CN110590747A - Compound, preparation method, pharmaceutical composition and application thereof - Google Patents

Compound, preparation method, pharmaceutical composition and application thereof Download PDF

Info

Publication number
CN110590747A
CN110590747A CN201810601884.4A CN201810601884A CN110590747A CN 110590747 A CN110590747 A CN 110590747A CN 201810601884 A CN201810601884 A CN 201810601884A CN 110590747 A CN110590747 A CN 110590747A
Authority
CN
China
Prior art keywords
compound
group
alkyl
substituted
heteroaryl
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.)
Granted
Application number
CN201810601884.4A
Other languages
Chinese (zh)
Other versions
CN110590747B (en
Inventor
杨磊
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.)
Longtaishen Pharmaceutical Technology Nanjing Co Ltd
Original Assignee
Longtaishen Pharmaceutical Technology Nanjing Co Ltd
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 Longtaishen Pharmaceutical Technology Nanjing Co Ltd filed Critical Longtaishen Pharmaceutical Technology Nanjing Co Ltd
Priority to CN201810601884.4A priority Critical patent/CN110590747B/en
Publication of CN110590747A publication Critical patent/CN110590747A/en
Application granted granted Critical
Publication of CN110590747B publication Critical patent/CN110590747B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Abstract

The invention discloses a compound, a preparation method, a pharmaceutical composition and application thereof. The compound I, stereoisomer or pharmaceutically acceptable salt thereof can be used as CDK7 kinase inhibitor, has high inhibitory activity, and can be used for treating various malignant tumors.

Description

Compound, preparation method, pharmaceutical composition and application thereof
Technical Field
The invention relates to a compound, a preparation method, a pharmaceutical composition and application thereof.
Background
Cyclin-dependent protein kinases (CDKs), a group of serine/threonine protein kinases, act as key regulators of the cell cycle in concert with cyclin. 20 CDK family members have been found in mammals. The CDK7 has been proved to have close relation with the occurrence and development of various malignant tumors, and the CDK7 inhibitor has the potential of treating various malignant tumors such as leukemia, breast cancer and the like.
Disclosure of Invention
The invention provides a compound, a preparation method, a pharmaceutical composition and application thereof. The compound can be used as a CDK7 kinase inhibitor, has high inhibitory activity and can be used for treating various malignant tumors.
The invention mainly solves the technical problems through the following technical scheme.
The invention provides a compound shown in a general formula I, a stereoisomer or pharmaceutically acceptable salt thereof,
wherein ring A is C3-C20Cycloalkyl (e.g. C)3-C6Cycloalkyl, preferably cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl), C6-C20Aryl (e.g. C)6-C14Aryl, preferably phenyl, naphthyl, anthryl or phenanthryl), C1-C20Heteroaryl (e.g. C)1-C10Heteroaryl, preferably pyridyl) or C2-C20A heterocycloalkyl group;
ring B is C6-C20Aryl (e.g. C)6-C14Aryl, preferably phenyl, naphthyl, anthryl or phenanthryl), C3-C20Cycloalkyl (e.g. C)3-C6Cycloalkyl, preferably cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl) or C1-C20Heteroaryl (e.g. C)1-C10Heteroaryl, preferably pyridyl);
R1is H or C1-C10Alkyl (e.g. C)1-C4Alkyl, preferably methyl, ethyl, propyl or butyl);
R2is H, C1-C10Alkyl (e.g. C)1-C4Alkyl, preferably methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl or tert-butyl), C2-C10Alkenyl (e.g. C)2-C4Alkenyl, preferably )、C2-C10Alkynyl (e.g. C)2-C4Alkynyl, preferably )、C6-C20Aryl or C3-C20Cycloalkyl (e.g. C)3-C6Cycloalkyl, preferably cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl);
R3is H, C6-C20Aryl or C1-C20A heteroaryl group;
or, R2And R3Taken together with the atoms to which they are attached form a 5-7 membered heteroaryl, a 5-7 membered cycloalkyl or a 5-7 membered heterocycloalkyl; the heteroatoms in said 5-7 membered heteroaryl or said 5-7 membered heterocycloalkyl are selected from N, O and S, the number of heteroatoms is 1-4;
m is 0, 1 or 2;
R4at any position, each R4Independently of one another, halogen, C1-C10Alkyl, hydroxy, C1-C10Alkoxy, amino, nitro, cyano, or C substituted by halogen1-C10An alkyl group;
R5is composed ofWherein n1 is 0 or 1; r5aAnd R5bIndependently is H or C1-C10An alkyl group; r5cIs H, halogen, C1-C10Alkyl radical, C1-C10Alkoxy, or substituted or unsubstituted C2-C20Heterocycloalkyl, said substituted C2-C20The substituent in the heterocycloalkyl group being C1-C10An alkyl group; said substituted or unsubstituted C2-C20Heterocyclic ringsCarbon atoms of alkyl radicals withConnecting;
q is 0, 1,2 or 3;
R6at any position, each R6Independently is halogen, substituted or unsubstituted C1-C10Alkyl, substituted or unsubstituted C1-C10Alkoxy, hydroxy, amino, nitro or cyano; said substituted C1-C10Alkyl or said substituted C1-C10Substituents in alkoxy are meant to be substituted by one or more (1-6, e.g. 1-3) of the following groups: wherein each R isaEach RbAnd each RcIndependently is H or C1-C10Alkyl, or Ra、RbAnd the heteroatoms connected with the heterocyclic group form a 4-8-membered heterocyclic group, wherein the 4-8-membered heterocyclic group refers to a 4-8-membered heterocyclic group (for example, the heteroatoms are selected from N, O and S) with 1-4 heteroatoms);
When R is5cIs H, halogen, C1-C10Alkyl or C1-C10When alkoxy, q is 1,2 or 3; each R6Independently is substituted C1-C10Alkyl or substituted C1-C10An alkoxy group;
L1is composed ofOr is absent; rlaAnd RlbIndependently H, C1-C10Alkyl or C substituted by halogen1-C10An alkyl group; or RlaAnd RlbTogether with the carbon atom to which they are attached form C3-C6A cycloalkyl group;the alkyl group of (a),The amino terminus of (a) is linked to the ring A; l is1Wherein, the carbon marked by the letter is S configuration chiral carbon, R configuration chiral carbon or achiral carbon;
L2is composed ofOr is absent;
wherein, said C1-C20Heteroaryl means a C having 1 to 4 heteroatoms selected from N, O and S1-C20A heteroaryl group; said C2-C20Heterocycloalkyl means a C having 1 to 4 heteroatoms selected from N, O and S2-C20A heterocycloalkyl group.
In a preferred embodiment of the invention, in the compounds of the formula I, R5cWherein said substituted or unsubstituted C2-C20The heterocycloalkyl structure is preferably as follows:wherein N is 1,2, 3, 4 or 5, and Z is C, O or N; when Z is O or N, R55Or R56Is absent; r51、R52、R53、R54、R55And R56Independently is H or C1-C10An alkyl group.
In a preferred embodiment of the invention, in the compounds of the formula I, R5cPreferably substituted C2-C20A heterocycloalkyl group.
In a preferred embodiment of the invention, in the compounds of the formula I, R5cPreferably substituted C2-C20Heterocycloalkyl, said substituent preferably being C1-C10Alkyl radical。
In a preferred embodiment of the invention, in the compounds of the formula I, R5cPreferably substituted C2-C20Heterocycloalkyl, the heteroatom being N, said substituent preferably being on N.
In a preferred embodiment of the invention, in the compounds of formula I, when R is5cIs substituted or unsubstituted C2-C20When it is heterocycloalkyl, said "C2-C20Heterocycloalkyl "is preferred
In a preferred embodiment of the invention, in the compounds of the formula I, R5cPreference is given to(e.g. in)、(e.g. in)、(e.g. in ) Or(e.g. in). Wherein, the carbon marked by the letter is S configuration chiral carbon, R configuration chiral carbon or non-chiral carbonAnd (4) carbon.
In a preferred embodiment of the invention, when R is6Is substituted C1-C10Alkyl or substituted C1-C10An alkoxy group; said substituted C1-C10Alkyl or said substituted C1-C10Substituents in alkoxy radicalsWhen the substitution is carried out,in, RaAnd RbIs C1-C10An alkyl group; wherein R isaAnd RbThe same or different.
In a preferred embodiment of the invention, when L1Is composed of(e.g. in) When the current is over; rlaAnd RlbOne is H and the other is C1-C10An alkyl group; l is1In the formula (I), the carbon marked by the letter is S configuration chiral carbon, R configuration chiral carbon or achiral carbon.
In a preferred embodiment of the invention, when L1Is composed of(e.g. in) And R islaAnd RlbOne is H and the other is C1-C10When it is alkyl; l is1In (b), the carbon marked with x is an R configuration chiral carbon.
In a preferred embodiment of the invention, when L1Is composed of(e.g. in) When the current is over; rlaAnd RlbOne is H and the other is C1-C10An alkyl group; l is1In (b), the carbon marked with a is an S-configuration chiral carbon.
In a preferred embodiment of the present invention, in the compound of formula I,
ring A is C6-C20Aryl (e.g. C)6-C14Aryl, preferably phenyl, naphthyl, anthryl or phenanthryl);
ring B is C6-C20Aryl (e.g. C)6-C14Aryl, preferably phenyl, naphthyl, anthryl or phenanthryl) or C1-C20Heteroaryl (e.g. C)1-C10Heteroaryl, preferably pyridyl);
R1is H;
R2is C1-C10Alkyl (e.g. C)1-C4Alkyl, preferably methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl or tert-butyl), C2-C10Alkenyl (e.g. C)2-C4Alkenyl, preferably )、C2-C10Alkynyl (e.g. C)2-C4Alkynyl, preferably) Or C3-C20Cycloalkyl (e.g. C)3-C6Cycloalkyl, preferably cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl);
R3is H;
m is 0;
R5is composed ofWherein R is5cIs H, or substituted or unsubstituted C2-C20Heterocycloalkyl (e.g. phenyl)Wherein, the carbon marked by the letter is S configuration chiral carbon, R configuration chiral carbon or achiral carbon);
q is 0 or 1; r6In any position, R6Is substituted C1-C10Alkyl or substituted C1-C10An alkoxy group; said substituted C1-C10Alkyl or said substituted C1-C10Substituents in alkoxy are meant to be substituted by one or more (1-6, e.g. 1-3) of the following groups: wherein each R isaEach RbAnd each RcIndependently is H or C1-C10Alkyl, or Ra、RbAnd the heteroatom to which they are attached, together form a 4-8 membered heterocyclic group (e.g.);
L1Is composed of(e.g. in);RlaAnd RlbIndependently H, C1-C10Alkyl or C substituted by halogen1-C10An alkyl group; l is1Wherein, the carbon marked by the letter is S configuration chiral carbon, R configuration chiral carbon or achiral carbon;
L2is absent.
In a preferred embodiment of the present invention, in the compound of formula I,
ring A is C6-C20Aryl (e.g. C)6-C14Aryl, preferably phenyl, naphthyl, anthryl or phenanthryl);
ring B is C6-C20Aryl (e.g. C)6-C14Aryl, preferably phenyl, naphthyl, anthryl or phenanthryl) or C1-C20Heteroaryl (e.g. C)1-C10Heteroaryl, preferably pyridyl);
R1is H;
R2is C3-C20Cycloalkyl (e.g. C)3-C6Cycloalkyl, preferably cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl);
R3is H;
m is 0;
R5is composed ofWherein R is5cIs substituted or unsubstituted C2-C20Heterocycloalkyl (e.g. phenyl)Wherein, the carbon marked by the letter is S configuration chiral carbon, R configuration chiral carbon or achiral carbon);
q is 0;
L1is composed of(e.g. in);RlaAnd RlbOne is H and the other is C1-C10An alkyl group; l is1Wherein, the carbon marked by the letter is S configuration chiral carbon, R configuration chiral carbon or achiral carbon;
L2is absent.
In a preferred embodiment of the present invention, the compound represented by the general formula I or a salt thereof is preferably any one of the following compounds: wherein, the carbon marked by the letter is S configuration chiral carbon, R configuration chiral carbon or achiral carbon;
a compound 11-1-P2 is prepared by chiral resolution of a compound 11-1-5 shown as follows by using an SFC device:
the resolution conditions and methods of the chiral resolution:
stationary phase (Column) chiral pak IC-H Daicel chemical Industries, Ltd,250 x30 mm i.d.,5 um; mobile phase A (Mobile phase A) Supercritical CO2Mobile phase B (mobile phase B) Ethanol (0.1% NH)3H2O); a: B60: 40at50ml/min (flow rate); column Temperature 38 deg.C; nozle Pressure 100 Bar; nozle temp. 60 deg.C; evaporator temp. 20 deg.C; the temperature of the Trimmer Temp is 25 ℃; wave length is UV 220 nm; rt 6.08 min.
A compound 11-3-P2 is prepared by chiral resolution of a compound 11-3-5 shown as the following by SFC equipment;
the conditions and methods of the chiral resolution:
stationary phase (Column) chiral pak IC-H Daicel chemical Industries, Ltd,250 x30 mm i.d.,5 um; mobile phase A (Mobile phase A) Supercritical CO2Mobile phase B (mobile phase B) Ethanol (0.1% NH)3H2O);A:B=60:40at50ml/min (flow rate); column Temperature 38 deg.C; nozle Pressure 100 Bar; nozle temp. 60 deg.C; evaporator temp. 20 deg.C; the temperature of the Trimmer Temp is 25 ℃; wave length is UV 220 nm; rt is 6.883 min.
The invention also provides a preparation method of the compound shown in the general formula I, which comprises the following steps: in a solvent, under the protection of gas and the action of alkali and a palladium catalyst, carrying out the reaction shown as the following on the compound shown as the general formula I-A and the compound shown as the general formula I-B;
wherein, in the compound shown in the general formula I-A, X is halogen (such as F, Cl, Br or I), M isWherein R isb1And Rb2Independently is hydroxy or C1-C10Alkoxy, or Rb1And Rb2Together with boron atoms (B) to formL2Absent, the remaining individual letter and radical definitions are as previously described.
In the preparation method of the compound shown in the general formula I, the reaction method and conditions are conventional in the field. The following conditions are preferred in the present invention:
the solvent is preferably an organic solvent or a mixed solvent of an organic solvent and water. The organic solvent is preferably an ether solvent, such as 1, 4-dioxane. When the solvent is a mixed solvent of an organic solvent and water, the amounts of the organic solvent and water are not particularly limited as long as the reaction is not affected, and the volume ratio of the organic solvent to the water is preferably 5: 1. The amount of the solvent to be used is not particularly limited as long as the reaction proceeds.
The gas is preferably an inert gas such as argon.
The base may be a base conventional in the art for such reactions, preferably an inorganic strong base, such as cesium carbonate. The base may be used in an amount conventional in such reactions in the art, and is preferably present in a molar ratio of 3:1 to the compound of formula I-A.
The palladium catalyst may be a palladium catalyst conventional for such reactions in the art, preferably Pd (dppf) Cl2. The palladium catalyst may be used in an amount conventional in such reactions in the art, and its molar ratio to the compound represented by formula I-A is preferably 0.1: 1.
The amount of the compound of formula I-A and the compound of formula I-B may be the amount conventionally used in such reactions in the art, and the molar ratio of the two is preferably 1: 1.2.
The temperature of the reaction may be a temperature conventional in the art for such reactions, preferably 100 ℃.
The progress of the reaction can be monitored by detection methods conventional in the art (e.g., TLC, GC, HNMR, or HPLC, etc.), preferably as the end point of the reaction when the disappearance of the compound of formula I-A and the compound of formula I-B is detected. The reaction time is preferably 1.5 hours.
After the reaction is finished, the reaction can be treated by adopting a post-treatment method which is conventional in the field.
In a preferred embodiment of the invention, in the compounds of formula I, R is1When the compound is H, the compound can be prepared by the following method, wherein the method comprises the following steps: in a solvent, carrying out deprotection reaction on a compound shown as a general formula I-C as shown in the specification;
wherein R is1Is H, Q is an amino protecting group (e.g., Boc or SEM); the remaining letter and radical definitions are as described above.
In the deprotection reaction, the solvent may be a solvent conventional in the art for such reactions, preferably a halogenated hydrocarbon solvent such as dichloromethane. The amount of the solvent to be used is not particularly limited as long as the reaction proceeds. The deprotection reaction is preferably carried out under the action of an acid. The acid is preferably an organic acid, such as trifluoroacetic acid. The temperature of the deprotection reaction may be a temperature conventional in the art for such reactions, preferably room temperature. The progress of the deprotection reaction can be monitored by detection methods conventional in the art (e.g., TLC, GC, HNMR, or HPLC, etc.), preferably as the end point of the reaction when the disappearance of the compound of formula I-C is detected. The reaction time is preferably 0.5 to 1 hour.
After the deprotection reaction is finished, the post-treatment method which is conventional in the field can be adopted for treatment. For example, spin-drying the solvent.
In the present invention, if the crude compound is obtained after the post-treatment, the crude compound can be separated and purified by conventional means such as preparative HPLC, preparative TLC or recrystallization.
In the deprotection reaction, when Q is an amino protecting group, and the amino protecting group can be removed under an acidic condition, the salt of the compound shown in the general formula I can be prepared. For example, when Q is Boc or SEM, it can be removed under the action of trifluoroacetic acid, and accordingly, the trifluoroacetate salt of the compound represented by the general formula I can be prepared.
In the invention, the compound shown in the general formula I can also be prepared into a compound shown in the general formula I, and the compound shown in the general formula I can be further prepared into other compounds shown in the general formula I by peripheral modification by adopting a conventional method in the field.
The invention also provides a pharmaceutical composition which comprises the compound shown in the general formula I, a stereoisomer or pharmaceutically acceptable salt thereof and pharmaceutically acceptable auxiliary materials.
The invention also provides an application of the compound shown in the formula I, a stereoisomer or a pharmaceutically acceptable salt thereof in preparing CDK kinase inhibitors.
In the present invention, said C3-C20Cycloalkyl is preferably C3-C6Cycloalkyl radical, said C3-C6Cycloalkyl is preferably cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl.
In the present invention, said C6-C20Aryl is preferably C6-C14Aryl is preferably phenyl, naphthyl, anthryl or phenanthryl.
In the present invention, said C1-C20The heteroaryl group is preferably selected from N, O and S, C having 1-4 heteroatoms1-C10A heteroaryl group. Said C1-C10Heteroaryl is preferably acridine, carbazole, cinnoline, carboline, quinoxaline, imidazole, pyrazole, pyrrole, indole, indoline, benzotriazole, benzimidazole, furan, thiophene, isothiazole, benzothiophene, dihydrobenzothiophene, benzofuran, isobenzofuran, benzoxazole, benzofurazan, benzopyrazole, quinoline, isoindole, isoquinoline, oxazole, oxadiazole, isoxazole, indole, pyrazine, pyridopyridine, tetrazolopyridine, pyridazine, pyridine, naphthyropyrimidine, pyrimidine, pyrrole, tetrazole, thiadiazole, thiazole, thiophene, triazole, quinazoline, tetrahydroquinoline, dihydrobenzimidazole, dihydrobenzofuran, dihydrobenzoxazole or dihydroquinoline.
In the present invention, said C2-C20The heterocycloalkyl group is preferably selected from N, O and S, C having 1 to 4 heteroatoms2-C10A heterocycloalkyl group. Said C2-C10The heterocycloalkyl group is preferably an oxazolinyl, oxetanyl, pyranyl, tetrahydropyranyl, azetidinyl, 1, 4-dioxanyl, piperazinyl, piperidinyl, pyrrolidinyl, morpholinyl, thiomorpholinyl, dihydrofuranyl, dihydroimidazolyl, dihydroindolyl, dihydroisoxazolyl, dihydroisothiazolyl, dihydrooxadiazolyl, dihydrooxazolyl, dihydropyrazinyl, dihydropyrazolyl, dihydropyridinyl, dihydropyrimidinyl, dihydropyrrolyl, dihydrotetrazolyl, dihydrothiadiazolyl, dihydrothiazolyl, dihydrothienyl, dihydrotriazolyl, dihydroazetidinyl, tetrahydrofuranyl or tetrahydrothienyl group.
In the present invention, said C1-C10Alkyl is preferably C1-C4An alkyl group. Said C1-C4The alkyl group is preferably a methyl group, an ethyl group, an n-propyl group, an isopropyl group, an n-butyl group, a tert-butyl group or an isobutyl group.
In the present invention, the 5-to 7-membered heteroaryl group is preferably a pyrrolyl group, a furyl group, a thienyl group, a pyrazolyl group, an imidazolyl group, an oxazolyl group, a thiazolyl group, an isoxazolyl group, a pyridyl group, a pyridazinyl group, a pyrimidinyl group or a pyrazinyl group.
In the present invention, the 5-to 7-membered cycloalkyl group is preferably cyclopentyl, cyclohexyl or cycloheptyl.
In the present invention, the 5-to 7-membered heterocycloalkyl group is preferably a pyranyl group, a tetrahydropyranyl group, a tetrahydrofuranyl group, a piperidyl group, a 1, 4-dioxanyl group, a piperazinyl group, a piperidyl group, a pyrrolidinyl group, a morpholinyl group, a thiomorpholinyl group, a dihydrofuranyl group, a dihydroimidazolyl group, a dihydroisoxazolyl group, a dihydroisothiazolyl group, a dihydrooxadiazolyl group, a dihydrooxazolyl group, a dihydropyrazinyl group, a dihydropyrazolyl group, a dihydropyridinyl group, a dihydropyrimidyl group, a dihydropyrrolyl group, a dihydrotetrazolyl group, a dihydrothiadiazolyl group, a dihydrothiazolyl group, a dihydrothienyl group, a dihydrotriazolyl group, a.
In the present invention, the halogen is preferably F, Cl, Br or I.
In the present invention, said C1-C10Alkoxy is preferably C1-C4An alkoxy group. Said C1-C4The alkoxy group is preferably methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, tert-butoxy or isobutoxy.
In the present invention, said C substituted by halogen1-C10In the alkyl group, the halogens are the same or different and are preferably F, Cl, Br or I. Said C substituted by halogen1-C10Alkyl is preferably C substituted by halogen1-C4Alkyl, for example trifluoromethyl.
In the present invention, said substituted or unsubstituted C2-C20In heterocycloalkyl radicals C2-C20Heterocycloalkyl groups are as defined above. R5cWherein said substituted or unsubstituted C2-C20The heterocycloalkyl structure is preferably as follows:wherein Z is C or N, and when Z is N, R55Or R56Is absent; r51、R52、R53、R54、R55And R56Independently is H or C1-C10An alkyl group; wherein C is1-C10Alkyl is as defined above.
The above preferred conditions can be arbitrarily combined to obtain preferred embodiments of the present invention without departing from the common general knowledge in the art.
The reagents and starting materials used in the present invention are commercially available.
In the present invention, room temperature means 10 to 30 ℃.
The positive progress effects of the invention are as follows: the CDK7 kinase inhibitor has high inhibitory activity and can be used for treating various malignant tumors.
Detailed Description
The invention is further illustrated by the following examples, which are not intended to limit the scope of the invention. The experimental methods without specifying specific conditions in the following examples were selected according to the conventional methods and conditions, or according to the commercial instructions.
In the following examples, abbreviations are explained:
DCM: dichloromethane; boc2O: di-tert-butyl dicarbonate; PE: petroleum ether; EA: ethyl acetate; rf: a ratio shift value; Dess-Martin: dess-martin oxidizer; DMF: n, N-dimethylformamide; DIEA: n, N-diisopropylethylamine; ACN: acetonitrile; LiHMDS: bis-trimethylsilyl amido lithium; THF: tetrahydrofuran; a dioxane: 1, 4-dioxane; TFA: trifluoroacetic acid; MeOH: methanol; prep-TLC: preparing thin-layer chromatography; DCE: 1, 2-dichloroethane; room temperature: 10-30 ℃; rt: room temperature; HATU: 2- (7-benzotriazole oxide) -N, N, N ', N' -tetramethyluronium hexafluorophosphate; prep-HPLC: preparing a high-performance liquid phase; b is2Pin2: pinacol ester diborate; SEM: (trimethylsilyl) ethoxymethyl; boc: tert-butoxycarbonyl group.
In the following examples, room temperature means 10-30 ℃; overnight means 8-15 hours, e.g., 12 hours; eq means equivalent; solvent ratio such as PE/EA refers to the volume ratio.
Example 1
(1) Preparation of Compound 4b
Compound 4a (5.0g,49.5mmol,1.0eq) was dissolved in DCM (50mL) and Boc was added portionwise at 0 deg.C2O (11.75g,54.45mmol,1.1eq), 0.5h at 0 ℃ and TLC showed the starting material was reacted. The reaction mixture was washed with saturated brine, dried over anhydrous sodium sulfate, and subjected to spin-drying to obtain compound 4b (8.5g, 85%). TLC PE/EA 4/1, I2,Rf(Compound 4a) ═ 0.1, Rf(compound 4b) ═ 0.3.
(2) Preparation of Compound 4
Compound 4b (8.5g,42.3mmol,1.0eq) was dissolved in DCM (150mL), Dess-Martin (19.7g,46.5mmol,1.1eq) was added in portions at 0 ℃ and then reacted at 0 ℃ for 1h, after warming to room temperature for 1h, TLC showed the starting material was reacted. The reaction was poured slowly into saturated NaHCO3In the solution, the organic phase is separated and saturated Na is used for the organic phase2SO3The solution was washed with brine, dried over anhydrous sodium sulfate, and subjected to spin-drying to obtain compound 4(5.0g, 62%). TLC PE/EA 4/1, I2,Rf(Compound 4b) ═ 0.3, Rf(compound 4) ═ 0.4.
(3) Preparation of Compound 3
Will (COCl)2(9.85mL,114.79mmol,1.5eq) was added dropwise at room temperature to a solution of Compound 2(15.0g,76.53mmol,1.0eq) in DCM (100mL), then 2 drops of DMF were added dropwise and reacted at room temperature for 2 h. The DCM was spun down and the residue was taken up three more times with DCM and spun dry. The residue was dissolved in DCM and slowly added dropwise to Compound 1(8.87g,51.27 m) at 0 deg.Cmol,0.67eq) and DIEA (26.36g,204mmol,2.67eq) in ACN (60mL), reacted at 0 ℃ for 1h after addition, and then warmed to room temperature overnight. Most of the solvent was removed by evaporation, EA was added and dissolved, and the mixture was washed with saturated brine, dried over anhydrous sodium sulfate, and subjected to spin-drying to obtain compound 3(5.5g, 30%). TLC PE/EA 1/1, 254nm, Rf(Compound 1) ═ 0.7, Rf(compound 3) ═ 0.2.
(4) Preparation of Compound 5
LiHMDS (12.8mL,1M,12.8mmol,1.5eq) was added dropwise to a THF (30mL) solution of compound 3(3.0g,8.54mmol,1.0eq) at-78 deg.C, the reaction was allowed to react at-78 deg.C for 1h, warmed to 0 deg.C for 1h, and then a THF solution of compound 4(2.5g,12.8mmol,1.5eq) was added dropwise to the system at 0 deg.C, allowed to react at 0 deg.C for 1h, and warmed to room temperature overnight. Saturated NH for reaction solution4After quenching with Cl solution, EA was added, washed with brine, dried over anhydrous sodium sulfate, and spin-dried, then applied to a column (PE/EA/DCM: 10/1/1-4/1/1) to obtain compound 5(2.0g, 59%). TLC PE/EA 4/1, 254nm, Rf(Compound 3) ═ 0.2, Rf(compound 5) ═ 0.7.
(5) Preparation of Compound 11-2-7
Mixing compound 5(200mg,0.5mmol,1.0eq), compound 6(291mg,0.6mmol,1.2eq), Cs2CO3(494mg,1.5mmol,3.0eq),Pd(dppf)Cl2(37mg, 0.05mmol,0.1eq) was added to the dioxane/H2In O (5mL/1mL), the mixture is heated to 100 ℃ under the protection of argon for reaction for 1.5h, and TLC shows that the reaction is finished. After the reaction was cooled to room temperature, EA (20mL) was added, washed with saturated brine, dried over anhydrous sodium sulfate, and spin-dried to give the crude product. The crude was dissolved in DCM (2mL), TFA (0.5mL) was added at RT and stirred for 1h at RT, and the reaction was monitored by LCMS. After the DCM was suspended, the residue was dissolved in water (10mL) and the organic phase was discarded after three back extractions of DCM. NaHCO for aqueous phase3The solid was adjusted to pH 7-8 and extracted three times with DCM. The organic phases were combined and dried over anhydrous sodium sulfate and the crude product obtained after spin drying was purified by prep-TLC (DCM/MeOH ═ 6/1 as developing solvent) to give compound 11-2-7(5mg, 2%).
TLC:DCM/MeOH)=6/1,254nm,Rf(Compound 5) ═ 0.9, Rf(compound 11-2-7) ═ 0.3. LCMS, M +1 is 471.3.1H NMR(400MHz,CD3OD)δ8.57(d,J=2.0Hz,1H),8.25(d,J=8.8Hz,1H),8.03(dd,J=8.6,2.4Hz,1H),7.66(s,1H),7.52(d,J=6.8Hz,1H),7.46-7.37(m,2H),6.99-6.94(m,1H),6.32(d,J=15.3Hz,1H),6.11(s,1H),3.91-3.88(m,1H),3.79-3.69(m,1H),3.10-2.99(m,1H),2.93-2.91(m,1H),2.19-2.17(m,1H),2.06-2.00(m,2H),1.87-1.78(m,3H),1.55(d,J=7.0Hz,3H),1.00-0.82(m,4H),0.69-0.66(m,2H).
Example 2
(1) Preparation of Compound 2
TFA (1mL) was added to a solution of compound 5(350mg,0.88mmol,1.0eq) in DCM (5mL) at room temperature and reacted for 1h at room temperature, TLC indicated complete reaction. With saturated NaHCO3The pH of the solution was adjusted to about 7.5, the organic phase was separated, the aqueous phase was extracted with DCM (2 × 10mL), the organic phases were combined, dried over anhydrous sodium sulfate and dried to give compound 2(260mg, 99%). TLC PE/EA 4/1, 254nm, Rf(compound 5) ═ 0.5, DCM/MeOH = 10/1, 254nm, Rf(compound 2) ═ 0.2.
(2) Preparation of Compound 3
Compound 2(260mg,0.88mmol,1.0eq) was dissolved in DCE (10mL) and HCHO (712mg, 37% wt in H) was added at room temperature2O,8.8mmol,10.0eq) and anhydrous MgSO4(1g) Stirred at room temperature for 5min, then NaBH (OAc)3(277mg,1.32mmol,1.5eq) was added to the reaction system in portions at 0 ℃ to react at 0 ℃ for 1h, then warmed to room temperature to react for 0.5h, TLC showed the reaction was complete. Filtration, cake washing with DCM, spin drying of the filtrate and column chromatography (DCM/MeOH-100/1-40/1) afforded compound 3(200mg, 73%). TLC DCM/MeOH-10/1, 254nm, Rf(Compound 2) ═ 0.2, Rf(compound 3) ═ 0.3.
(3) Preparation of Compound 11-1-5
Mixing compound 3(150mg,0.48mmol,1.0eq), compound 6(279mg,0.58mmol,1.2eq) and Cs2CO3(470mg,1.45mmol,3.0eq) was dissolved in dioxane (10mL) and water (2mL), Pd (dppf) Cl was added2(35mg,0.048mmol,0.1eq), argon replacement, heating to 100 ℃ for 1.5h, TLC showed the reaction was complete. After the reaction was cooled to room temperature, EA (20mL) was added, washed with saturated brine, dried over anhydrous sodium sulfate, and then spin-dried to obtain the crude product. The crude product was dissolved in DCM (2mL), TFA (0.5mL) was added at RT and stirred for 1h at RT, and LC-MS monitored for completion of the reaction. The DCM was suspended dry and dissolved in water (10mL), back extracted 3 times with MTBE and the organic phase discarded. NaHCO for aqueous phase3The solid was adjusted to pH 7-8 and extracted three times with DCM, the organic phases were combined and dried over anhydrous sodium sulphate and spun to give crude 200mg, 100mg of which was purified by prep-TLC (DCM/MeOH-6/1 as developing solvent) to give compound 11-1-5(25mg, 20%).
TLC:DCM/MeOH=5/1,254nm,Rf(Compound 3) ═ 0.6, Rf(compound 11-1-5) ═ 0.3. LCMS M + 1-485.3.1H NMR(400MHz,DMSO-d6)δ12.03(s,1H),10.76(s,1H),10.42(s,1H),8.63(d,J=2.4Hz,1H),8.29(d,J=8.8Hz,1H),8.08(dd,J=8.8,2.4Hz,1H),7.71(s,1H),7.57(d,J=7.6Hz,1H),7.47-7.33(m,2H),6.71-6.70(m,1H),6.46(d,J=15.6Hz,1H),6.15(s,1H),3.97-3.86(m,1H),3.05(s,1H),2.76(s,1H),2.22(s,3H),2.01(s,1H),1.86-1.67(m,3H),1.59(s,1H),1.43(d,J=6.8Hz,3H),1.24(s,1H),0.88(d,J=8.4Hz,2H),0.62(d,J=3.2Hz,2H).
Example 3
(1) Preparation of Compound 4b-1
Compound 4a-1(5.0g,49.5mmol,1.0eq) was dissolved in DCM (50mL) and Boc was added portionwise at 0 deg.C2O (11.75g,54.45mmol,1.1eq), 0 ℃ for 0.5h, TLC showed the starting material was reacted. The reaction mixture was washed with saturated brine, dried over sodium sulfate, and spin-dried to obtain compound 4b-1(8.5g, 85%) through a column (PE/EA ═ 10/1). TLC PE/EA 4/1, I2,Rf(Compound 4a-1) ═ 0.1, Rf(compound 4b-1) ═ 0.3.
(2) Preparation of Compound 4-1
Compound 4b-1(8.5g,42.3mmol,1.0eq) was dissolved in DCM (150mL) and Dess-Martin (19.7g,46.5mmol,1.1eq) was added in portions at 0 deg.C, reacted at 0 deg.C for 1h, then warmed to room temperature for 1h and TLC showed the starting material was reacted. The reaction was poured slowly into saturated NaHCO3Separating organic phase from the solution, and adding saturated Na2SO3The solution was washed, washed with brine, dried over sodium sulfate, and spin-dried to obtain compound 4-1(7.0g, 82%) through a column (PE/EA ═ 10/1). TLC PE/EA 4/1, I2,Rf(Compound 4b-1) ═ 0.3, Rf(compound 4-1) ═ 0.4.
(3) Preparation of Compound 3
Will (COCl)2(6.6mL,76.5mmol,1.5eq) was added dropwise to a solution of Compound 2(10.0g,51mmol,1.0eq) in DCM (80mL) at RT, then 2 drops of DMF were added dropwise and the reaction was carried out for 2-3h at RT. The DCM was spun off, taken up several times with DCM and then dissolved with DCM, and compound 1(5.92g,34.1mmol,0.67eq) and DIEA (17.6g,136.2mmol,2.67eq) were added slowly dropwise to a solution of compound 1 (60mL) in acetonitrile at 0 deg.C, reacted for 1h at 0 deg.C and then allowed to warm to room temperature overnight. Removing most of the solvent by spinning, adding EA to dissolve the rest, washing with saturated saline solution, drying with sodium sulfate, spin-drying,chromatography (PE/EA ═ 3/1) afforded compound 3(3.0g, 25%). TLC PE/EA 1/1, 254nm, Rf(Compound 1) ═ 0.7, Rf(compound 3) ═ 0.2.
(4) Preparation of Compound 5-1
LiHMDS (12.8mL,1M,12.8mmol,1.5eq) was added dropwise to a THF (30mL) solution of compound 3(3.0g,8.54mmol,1.0eq) at-78 deg.C, the reaction was allowed to react at-78 deg.C for 1h, warmed to 0 deg.C for 1h, and then a THF solution of compound 4-1(2.5g,12.8mmol,1.5eq) was added dropwise to the system at 0 deg.C for 1h, and then warmed to room temperature overnight. With saturated NH4The Cl solution was quenched, EA was added, washed with brine, dried over sodium sulfate, spun dry, and column filtered (PE/EA/DCM: 10/1/1, 4/1/1) to give compound 5-1(2.0g, 59%). TLC PE/EA 4/1, 254nm, Rf(Compound 3) ═ 0.2, Rf(compound 5-1) ═ 0.7.
(5) Preparation of Compound 11-4-7
Mixing 5-1(137mg,0.35mmol,1.0eq), 6(200mg,0.42mmol,1.2eq), Cs2CO3(339mg,1.0mmol,3.0eq),Pd(dppf)Cl2(20mg,0.03mmol,0.08eq) was added to the dioxane/H2Reacting with O (5mL/1mL) under argon atmosphere at 120 deg.C for 2h, separating with EA and water, washing the organic phase with water and saturated saline solution, drying with sodium sulfate, rotary evaporating to dry, adding 3N dioxane hydrochloride solution, stirring at room temperature for 1h, adding NaHCO3The reaction was dried by rotary drying to pH greater than 7, EA was added, the solid was filtered off, the EA was dried and purified by prep-TLC (DCM/MeOH ═ 7:1 as developing solvent) to give compound 11-4-7(10mg, yield: 6%).
TLC:DCM/MeOH)=10/1,254nm,Rf(Compound 6) ═ 0.9, Rf(compound 11-4-7) ═ 0.3. LCMS M + 1-471.4.1H NMR(400MHz,DMSO-d6)δ12.02(br s,1H),10.79(s,1H),10.40(s,1H),8.62(d,J=2.0Hz,1H),8.27(d,J=8.8Hz,1H),8.07(dd,J=8.8,2.4Hz,1H),7.70(s,1H),7.56(d,J=7.6Hz,1H),7.44-7.36(m,2H),6.88-6.83(m,1H),6.49(d,J=15.2Hz,1H),6.12(s,1H),4.44(s,1H),3.92-3.86(m,2H),3.08-2.92(m,2H),2.10-1.95(m,2H),1.86-1.73(m,2H),1.60-1.52(m,1H),1.43(d,J=6.8Hz,3H),0.88-0.85(m,2H),0.63-0.59(m,2H).
Example 4
(1) Preparation of Compound 2-a
TFA (1mL) was added to a solution of compound 5-1(350mg,0.88mmol,1.0eq) in DCM (5mL) at room temperature and reacted for 1h at room temperature. TLC showed complete reaction, saturated NaHCO3Adjusting pH of the solution to about 7.5, separating to obtain organic phase, extracting the aqueous phase with DCM (2X 10mL), combining the organic phase and Na2SO4Drying and spin-drying gave compound 2-a (260mg, 99%). TLC PE/EA 4/1, 254nm, Rf(compound 5-1) ═ 0.5, DCM/MeOH = 10/1, 254nm, Rf(compound 2) ═ 0.2.
(2) Preparation of Compound 3
Compound 2-a (260mg,0.88mmol,1.0eq) was dissolved in DCE (10mL) and HCHO (712mg, 37% wt in H) was added at room temperature2O,8.8mmol,10.0eq) and anhydrous MgSO4(1g) Stirred at room temperature for 5min, then NaBH (OAc)3(277mg,1.32mmol,1.5eq) was added to the reaction system in portions at 0 ℃ and reacted at 0 ℃ for 1h, then warmed to room temperature and reacted for 0.5 h. TLC showed the reaction was complete, filtered, the filter cake was washed with DCM, the filtrate was spun dry and passed through a column (DCM/MeOH-100/1-40/1) to give compound 3-a (200mg, 73%). TLC DCM/MeOH-10/1, 254nm, Rf(Compound 2-a) ═ 0.2, Rf(compound 3-a) ═ 0.3.
(3) Preparation of Compound 11-3-5
Mixing the compound 3-a (200mg,0.64mmol,1.0eq), the compound 6(372mg,0.77mmol,1.2eq) and Cs2CO3(631mg,1.93mmol,3.0eq) was dissolved in dioxane (10mL) in water (2mL) and Pd (dppf) Cl was added2(47mg,0.064mmol,0.1eq), argon replacement, heating to 100 ℃ for 1.5h, and TLC showed the reaction was complete. After the reaction was cooled to room temperature, EA (20mL) was added, washed with saturated brine and anhydrous Na2SO4Drying and spin-drying to obtain a crude product. The crude product was dissolved in DCM (2mL), TFA (0.5mL) was added at room temperature and stirred at room temperature for 1h, LC-MS monitored reaction completion, DCM was spin-dried, then it was dissolved in water (10mL), back-extracted with DCM 2-3 times, then NaHCO3The solid was adjusted to pH 8, extracted several times with DCM (8 × 10mL), the organic phases combined, anhydrous Na2SO4Drying and spin-drying gave the crude product which was purified by prep-TLC (DCM/MeOH ═ 6/1 as developing solvent) to give compound 11-3-5(35mg, 11%).
TLC:DCM/MeOH=5/1,254nm,Rf(Compound 3-a) ═ 0.6, Rf(compound 11-3-5) ═ 0.3. LCMS M + 1-485.3.1H NMR(400MHz,DMSO-d6)δ12.02(s,1H),10.80(s,1H),10.40(s,1H),8.63(d,J=2.0Hz,1H),8.28(d,J=8.8Hz,1H),8.08(dd,J=8.8,2.4Hz,1H),7.70(s,1H),7.57(d,J=7.6Hz,1H),7.44-7.36(m,2H),6.75-6.71(m,1H),6.50(d,J=15.6Hz,1H),6.14(s,1H),3.93-3.91(m,1H),3.18(br s,1H),2.34(br s,3H),2.06(br s,1H),1.84-1.78(m,3H),1.66(s,1H),1.43(d,J=6.8Hz,3H),1.23(s,1H),0.88-0.86(m,2H),0.61-0.60(m,2H).
Example 5
(1) Preparation of Compound 2
LDA (2M in THF,315mL,0.63mol,3.0eq) was added dropwise to a solution of Compound 1(45.0g,0.21mol,1.0eq) in THF (500mL) at 0 deg.C under Ar protection. After the dropwise addition, the reaction was carried out at 0 ℃ for 30 min. Isoiodopropane (114g,0.67mol,3.2eq) was slowly added dropwise. Stirred at room temperature for 30 min. The reaction was quenched with hydrochloric acid. The reaction mixture was concentrated under reduced pressure to remove the solvent THF. The residue was extracted with EtOAc (3X 200mL), and the extracts were combined and washed with 2N HCl (2X 200 mL). The organic phase was extracted with 10% NaOH (2 × 300mL) and the aqueous phases were combined and adjusted to pH ≈ 7 with hydrochloric acid. EtOAc (3X 500mL) extraction and combination of extracts were washed with water (2X 500mL) and saturated brine (2X 500 mL). Dried over anhydrous sodium sulfate and spin-dried to give compound 2 as a pale yellow oil (45.0g, 84%).
(2) Preparation of Compound 4
Compound 2(2.3g,9.0mmol,2.0eq), compound 3(1.0g,4.5mmol,1.0eq), HATU (3.4g,9.0mmol,2.0eq) and DIEA (1.7g,13.4mmol,3.0eq) were dissolved in DMF (25 mL). The reaction was stirred at 60 ℃ for 16 h. Dilution with water, extraction with EtOAc, concentration of the extract under reduced pressure, and purification on silica gel (PE/EtOAc. 10/1) afforded compound 4(1.9g, 46%) as a colorless oil. TLC, PE/EtOAc 2:1, 254nm, Rf (compound 3) 0.1, and Rf (compound 4) 0.7.
(3) Preparation of Compound 5
Mixing compound 4(680mg,1.47mmol,1.0eq), B2Pin2(470mg,1.76mmol,1.2eq), KOAc (432mg,4.41mmol,3.0eq) and Pd (dppf) Cl2(52mg,0.07mmol,0.05eq) was added to dioxane (10 mL). Stirring and reacting for 6 hours at 100 ℃ under the protection of argon. The reaction was concentrated under reduced pressure and purified by column chromatography (PE/EtOAc 10:1) to give compound 5(674mg, 90%) as a colorless oil.
(4) Preparation of Compound 11-5
Mixing compound 5(140mg,0.27mmol,1.1eq), compound 6(78mg,0.25mmol,1.0eq), Cs2CO3(313mg,0.82mmol,3.0eq),Pd(dppf)Cl2(18mg,0.025mmol,0.1eq) in Dioxane/H2O (2mL/0.5 mL). Stirring and reacting for 2h at 100 ℃ under the protection of argon. The reaction was concentrated under reduced pressure, and the residue was diluted with EtOAc, washed with water and concentrated under reduced pressure. Obtained CH for solid2Cl2Dissolved (5mL), and TFA (1mL) was added dropwise. The reaction was stirred at room temperature for 1 h. Sat. NaHCO for reaction solution3Washing with anhydrous Na2SO4Drying, concentrating under reduced pressure, and purifying by crude prep-TLC (CH)2Cl2MeOH 10:1) purification followed by prep-HPLC (mobile phase: 0.1% TFA/H2O/CH3CN) to give compound 11-5 as a white solid, TFA salt (5.7mg, 4%).
LCMS:M+1=513.3。1H NMR(400MHz,DMSO-d6)δ11.05(s,1H),10.46(s,1H),9.99(br,1H),8.65(d,J=2.0Hz,1H),8.30(d,J=8.4Hz,1H),8.12(dd,J=8.8,2.4Hz,1H),7.70(s,1H),7.59(d,J=7.6Hz,1H),7.45-7.37(m,2H),6.85-6.79(m,1H),6.70(d,J=15.2Hz,1H),6.13(s,1H),4.11-4.02(m,1H),3.73-3.65(m,1H),3.37(d,J=10.8Hz,1H),3.18-3.10(m,1H),2.83(d,J=4.0Hz,3H),2.44-2.37(m,1H),2.33-2.28(m,1H),2.13-1.77(m,4H),1.00(d,J=6.4Hz,3H),0.90-0.84(m,2H),0.69(d,J=6.4Hz,3H),0.65-0.58(m,2H);19F NMR(376MHz,DMSO-d6):δ-74.05。
Example 6
Preparation of Compound 11-6
Intermediate compound 5(200mg,0.39mmol,1.0eq) of example 5, compound 2(122mg,0.39mmol,1.0eq), K2CO3(163mg,1.18mmol,3.0eq) and Pd (dppf) Cl2(29mg,0.04mmol,0.1eq) in dioxane/H2O (2mL/0.5 mL). Stirring and reacting for 6h at 80 ℃ under the protection of argon. The reaction was concentrated under reduced pressure, the residue was diluted with EtOAc and washed with water and anhydrous Na2SO4Drying, and concentrating under reduced pressure. Obtained CH for solid2Cl2Dissolved (5mL), and TFA (1mL) was added dropwise. The reaction was stirred at room temperature for 1 h. Sat. NaHCO for reaction solution3Washing with anhydrous Na2SO4Drying, concentrating under reduced pressure, and purifying by crude prep-TLC (CH)2Cl2MeOH 10:1) purification followed by prep-HPLC (mobile phase: 0.1% TFA/H2O/CH3CN) to give compound 11-6 as a white solid, TFA salt (35.7mg, 15%).
LCMS:M+1=513.4。1H NMR(400MHz,DMSO-d6)δ11.05(s,1H),10.47(s,1H),10.01(br,1H),8.65(d,J=2.0Hz,1H),8.30(d,J=8.8Hz,1H),8.12(dd,J=8.4,2.0Hz,1H),7.70(s,1H),7.59(d,J=7.6Hz,1H),7.45-7.37(m,2H),6.85-6.79(m,1H),6.70(d,J=15.2Hz,1H),6.13(s,1H),4.11-4.02(m,1H),3.73-3.65(m,1H),3.37(d,J=10.8Hz,1H),3.18-3.09(m,1H),2.83(d,J=4.4Hz,3H),2.44-2.38(m,1H),2.33-2.27(m,1H),2.12-1.77(m,4H),1.00(d,J=6.4Hz,3H),0.90-0.85(m,2H),0.69(d,J=6.8Hz,3H),0.65-0.59(m,2H)。19F NMR(376MHz,DMSO-d6):δ-74.39。
Example 7
(1) Preparation of Compound 2
Compound 1(540mg,1.82mmol,1.0eq) was dissolved in DCE (20 mL). Acetone (530mg,9.12mmol,5.0eq), NaBH (OAc) was added at room temperature3(773mg,3.65mmol,2.0eq), anhydrous MgSO4(1.0g) and acetic acid (1 drop). The reaction was carried out at room temperature for 2 h. Filtering the reaction solution, concentrating the filtrate under reduced pressure, and purifying with Column (CH)2Cl2MeOH: 100:1 to 30:1) gave compound 2 as a brown oil (187mg, 30%). TLC CH2Cl2:MeOH=20:1,254nm,Rf(Compound 1) ═ 0.4, Rf(compound 2) ═ 0.5.
(2) Preparation of Compounds 11-7
Compound 2(100mg,0.30mmol,1.0eq), compound 3(142mg,0.30mmol,1.0eq), K2CO3(123mg,0.89mmol,3.0eq) and Pd (dppf) Cl2(22mg,0.03mmol,0.1eq) in dioxane/H2O (6.0mL/1.5 mL). Stirring and reacting for 2h at 80 ℃ under the protection of argon. The reaction solution was concentrated under reduced pressure, diluted with EtOAc, washed with water and dried over Na2SO4Drying, and concentrating under reduced pressure. Obtained CH for solid2Cl2Dissolved (5mL), and TFA (1mL) was added dropwise. The reaction was stirred at room temperature for 1 h. The reaction was concentrated under reduced pressure and the crude prep-HPLC (mobile phase: 0.1% TFA/H2O/CH3CN) to give compound 11-7 as a white solid, TFA salt (19.91mg, 13%).
LCMS:M+1=513.5。1H NMR(400MHz,DMSO-d6)δ10.70(s,1H),10.41(s,1H),8.62(d,J=2.0Hz,1H),8.27(d,J=8.8Hz,1H),8.21(s,1H),8.08(dd,J=8.8,2.4Hz,1H),7.70(s,1H),7.56(d,J=7.6Hz,1H),7.42(t,J=7.6Hz,1H),7.37(d,J=7.6Hz,1H),6.77-6.71(m,1H),6.44(d,J=15.6Hz,1H),6.12(s,1H),3.93-3.88(m,1H),2.89-2.78(m,3H),2.54-2.51(m,1H),1.99-1.89(m,1H),1.85-1.78(m,1H),1.74-1.67(m,2H),1.58-1.50(m,1H),1.43(d,J=7.2Hz,3H),1.05(d,J=6.4Hz,3H),0.95(d,J=6.4Hz,3H),0.90-0.85(m,2H),0.63-0.59(m,2H)。19F NMR(376MHz,DMSO-d6):δ-73.41。
Example 8
(1) Preparation of Compound 2
Compound 1(351mg,1.19mmol,1.0eq) was dissolved in DCE (5 mL). Acetone (344mg,5.93mmol,5.0eq), NaBH (OAc) was added at room temperature3(502mg,2.37mmol,2.0eq), anhydrous MgSO4(500mg) and acetic acid (1 drop). The reaction was carried out at room temperature for 2 h. Adding CH into the reaction solution2Cl2Diluting, washing with water, and removing anhydrous Na2SO4Drying, concentrating under reduced pressure, and purifying with Column (CH)2Cl2MeOH: 50:1 to 20:1) gave compound 2 as a brown oil (78mg, 19%). TLC CH2Cl2:MeOH=20:1,254nm,Rf(Compound 2) ═ 0.4, Rf(compound 3) ═ 0.5.
(2) Preparation of Compounds 11-8
Compound 2(78mg,0.23mmol,1.0eq), compound 3 (E: (E))111mg,0.23mmol,1.0eq),K2CO3(96mg,0.69mmol,3.0 eq.) and Pd (dppf) Cl2(17mg,0.023mmol,0.1eq) in dioxane/H2O (4mL/1 mL). Stirring and reacting for 2h at 80 ℃ under the protection of argon. Filtering the reaction solution to obtain filtrate CH2Cl2Diluting, washing with water, and removing anhydrous Na2SO4Drying, and concentrating under reduced pressure. Obtained CH for solid2Cl2Dissolved (5mL), and TFA (1mL) was added dropwise. The reaction was stirred at room temperature for 1 h. The reaction was concentrated under reduced pressure and the crude prep-HPLC (mobile phase: 0.1% TFA/H2O/CH3CN) to yield compound 11-8 as a yellow solid, TFA salt (25.88mg, 22%).
LCMS:M+1=513.4。1H NMR(400MHz,DMSO-d6)δ11.00(s,1H),10.44(s,1H),9.80(br,1H),8.65(d,J=2.4Hz,1H),8.27(d,J=8.4Hz,1H),8.11(dd,J=8.8,2.4Hz,1H),7.70(s,1H),7.57(d,J=7.6Hz,1H),7.45-7.37(m,2H),6.91-6.85(m,1H),6.68(d,J=15.2Hz,1H),6.12(s,1H),4.39-4.31(m,1H),3.94-3.89(m,1H),3.59-3.54(m,1H),3.48-3.42(m,1H),3.31-3.22(m,1H),2.33-2.20(m,1H),2.05-1.98(m,2H),1.93-1.78(m,2H),1.44(d,J=6.8Hz,3H),1.30(dd,J=16.4,6.4Hz,6H),0.89-0.86(m,2H),0.63-0.61(m,2H)。19F NMR(376MHz,DMSO-d6):δ-74.54。
Example 9
(1) Preparation of Compound 3
At room temperature (COCl)2(6.6g,52.32mmol,1.8eq) was added dropwise to CH of Compound 2(6.8g,34.88mmol,1.2eq)2Cl2(50mL), 2 drops of DMF was added dropwise and the reaction was carried out at room temperature for 2 hours. CH (CH)2Cl2After removal of the residue by CH2Cl2(10mL) after dissolution was added slowly dropwise to a solution of compound 1(5.0g,29.07mmol,1.0eq) and DIEA (15.0g,116.27mmol,4.0eq) in MeCN (50mL) at 0 ℃. After the dropwise addition, the reaction was carried out at room temperature for 5 hours. The reaction was quenched with water. Adding CH2Cl2Diluted (100mL), washed with saturated brine (2X 100mL), dried over anhydrous sodium sulfate, and then applied to a column (PE/EA: 5/1) to obtain a yellow solidProduct 3(3.9g, 38%). TLC PE/EA 1/1, 254nm, Rf(Compound 1) ═ 0.7, Rf(compound 3) ═ 0.2.
(2) Preparation of Compound 5
Li HMDS (1M in THF,7.71mL,7.71mmol,1.5eq) was added dropwise to a solution of compound 3(1.8g,5.14mmol,1.0eq) in THF (20mL) at-78 deg.C under protection of Ar. The reaction solution reacts for 1h at-78 ℃, and then the temperature is raised to 0 ℃ for reaction for 1 h. A solution of Compound 4(1.5g,7.71mmol,1.5eq) in THF (5mL) was added dropwise at 0 deg.C. The reaction was stirred at room temperature overnight. Saturated NH for reaction4And (4) quenching the Cl solution. EtOAc (30mL) was added, washed with saturated brine (2 × 30mL), dried over anhydrous sodium sulfate, and then applied to a column (PE/EA: 4/1-2/1) to give compound 5(1.2g, 59%) as a pale yellow solid. TLC PE/EA 4/1, 254nm, Rf(Compound 3) ═ 0.2, Rf(compound 5) ═ 0.7.
(3) Preparation of Compound 6
TFA (2mL) was added dropwise to CH of Compound 5(1.2g,3.04mmol) at room temperature2Cl2(10mL) in solution. The reaction was carried out at room temperature for 1 hour. The reaction was concentrated under reduced pressure, and the residue was diluted with water (20mL) and then with saturated NaHCO3The pH of the solution is adjusted to be about 9. CH (CH)2Cl2(3X 20mL) and the combined organic phases are dried over anhydrous Na2SO4And (5) drying and spin-drying. The resulting solid was washed with (PE: EtOAc ═ 1:1) to give compound 6(500mg, 56%) as a white solid.
(4) Preparation of Compound 7
Compound 6(200mg,0.68mmol,1.0eq) was dissolved in DCE (5 mL). Aq.37% (wt%) HCHO (550mg,6.78mmol,10.0eq) and anhydrous MgSO were added at room temperature4(300 mg). Stir at room temperature for 10 min. AddingEntering NaBH (OAc)3(215mg,1.05mmol,1.5eq) at room temperature for 2 h. Concentrating the reaction solution under reduced pressure, and purifying with Column (CH)2Cl2MeOH ═ 10:1) to give compound 7(150mg, 72%) as a pale yellow solid.
(5) Preparation of Compounds 11-9
Mixing compound 7(50mg,0.16mmol,1.0eq), compound 8(93mg,0.19mmol,1.2eq) and K2CO3(67mg,0.49mmol,3.0eq) was dissolved in dioxane (5mL) and water (1 mL). Adding Pd (dppf) Cl2(12mg,0.02mmol,0.1 eq). Heating to 80 ℃ under the protection of argon and reacting for 2 h. Saturated NH for reaction4And (4) quenching the Cl solution. Adding CH to the reaction solution2Cl2(20mL), washed with saturated brine (2X 20mL), anhydrous Na2SO4And (5) drying and spin-drying. The resulting solid is dissolved in CH2Cl2To (5mL) was added TFA (1mL) dropwise. The reaction was stirred at room temperature for 1 h. The reaction was concentrated under reduced pressure, the residue was dissolved in water (10mL), saturated NaHCO3The pH is adjusted to be approximately equal to 9. CH (CH)2Cl2(2X 20mL) and the combined extracts were extracted with anhydrous Na2SO4Drying, spin-drying, and passing through Column (CH)2Cl2MeOH ═ 5: 1). The solid obtained was then subjected to a C18 column (MeCN/H)2O) to yield the product 11-9 as a white solid (12mg, 15%).
LCMS:[M+1]=484.4。1H NMR(400MHz,DMSO-d6)δ12.02(br,1H),10.41(s,1H),10.32(br,1H),7.76(d,J=8.4Hz,2H),7.65(s,1H),7.61(d,J=8.4Hz,2H),7.50(d,J=8.0Hz,1H),7.38(t,J=8.0Hz,1H),7.31(d,J=7.6Hz,1H),6.75-6.69(m,1H),6.42(d,J=13.6Hz,1H),6.13(s,1H),3.92-3.86(m,1H),2.50-2.38(m,5H),2.17-2.07(m,2H),1.91-1.71(m,4H),1.42(d,J=7.2Hz,3H),0.89-0.85(m,2H),0.63-0.59(m,2H)。
Example 10
(1) Preparation of Compound 3
LiHMDS (1M in THF,7.71mL,7.71mmol,1.5eq) was added dropwise to a solution of compound 1(1.8g,5.14mmol,1.0eq) in THF (20mL) at-78 deg.C under protection of Ar. The reaction solution reacts for 1h at-78 ℃, and then the temperature is raised to 0 ℃ for reaction for 1 h. A solution of Compound 2(1.5g,7.71mmol,1.5eq) in THF (5mL) was added dropwise at 0 deg.C. The reaction was stirred at room temperature overnight. Saturated NH for reaction4And (4) quenching the Cl solution. EtOAc (30mL) was added, washed with saturated brine (2 × 30mL), dried over anhydrous sodium sulfate, and then applied to a column (PE/EA: 4/1-2/1) to give compound 3(1.2g, 59%) as a pale yellow solid. TLC PE/EA 4/1, 254nm, Rf(Compound 1) ═ 0.2, Rf(compound 3) ═ 0.7.
(2) Preparation of Compound 4
TFA (2mL) was added dropwise to CH of Compound 3(1.2g,3.04mmol) at room temperature2Cl2(10mL) in solution. The reaction was carried out at room temperature for 1 hour. The reaction was concentrated under reduced pressure, and the residue was diluted with water (20mL) and then with saturated NaHCO3The pH of the solution is adjusted to be about 9. CH (CH)2Cl2(3X 20mL) and the combined organic phases are dried over anhydrous Na2SO4And (5) drying and spin-drying. The resulting solid was washed with (PE: EtOAc ═ 1:1) to give compound 4(600mg, 67%) as a white solid.
(3) Preparation of Compound 5
Compound 4(200mg,0.68mmol,1.0eq) was dissolved in DCE (5 mL). Aq.37% (wt%) HCHO (550mg,6.78mmol,10.0eq) and anhydrous MgSO were added at room temperature4(300 mg). Stir at room temperature for 10 min. Addition of NaBH (OAc)3(215mg,1.05mmol,1.5eq) at room temperature for 2 h. Concentrating the reaction solution under reduced pressure, and purifying with Column (CH)2Cl2MeOH ═ 10:1) to give compound 5(145mg, 69%) as a pale yellow solid.
(4) Preparation of Compounds 11-10
Mixing compound 5(75mg,0.24mmol,1.0eq), compound 6(140mg,0.29mmol,1.2eq) and Cs2CO3(237mg,0.73mmol,3.0eq) was dissolved in dioxane (5mL) and water (1 mL). Adding Pd (dppf) Cl2(18mg,0.02mmol,0.1 eq). Heating to 100 ℃ under the protection of argon and reacting for 2 h. Saturated NH for reaction4And (4) quenching the Cl solution. Adding CH to the reaction solution2Cl2(20mL), washed with saturated brine (2X 20mL), anhydrous Na2SO4And (5) drying and spin-drying. The resulting solid is dissolved in CH2Cl2To (5mL) was added TFA (1mL) dropwise. The reaction was stirred at room temperature for 1 h. The reaction was concentrated under reduced pressure, the residue was dissolved in water (10mL), saturated NaHCO3The pH is adjusted to be approximately equal to 9. CH (CH)2Cl2(2X 20mL) and the combined extracts were extracted with anhydrous Na2SO4Drying, spin-drying and then prep-TLC (CH)2Cl2MeOH ═ 10:1) purified to give the product 11-10(15mg, 13%) as a yellow solid.
LCMS:[M+1]=484.4。1H NMR(400MHz,DMSO-d6)δ12.03(br,1H),10.41(s,1H),10.28(br,1H),7.77(d,J=8.8Hz,2H),7.65(s,1H),7.61(d,J=8.8Hz,2H),7.50(d,J=7.6Hz,1H),7.38(t,J=7.6Hz,1H),7.32(d,J=7.2Hz,1H),6.72-6.66(m,1H),6.37(d,J=15.2Hz,1H),6.13(s,1H),3.92-3.86(m,1H),2.50-2.33(m,5H),2.10-1.96(m,2H),1.85-1.77(m,3H),1.67-1.60(m,1H),1.42(d,J=6.8Hz,3H),0.89-0.85(m,2H),0.63-0.59(m,2H)。
Example 11
(1) Preparation of Compound 3
Compound 1(768mg,2.48mmol,1.0eq), compound 2(1.47g,4.95mmol,2.0eq), K2CO3(1.03g,7.44mmol,3.0 eq.) and Pd (dppf) Cl2(181mg,0.025mmol,0.1eq) in dioxane/H2O (8mL/2 mL). Stirring and reacting for 16h at 80 ℃ under the protection of argon. The reaction was diluted with water (30mL) and extracted with EtOAc (3X 15 mL). Mixing the extractive solutions, anhydrous Na2SO4Drying, concentrating under reduced pressureConcentrating, purifying with Column (CH)2Cl2MeOH 50/1) yielded compound 3 as a yellow solid (630mg, 65%).
(2) Preparation of Compound 4
To a solution of compound 3(120mg,0.31mmol,1.0eq) in MeOH (10mL) was added dropwise 1M aq naoh (0.91mL,0.91mmol,3.0 eq). The reaction was stirred at room temperature overnight. The reaction was concentrated under reduced pressure to remove solvent MeOH. Adjusting the pH of the residual solution to 2-3 with 1M aq. HCl, and then adding CH2Cl2MeOH (10/1,3X 10mL) extraction. The combined extracts were washed with saturated brine (10mL) and anhydrous Na2SO4Dried and concentrated under reduced pressure to give compound 4 as a yellow solid (100mg, 87%).
(3) Preparation of Compound 5
Compound 4(100mg,0.264mmol,1.0eq), compound 5(134mg,0.528mmol,2.0eq), HATU (150mg,0.395mmol,1.5eq) and DIEA (102mg,0.791mmol,3.0eq) were dissolved in DMF (10 mL). The reaction was stirred overnight at50 ℃. Water was added for dilution and EtOAc extraction. Mixing the extractive solutions, washing with saturated saline, and adding anhydrous Na2SO4Drying and concentration under reduced pressure gave compound 5 as a yellow solid (150mg, 93%).
(4) Preparation of Compounds 11-13
To compound 6(150mg,0.24mmol,1.0eq) in CH2Cl2To the solution (3mL) was added TFA (3mL) dropwise. The reaction was stirred at room temperature overnight. The reaction was concentrated under reduced pressure and the crude prep-HPLC (mobile phase: 0.1% TFA/H2O/CH3CN) to give compound 11-13 as a pale yellow solid, TFA salt (32mg, 27%).
LCMS:M+1=485.4。1H NMR(400MHz,DMSO-d6)δ11.03(s,1H),10.57(s,1H),10.00(br,1H),8.67(d,J=2.0Hz,1H),8.30(d,J=8.8Hz,1H),8.12(dd,J=8.4,2.4Hz,1H),7.72(s,1H),7.59(d,J=7.6Hz,1H),7.46-7.39(m,2H),6.85-6.79(m,1H),6.68(d,J=15.2Hz,1H),6.55(s,1H),5.45(s,1H),5.04(s,1H),4.11-4.03(m,1H),3.97-3.92(m,1H),3.71-3.66(m,1H),3.17-3.12(m,1H),2.83(d,J=3.6Hz,3H),2.36-2.27(m,1H),2.14-2.07(m,1H),2.03-1.94(m,1H),1.98(s,3H),1.92-1.84(m,1H),1.46(d,J=6.8Hz,3H)。19F NMR(376MHz,DMSO-d6):δ-74.30。
Example 12
(1) Preparation of Compound 2
To a solution of compound 1(25.0g,0.22mol,1.0eq) in THF (250mL) under Ar protection was added NaH (60%, 9.7g,0.24mol,1.1eq) in portions. After stirring at room temperature for 10min SEMCl (44.2g,0.27mol,1.2eq) was added dropwise. Stir at room temperature overnight. Saturated NH for reaction4And (4) quenching by using a Cl aqueous solution. The reaction was diluted with EtOAc, washed with water and anhydrous Na2SO4Drying, concentration under reduced pressure and column purification (PE/EtOAc ═ 5/1) gave compound 2(36.3g, 67%) as a white solid.
(2) Preparation of Compound 3
LDA (2M in THF,90mL,179.5mmol,1.2eq) was added dropwise to a solution of Compound 2(36.3g,149.6mmol,1.0eq) in THF (400mL) at-78 deg.C under Ar protection. Stirring and reacting at-78 ℃ for 1h, and then dropwise adding I2(45.8g,179.5mmol,1.2eq) in THF. Stirred at room temperature for 2 h. Saturated Na for reaction2S2O3Aqueous solution and saturated NH4And (4) quenching by using a Cl aqueous solution. The reaction was diluted with EtOAc, washed with water and anhydrous Na2SO4Dried, concentrated under reduced pressure, and purified by column chromatography (PE/EtOAc ═ 10/1) to give compound 3(33.6g, 61%) as a yellow solid. TLC PE/EtOAc-5/1, 254nm, Rf(Compound 2) ═ 0.2, Rf(compound 3) ═ 0.5.
(3) Preparation of Compound 5
Compound 3(4.0g,10.83mmol,1.0eq), compound 4(1.82g,10.83mmol,1.0eq), K2CO3(4.49g,32.5mmol,3.0eq),Pd(dppf)Cl2(792mg,1.08mmol,0.1eq) in dioxane/H2O (24mL/6 mL). Stirring and reacting for 3 hours at 80 ℃ under the protection of argon. The reaction was concentrated under reduced pressure, the residue was diluted with EtOAc, washed with water, concentrated under reduced pressure, and purified on silica gel (PE/EtOAc ═ 60/1) to give compound 5(2.5g, 83%) as a yellow oil. TLC PE/EtOAc-8/1, 254nm, Rf(Compound 3) ═ 0.4, Rf(compound 5) ═ 0.5.
(4) Preparation of Compound 6
Compound 5(2.03g,7.17mmol,1.0eq), reduced iron powder (4.0g,71.73mmol,10.0eq) and ammonium chloride (7.67g,143.46mmol,20.0eq) were dissolved in ethanol (12mL) and water (6 mL). The reaction was stirred for 1h while heating to 70 ℃. Filtration and spin-drying of the filtrate gave compound 6 as a yellow oil (1.7g, 95%).
(5) Preparation of Compound 9
Compound 7(620mg,2.0mmol,1.0eq), compound 8(1.0g,3.6mmol,1.8eq), K2CO3(828mg,6.0mmol,3.0eq) and Pd (dppf) Cl2(146mg,0.02mmol,0.1eq) in dioxane/H2O (14mL/3 mL). Stirring and reacting for 16h at 80 ℃ under the protection of argon. The reaction was concentrated under reduced pressure, the residue was diluted with EtOAc and washed with water and anhydrous Na2SO4Drying, concentrating under reduced pressure, and purifying with Column (CH)2Cl2MeOH 200/1-20/1) gave compound 9 as a brown oil (310mg, 31%).
(6) Preparation of Compound 10
To a solution of compound 9(100mg,0.25mmol,1.0eq) in MeOH (2mL) was added dropwise a solution of NaOH (31mg,0.76mmol,3.0eq) in water (2 mL). The reaction was stirred at room temperature overnight. The reaction was concentrated under reduced pressure to remove solvent MeOH. The residue was adjusted to pH 5-6 with 2M aq. hcl and concentrated to give compound 10 as a brown oil (crude, 96mg, 100%).
(7) Preparation of Compound 11
Compound 10(96mg,0.25mmol,1.0eq), compound 6(77mg,0.30mmol,1.2eq), HATU (144mg,0.38mmol,1.5eq) and DIEA (98mg,0.76mmol,3.0eq) were dissolved in DMF (2 mL). The reaction was stirred at room temperature overnight. Diluting with water, extracting with EtOAc, and extracting with anhydrous Na2SO4Drying and concentration under reduced pressure gave compound 11 as a brown oil (crude, 155mg, 100%).
(8) Preparation of Compounds 11-14
To compound 11(155mg,0.25mmol,1.0eq) in CH2Cl2To the solution (2mL) was added TFA (2mL) dropwise. The reaction was stirred at room temperature overnight. The reaction was concentrated under reduced pressure and the crude prep-HPLC (mobile phase: 0.1% TFA/H2O/CH3CN) to give compound 11-14 as an off-white solid, TFA salt (27.58mg, 22%). LCMS M +1 ═ 485.4
1H NMR(400MHz,DMSO-d6)δ11.03(s,1H),10.56(s,1H),9.97(br,1H),8.66(d,J=2.0Hz,1H),8.28(d,J=8.4Hz,1H),8.11(dd,J=8.8,2.4Hz,1H),7.72(s,1H),7.60(d,J=7.6Hz,1H),7.46-7.38(m,2H),6.85-6.79(m,1H),6.70(d,J=15.2Hz,1H),6.55(s,1H),5.45(s,1H),5.03(s,1H),4.11-4.02(m,1H),3.97-3.91(m,1H),3.19-3.10(m,1H),2.82(d,J=4.4Hz,3H),2.71-2.66(m,1H),2.33-2.29(m,1H),2.14-2.07(m,1H),2.03-1.98(m,1H),1.98(s,3H),1.94-1.86(m,1H),1.46(d,J=7.2Hz,3H).19F NMR(376MHz,DMSO-d6):δ-73.78.
Example 13
(1) Preparation of Compound 3
Compound 1(180mg,0.422mmol,1.0eq), compound 2(140mg,0.633mmol,1.5eq), HATU (240mg,0.633mmol,1.5eq) and DIEA (163mg,1.266mmol,3.0eq) were dissolved in DMF (6 mL). The reaction was stirred overnight at50 ℃. Water was added for dilution and extracted with EtOAc (3X10 mL). Mixing the extractive solutions, washing with saturated saline, and adding anhydrous Na2SO4Dried and concentrated under reduced pressure to give compound 3(150mg, 53%) as a yellow solid.
(2) Preparation of Compounds 11-15
To compound 3(150mg,0.25mmol,1.0eq) in CH2Cl2To the solution (4mL) was added TFA (4mL) dropwise. The reaction was stirred at room temperature for 6 h. The reaction was concentrated under reduced pressure and the crude prep-HPLC (mobile phase: 0.1% TFA/H2O/CH3CN) to give compound 11-15 as a pale yellow solid, TFA salt (32mg, 27%).
LCMS:M+1=469.4.1H NMR(400MHz,DMSO-d6)δ11.03(s,1H),10.72(s,1H),9.94(br,1H),8.67(d,J=2.4Hz,1H),8.28(d,J=8.8Hz,1H),8.11(dd,J=8.8,2.4Hz,1H),7.71(s,1H),7.58(d,J=7.6Hz,1H),7.44(t,J=7.6Hz,1H),7.39(d,J=7.6Hz,1H),6.85-6.79(m,1H),6.70(d,J=15.2Hz,1H),6.63(s,1H),4.50(s,1H),4.11-4.03(m,1H),3.97-3.91(m,1H),3.69-3.65(m,1H),3.19-3.10(m,1H),2.83(d,J=4.0Hz,3H),2.36~2.28(m,1H),2.14-2.07(m,1H),2.03-1.97(m,1H),1.94-1.86(m,1H),1.46(d,J=6.8Hz,3H).19F NMR(376MHz,DMSO-d6):δ-73.82.
Example 14
(1) Preparation of Compound 3
To a solution of compound 1(3.0g,8.12mmol,1.0eq), compound 2(1.2g,12.19mmol,1.5eq) in THF (30mL) was added CuI (155mg,0.81mmol,0.1eq), PdCl2(PPh3)2(570mg,0.81mmol,0.1eq) and Et3N (3.3g,32.50mmol,4.0 eq). Stirring for 2h at room temperature under the protection of Ar. The reaction solution was diluted with EtOAc, washed with water and dried over Na2SO4Dried, concentrated under reduced pressure, and purified on silica gel column (PE/EtOAc ═ 20/1) to give compound 3(1.9g, 69%) as a yellow oil. TLC PE/EtOAc-10/1, 254nm, Rf(Compound 1) ═ 0.2, Rf(compound 3) ═ 0.6.
(2) Preparation of Compound 4
To a solution of compound 3(1.9g,5.6mmol,1.0eq) in MeOH (30mL) was added K2CO3(1.2g,8.4mmol,1.5 eq). Stirred at room temperature for 2 h. Reaction solution CH2Cl2Dilution, filtration and concentration of the filtrate under reduced pressure gave compound 4 as a yellow oil (1.24g, 83%).
(3) Preparation of Compound 5
Compound 4(1.24g,4.64mmol,1.0eq), reduced iron powder (2.6g,46.4mmol,10.0eq) and ammonium chloride (5.0g,92.8mmol,20.0eq) were dissolved in ethanol (20mL) and water (10 mL). The reaction was heated to 60 ℃ and stirred for 2 h. Filtration and spin-drying of the filtrate gave compound 5 as a yellow oil (1.0g, 91%).
(4) Preparation of Compounds 11-16
Compound 5(58mg,0.24mmol,1.2eq), compound 6(77mg,0.20mmol,1.0eq), HATU (116mg,0.30mmol,1.5eq) and DIEA (79mg,0.61mmol,3.0eq) were dissolved in DMF (2 mL). The reaction was stirred overnight at50 ℃. Diluting with water, EtOAcExtracted with anhydrous Na2SO4Drying, and concentrating under reduced pressure. Residue plus CH2Cl2Dissolved (2mL), and TFA (2mL) was added dropwise. The reaction was stirred at room temperature overnight. The reaction was concentrated under reduced pressure and the crude prep-HPLC (mobile phase: 0.1% TFA/H2O/CH3CN) to yield compound 11-16 as a yellow solid, TFA salt (5.23mg, 4%).
LCMS:M+1=469.4.1H NMR(400MHz,DMSO-d6)δ11.03(s,1H),10.72(s,1H),10.04(br,1H),8.66(d,J=2.4Hz,1H),8.28(d,J=8.8Hz,1H),8.12(dd,J=8.4,2.4Hz,1H),7.71(s,1H),7.59(d,J=7.6Hz,1H),7.44(t,J=7.6Hz,1H),7.39(d,J=7.6Hz,1H),6.87-6.81(m,1H),6.67(d,J=15.2Hz,1H),6.63(s,1H),4.50(s,1H),4.11-4.02(m,1H),3.71-3.64(m,1H),3.18-3.09(m,1H),2.82(d,J=4.8Hz,3H),2.70-2.66(m,1H),2.33-2.29(m,1H),2.12-2.05(m,1H),2.03-1.97(m,1H),1.94-1.86(m,1H),1.46(d,J=7.2Hz,3H).19F NMR(376MHz,DMSO-d6):δ-74.18.
Example 15
(1) Preparation of Compound 2b
Dissolving the compound 2a (5.0g,21.8mmol,1.0eq) in THF (60mL), cooling the reaction system to 0 deg.C, and adding BH3THF (26.2ml, 26.2mmol,1.2eq) was added to the reaction and allowed to warm to room temperature and stir for 2 h. Saturated NaHCO for reaction3Quench, EtOAc (2X30 mL) extraction, saturated brine wash, Na2SO4Drying and spin-drying to obtain compound 2b (4.8g, yield: 100%). TLC DCM/MeOH 10/1, I2,Rf(Compound 2a) ═ 0.2, Rf(compound 2b) ═ 0.4.
(2) Preparation of Compound 2
Dess-Matin (10.4g, 24.5mmol, 1.1eq) was added to Compound 2b (4.8g,22.3mmol,1.0eq) in DCM (60 mL). The reaction was carried out at room temperature for 2 h. The reaction solution was poured into saturated NaHCO3Extracted with EtOAc (2 × 30 mL). The organic phases were combined, washed with brine and Na2SO4Drying, spin-drying and column chromatography (PE/EA-8/1) gave compound 2(3.9g, yield: 82%). TLC PE/EA 2/1, I2,Rf(Compound 2b) ═ 0.2, Rf(compound 2) ═ 0.4.
(3) Preparation of Compound 3
LiHMDS (1M in THF,1.5mL,1.5mmol,1.5eq) was added to compound 1(352mg,1.0mmol,1.0eq) in THF (10mL) at-78 ℃. The reaction is carried out at minus 78 ℃ for 1h and at 0 ℃ for 1 h. A solution of compound 2(256mg,1.2mmol,1.2eq) in THF (3mL) was added to the reaction at 0 deg.C. The temperature is raised to room temperature and stirred for 2 h. Pouring the reaction solution into NH4In an aqueous Cl solution. EtOAc (3X 20mL) extraction, organic phase saturated brine wash, Na2SO4Dried, spun-dried and chromatographed (PE/EA/DCM-15/1/1) to give compound 3(280mg, yield: 68%). TLC PE/EA 1/1, 254nm, Rf(Compound 1) ═ 0.2, Rf(compound 3) ═ 0.6.
(4) Preparation of Compound 4
TFA (2mL) was added to Compound 3(440mg,1.08mmol,1.0eq) in DCM (5mL) and stirred at room temperature for 2 h. The solvent was spun off and the residue was dissolved in DCM. With saturated NaHCO3The solution was adjusted to pH 8. The organic phase was separated, washed with saturated brine and Na2SO4Drying and spin-drying to obtain compound 4(300mg, yield: 90%). TLC PE/EA 4/1, 254nm, Rf(compound 3) ═ 0.4, DCM/MeOH = 10/1, 254nm, Rf(compound 4) ═ 0.2.
(5) Preparation of Compound 5
Compound 4(300mg,0.97mmol,1.0eq)HCHO (37% in water, 728mg,9.7mmol,10.0eq) and MgSO4(2g) Dissolve in DCE (10 mL). Stir at room temperature for 10 min. Reacting NaBH (OAc)3(310mg,1.46mmol,1.5eq) was added to the reaction in portions at 0 ℃. The reaction is carried out for 1h at the temperature of 0 ℃ and 0.5h at the room temperature. The reaction was filtered, the filtrate was dried by spinning, and the filtrate was purified by column chromatography (DCM/MeOH: 50/1) to give compound 5(200mg, yield: 63%). TLC DCM/MeOH-10/1, 254nm, Rf(Compound 4) ═ 0.2, Rf(compound 5) ═ 0.3.
(6) Preparation of Compound 7
Compound 5(200mg,0.62mmol,1.0eq), compound 6(481.39mg,1.24mmol,2.0eq), K2CO3(257mg,1.86mmol,3.0eq) and Pd (dppf) Cl2(45mg,0.062mmol,0.1eq) was dissolved in dioxane (10mL) and H2O (2 mL). Heating to 80 ℃ for reaction for 2 h. The reaction was poured into EtOAc (20mL), washed with saturated brine, Na2SO4Dry, spin dry, and column pass (DCM/MeOH ═ 20/1) to give compound 7(60mg, yield: 32%). TLC DCM/MeOH-8/1, 254nm, Rf(Compound 5) ═ 0.4, Rf(compound 7) ═ 0.3.
(7) Preparation of Compounds 11-19
TFA (2mL) was added to Compound 7(90mg,0.15mmol,1.0eq) in DCM (5 mL). Stir at room temperature for 1 h. The solvent was spun off and the residue was dissolved in DCM. With saturated NaHCO3The solution was adjusted to pH 8. Separating the organic phase from Na2SO4And (5) drying and spin-drying. Crude prep-TLC (DCM/MeOH ═ 6/1) purified to give compounds 11-19(16mg, yield: 21%).
LCMS:M+1=499.4.1H NMR(400MHz,DMSO-d6)δ12.10(br s,1H),10.85(s,1H),10.49(s,1H),8.69(d,J=2.0Hz,1H),8.35(d,J=8.8Hz,1H),8.16-8.13(m,1H),7.78(s,1H),7.63(d,J=7.2Hz,1H),7.50-7.42(m,2H),6.83-6.77(m,1H),6.52(d,J=15.2Hz,1H),6.20(s,1H),4.00-3.95(m,1H),2.93(d,J=9.6Hz,1H),2.71(br s,1H),2.21(s,3H),2.11(br s,1H),1.84-1.24(m,13H),0.88-0.86(m,2H),0.62-0.60(m,2H).
Example 16
(1) Preparation of Compound 2b
Compound 2a (5.0g,21.8mmol,1.0eq), N, O-dimethylhydroxylamine hydrochloride (2.6g,26.2mmol,1.2eq), DIPEA (7.6mL,43.6mmol,1.5eq), EDCI (6.3g,32.7mmol,1.5eq) and HOBT (5.3g,32.7mmol,1.5eq) were dissolved in DCM (50 mL). Stirred at room temperature for 3 h. Washing the reaction solution with water, washing with saturated brine, and Na2SO4Drying, spin-drying and column chromatography (PE/EA-5/1) gave compound 2b (5.3g, yield: 89%). TLC PE/EA 1/1, I2,Rf(Compound 2a) ═ 0.2, Rf(compound 2b) ═ 0.4.
(2) Preparation of Compound 2
Mixing LiAlH4(1.25g,33.0mmol,1.5eq) was added portionwise to compound 2b (6.0g,22.0mmol,1.0eq) in THF (50mL) at 0 deg.C. The reaction is carried out for 1h at 0 ℃. The reaction solution was poured into ice water and filtered. The filtrate was extracted with EtOAc (3 × 100 mL). The organic phases were combined, washed with brine and Na2SO4Drying, spin-drying and column chromatography (PE/EA: 8/1) gave compound 2(4.4g, yield: 95%). TLC PE/EA 4/1, I2,Rf(Compound 2b) ═ 0.2, Rf(compound 2) ═ 0.4.
(3) Preparation of Compound 3
LiHMDS (1M in THF,2.13mL,2.13mmol,1.5eq) was added to compound 1(500mg,1.42mmol,1.0eq) in THF (10mL) at-78 ℃. The reaction is carried out at minus 78 ℃ for 1h and at 0 ℃ for 1 h. Compound 2(455mg,2.13mmol,1.5eq) was dissolved in THF (3mL)The solution was added to the reaction system at 0 ℃. The temperature is raised to room temperature and stirred for 2 h. Pouring the reaction solution into NH4In an aqueous Cl solution. EtOAc (3X 20mL) extraction, organic phase saturated brine wash, Na2SO4Dried, spun-dried and chromatographed (PE/EA/DCM-15/1/1) to give compound 3(400mg, yield: 70%). TLC PE/EA 1/1, 254nm, Rf(Compound 1) ═ 0.2, Rf(compound 3) ═ 0.6.
(4) Preparation of Compound 4
TFA (1mL) was added to Compound 3(400mg,0.975mmol,1.0eq) in DCM (3mL) and stirred at room temperature for 2 h. The solvent was spun off and the residue was dissolved in DCM. With saturated NaHCO3The solution was adjusted to pH 8. Separating the organic phase, washing with saturated brine, Na2SO4Drying and spin-drying gave Compound 4(287mg, yield: 95%). TLC PE/EA 4/1, 254nm, Rf(compound 3) ═ 0.4; DCM/MeOH 10/1, 254nm, Rf(compound 4) ═ 0.2.
(5) Preparation of Compound 5
Compound 4(390mg,1.26mmol,1.0eq), HCHO (37% in water, 1.0g,12.6mmol,10.0eq) and MgSO4(1.0g,8.33mmol,6.5eq) was dissolved in DCE (5 mL). Stir at room temperature for 10 min. Reacting NaBH (OAc)3(400mg,1.89mmol,1.5eq) was added to the reaction in portions at 0 ℃. The reaction is carried out for 1h at the temperature of 0 ℃ and 0.5h at the room temperature. The reaction was filtered, the filtrate was dried by spinning, and the filtrate was purified by column chromatography (DCM/MeOH-50/1) to give compound 5(260mg, yield: 63%). TLC DCM/MeOH-10/1, 254nm, Rf(Compound 4) ═ 0.2, Rf(compound 5) ═ 0.3.
(6) Preparation of Compound 7
Compound 5(100mg,0.31mmol,1.0eq), compound 6(149mg,0.31mmol,1.0eq), K2CO3(127mg,0.92mmol,3.0 eq.) and Pd (dppf) Cl2(25mg,0.031mmol,0.1eq) in dioxane (10mL) and H2O (2 mL). Heating to 80 ℃ for reaction for 2 h. The reaction was poured into EtOAc (20mL), washed with saturated brine, and Na2SO4Dry, spin dry, and column pass (DCM/MeOH ═ 20/1) to give compound 7(60mg, yield: 32%). TLC DCM/MeOH-8/1, 254nm, Rf(Compound 7) ═ 0.4, Rf(compound 5) ═ 0.3.
(7) Preparation of Compounds 11-20
TFA (0.5mL) was added to Compound 7(60mg,0.1mmol,1.0eq) in DCM (1 mL). Stir at room temperature for 1 h. The solvent was spun off and the residue was dissolved in DCM. With saturated NaHCO3The solution was adjusted to pH 8. Separating the organic phase from Na2SO4And (5) drying and spin-drying. The crude Prep-TLC (DCM/MeOH ═ 6/1) was purified to give compounds 11-20(13mg, yield: 26%).
LCMS:M+1=499.4。1H NMR(400MHz,DMSO-d6)δ12.05(s,1H),10.83(s,1H),10.43(s,1H),8.64(s,1H),8.29(d,J=8.8Hz,1H),8.10-8.08(m,1H),7.71(s,1H),7.58(d,J=7.2Hz,1H),7.45-7.37(m,2H),6.74(d,J=9.2Hz,1H),6.49(d,J=15.2Hz,1H),6.14(s,1H),3.92(d,J=6.8Hz,1H),2.22(s,3H),1.84-1.24(m,13H),0.88(d,J=7.2Hz,2H),0.62(d,J=3.2Hz,2H)。
Example 17
The compounds 11-1 to 5(300mg, 96% purity by LC-MS) of example 2 were subjected to chiral resolution using an SFC apparatus (resolution conditions and method: stationary phase (Column): ChiralPak IC-H Daicel chemical Industries, Ltd,250 x30 mm I.D.,5 um; mobile phase A: (Mobile phase A): Supercrystalline CO2Mobile phase B (mobile phase B) Ethanol (0.1% NH)3H2O); a: B60: 40at50ml/min (flow rate); 38 ℃ for Column Temperature(ii) a Nozle Pressure 100 Bar; nozle temp. 60 deg.C; evaporator temp. 20 deg.C; TrimmerTemp at 25 deg.C; wave length: UV 220nm) to give two yellow solids, Peak 1 and Peak 2.
Peak 1,11-1-P1
Yellow solid (Rt 5.05min,75.82mg, yield: 25%, solidified by1H NMR and LC-MS);
LCMS:M+1=485.4;
1H NMR(400MHz,DMSO-d6)δ12.03(s,1H),10.74(s,1H),10.42(s,1H),8.62(d,J=2.0Hz,1H),8.28(d,J=8.4Hz,1H),8.07(dd,J=8.8,2.4Hz,1H),7.70(s,1H),7.57(d,J=7.2Hz,1H),7.44-7.36(m,2H),6.72-6.66(m,1H),6.44(d,J=15.6Hz,1H),6.14(s,1H),3.92-3.89(m,1H),3.02-3.00(m,1H),2.73-2.71(m,1H),2.22(s,3H),2.11-2.03(m,1H),1.98-1.97(m,1H),1.82-1.75(m,3H),1.59-1.50(m,1H),1.42(d,J=6.8Hz,3H),0.87(d,J=8.4Hz,2H),0.62(d,J=3.2Hz,2H).
Peak 2,11-1-P2
Yellow solid (Rt 6.08min,72.05mg, yield: 24%, solidified by1H NMR and LC-MS)
LCMS:M+1=485.4
1H NMR(400MHz,DMSO-d6)δ12.03(s,1H),10.74(s,1H),10.42(s,1H),8.62(d,J=2.0Hz,1H),8.28(d,J=8.4Hz,1H),8.07(dd,J=8.8,2.4Hz,1H),7.70(s,1H),7.57(d,J=7.6Hz,1H),7.44-7.36(m,2H),6.72-6.66(m,1H),6.44(d,J=15.6Hz,1H),6.14(s,1H),3.93-3.89(m,1H),3.02-3.00(m,1H),2.73-2.69(m,1H),2.22(s,3H),2.11-2.03(m,1H),1.98-1.97(m,1H),1.82-1.75(m,3H),1.59-1.50(m,1H),1.42(d,J=6.8Hz,3H),0.87(d,J=8.4Hz,2H),0.62(d,J=3.2Hz,2H).
Example 18
Chiral resolution of the compounds 11-3-5 of example 4(300mg, 95% purity by LC-MS) was carried out using an SFC apparatus (resolution conditions and method: Column: ChiralPak IC-H Daicel chemical Industries, Ltd, 250: 30mm I.D.,5 um; mobile phase A (Mobile phase A): Supercriticica)l CO2Mobile phase B (mobile phase B) Ethanol (0.1% NH)3H2O); a: B60: 40at50ml/min (flow rate); column Temperature 38 deg.C; nozle Pressure 100 Bar; nozle temp. 60 deg.C; evaporator temp. 20 deg.C; the temperature of the Trimmer Temp is 25 ℃; wavelet at 220nm) to obtain two yellow solids, Peak 1 and Peak 2.
Peak 1,11-3-P1
Yellow solid(Rt=5.719min,71.78mg,yield:24%,confirmed by 1H NMR and LC-MS);
LCMS:M+1=485.4;
1H NMR(400MHz,DMSO-d6)δ11.99(br s,1H),10.74(s,1H),10.45(s,1H),8.62(d,J=2.0Hz,1H),8.28(d,J=8.4Hz,1H),8.07(dd,J=8.8,2.4Hz,1H),7.70(s,1H),7.56(d,J=7.6Hz,1H),7.44-7.36(m,2H),6.72-6.66(m,1H),6.44(d,J=15.6Hz,1H),6.12(s,1H),3.93-3.91(m,1H),3.05-3.02(m,1H),2.75-2.71(m,1H),2.20-2.13(m,4H),2.07-1.96(m,1H),1.83-1.70(m,3H),1.69-1.58(m,1H),1.43(d,J=6.8Hz,3H),0.88-0.86(m,2H),0.61-0.60(m,2H).
Peak 2,11-3-P2
Yellow solid(Rt=6.883min,65.67mg,yield:22%,confirmed by 1H NMR and LC-MS);
LCMS:M+1=485.3;
1H NMR(400MHz,DMSO-d6)δ12.03(s,1H),10.74(s,1H),10.42(s,1H),8.62(d,J=2.4Hz,1H),8.28(d,J=8.8Hz,1H),8.07(dd,J=8.8,2.4Hz,1H),7.70(s,1H),7.56(d,J=7.6Hz,1H),7.44-7.36(m,2H),6.72-6.66(m,1H),6.44(d,J=15.6Hz,1H),6.14(s,1H),3.93-3.89(m,1H),3.05-3.00(m,1H),2.73-2.71(m,1H),2.20-2.13(m,4H),2.00-1.97(m,1H),1.82-1.70(m,3H),1.69-1.58(m,1H),1.43(d,J=6.8Hz,3H),0.88-0.86(m,2H),0.61-0.60(m,2H).
Effect example 1
Test methods for CDK7 inhibitory activity refer to WO 2015058140.
CDK7 inhibition by compounds at 200nM and 10nM concentrations was determined using commercially available reagents such as CDK7/CyclinH/MAT1(Millipore, Cat. No. 14-476K, Lot. No. 1634571, His-CDK7+ GST-MAT1+ cyclinH) and CTD3 peptide (GL Biochem, Cat. No. 346885). The results are shown in Table 1.
TABLE 1 CDK7 inhibitory Activity
Effect example 2
Test methods for CDK7 inhibitory activity refer to WO 2015058140.
IC determination of CDK7 inhibition by the Compounds of examples 17 and 18 by CDK7 was determined using commercially available reagents CDK7/CyclinH/MAT1(Millipore, Cat. No. 14-476K, Lot. No. 1634571, His-CDK7+ GST-MAT1+ cyclinH) and CTD3 peptide (GL Biochem, Cat. No.346885)50. The results are shown in Table 2.
Compound (I) IC50(nM)
11-1-P1 584
11-1-P2 13
11-3-P1 487
11-3-P2 8.1

Claims (11)

1. A compound shown in a general formula I, a stereoisomer or pharmaceutically acceptable salt thereof,
wherein the content of the first and second substances,
ring A is C3-C20Cycloalkyl radical, C6-C20Aryl radical, C1-C20Heteroaryl or C2-C20A heterocycloalkyl group;
ring B is C6-C20Aryl radical, C3-C20Cycloalkyl or C1-C20A heteroaryl group;
R1is H or C1-C10An alkyl group;
R2is H, C1-C10Alkyl radical, C2-C10Alkenyl radical, C2-C10Alkynyl, C6-C20Aryl or C3-C20A cycloalkyl group;
R3is H, C6-C20Aryl or C1-C20A heteroaryl group;
or, R2And R3Taken together with the atoms to which they are attached form a 5-7 membered heteroaryl, a 5-7 membered cycloalkyl or a 5-7 membered heterocycloalkyl; the heteroatoms in said 5-7 membered heteroaryl or said 5-7 membered heterocycloalkyl are selected from N, O and S, the number of heteroatoms is 1-4;
m is 0, 1 or 2;
R4at any position, each R4Independently of one another, halogen, C1-C10Alkyl, hydroxy, C1-C10Alkoxy, amino, nitro, cyano, or C substituted by halogen1-C10An alkyl group;
R5is composed ofWherein n1 is 0 or 1; r5aAnd R5bIndependently is H or C1-C10An alkyl group; r5cIs H, halogen, C1-C10Alkyl radical, C1-C10Alkoxy, or substituted or unsubstituted C2-C20Heterocycloalkyl, said substituted C2-C20The substituent in the heterocycloalkyl group being C1-C10An alkyl group; said substituted or unsubstituted C2-C20Carbon atoms of heterocycloalkyl radicals withConnecting;
q is 0, 1,2 or 3;
R6at any position, each R6Independently is halogen, substituted or unsubstituted C1-C10Alkyl, substituted or unsubstituted C1-C10Alkoxy, hydroxy, amino, nitro or cyano; said substituted C1-C10Alkyl or said substituted C1-C10Substituents in alkoxy are meant to be substituted by one or more of the following groups: wherein each R isaEach RbAnd each RcIndependently is H or C1-C10Alkyl, or Ra、RbAnd the heteroatoms connected with the heterocyclic group form a 4-8-membered heterocyclic group, wherein the 4-8-membered heterocyclic group refers to a 4-8-membered heterocyclic group with 1-4 heteroatoms and the heteroatoms selected from N, O and S;
when R is5cIs H, halogen, C1-C10Alkyl or C1-C10When alkoxy, q is 1,2 or 3; each R6Independently is substituted C1-C10Alkyl or substituted C1-C10An alkoxy group;
L1is composed ofOr is absent; rlaAnd RlbIndependently H, C1-C10Alkyl or C substituted by halogen1-C10An alkyl group; or RlaAnd RlbTogether with the carbon atom to which they are attached form C3-C6A cycloalkyl group;the alkyl group of (a),The amino terminus of (a) is linked to the ring A; l is1Wherein, the carbon marked by the letter is S configuration chiral carbon, R configuration chiral carbon or achiral carbon;
L2is composed ofOr is absent;
wherein, said C1-C20Heteroaryl means a C having 1 to 4 heteroatoms selected from N, O and S1-C20A heteroaryl group; said C2-C20Heterocycloalkyl means a C having 1 to 4 heteroatoms selected from N, O and S2-C20A heterocycloalkyl group.
2. The compound of formula I, a stereoisomer or pharmaceutically acceptable salt thereof according to claim 1, wherein, when ring a is C6-C20When aryl, said C6-C20Aryl is C6-C14An aryl group;
and/or, when ring A is C3-C20When there is a cycloalkyl group, said C3-C20Cycloalkyl being C3-C6A cycloalkyl group;
and/or, when ring A is C1-C20When it is heteroaryl, said C1-C20Heteroaryl is C1-C10A heteroaryl group;
and/or, when ring B is C6-C20When aryl, said C6-C20Aryl is C6-C14An aryl group;
and/or, when ring B is C3-C20When there is a cycloalkyl group, said C3-C20Cycloalkyl being C3-C6A cycloalkyl group;
and/or, when ring B is C1-C20When it is heteroaryl, said C1-C20Heteroaryl is C1-C10A heteroaryl group;
and/or when R1Is C1-C10When alkyl, said C1-C10Alkyl is C1-C4An alkyl group;
and/or when R2Is C1-C10When alkyl, said C1-C10Alkyl is C1-C4An alkyl group;
and/or when R2Is C2-C10When alkenyl, said C2-C10Alkenyl is C2-C4An alkenyl group;
and/or when R2Is C2-C10When it is alkynyl, said C2-C10Alkynyl is C2-C4An alkynyl group;
and/or when R2Is C3-C20When there is a cycloalkyl group, said C3-C20Cycloalkyl being C3-C6A cycloalkyl group;
and/or, R5cWherein said substituted or unsubstituted C2-C20The heterocycloalkyl structure is as follows:wherein N is 1,2, 3, 4 or 5, and Z is C, O or N; when Z is O or N, R55Or R56Is absent; r51、R52、R53、R54、R55And R56Independently is H or C1-C10An alkyl group;
and/or when R6To getSubstituted C1-C10Alkyl or substituted C1-C10An alkoxy group; said substituted C1-C10Alkyl or said substituted C1-C10Substituents in alkoxy radicalsWhen the substitution is carried out,in, RaAnd RbIs C1-C10An alkyl group; wherein R isaAnd RbThe same or different;
and/or when L1Is composed ofWhen the current is over; rlaAnd RlbOne is H and the other is C1-C10An alkyl group; l is1In the formula (I), the carbon marked by the letter is S configuration chiral carbon, R configuration chiral carbon or achiral carbon.
3. The compound of formula I, a stereoisomer or pharmaceutically acceptable salt thereof according to claim 2, wherein, when ring a is C6-C20When aryl, said C6-C20Aryl is phenyl, naphthyl, anthryl or phenanthryl;
and/or, when ring A is C3-C20When there is a cycloalkyl group, said C3-C20Cycloalkyl is cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl;
and/or, when ring A is C1-C20When it is heteroaryl, said C1-C20Heteroaryl is pyridyl;
and/or, when ring B is C6-C20When aryl, said C6-C20Aryl is phenyl, naphthyl, anthryl or phenanthryl;
and/or, when ring B is C1-C20When it is heteroaryl, said C1-C20Heteroaryl is pyridyl;
and/or, when ring B is C3-C20When there is a cycloalkyl group, said C3-C20Cycloalkyl is cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl;
and/or when R1Is C1-C10When alkyl, said C1-C10Alkyl is methyl, ethyl, propyl or butyl;
and/or when R2Is C1-C10When alkyl, said C1-C10Alkyl is methyl, ethyl, propyl or butyl;
and/or when R2Is C2-C10When alkenyl, said C2-C10Alkenyl is
And/or when R2Is C2-C10When it is alkynyl, said C2-C10Alkynyl is
And/or when R2Is C3-C20When there is a cycloalkyl group, said C3-C20Cycloalkyl is cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl;
and/or when R5cIs substituted C2-C20When the heterocyclic alkyl is substituted by C1-C10An alkyl group;
and/or when R5cIs substituted C2-C20When heterocycloalkyl, the heteroatom is N and the substituent is on N.
4. A compound of formula I according to claim 3, a stereoisomer or a pharmaceutically acceptable salt thereof,
when R is5cIs substituted or unsubstituted C2-C20When it is heterocycloalkyl, said "C2-C20Heterocycloalkyl "is
5. The compound of formula I in claim 1, a stereoisomer or pharmaceutically acceptable salt thereof, wherein R is5cIs composed of
And/or, L1Is composed of
6. The compound of formula I, a stereoisomer or pharmaceutically acceptable salt thereof, as claimed in claim 1, wherein ring a is C6-C20An aryl group;
ring B is C6-C20Aryl or C1-C20A heteroaryl group;
R1is H;
R2is C1-C10Alkyl radical, C2-C10Alkenyl radical, C2-C10Alkynyl or C3-C20A cycloalkyl group;
R3is H;
m is 0;
R5is composed ofWherein R is5cIs H, or substituted or unsubstituted C2-C20HeterocycloalkanesA group;
q is 0 or 1; r6In any position, R6Is substituted C1-C10Alkyl or substituted C1-C10An alkoxy group; said substituted C1-C10Alkyl or said substituted C1-C10Substituents in alkoxy are meant to be substituted by one or more of the following groups: wherein each R isaEach RbAnd each RcIndependently is H or C1-C10Alkyl, or Ra、RbAnd the hetero atom to which they are attached together form a 4-to 8-membered heterocyclic group, e.g.
L1Is composed ofRlaAnd RlbIndependently H, C1-C10Alkyl or C substituted by halogen1-C10An alkyl group; l is1Wherein, the carbon marked by the letter is S configuration chiral carbon, R configuration chiral carbon or achiral carbon;
L2is absent.
7. The compound of formula I, a stereoisomer or pharmaceutically acceptable salt thereof, as claimed in claim 1, wherein ring a is C6-C20An aryl group;
ring B is C6-C20Aryl or C1-C20A heteroaryl group;
R1is H;
R2is C3-C20A cycloalkyl group;
R3is H;
m is 0;
R5is composed ofWherein R is5cIs substituted or unsubstituted C2-C20A heterocycloalkyl group;
q is 0;
L1is composed ofRlaAnd RlbOne is H and the other is C1-C10An alkyl group; l is1Wherein, the carbon marked by the letter is S configuration chiral carbon, R configuration chiral carbon or achiral carbon;
L2is absent.
8. The compound of formula I, a stereoisomer or a pharmaceutically acceptable salt thereof according to any one of claims 1 to 6, wherein the compound of formula I or a salt thereof is any one of the following compounds: the carbon marked by the letter is S configuration chiral carbon, R configuration chiral carbon or achiral carbon;
9. a process for producing a compound represented by the general formula I as claimed in claim 1, which comprises the following process (1) or process (2);
wherein the method (1) comprises the following steps: in a solvent, under the protection of gas and the action of alkali and a palladium catalyst, carrying out the reaction shown as the following on the compound shown as the general formula I-A and the compound shown as the general formula I-B;
wherein, in the compound shown in the general formula I-A, X is halogen, M isWherein R isb1And Rb2Independently is hydroxy or C1-C10Alkoxy, or Rb1And Rb2Together with boron atoms formingL2Absent, each of the remaining letters and groups are as defined in claim 1;
method (2): in the compounds of the formula I, R1When the compound is H, the compound can be prepared by the following method, wherein the method comprises the following steps: in a solvent, carrying out deprotection reaction on a compound shown as a general formula I-C as shown in the specification;
wherein R is1Is H, Q is an amino protecting group; the remaining letter and group definitions are as defined in claim 1.
10. A pharmaceutical composition comprising a compound of formula I as described in any one of claims 1-8, a stereoisomer or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
11. Use of a compound of general formula I according to any one of claims 1 to 8, a stereoisomer or a pharmaceutically acceptable salt thereof in the preparation of a CDK7 kinase inhibitor.
CN201810601884.4A 2018-06-12 2018-06-12 Compound, preparation method, pharmaceutical composition and application thereof Active CN110590747B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201810601884.4A CN110590747B (en) 2018-06-12 2018-06-12 Compound, preparation method, pharmaceutical composition and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201810601884.4A CN110590747B (en) 2018-06-12 2018-06-12 Compound, preparation method, pharmaceutical composition and application thereof

Publications (2)

Publication Number Publication Date
CN110590747A true CN110590747A (en) 2019-12-20
CN110590747B CN110590747B (en) 2022-03-22

Family

ID=68848962

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201810601884.4A Active CN110590747B (en) 2018-06-12 2018-06-12 Compound, preparation method, pharmaceutical composition and application thereof

Country Status (1)

Country Link
CN (1) CN110590747B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023224961A1 (en) * 2022-05-16 2023-11-23 Exelixis, Inc. Cancer therapy using a combination of a cdk7 inhibitor with an oral serd

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105849099A (en) * 2013-10-18 2016-08-10 达纳-法伯癌症研究所股份有限公司 Polycyclic inhibitors of cyclin-dependent kinase 7 (CDK7)
CN108024970A (en) * 2015-06-04 2018-05-11 奥瑞基尼探索技术有限公司 The Hete rocyclic derivatives by substitution as CDK inhibitor

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105849099A (en) * 2013-10-18 2016-08-10 达纳-法伯癌症研究所股份有限公司 Polycyclic inhibitors of cyclin-dependent kinase 7 (CDK7)
CN108024970A (en) * 2015-06-04 2018-05-11 奥瑞基尼探索技术有限公司 The Hete rocyclic derivatives by substitution as CDK inhibitor

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023224961A1 (en) * 2022-05-16 2023-11-23 Exelixis, Inc. Cancer therapy using a combination of a cdk7 inhibitor with an oral serd

Also Published As

Publication number Publication date
CN110590747B (en) 2022-03-22

Similar Documents

Publication Publication Date Title
CN105837576B (en) BTK inhibitor
CN106061976B (en) Multi-fluorinated compounds are as bruton's tyrosine kinase inhibitor
CN112574224A (en) KRAS G12C inhibitor and application thereof
CN109071538A (en) Pyridine derivate and its preparation method and application as ASK1 inhibitor
CN101842361B (en) Pyrazinone derivatives and their use in the treatment of lung diseases
JP6419990B2 (en) Benzimidazole derivatives as bromodomain inhibitors
CN101903372B (en) Heteroaryl derivatives as orexin receptor antagonists
KR100929941B1 (en) pyrazine-2-carboxamide derivatives as an antagonist
CN106661000A (en) Egfr inhibitor, and preparation and application thereof
CN101896461A (en) Gamma secretase modulators
CN107759573A (en) Hepatitis C virus inhibitor and its pharmaceutical applications
CN106103432B (en) Substituted thiazole or oxazole P2X7 receptor antagonist
WO2019208812A1 (en) Benzisoxazole compound
CN107709298A (en) Spiral shell [indoline of cyclobutane 1,3 '] 2 ' ketone derivatives as bromine domain inhibitor
CN107001371B (en) Oxipurinol class compound and its application
CN111433196A (en) Novel bradykinin B2 receptor antagonists and uses thereof
CN113166083A (en) Succinate and fumarate acid addition salts of piperazine derivatives as glycosidase inhibitors
CN111233863B (en) Hydroxyl purine compound and application thereof
CN104379568A (en) Quinazolinedione derivative
KR20160143668A (en) Piperidine derivatives as orexin receptor antagonist
CN109575013A (en) Triazole and pyrimidine, triazole and pyridine compounds and combinations thereof are for treating the disease of PRC2 mediation
CN114269748A (en) Imidazo [1,2-c ] pyrimidine derivatives as PRC2 inhibitors for the treatment of cancer
CN111356695A (en) Novel tricyclic compounds
CN110590747B (en) Compound, preparation method, pharmaceutical composition and application thereof
CN101798302A (en) Method and technology for synthesizing and producing antibiotic medicament namely 1-(o-fluorophenyl) dihydropyridone

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant