steps A: will contain 6.52g (40.0mmol) 4-amino-2 in the 200mL pyridine, the solution of 6-dichloropyridine is handled with 22.8g (200mmol) methylsulfonyl chloride and was heated 72 hours down at 60 ℃.Cooling reaction mixture and evaporating solvent.The gained resistates is dissolved in the methylene dichloride of 200mL again and uses NaHCO 3(50mL), water (3 * 50mL) and salt solution (50mL) washing.Organic phase MgSO 4Drying is filtered and is concentrated.Obtain the disulfonic acid amide that required sulphonamide (5.30g, 55%) and 3.44g (27%) do not expect by silica gel chromatography purifying (20% EtOAc/ hexane).LCMS[M+H]=241.1
Step B: the solution of 5.00g (20.7mmol) sulphonamide that will in 12.5g (176mmol) cyclopropylamine, contain steps A under 125 ℃ at the sealed tube internal heating.After 72 hours, cool off this reaction mixture and evaporate the brown resistates, obtain required aminopyridine (3.42g, 60%) by silica gel chromatography purifying (60% EtOAc/ hexane).LCMS[M+H]=276.1
Step C: in 0 ℃ of solution of aminopyridine that in 15mL DMF, contains 1.00g (3.62mmol) step B and 1.13g (7.98mmol) methyl iodide, add 191mg (7.98mmol) sodium hydride.This solution is at room temperature stirred months 17 hours and end with 5mL water.With reaction mixture with 100mL ether dilution and water (6 * 10mL), use salt solution (10mL) washing subsequently.Organic phase MgSO 4Drying is filtered and is concentrated.Obtain this aminopyridine that methylates (1.00g, 91%) by silica gel chromatography purifying (30% EtOAc/Hex).LCMS[M+H]=304.1
Step D: (773mg 6.58mmol), adds Pd (PPh3) 4mg, 0.658mmol) subsequently to obtain adding in the solution zinc cyanide to the chloropyridine that contains 1.00g (3.29mmol) step C in 15mL DMF.This solution heated 24 hours down at 90 ℃, made this reaction mixture cooling subsequently and with ether dilution (100mL), used H subsequently 2O (6 * 10mL) washings.Organic phase MgSO 4Drying is filtered and is concentrated.Obtain required cyanopyridine (2.66g, 81%) by silica gel chromatography purifying (30% EtOAc/Hex).LCMS[M+H]=295.2
Step e: at 9: 1 EtOH/H 2Contain among the O (10mL) in steps the cyanopyridine of D (add in the 150mg, solution 0.51mmol) solid KOH (286mg, 5.1mmol).This solution is heated to 85 ℃ reaches 16 hours.The cooling reaction mixture is used 3N HCl acidifying, and (3 * 30mL) extract with EtOAc.The organic layer that merges salt water washing (1 * 50mL), use MgSO 4Drying is filtered and is concentrated.Resistates need not to be further purified and can use.In the solution of this carboxylic acid in DCM (5mL), add the hydrazides intermediate compound I (140mg, 0.48mmol), DIPEA (186mg, 1.43mmol) and bop reagent (212mg, 0.48mmol).This reaction was at room temperature stirred 1 hour and directly was carried in silicagel column (50%EtOAc/ hexane) and obtains required hydrazides (150mg, 53%) LCMS[M+H]=589.1
Step F: to the hydrazides of step e (150mg, 0.255mmol) 1, add in the 2-dichloroethane solution (3mL) Burgess reagent (182mg, 0.764mmol).With this solution 140 ℃ of following microwave treatment 8 minutes.Obtain required  diazole (90mg, 62%) by the silica gel chromatography purifying.LCMS[M+H]=571.1?H?NMR(CDCl 3)δ7.29-7.19(m,5H),7.05(m,2H),6.79(s,1H),5.23(bs,1H),3.54-3.41(m,4H),3.38(s,3H),3.17(s,3H),2.92(s,3H),1.76(m,3H),1.41(br?s,9H),1.24-1.13(m,1H),0.50(m,2H),0.26(m,2H)。
Step G: (add TFA (1.5mL) in the 50mg, DCM 0.083mmol) (5mL) solution to the  of step e diazole.This solution stirring 30 is placed and concentrated.Obtain 50mg (100%) required compound with the ether development, it is a tfa salt.LCMS[M+H]=471.1
1H?NMR(CD 3OD)δ7.41(s,1H),7.37-7.25(m,3H),7.05(m,2H),6.75(s,1H),3.54-3.41(m,4H),3.38(s,3H),3.17(s,3H),2.94(s,3H),1.86(m,3H),1.24-1.13(m,1H),0.50(m,2H),0.26(m,2H)。
Following abbreviation is applicable in full:
Me: methyl
Bu: butyl
I-Bu: isobutyl-
T-Bu: the tertiary butyl
Et: ethyl
Pr: propyl group
I-Pr: sec.-propyl
Ar: aryl
Ph: phenyl
Py: pyridine
Ac: ethanoyl
EDC: ethyl-3-(3-dimethylaminopropyl)-carbodiimide
HOAt:1-hydroxyl-7-azepine-benzotriazole
HOBt: hydroxybenzotriazole
DMF:N, N '-dimethyl formamide
THF: tetrahydrofuran (THF)
DMSO: dimethyl sulfoxide (DMSO)
EDTA: ethylenediamine tetraacetic acid (EDTA)
Boc: tert-butoxycarbonyl
BOP: benzotriazole-1-base oxygen base-three (dimethylamino) 
CHAPS:3-[(3-chloro-acid amide base propyl group) Dimethyl Ammonium]-2-hydroxyl-1-and sulfonate
TEA: triethylamine
TFA: trifluoroacetic acid
NaHMDS: hexamethyldisilazane
The NCS:N-chloro-succinimide
DCE: ethylene dichloride
DIPEA: diisopropylethylamine
DCM: methylene dichloride
DIC:N, N '-DIC
DCA:1, the 2-ethylene dichloride
DMA:N, the N-N,N-DIMETHYLACETAMIDE
Aq: moisture
Rt: room temperature
HPLC: high efficiency liquid chromatography
Though the present invention describes and explanation with reference to some embodiments, it will be understood by those skilled in the art that many adaptations, change, modification, the method that substitutes, omits and add and scheme can obtain not breaking away under aim of the present invention and the scope.So, of the present invention be service by claims limit and these type of claims as much as possible from broadly carrying out reasonable dismissal.

Claims (22)

1. the compound of formula (I):
Wherein: X is selected from group
Figure A2005800118430002C2
R wherein 11And R 12Be independently selected from
(a) hydrogen,
(b)-C 1-10Alkyl,
(c) C 2-10Alkenyl,
(d) C 2-10Alkynyl group,
(e)-C 3-12Cycloalkyl and
(f) be selected from the aryl of phenyl and naphthyl;
This alkyl wherein, cycloalkyl, alkenyl, alkynyl group or aryl are not substituted or are replaced by one or more following groups
(i) halogen,
(ii)-OH,
(iii)-CN,
(iv)-C 1-10Alkyl,
(v)-C 3-12Cycloalkyl and
(vi)-O-C 1-10Alkyl,
Y 1Be N, and Y 2With Y4 be respectively CH, or
Y 2Be N, and Y 1And Y 3Be respectively CH, or
Y 3Be N, and Y 1And Y 2Be respectively CH;
A is selected from group
(1) hydrogen,
(2)-C 1-10Alkyl and
(3)-C 2-10Alkenyl,
Wherein this alkyl or alkenyl are not replace or replaced by one or more following groups
(a) halogen,
(b)-C 3-12Cycloalkyl,
(c)-OH,
(d)-CN,
(e)-O-C 1-10Alkyl,
(f) phenyl, or
(g) heteroaryl is selected from pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidyl, pyrryl, tetrazyl, furyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl,  azoles base, different  azoles base, thiazolyl, the  di azoly, indyl, quinolyl, isoquinolyl, benzimidazolyl-, benzofuryl, indoles alkynyl (indynyl) and benzoxazol base
With this phenyl and heteroaryl be not replace or replaced by one or more following groups
(i) halogen,
(ii)-OH,
(iii)-CN,
(iv)-O-C 1-10Alkyl,
(v)-C 1-10Alkyl, or
(vi)-C 3-12Cycloalkyl;
Q is-C 0-3Alkyl, wherein this alkyl is not replace or replaced by one or more following groups
(1) halogen,
(2)-C 3-12Cycloalkyl,
(3)-OH,
(4)-CN,
(5)-O-C 1-10Alkyl and
(6)-C 1-10Alkyl;
R 1Be the aryl that (1) is selected from phenyl and naphthyl,
(2) heteroaryl is selected from pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidyl, pyrryl, tetrazyl, furyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl,  azoles base, different  azoles base, thiazolyl, the  di azoly, indyl, quinolyl, isoquinolyl, benzimidazolyl-, benzofuryl, indoles alkynyl and benzoxazol base
(3)-C 1-10Alkyl and
(4) C 3-8Cycloalkyl, this cycloalkyl randomly with C 6-10It is aryl-condensed,
This alkyl wherein, cycloalkyl, aryl or heteroaryl are not replace or replaced by one or more following groups
(a) halogen,
(b)-C 1-10Alkyl, wherein this alkyl is not replace or replaced by halogen,
(c)-OH,
(d)-CN,
(e)-O-C 1-10Alkyl,
(f)-C 3-12Cycloalkyl and
(g)-NR 8R 9
R 2Be selected from
(1)-OH and
(2)-NR 8R 9, R wherein 8And R 9Be selected from
(a) hydrogen,
(b) C 1-10Alkyl and
(c) C0-6 alkyl-C 6-10Aryl,
Or work as R 2Be NR 8R 9, and R 8When being hydrogen with A, Q then, R 1And R 9Formation-CH can link together 2CH 2CH 2-, work as R 2Be NR 8R 9The time, then Q, R 1Can link together with A constitutes 4 or 5 carbon alkyl chains, and wherein the one or more carbon atoms in the alkyl chain can be used N, O or S atom, or SO 2Group substitutes;
R 4Be selected from group
(1)-C 1-10Alkyl, or
(2)-C 3-12Cycloalkyl, wherein this alkyl and cycloalkyl are not substituted or are replaced by one or more following groups
(a) halogen,
(b)-OH,
(c)-CN,
(d)-O-C 1-10Alkyl,
(e)-C 1-10Alkyl,
(f)-C 3-12Cycloalkyl,
(g) be selected from the aryl of phenyl and naphthyl, or
(h) heteroaryl is selected from pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidyl, pyrryl, tetrazyl, furyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl,  azoles base, different  azoles base, thiazolyl, the  di azoly, indyl, quinolyl, isoquinolyl, benzimidazolyl-, benzofuryl, indoles alkynyl and benzoxazol base
With this aryl and heteroaryl be not replace or replaced by one or more following groups
(i) halogen,
(ii)-OH,
(iii)-CN,
(iv)-O-C 1-10Alkyl,
(v)-C 3-12Cycloalkyl, or
(vi)-C 1-10Alkyl;
R 7Be selected from group
(1) hydrogen,
(2)-C 1-10Alkyl and
(3) be selected from the aryl of phenyl and naphthyl;
Wherein this alkyl and aryl are not replace or replaced by one or more following groups
(a) halogen,
(b)-OH,
(c)-CN,
(d)-O-C 1-10Alkyl,
(e)-C 3-12Cycloalkyl,
(f) be selected from the aryl of phenyl and naphthyl, or
(g) heteroaryl is selected from pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidyl, pyrryl, tetrazyl, furyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl,  azoles base, different  azoles base, thiazolyl,  di azoly, indyl, quinolyl, isoquinolyl, benzimidazolyl-, benzofuryl, indoles alkynyl and benzoxazol base;
This cycloalkyl wherein, aryl or heteroaryl are not replace or replaced by one or more following groups
(i) halogen,
(ii)-OH,
(iii)-CN,
(iv)-O-C 1-10Alkyl,
(v)-C 3-12Cycloalkyl, or
(vi) be selected from the aryl of phenyl and naphthyl;
Or R 4And R 7Formation-CH can link to each other 2CH 2CH 2-;
R 5And R 6Be independently selected from
(1) hydrogen,
(2)-C 1-10Alkyl,
(3)-C 2-10Alkenyl,
(4)-C 2-10Alkynyl group and
(5)-C 1-10Alkyl-C 3-12Cycloalkyl;
This alkyl wherein, cycloalkyl, alkenyl or alkynyl group are not replace or replaced by one or more following groups
(a) halogen,
(b)-OH,
(c)-CN,
(d)-C 1-10Alkyl
(e)-C 3-12Cycloalkyl,
(f)-O-C 1-10Alkyl,
(g) heteroaryl is selected from pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidyl, pyrryl, tetrazyl, furyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl,  azoles base, different  azoles base, thiazolyl, the  di azoly, indyl, quinolyl, isoquinolyl, benzimidazolyl-, benzofuryl, indoles alkynyl and benzoxazol base
Wherein this heteroaryl can not replace or be replaced by halogen;
(h) phenyl, or
(i)-NR 8R 9
Or R 5And R 6The continuous 4-6 person of formation encircles with the nitrogen-atoms that they connected, and it does not replace or is replaced by one or more following groups
(a)-C 1-10Alkyl,
(b)-C 3-12Cycloalkyl,
(c)-(CH 2) positive phenyl,
(d)-C 2-10Alkenyl, or
(e)-C 2-10Alkynyl group, this alkyl wherein, alkenyl and alkynyl group are not replace or replaced by one or more following groups
(i) halogen,
(ii)-OH,
(iii)-CN,
(iv)-O-C 1-10Alkyl, or
(v)-C 3-12Cycloalkyl;
With this cycloalkyl and phenyl be not replace or replaced by one or more following groups
(i) halogen,
(ii)-C 1-10Alkyl,
(iii)-OH,
(iv)-CN,
(v)-C 3-12Cycloalkyl, or
(vi)-O-C 1-10Alkyl;
N is 0,1,2,3 or 4;
With its pharmaceutically acceptable salt, with and each enantiomer and diastereomer.
2. the compound of claim 1, wherein Y 1Be N and Y 2And Y 3Be respectively CH.
3. the compound of claim 1, wherein R 1Be that phenyl and Q are CH 2
4. the compound of claim 1, wherein R 2Be-NR 8R 9
5. the compound of claim 1, wherein A is C 1-6Alkyl.
6. the compound of claim 1, wherein R 4And R 7Be C 1-10Alkyl.
7. the compound of claim 1, wherein X is that the  diazole is selected from
8. the compound of claim 1, wherein X is that the  azoles is selected from
Figure A2005800118430008C2
9. the compound of claim 1, wherein X is that thiazole is selected from
Figure A2005800118430008C3
10. the compound of claim 1, it is the compound of formula (II)
A wherein, X, Y 1, Y 2, Y 3, Q, R 1, R 2, R 4, R 5And R 7As the definition of above-mentioned claim 1, and its pharmaceutically acceptable salt, with and each enantiomer and diastereomer.
11. the compound of claim 10, wherein Y 1Be N and Y 2And Y 3Be respectively CH.
12. the compound of claim 10, wherein R 1Be that phenyl and Q are CH 2
13. the compound of claim 10, wherein R 2Be NR 8R 9
14. the compound of claim 10, wherein X is that the  diazole is selected from
Figure A2005800118430009C1
15. the compound of claim 10, wherein X is that the  azoles is selected from
Figure A2005800118430009C2
16. the compound of claim 10, wherein X is that thiazole is selected from
Figure A2005800118430009C3
17. the compound of claim 10, wherein R 5Be hydrogen or C 1-10Alkyl, wherein this C 1-10Alkyl does not replace or is replaced by one or more following groups:
(1) halogen,
(2)-OH,
(3)-CN,
(4) phenyl,
(5)-OC 1-10Alkyl, or
(6)-NR 8R 9
18. the compound of claim 17, wherein R 5Be C 1-10Alkyl, wherein this C 1-10Alkyl does not replace or is replaced by one or more halogens.
19. the compound of formula (III)
Figure A2005800118430010C1
A wherein, X, Q, R 1, R 2, R 4, R 5, R 6And R 7As the definition of above-mentioned claim 1, with and pharmaceutically acceptable salt and its each enantiomer and diastereomer.
20. the compound of claim 1, it is selected from group
Figure A2005800118430010C2
Figure A2005800118430012C1
Figure A2005800118430013C1
Figure A2005800118430014C1
Figure A2005800118430015C1
Figure A2005800118430016C1
Figure A2005800118430018C1
Figure A2005800118430019C1
21. pharmaceutical composition wherein contains compound or its pharmaceutically acceptable salt and the pharmaceutical acceptable carrier of the claim 1 for the treatment of significant quantity.
22. the method for a treatment Alzheimer in its patient of needs comprises compound or its pharmaceutically acceptable salt to the claim 1 of this patient's administering therapeutic significant quantity.
CNA2005800118430A 2004-04-20 2005-04-20 2, 4, 6-substituted pyridyl derivative compounds useful as beta-secretase inhibitors for the treatment of Alzheimer's disease Pending CN1942467A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US56361204P 2004-04-20 2004-04-20
US60/563,612 2004-04-20
US60/630,539 2004-11-23
US60/653,037 2005-02-15

Publications (1)

Publication Number Publication Date
CN1942467A true CN1942467A (en) 2007-04-04

Family

ID=37959788

Family Applications (1)

Application Number Title Priority Date Filing Date
CNA2005800118430A Pending CN1942467A (en) 2004-04-20 2005-04-20 2, 4, 6-substituted pyridyl derivative compounds useful as beta-secretase inhibitors for the treatment of Alzheimer's disease

Country Status (1)

Country Link
CN (1) CN1942467A (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105348112A (en) * 2015-11-06 2016-02-24 上海皓元化学科技有限公司 Technological synthesis method of 1-amino cyclopropyl acetylene
CN116106455A (en) * 2023-03-28 2023-05-12 上海赛默罗生物科技有限公司 Method for detecting related substances in substituted nicotinamide medicines

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105348112A (en) * 2015-11-06 2016-02-24 上海皓元化学科技有限公司 Technological synthesis method of 1-amino cyclopropyl acetylene
CN116106455A (en) * 2023-03-28 2023-05-12 上海赛默罗生物科技有限公司 Method for detecting related substances in substituted nicotinamide medicines

Similar Documents

Publication Publication Date Title
CN1942457A (en) 1,3,5-substituted phenyl derivative compounds useful as beta-secretase inhibitors for the treatment of alzheimer's disease
CN1049434C (en) Aromatic Amidine derivatives and salts thereof
CN1313450C (en) Tachykinin receptor antagonists
CN1036520C (en) 1,4-diazepine derivative and its pharmaceutical use
CN1434805A (en) Selective neurokinin antagonists
CN1923812A (en) New compounds
CN101068545A (en) Spiropiperidine compounds useful as beta-secretase inhibitors for the treatment of alzhermer disease
CN1285588C (en) Farnesyl protein transferase inhibitors
CN1898199A (en) Phenylamide and pyridylamide beta-secretase inhibitors for the treatment of alzheimer's disease
CN1378544A (en) Substituted azoles compounds
CN1753672A (en) 4,5-diarylthiazole derivatives as cb-1 ligands
CN1255401C (en) Aroyl-piperazine derivatives, their preparation and their use as tachykinin antagonists
CN1913886A (en) Heterocyclic aspartyl protease inhibitors
CN1918137A (en) Tetrazole compounds and their use as metabotropic glutamate receptor antagonists
CN1088917A (en) The benzylamino nitrogen containing non-aromatic heterocycles that replaces
CN1993363A (en) 1,3-disubstituted heteroaryl nmda/nr2b antagonists
CN101061119A (en) Macrocyclic aminopyridyl beta-secretase inhibitors for the treatment of alzheimer's disease
CN1221534C (en) Substited 8-anylquinoline phosphodiesterase-4 inhibitors
CN1064042C (en) Intermediate of benzazepine derivative
CN1296354C (en) Pharmaceutically active pyrrolidine derivatives as BAX inhibitors
CN1111528C (en) Bridged piperazine derivatives as neurokinin antagonists
CN1138764C (en) Isoxazole derivatives
CN1639155A (en) Pyrrolidine oxadiazole- and thiadiazole derivatives
CN1942467A (en) 2, 4, 6-substituted pyridyl derivative compounds useful as beta-secretase inhibitors for the treatment of Alzheimer's disease
CN1064350C (en) Aryl and heteroaryl alkoxynaphtalene derivatives

Legal Events

Date Code Title Description
C06 Publication
PB01 Publication
C10 Entry into substantive examination
SE01 Entry into force of request for substantive examination
C02 Deemed withdrawal of patent application after publication (patent law 2001)
WD01 Invention patent application deemed withdrawn after publication

Open date: 20070404