CN109928971B - Aryl substituted aminotetrahydropyrans and uses thereof - Google Patents

Aryl substituted aminotetrahydropyrans and uses thereof Download PDF

Info

Publication number
CN109928971B
CN109928971B CN201910191868.7A CN201910191868A CN109928971B CN 109928971 B CN109928971 B CN 109928971B CN 201910191868 A CN201910191868 A CN 201910191868A CN 109928971 B CN109928971 B CN 109928971B
Authority
CN
China
Prior art keywords
mmol
compound
added
tetrahydropyran
methanol
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.)
Active
Application number
CN201910191868.7A
Other languages
Chinese (zh)
Other versions
CN109928971A (en
Inventor
黎健豪
顾峥
唐万军
康盼盼
张宗远
邓新山
王绪礼
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.)
Sunshine Lake Pharma Co Ltd
Original Assignee
Guangdong HEC Pharmaceutical
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 Guangdong HEC Pharmaceutical filed Critical Guangdong HEC Pharmaceutical
Priority to CN201910191868.7A priority Critical patent/CN109928971B/en
Publication of CN109928971A publication Critical patent/CN109928971A/en
Application granted granted Critical
Publication of CN109928971B publication Critical patent/CN109928971B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Landscapes

  • Pyridine Compounds (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention relates to an aryl substituted aminotetrahydropyran compound and application thereof, and further relates to a pharmaceutical composition containing the compound. The compounds of the invention or the pharmaceutical compositions may be used as dipeptidyl peptidase-IV (DPP-IV) inhibitors.

Description

Aryl substituted aminotetrahydropyrans and uses thereof
Technical Field
The invention belongs to the technical field of medicines, and particularly relates to an aryl substituted amino tetrahydropyran compound and application thereof, and further relates to a pharmaceutical composition containing the compound. The compounds or the pharmaceutical compositions may be used as dipeptidyl peptidase-IV (DPP-IV) inhibitors.
Background
Diabetes is a disease process in which blood glucose rises due to a variety of pathogenic factors. There are generally two types of diabetes. Type I diabetes, also known as or Insulin Dependent Diabetes Mellitus (IDDM), is primarily characterized by damage to pancreatic beta cells by autoimmune processes. Type II diabetes, also known as non-insulin dependent diabetes mellitus (NIDDM), is a long-term, progressive metabolic disease characterized by hyperglycemia, hyperlipidemia, and insulin resistance. Typical symptoms are polyuria, polydipsia and polyphagia. Type II diabetes is more common and is expected to affect 30 million people worldwide in 2025.
Incretins play an important role in the regulation of blood glucose balance in normal and pathological states. Glucagon peptide (GLP-1) and glucose-dependent insulinotropic polypeptide in incretins are released from the intestinal tract after food intake and have an effect on insulin secretion. GLP-1 reduces hyperglycemia in a glucose-dependent manner by simultaneously promoting insulin secretion and reducing glucagon secretion. At the same time, GLP-1 can slow gastric emptying and act as a satiety agent to suppress appetite and reduce body weight. GLP-1 has a trophic effect on beta cells in animal experiments, thus increasing the likelihood of potential to improve disease processes by increasing beta cell density and function. Recent studies have shown that GLP-1 has a benefit on the cardiovascular system, which is important because cardiovascular complications are one of the leading causes of death in the diabetic population. In summary, GLP-1 has many benefits, which make it quite different from existing therapeutic approaches. GLP-1, however, is rapidly degraded and inactivated by the widely distributed serine protease dipeptidyl peptidase-IV (DPP-IV) in the body. Thus, much interest is now focused on the development of long-acting analogues of GLP-1 and dipeptidyl peptidase-IV inhibitors that are resistant to peptidase degradation.
Dipeptidyl peptidase-IV is a glycoprotein and serine exopeptidase within the cell membrane that cleaves X-proline dipeptides from the N-terminus of polypeptides. DPP-IV is widely distributed in various tissues of humans, including endothelial cells of the intestinal tract and intestinal mucosal vessels. DPP-IV also expresses T-lymphocytes in the circulatory system, which is equivalent to the T-cell activation marker CD26, and data obtained from the systemic model also indicate that DPP-IV enzyme has potential co-activation during T-cell activation. Furthermore, DPP-IV also exhibits degradation of many immunomodulatory, endocrine and neuro polypeptides.
DPP-IV inhibitors may cause an increase in undegraded biologically active GLP-1 levels, and may provide an alternative treatment for type II diabetes. Furthermore, DPP-IV inhibitors are orally active compared to incretin analogues. Inhibiting DPP-IV activity may improve glycemic control. The DPP-IV inhibitors have the advantage that insulin release can be increased without increasing the risk of hypoglycemia. However, as a member of the exopeptidase family, DPP-IV inhibitors require high selectivity to inhibit only DPP-IV activity without affecting other DPPs.
The principle of treatment of type II diabetes with DPP-IV inhibitors has been strongly demonstrated. Many DPP-IV inhibitors have been in various stages of clinical development and there are a number of drugs marketed, including sitagliptin in 2006, vildagliptin in 2007, saxagliptin in 2009 and alogliptin in 2010, etc. However, there is still a need for DPP-IV inhibitors with advantageous activity, stability, selectivity and pharmacokinetic properties.
Disclosure of Invention
The invention provides aryl substituted aminotetrahydropyrans with better dipeptidyl peptidase-IV (DPP-IV) activity, a pharmaceutical composition thereof and application of the compounds or the pharmaceutical composition in preparing medicines for treating, preventing or relieving diabetes mellitus, diabetic retinopathy, diabetic neuropathy, diabetic nephropathy, insulin resistance, hyperglycemia, hyperinsulinemia, obesity, hypertriglyceridemia, syndrome X, atherosclerosis or hypertension.
In one aspect, the present invention relates to a compound which is a compound of formula (I) or a stereoisomer, a geometric isomer, a tautomer, a nitroxide, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug thereof of a compound of formula (I),
Wherein,
R 1a 、R 1b and R is 1c Each independently is H, deuterium, F, cl, br, I, -CN, -NO 2 、-OH、-NH 2 、C 1-6 Alkyl, C 1-6 Alkoxy, C 1-6 Haloalkyl or C 1-6 Haloalkoxy groups;
w is
L is C 1-4 An alkyl group;
each R is 5 、R 7 、R 8 、R 10 And R is 14 Is independently H, deuterium, -CN, C 1-6 Alkyl, C 1-6 Haloalkyl, C 3-6 Cycloalkyl, heterocyclyl of 3-6 atoms, C 6-10 Aryl, heteroaryl consisting of 5-6 atoms, -S (=o) 2 R a 、-C(=O)R b 、-OC(=O)R b 、-C(=O)OR c 、-S(=O) 2 NHR d or-C (=o) NHR d The method comprises the steps of carrying out a first treatment on the surface of the Wherein said C 1-6 Alkyl, C 1-6 Haloalkyl, C 3-6 Cycloalkyl, heterocyclyl of 3-6 atoms, C 6-10 Aryl and heteroaryl consisting of 5-6 atoms are each independently unsubstituted or substituted with 1, 2, or 3R x Substituted;
R 12 is H, deuterium, -CN, C 2-6 Alkyl, C 1-6 Haloalkyl, C 3-6 Cycloalkyl, heterocyclyl of 3-6 atoms, C 6-10 Aryl, heteroaryl consisting of 5-6 atoms, -S (=o) 2 R e 、-C(=O)R f 、-OC(=O)R b 、-C(=O)OR c 、-S(=O) 2 NHR d or-C (=o) NHR d The method comprises the steps of carrying out a first treatment on the surface of the Wherein said C 2-6 Alkyl, C 1-6 Haloalkyl, C 3-6 Cycloalkyl, heterocyclyl of 3-6 atoms, C 6-10 Aryl and heteroaryl consisting of 5-6 atoms are each independently unsubstituted or substituted with 1, 2, or 3R x Substituted;
each R is a 、R b 、R c And R is d Independently C 1-6 Alkyl, C 1-6 Haloalkyl, C 3-6 Cycloalkyl, heterocyclyl of 3-6 atoms, C 6-10 Aryl or heteroaryl of 5-6 atoms, wherein said C 1-6 Alkyl, C 1-6 Haloalkyl, C 3-6 Cycloalkyl, heterocyclyl of 3-6 atoms, C 6-10 Aryl and heteroaryl consisting of 5-6 atoms are each independently unsubstituted or substituted with 1, 2, or 3R x Substituted;
R e and R is f Each independently is C 2-6 Alkyl, C 4-6 Cycloalkyl, heterocyclyl of 3-6 atoms, C 6-10 Aryl or heteroaryl of 5-6 atoms, wherein said C 2-6 Alkyl, C 4-6 Cycloalkyl, heterocyclyl of 3-6 atoms, C 6-10 Aryl and heteroaryl consisting of 5-6 atoms are each independently unsubstituted or substituted with 1, 2, or 3R x Substituted;
each R is 4a 、R 4b 、R 4c 、R 4d 、R 6a 、R 6b 、R 6c 、R 6d 、R 6 、R 9 、R 11 And R is 13 Independently is H, deuterium, F, cl, br, I, =o, -OH, -CN, -NH 2 、-NO 2 、C 1-6 Alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 1-6 Alkoxyacyl, C 1-6 Alkanoyl, C 3-6 Cycloalkyl, heterocyclyl of 3-6 atoms, C 6-10 Aryl or heteroaryl of 5-6 atoms, wherein said C 1-6 Alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 1-6 Alkoxyacyl groupRadical, C 1-6 Alkanoyl, C 3-6 Cycloalkyl, heterocyclyl of 3-6 atoms, C 6-10 Aryl and heteroaryl consisting of 5-6 atoms are each independently unsubstituted or substituted with 1, 2, or 3R y Substituted;
each R is x And R is y Independently deuterium, F, cl, br, I, -OH, -CN, C 1-6 Alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 1-6 Alkanoyl, C 1-6 Alkoxyacyl or C 1-6 An alkylsulfonyl group;
each n, t or s is independently 1, 2, 3 or 4;
q, p and u are each independently 0, 1, 2 or 3.
In some embodiments, each R 5 、R 7 、R 8 、R 10 And R is 14 Is independently H, deuterium, -CN, C 1-4 Alkyl, C 1-4 Haloalkyl, C 3-6 Cycloalkyl, heterocyclyl of 5-6 atoms, C 6-10 Aryl, heteroaryl consisting of 5-6 atoms, -S (=o) 2 R a 、-C(=O)R b 、-OC(=O)R b 、-C(=O)OR c 、-S(=O) 2 NHR d or-C (=o) NHR d The method comprises the steps of carrying out a first treatment on the surface of the Wherein said C 1-4 Alkyl, C 1-4 Haloalkyl, C 3-6 Cycloalkyl, heterocyclyl of 5-6 atoms, C 6-10 Aryl and heteroaryl consisting of 5-6 atoms are each independently unsubstituted or substituted with 1, 2, or 3R x Substituted.
In some embodiments, R 12 Is H, deuterium, -CN, C 2-4 Alkyl, C 1-4 Haloalkyl, C 3-6 Cycloalkyl, heterocyclyl of 5-6 atoms, C 6-10 Aryl, heteroaryl consisting of 5-6 atoms, -S (=o) 2 R e 、-C(=O)R f 、-OC(=O)R b 、-C(=O)OR c 、-S(=O) 2 NHR d or-C (=o) NHR d The method comprises the steps of carrying out a first treatment on the surface of the Wherein said C 2-4 Alkyl, C 1-4 Haloalkyl, C 3-6 Cycloalkyl, heterocyclyl of 5-6 atoms, C 6-10 Aryl and 5-6 atomsEach independently unsubstituted or substituted with 1, 2, or 3R x Substituted.
In some embodiments, each R 5 、R 7 、R 8 、R 10 And R is 14 Independently H, deuterium, -CN, methyl, ethyl, n-propyl, isopropyl, n-butyl, tert-butyl, trifluoromethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, heterocyclyl consisting of 5-6 atoms, phenyl, heteroaryl consisting of 5-6 atoms, -S (=O) 2 R a 、-C(=O)R b 、-OC(=O)R b 、-C(=O)OR c 、-S(=O) 2 NHR d or-C (=o) NHR d The method comprises the steps of carrying out a first treatment on the surface of the Wherein the methyl, ethyl, n-propyl, isopropyl, n-butyl, tert-butyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, heterocyclyl consisting of 5-6 atoms, phenyl and heteroaryl consisting of 5-6 atoms are each independently unsubstituted or substituted with 1, 2, or 3R x Substituted.
In some embodiments, R 12 Is H, deuterium, -CN, ethyl, n-propyl, isopropyl, n-butyl, tert-butyl, trifluoromethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, heterocyclyl consisting of 5-6 atoms, phenyl, heteroaryl consisting of 5-6 atoms, -S (=O) 2 R e 、-C(=O)R f 、-OC(=O)R b 、-C(=O)OR c 、-S(=O) 2 NHR d or-C (=o) NHR d The method comprises the steps of carrying out a first treatment on the surface of the Wherein the ethyl, n-propyl, isopropyl, n-butyl, tert-butyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, heterocyclyl consisting of 5-6 atoms, phenyl and heteroaryl consisting of 5-6 atoms are each independently unsubstituted or substituted with 1, 2, or 3R x Substituted.
In some embodiments, each R a 、R b 、R c And R is d Independently C 1-4 Alkyl, C 1-4 Haloalkyl, C 3-6 Cycloalkyl, heterocyclyl of 5-6 atoms, C 6-10 Aryl or heteroaryl of 5-6 atoms, wherein said C 1-4 Alkyl, C 1-4 Haloalkyl, C 3-6 Cycloalkyl, 5-6 atoms Constituent heterocyclic groups, C 6-10 Aryl and heteroaryl consisting of 5-6 atoms are each independently unsubstituted or substituted with 1, 2, or 3R x Substituted.
In some embodiments, R e And R is f Each independently is C 2-4 Alkyl, C 4-6 Cycloalkyl, heterocyclyl of 5-6 atoms, C 6-10 Aryl or heteroaryl of 5-6 atoms, wherein said C 2-4 Alkyl, C 4-6 Cycloalkyl, heterocyclyl of 5-6 atoms, C 6-10 Aryl and heteroaryl consisting of 5-6 atoms are each independently unsubstituted or substituted with 1, 2, or 3R x Substituted.
In some embodiments, each R a 、R b 、R c And R is d Independently methyl, ethyl, n-propyl, isopropyl, n-butyl, tert-butyl, trifluoromethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, piperidinyl, piperazinyl, tetrahydrofuranyl, dihydrofuranyl, morpholinyl, thiomorpholinyl, phenyl, pyrrolyl, pyridinyl, pyrimidinyl, thiazolyl, thienyl, furanyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, pyrazinyl, or pyridazinyl, wherein the methyl, ethyl, n-propyl, isopropyl, n-butyl, tert-butyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, pyrrolidinyl, pyrazolidinyl, imidazolyl, piperidinyl, piperazinyl, tetrahydrofuranyl, dihydrofuranyl, morpholinyl, thiomorpholinyl, phenyl, pyrrolyl, pyridinyl, pyrimidinyl, thiazolyl, thienyl, furanyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, pyridazinyl, and pyridazinyl are each independently substituted with 1 or 2R x Substituted.
In some embodiments, R e And R is f Each independently is ethyl, n-propyl, isopropyl, n-butyl, tert-butyl, cyclobutyl, cyclopentyl, cyclohexyl, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, piperidinyl, piperazinyl, tetrahydrofuranyl, dihydrofuroA pyranyl, morpholinyl, thiomorpholinyl, phenyl, pyrrolyl, pyridinyl, pyrimidinyl, thiazolyl, thiophenyl, furanyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, pyrazinyl, or pyridazinyl group, wherein the ethyl, n-propyl, isopropyl, n-butyl, tert-butyl, cyclobutyl, cyclopentyl, cyclohexyl, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, piperidinyl, piperazinyl, tetrahydrofuranyl, dihydrofuranyl, morpholinyl, thiomorpholinyl, phenyl, pyrrolyl, pyridinyl, pyrimidinyl, thiazolyl, thiophenyl, furanyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, pyrazinyl, and pyridazinyl group are each independently unsubstituted or substituted with 1, 2, or 3R x Substituted.
In some embodiments, each R 4a 、R 4b 、R 4c 、R 4d 、R 6a 、R 6b 、R 6c 、R 6d 、R 6 、R 9 、R 11 And R is 13 Independently is H, deuterium, F, cl, br, I, =o, -OH, -CN, -NH 2 、-NO 2 、C 1-4 Alkyl, C 1-4 Haloalkyl, C 1-4 Alkoxy, C 1-4 Alkoxyacyl, C 1-4 Alkanoyl, C 3-6 Cycloalkyl, heterocyclyl of 5-6 atoms, C 6-10 Aryl or heteroaryl of 5-6 atoms, wherein said C 1-4 Alkyl, C 1-4 Haloalkyl, C 1-4 Alkoxy, C 1-4 Alkoxyacyl, C 1-4 Alkanoyl, C 3-6 Cycloalkyl, heterocyclyl of 5-6 atoms, C 6-10 Aryl and heteroaryl consisting of 5-6 atoms are each independently unsubstituted or substituted with 1, 2, or 3R y Substituted.
In some embodiments, each R 4a 、R 4b 、R 4c 、R 4d 、R 6a 、R 6b 、R 6c 、R 6d 、R 6 、R 9 、R 11 And R is 13 Independently is H, deuterium, F, cl, br, I, =o, -OH, -CN, -NH 2 、-NO 2 Methyl, ethyl, n-propyl, isopropyl, trifluoromethyl, methoxy, ethoxy, 1-propoxy, 2-propoxy, methoxyacyl, ethoxyacyl, acetyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, heterocyclyl consisting of 5 to 6 atoms, phenyl or heteroaryl consisting of 5 to 6 atoms, wherein the methyl, ethyl, n-propyl, isopropyl, methoxy, ethoxy, 1-propoxy, 2-propoxy, methoxyacyl, ethoxyacyl, acetyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, heterocyclyl consisting of 5 to 6 atoms, phenyl and heteroaryl consisting of 5 to 6 atoms are each independently unsubstituted or substituted with 1, 2, or 3 substituents R y Substituted.
In some embodiments, each R x And R is y Independently deuterium, F, cl, br, I, -OH, -CN, C 1-4 Alkyl, C 1-4 Haloalkyl, C 1-4 Alkoxy, C 1-4 Alkanoyl, C 1-4 Alkoxyacyl or C 1-4 An alkylsulfonyl group.
In some embodiments, each R x And R is y And independently is deuterium, F, cl, br, I, -OH, -CN, methyl, ethyl, n-propyl, isopropyl, trifluoromethyl, methoxy, ethoxy, 1-propoxy, 2-propoxy, n-butoxy, t-butoxy, acetyl, methoxyacyl, ethoxyacyl, methylsulfonyl, ethylsulfonyl, n-propylsulfonyl or isopropylsulfonyl.
In some embodiments, R 1a 、R 1b And R is 1c Each independently is H, deuterium, F, cl, br, I, -CN, -NO 2 、-OH、-NH 2 、C 1-4 Alkyl, C 1-4 Alkoxy, C 1-4 Haloalkyl or C 1-4 Haloalkoxy groups.
In some embodiments, R 1a 、R 1b And R is 1c Each independently is H, deuterium, F, cl, br, I, -CN, -NO 2 、-OH、-NH 2 Methyl, ethyl, n-propyl, isopropyl, methoxy, ethoxy, trifluoromethyl or trifluoromethoxy.
In some embodiments, L is methyl, ethyl, or n-propyl.
In some embodiments, the compounds of the present invention are stereoisomers, geometric isomers, tautomers, nitroxides, hydrates, solvates, metabolites, pharmaceutically acceptable salts or prodrugs thereof, of the compounds of formula (II),
Wherein R is 1a 、R 1b 、R 1c And W has the meaning described in the present invention.
In some embodiments, the compounds of the present invention are compounds that are one of,
or a stereoisomer, geometric isomer, tautomer, nitroxide, hydrate, solvate, metabolite, pharmaceutically acceptable salt or prodrug thereof.
In another aspect, the invention relates to a pharmaceutical composition comprising a compound of the invention.
In some embodiments, the pharmaceutical compositions of the present invention further comprise a pharmaceutically acceptable carrier, excipient, adjuvant, vehicle, or combination thereof.
In some embodiments, the pharmaceutical compositions of the present invention further comprise one or more additional active ingredients selected from ACE inhibitors, renin inhibitors, angiotensin II receptor antagonists, beta-receptor blockers, acetylsalicylic acid, diuretics, calcium antagonists, statins, digitalis derivatives, calcium sensitizers, nitrates and antithrombotics.
In another aspect, the present invention relates to the use of a compound according to the invention or a pharmaceutical composition according to the invention for the preparation of a dipeptidyl peptidase-IV (DPP-IV) inhibitor drug.
In another aspect, the invention relates to the use of a compound according to the invention or a pharmaceutical composition according to the invention for the preparation of a medicament for the prevention, treatment or alleviation of diabetes, diabetic retinopathy, diabetic neuropathy, diabetic nephropathy, insulin resistance, hyperglycemia, hyperinsulinemia, obesity, hypertriglyceridemia, syndrome X, atherosclerosis or hypertension.
Detailed description of the invention
Definitions and general terms
Reference will now be made in detail to certain embodiments of the invention, examples of which are illustrated in the accompanying structural and chemical formulas. The invention is intended to cover all alternatives, modifications and equivalents, which may be included within the scope of the invention as defined by the appended claims. Those skilled in the art will recognize that many methods and materials similar or equivalent to those described herein can be used in the practice of the present invention. The present invention is in no way limited to the methods and materials described herein. In the event of one or more of the incorporated references, patents and similar materials differing from or contradictory to the present application (including but not limited to defined terms, term application, described techniques, etc.), the present application controls.
It should further be appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable sub-combination.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. All patents and publications referred to herein are incorporated by reference in their entirety.
The following definitions as used herein should be applied unless otherwise indicated. For the purposes of the present invention, chemical elements are in accordance with CAS version of the periodic Table of the elements, and handbook of chemistry and physics, 75 th edition, 1994. In addition, general principles of organic chemistry may be referenced to the descriptions in "Organic Chemistry", thomas Sorrell, university Science Books, sausalato:1999, and "March's Advanced Organic Chemistry" by Michael b.smith and Jerry March, john Wiley & Sons, new york:2007, the entire contents of which are incorporated herein by reference.
The articles "a," "an," and "the" are intended to include "at least one" or "one or more" unless the context clearly dictates otherwise or otherwise. Thus, as used herein, these articles refer to one or to more than one (i.e., to at least one) object. For example, "a component" refers to one or more components, i.e., more than one component is contemplated as being employed or used in embodiments of the described embodiments.
The term "patient" as used herein refers to a human (including adults and children) or other animals. In some embodiments, "patient" refers to a human.
The term "comprising" is an open-ended expression, i.e., including what is indicated by the invention, but not excluding other aspects.
"stereoisomers" refer to compounds having the same chemical structure but different arrangements of atoms or groups in space. Stereoisomers include enantiomers, diastereomers, conformational isomers (rotamers), geometric isomers (cis/trans), atropisomers, and the like.
"enantiomer" refers to two isomers of a compound that do not overlap but are in mirror image relationship to each other.
"diastereoisomers" refers to stereoisomers which have two or more chiral centers and whose molecules are not mirror images of each other. Diastereomers have different physical properties, such as melting point, boiling point, spectral properties, and reactivity. The diastereomeric mixture may be separated by high resolution analytical procedures such as electrophoresis and chromatography, e.g., HPLC.
The stereochemical definitions and rules used in the present invention generally follow S.P. Parker, ed., mcGraw-Hill Dictionary of Chemical Terms (1984) McGraw-Hill Book Company, new York; and Eliel, e.and Wilen, s., "Stereochemistry of Organic Compounds", john Wiley & Sons, inc., new York,1994.
Any asymmetric atom (e.g., carbon, etc.) of the disclosed compounds may exist in racemic or enantiomerically enriched form, such as in the (R) -, (S) -or (R, S, R) -configuration. In certain embodiments, each asymmetric atom has at least 50% enantiomeric excess, at least 60% enantiomeric excess, at least 70% enantiomeric excess, at least 80% enantiomeric excess, at least 90% enantiomeric excess, at least 95% enantiomeric excess, or at least 99% enantiomeric excess in the (R) -or (S) -configuration.
The resulting mixture of any stereoisomers may be separated into pure or substantially pure geometric isomers, enantiomers, diastereomers, e.g., by chromatography and/or fractional crystallization, depending on the differences in the physicochemical properties of the components.
The term "tautomer" or "tautomeric form" refers to structural isomers having different energies that can be interconverted by a low energy barrier (low energy barrier). If tautomerism is possible (e.g., in solution), chemical equilibrium of the tautomers can be achieved. For example, proton tautomers (also known as proton transfer tautomers (prototropic tautomer)) include interconversions by proton transfer, such as keto-enol isomerisation and imine-enamine isomerisation. Valence tautomers (valance tautomers) include interconversions by recombination of some of the bond-forming electrons. Specific examples of keto-enol tautomerism are tautomerism of pentane-2, 4-dione and 4-hydroxypent-3-en-2-one tautomer. Another example of tautomerism is phenol-ketone tautomerism. One specific example of phenol-ketone tautomerism is the interconversion of pyridin-4-ol and pyridin-4 (1H) -one tautomers. Unless otherwise indicated, all tautomeric forms of the compounds of the invention are within the scope of the invention.
The term "prodrug" as used herein means a compound that is converted in vivo to a compound of formula (I) or formula (II). Such conversion is effected by hydrolysis of the prodrug in the blood or enzymatic conversion to the parent structure in the blood or tissue. The prodrug of the invention can be ester, and in the prior invention, the ester can be phenyl ester, aliphatic (C 1-24 ) Esters, acyloxymethyl esters, carbonates, carbamates and amino acid esters. For example, one compound of the invention may contain a hydroxyl group, i.e., it may be acylated to provide the compound in a prodrug form. Other prodrug forms include phosphates, such as those obtained by phosphorylation of a hydroxyl group on the parent. For a complete discussion of prodrugs, reference may be made to the following documents: higuchi and V.stilla, pro-drugs as Novel Delivery Systems, vol.14of the A.C.S. symposium Series, edward B.Roche, ed., bioreversible Carriers in Drug Design, american Pharmaceutical Association and Pergamon Press,1987,J.Rautio et al, prodrug: design and Clinical Applications, nature Review Drug Discovery,2008,7,255-270,and S.J.Hecker et al, prodrugs of Phosphates and Phosphonates, journal of Medicinal Chemistry,2008,51,2328-2345.
"metabolite" refers to a product obtained by metabolizing a specific compound or salt thereof in vivo. The metabolites of a compound may be identified by techniques well known in the art and their activity may be characterized by employing the assay methods as described herein. Such products may be obtained by oxidation, reduction, hydrolysis, amidization, deamination, esterification, degreasing, enzymatic cleavage, etc. of the administered compound. Accordingly, the present invention includes metabolites of compounds, including metabolites produced by contacting a compound of the present invention with a mammal for a period of time sufficient.
Pharmaceutically acceptable used in the present inventionThe term "salts" refers to organic and inorganic salts of the compounds of the present invention. Pharmaceutically acceptable salts are well known in the art, as in the literature: S.M. Berge et al describe pharmaceutically acceptable salts in detail in J.pharmaceutical Sciences,1977,66:1-19. Pharmaceutically acceptable non-toxic acid forming salts include, but are not limited to, mineral acid salts such as hydrochloride, hydrobromide, phosphate, sulfate, perchlorate; organic acid salts such as acetates, oxalates, maleates, tartrates, citrates, succinates, malonates; or by other methods described in the book literature, such as ion exchange. Other pharmaceutically acceptable salts include adipic acid salts, alginates, ascorbates, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphoric acid salts, cyclopentylpropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumaric acid salts, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, caproate, hydroiodic acid salts, 2-hydroxy-ethanesulfonate, lactobionic acid salts, lactate, laurate, lauryl sulfate, malate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, palmitate, pamoate, pectate, persulfate, 3-phenylpropionate, picrate, pivalate, propionate, stearate, thiocyanate, p-toluenesulfonate, undecanoate, valerate, and the like. Salts obtained by reaction with suitable bases include alkali metals, alkaline earth metals, ammonium and N + (C 1-4 Alkyl group 4 Is a salt of (a). The present invention also contemplates quaternary ammonium salts formed from any of the compounds containing a group of N. The water-soluble or oil-soluble or dispersible product may be obtained by quaternization. Alkali or alkaline earth metals that may form salts include sodium, lithium, potassium, calcium, magnesium, and the like. The pharmaceutically acceptable salts further include suitable, non-toxic ammonium, quaternary ammonium salts and counter-ion forming amine cations, such as halides, hydroxides, carboxylates, sulphates, phosphates, nitrates, C 1-8 Sulfonate and fragranceA sulfonate.
"solvate" according to the present invention refers to an association of one or more solvent molecules with a compound according to the present invention. Solvents that form solvates include, but are not limited to, water, isopropanol, ethanol, methanol, dimethylsulfoxide, ethyl acetate, acetic acid, and aminoethanol. The term "hydrate" refers to an association of solvent molecules that are water.
"nitroxide" in the present invention means that when a compound contains several amine functions, 1 or more than 1 nitrogen atom can be oxidized to form an N-oxide. Specific examples of N-oxides are N-oxides of tertiary amines or N-oxides of nitrogen atoms of nitrogen-containing heterocycles. The corresponding amine may be treated with an oxidizing agent such as hydrogen peroxide or a peracid (e.g., peroxycarboxylic acid) to form an N-oxide (see Advanced Organic Chemistry, wiley Interscience, 4 th edition, jerry March, pages). In particular, the N-oxides can be prepared by the method L.W.Deady (Syn.Comm.1977, 7, 509-514) in which an amine compound is reacted with m-chloroperoxybenzoic acid (MCPBA), for example in an inert solvent such as methylene chloride.
The compounds of the invention may be optionally substituted with one or more substituents, as described in the present invention, such as the compounds of the general formula above, or as specific examples within the examples, subclasses, and classes of compounds encompassed by the invention.
The term "optional" or "optionally" means that the subsequently described event or circumstance may, but need not, occur, i.e., that the description includes instances where said event or circumstance occurs and instances where it does not.
In addition, unless explicitly indicated otherwise, the descriptions used in this disclosure of the manner in which each … is independently "and" … is independently "and" … is independently "are to be construed broadly as meaning that particular items expressed between the same symbols in different groups do not affect each other, or that particular items expressed between the same symbols in the same groups do not affect each other. Likewise, the broad sense of the above should also be taken with respect to "independent" in the description of the manner "… independent optionally".
In the various parts of the present specification, substituents of the presently disclosed compounds are disclosed in terms of the type or scope of groups. It is specifically noted that the present invention includes each individual subcombination of the individual members of these group classes and ranges. For example, the term "C 1-6 Alkyl "means in particular independently disclosed C 1 Alkyl (methyl), C 2 Alkyl (ethyl), C 3 Alkyl, C 4 Alkyl, C 5 Alkyl and C 6 An alkyl group; "C 1-4 Alkyl "refers specifically to independently disclosed C 1 Alkyl (methyl), C 2 Alkyl (ethyl), C 3 Alkyl (i.e. propyl, including n-propyl and isopropyl), C 4 Alkyl (i.e., butyl, including n-butyl, isobutyl, sec-butyl, and tert-butyl).
In the various parts of the invention, linking substituents are described. When the structure clearly requires a linking group, the markush variables recited for that group are understood to be linking groups. For example, if the structure requires a linking group and the markush group definition for that variable enumerates an "alkyl" or "aryl" group, it will be understood that the "alkyl" or "aryl" represents a linked alkylene group or arylene group, respectively.
The term "alkyl" or "alkyl group" as used herein means a saturated, straight or branched, monovalent hydrocarbon group containing from 1 to 20 carbon atoms, wherein the alkyl group may be optionally substituted with one or more substituents described herein. In some embodiments, the alkyl group contains 1 to 12 carbon atoms; in other embodiments, the alkyl group contains 1 to 6 carbon atoms, i.e., C 1-6 An alkyl group; in other embodiments, the alkyl group contains 2 to 6 carbon atoms, i.e., C 2-6 An alkyl group; in still other embodiments, the alkyl group contains 1 to 4 carbon atoms, i.e., C 1-4 An alkyl group; in some embodiments, the alkyl group contains 1 to 3 carbon atoms, i.e., C 1-3 An alkyl group; in some embodiments, the alkyl group contains 2 to 6 carbon atoms, i.e., C 2-6 An alkyl group. In some embodiments, C as described in the present invention 1-6 The alkyl group may beC 1-4 An alkyl group; in other embodiments, C as described in the present invention 1-6 Alkyl can be C 1-2 An alkyl group.
Examples of alkyl groups include, but are not limited to, methyl, ethyl, propyl (including n-propyl and isopropyl), butyl (including n-butyl, isobutyl, sec-butyl, tert-butyl), n-pentyl, 2-pentyl, 3-pentyl, 2-methyl-2-butyl, 3-methyl-1-butyl, 2-methyl-1-butyl, n-hexyl, 2-hexyl, 3-hexyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 3-methyl-3-pentyl, 2, 3-dimethyl-2-butyl, 3-dimethyl-2-butyl, n-heptyl, n-octyl, and the like.
The term "alkoxy" means that the alkyl group is attached to the remainder of the molecule through an oxygen atom, wherein the alkyl group has the meaning as described herein. Examples of alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy (including 1-propoxy or 2-propoxy), butoxy (including n-butoxy, isobutoxy, sec-butoxy, tert-butoxy), and the like.
The term "haloalkyl" or "haloalkoxy" means that an alkyl or alkoxy group is substituted with one or more halogen atoms, examples of which include, but are not limited to, trifluoromethyl, trifluoromethoxy, chloroethyl (e.g., 2-chloroethyl), trifluoroethyl (e.g., 2-trifluoroethyl), 2-difluoroethyl, 2-chloro-1-methylethyl, and the like.
The term "alkoxyacyl" means that the alkoxy group is attached to the remainder of the molecule via-C (=o) -i.e. -C (=o) -alkoxy, wherein the alkoxy group has the meaning as described in the present invention. In some embodiments, the alkoxyacyl group contains 1 to 6 carbon atoms, i.e., C 1-6 An alkoxy acyl group; in other embodiments, the alkoxyacyl group contains 1 to 4 carbon atoms, i.e., C 1-4 An alkoxy acyl group; in still other embodiments, the alkoxyacyl group contains 1 to 2 carbon atoms, i.e., C 1-2 An alkoxy acyl group. Examples include, but are not limited to, methoxyacyl (-C (=O) O-CH 3 ) Ethoxyacyl (-C (=O) O-CH 2 CH 3 )、-C(=O)O-CH(CH 3 ) 2
The term "alkanoyl" means an alkyl group attached to the remainder of the molecule through-C (=o) -i.e. -C (=o) -alkyl, wherein the alkyl group has the meaning as described herein. In some embodiments, the alkanoyl group contains 1-6 carbon atoms, i.e., C 1-6 An alkanoyl group; in other embodiments, the alkanoyl group contains 1-4 carbon atoms, i.e., C 1-4 An alkanoyl group; in still other embodiments, the alkanoyl group contains 1-2 carbon atoms, i.e., C 1-2 Alkanoyl. Examples include, but are not limited to, acetyl (-C (=O) CH 3 )。
The term "alkylsulfonyl" denotes the passage of an alkyl group through-S (=o) 2 To the remainder of the molecule, i.e. -S (=o) 2 -alkyl, wherein the alkyl group has the meaning as described in the present invention. In some embodiments, the alkylsulfonyl group contains 1 to 6 carbon atoms, i.e., C 1-6 An alkylsulfonyl group; in some embodiments, the alkylsulfonyl group contains 1 to 4 carbon atoms, i.e., C 1-4 An alkylsulfonyl group; in some embodiments, the alkylsulfonyl group contains 1 to 2 carbon atoms, i.e., C 1-2 An alkylsulfonyl group. Examples include, but are not limited to, methylsulfonyl (-S (=o) 2 CH 3 ) Ethylsulfonyl (-S (=o) 2 CH 2 CH 3 ) N-propylsulfonyl (-S (=O) 2 CH 2 CH 2 CH 3 ) Isopropyl sulfonyl (-S (=o) 2 CH(CH 3 ) 2 )。
The term "amino" denotes the group-NH 2
The term "carboxyl" denotes the group-COOH.
The term "hydroxy" denotes the group-OH.
The term "cyano" denotes the group-CN.
The term "nitro" denotes the group-NO 2
The term "oxo" represents the group=o.
The term "halogen" refers to fluorine (F), chlorine (Cl), bromine (Br) or iodine (I).
The term "cycloalkyl" means a saturated monocyclic, bicyclic or tricyclic ring system containing 3 to 12 ring carbon atoms. In some embodiments, cycloalkyl groups contain 3 to 10 ring carbon atoms, e.g., C 3-10 Cycloalkyl; in other embodiments, cycloalkyl groups contain 3 to 8 ring carbon atoms, e.g., C 3-8 Cycloalkyl; in other embodiments, cycloalkyl groups contain 3 to 6 ring carbon atoms, e.g., C 3-6 Cycloalkyl; in other embodiments, cycloalkyl groups contain 4 to 6 ring carbon atoms, e.g., C 4-8 Cycloalkyl groups. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, and the like; wherein, C is as follows 3-6 Cycloalkyl includes cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl; the C is 4-6 Cycloalkyl groups include cyclobutyl, cyclopentyl and cyclohexyl. The cycloalkyl group may be optionally substituted with one or more substituents described herein.
The term "heterocyclyl" refers to a saturated or partially unsaturated mono-, bi-or tricyclic ring system containing 3 to 12 ring atoms, wherein at least one ring atom is selected from the group consisting of nitrogen, sulfur and oxygen atoms; wherein the heterocyclic group is non-aromatic and does not contain any aromatic ring. Unless otherwise indicated, a heterocyclic group may be a carbon or nitrogen group, and-CH 2 The group may optionally be replaced by-C (=o) -. The sulfur atom of the ring may optionally be oxidized to an S-oxide. The nitrogen atom of the ring may optionally be oxidized to an N-oxide. The term "heterocyclyl" may be used interchangeably with the term "heterocycle". The heterocyclyl group may be optionally substituted with one or more substituents described herein. Examples of heterocyclyl groups include, but are not limited to: oxiranyl, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, tetrahydrofuranyl, dihydrofuranyl, and the like. The heterocyclyl group may be optionally substituted with one or more substituents described herein.
The term "aryl" means a radical containing 6 to 14 ring atoms, orMonocyclic, bicyclic and tricyclic carbocyclic ring systems of 6 to 12 ring atoms, or 6 to 10 ring atoms, wherein at least one ring is aromatic and has one or more points of attachment to the remainder of the molecule. The term "aryl" may be used interchangeably with the term "aromatic ring" or "aromatic ring". Unless otherwise indicated, the group "C 6-10 Aryl "means an aryl group containing 6 to 10 ring carbon atoms. Examples of aryl groups may include phenyl, 2, 3-dihydro-1H-indenyl, naphthyl and anthracenyl. The aryl group may be optionally substituted with one or more substituents described herein.
The term "heteroaryl" means monocyclic, bicyclic and tricyclic ring systems containing 5 to 12 ring atoms, or 5 to 10 ring atoms, or 5 to 6 ring atoms, wherein at least one ring is aromatic and at least one ring contains 1, 2,3 or 4 ring heteroatoms selected from nitrogen, oxygen, sulfur, and wherein said heteroaryl has one or more attachment points attached to the remainder of the molecule. When the-CH is present in the heteroaryl group 2 -said-CH, when a group is 2 The group may optionally be replaced by-C (=o) -. Unless otherwise indicated, the heteroaryl group may be attached to the remainder of the molecule (e.g., the host structure in the formula) at any reasonable point (which may be C in CH, or N in NH). The term "heteroaryl" may be used interchangeably with the term "heteroaromatic ring" or "heteroaromatic compound". In some embodiments, heteroaryl is a heteroaryl consisting of 5 to 10 atoms, meaning that the heteroaryl contains 1 to 9 ring carbon atoms and 1, 2,3, or 4 ring heteroatoms selected from O, S and N; in other embodiments, heteroaryl is a heteroaryl consisting of 5-6 atoms, meaning that the heteroaryl contains 1-5 ring carbon atoms and 1, 2,3, or 4 ring heteroatoms selected from O, S and N. Examples include, but are not limited to, furyl, imidazolyl, isoxazolyl, oxazolyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, pyridyl, pyrimidinyl, pyridazinyl, pyrazinyl, thienyl, thiazolyl, and the like. The heteroaryl group may be optionally substituted with one or more substituents described herein.
As described herein, a ring system in which a substituent R is attached to a central ring by a bond represents that the substituent R may be substituted only at any substitutable or any reasonable position on the ring to which it is attached. For example, formula b represents that substituent R may be substituted at any position on the C-ring that may be substituted, but may not be substituted on the D-ring, as shown in formulas b-1 to b-3:
the term "pharmaceutical composition" means a mixture of one or more of the compounds described herein or a physiologically/pharmaceutically acceptable salt or prodrug thereof with other chemical components, such as physiologically/pharmaceutically acceptable carriers, excipients, auxiliaries, vehicles, diluents, binders, fillers, and the like, and additional therapeutic agents such as antidiabetic agents, antihyperglycemic agents, antiobesity agents, antihypertensive agents, antiplatelet agents, antiatherosclerotic agents, or lipid lowering agents. The purpose of the pharmaceutical composition is to facilitate the administration of the compound to the organism.
The term "pharmaceutically acceptable" refers to molecular entities and compositions that are physiologically tolerable and do not generally produce allergies or similar inappropriate reactions, such as gastrointestinal discomfort, dizziness, etc., when administered to humans. Preferably, the term "pharmaceutically acceptable" as used herein refers to use in animals, particularly in humans, approved by the federal regulatory agency or a state government or listed in the U.S. pharmacopeia or other generally recognized pharmacopeia.
The term "carrier" refers to a diluent, adjuvant, excipient, or matrix with which the compound is administered. These pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water and aqueous solutions (e.g., saline solution, aqueous dextrose solution, aqueous glycerol solution) are preferably employed as carriers, particularly injectable solutions. Suitable drug carriers are described in "Remington's Pharmaceutical Sciences" of e.w. martin.
The term "syndrome X", also known as a condition, disease of metabolic syndrome, the condition of which is described in detail in Johannsson et al, j.clin.endocrinol.metab.,1997,82,727-734.
The expression "compound of the present invention", "compound described in the present invention" or the like as used in the present invention refers to a compound represented by the formula (I) or the formula (II) in the present invention.
The term "treating" as used herein refers in some embodiments to ameliorating a disease or disorder (i.e., slowing or preventing or alleviating the progression of the disease or at least one clinical symptom thereof). In other embodiments, "treating" refers to moderating or improving at least one physical parameter, including physical parameters that may not be perceived by the patient. In other embodiments, "treating" refers to modulating a disease or disorder physically (e.g., stabilizing a perceived symptom) or physiologically (e.g., stabilizing a parameter of the body) or both. In other embodiments, "treating" refers to preventing or delaying the onset, or exacerbation of a disease or disorder.
Any formulae given herein are also intended to represent non-isotopically enriched forms as well as isotopically enriched forms of such compounds. Isotopically enriched compounds have structures depicted by the formulae given herein except that one or more atoms are replaced by an atom having a selected atomic or mass number. Exemplary isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, and chlorine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 15 N, 17 O, 18 O, 18 F, 31 P, 32 P, 35 S, 36 Cl and Cl 125 I。
In another aspect, the compounds of the invention include isotopically enriched compounds defined in the invention, e.g., wherein a radioisotope, such as 3 H, 14 C and C 18 F, or wherein non-radioactive is presentRadioisotopes, e.g. 2 H and 13 C. such isotopically enriched compounds are useful in metabolic studies (using 14 C) Reaction kinetics studies (using, for example 2 H or 3 H) Detection or imaging techniques, such as Positron Emission Tomography (PET) or Single Photon Emission Computed Tomography (SPECT) including drug or substrate tissue distribution assays, or may be used in radiation therapy of a patient. 18 F-enriched compounds are particularly desirable for PET or SPECT studies. Isotopically enriched compounds of formula (I) can be prepared by conventional techniques familiar to those skilled in the art or by describing the examples and processes of preparation of the present invention using a suitable isotopically labelled reagent in place of the one previously used unlabelled reagent.
In addition, heavier isotopes are in particular deuterium (i.e., 2 substitution of H or D) may provide certain therapeutic advantages, which are brought about by a higher metabolic stability. For example, increased in vivo half-life or reduced dosage requirements or improved therapeutic index. It is to be understood that deuterium in the context of the present invention is considered as a substituent for a compound of formula (I) or formula (II). The concentration of such heavier isotopes, particularly deuterium, can be defined by an isotopic enrichment factor. The term "isotopically enriched factor" as used herein refers to the ratio between the isotopic abundance and the natural abundance of a specified isotope. If a substituent of a compound of the invention is designated as deuterium, the compound has an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation at each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium incorporation), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation). Pharmaceutically acceptable solvates of the invention include those wherein the crystallization solvent may be isotopically substituted, e.g. D 2 O, acetone-d 6 、DMSO-d 6 Those solvates of (a).
All tautomeric forms of the compounds of the invention are included within the scope of the invention unless otherwise indicated. In addition, unless otherwise indicated, the structural formulae of the compounds described herein include enriched isotopes of one or more different atoms.
Abbreviations for any protecting groups, amino acids and other compounds used in the present invention are, unless otherwise indicated, based on their commonly used, accepted abbreviations or with reference to IUPAC-IUBCommission on Biochemical Nomenclature (see biochem.1972, 11:942-944).
Pharmaceutical compositions, formulations, administration and uses of the compounds of the invention
In another aspect, the invention features a pharmaceutical composition that includes an aryl-substituted aminotetrahydropyran compound of formula (I) or formula (II), a compound listed herein, or a compound of the examples, and a pharmaceutically acceptable carrier, excipient, adjuvant, vehicle, or combination thereof. The amount of the compound in the compositions of the invention is effective to treat or ameliorate a condition associated with dipeptidyl peptidase-IV (DPP-IV) in a patient.
As described herein, the pharmaceutically acceptable compositions of the present invention further comprise a pharmaceutically acceptable carrier, excipient, adjuvant, vehicle, or combination thereof, as used herein, including any solvents, diluents, or other liquid excipients, dispersing or suspending agents, surfactants, isotonic agents, thickening agents, emulsifying agents, preservatives, solid binders or lubricants, and the like, suitable for the particular target dosage form. As described in the following documents: in Remington, the Science and Practice of Pharmacy,21st edition,2005,ed.D.B.Troy,Lippincott Williams&Wilkins,Philadelphia,and Encyclopedia of Pharmaceutical Technology,eds.J.Swarbrick and J.C.Boylan,1988-1999,Marcel Dekker,New York, in combination with the teachings of the literature herein, shows that different carriers can be used In the formulation of pharmaceutically acceptable compositions and their well-known methods of preparation. In addition to the extent to which any conventional carrier vehicle is incompatible with the compounds of the present invention, such as any adverse biological effects produced or interactions with any other component of the pharmaceutically acceptable composition in a deleterious manner, their use is also contemplated by the present invention.
Materials that may be used as pharmaceutically acceptable carriers include, but are not limited to, ion exchangers, aluminum stearate, lecithin, serum proteins, such as human serum proteins, buffer substances, such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silicon, magnesium trisilicate, polyvinylpyrrolidone, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, lanolin, sugars, such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; a gum powder; malt; gelatin; talc powder; adjuvants such as cocoa butter and suppository waxes; oils such as peanut oil, cotton seed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycol compounds such as propylene glycol and polyethylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic salt; ringer's solution; ethanol, phosphate buffer, and other non-toxic suitable lubricants such as sodium lauryl sulfate and magnesium stearate, coloring agents, releasing agents, coating materials, sweetening, flavoring and perfuming agents, preserving and antioxidant agents.
The pharmaceutical compositions of the present invention may be administered directly or in pharmaceutical compositions or pharmaceutical forms along with suitable carriers or excipients, as is well known in the art. The methods of treatment of the present invention may comprise administering to a subject in need thereof an effective amount of a compound of the present invention. In some embodiments, the subject is a mammalian subject, and in some preferred embodiments, the subject is a human subject.
The effective amount of a compound, pharmaceutical composition or medicament of the present invention can be readily determined by routine experimentation, and the most effective and convenient route of administration and most appropriate formulation can also be determined by routine experimentation.
The pharmaceutical dosage forms of the compounds of the present invention may be provided in the form of immediate release, controlled release, sustained release or target drug release systems. For example, common dosage forms include solutions and suspensions, (micro) emulsions, ointments, gels and patches, liposomes, tablets, dragees, soft or hard shell capsules, suppositories, ovules, implants, amorphous or crystalline powders, aerosols and freeze-dried formulations. Depending on the route of administration used, special devices may be required to administer or administer the drug, such as syringes and needles, inhalers, pumps, injection pens, applicators, or Special bottles (Special flash). Pharmaceutical dosage forms often consist of a drug, excipients and a container/sealing system. One or more excipients (also known as inactive ingredients) may be added to the compounds of the present invention to improve or promote the manufacture, stability, administration and safety of the drug, and may provide a means to achieve a desired drug release profile. Thus, the type of excipient added to a drug may depend on various factors, such as the physical and chemical characteristics of the drug, the route of administration, and the manufacturing steps. Pharmaceutically acceptable excipients are present in this field and include those listed in the various pharmacopoeias. (see U.S. Pharmacopeia (U.S.Pharmacopeia, USP), japanese Pharmacopeia (Japanese Pharmacopoeia, JP), european Pharmacopeia (European Pharmacopoeia, EP) and British Pharmacopeia (British pharmacopoeia, BP); U.S. food and drug administration (the U.S. food and Drug Administration, www.fda.gov) drug evaluation and research center (Center for Drug Evaluation and Research, CEDR) publications, for example, inactive ingredient guide (Inactive Ingredient Guide, 1996); pharmaceutical additives handbook written by Ash and Ash (Handbook of Pharmaceutical Additives,2002, incorporated information resources, inc. (Synapse Information Resources, inc., endiott NY; etc.).
Pharmaceutical dosage forms of the compounds of the present invention may be manufactured by any of the methods well known in the art, for example by conventional mixing, sieving, dissolving, melting, granulating, dragee-making, tabletting, suspending, extruding, spray-drying, grinding, emulsifying, (nano/micro) encapsulating, packaging, or lyophilizing processes. As noted above, the compositions of the present invention may include one or more physiologically acceptable inactive ingredients that facilitate processing of the active molecule into a formulation for pharmaceutical use.
Suitable formulations will depend upon the route of administration desired. For example, for intravenous injection, the composition may be formulated in aqueous solution, if necessary with physiologically compatible buffers including, for example, phosphates, histidines or citrates for adjusting the pH of the formulation, and tonicity agents such as sodium chloride or dextrose. For transmucosal or nasal administration, semi-solid, liquid formulations or patches, possibly containing permeation enhancers, may be preferred; such penetrants are generally known in the art. For oral administration, the compounds may be formulated in liquid or solid dosage forms and as immediate release or controlled release/sustained release formulations. Suitable dosage forms for oral ingestion by an individual include tablets, pills, dragees, hard and soft shell capsules, liquids, gels, syrups, slurries, suspensions and emulsions. The compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
Solid oral dosage forms may be obtained using excipients including fillers, disintegrants, binders (dry and wet), dissolution retarders, lubricants, glidants, anti-sticking agents, cationic exchange resins, wetting agents, antioxidants, preservatives, colorants, and flavoring agents. These excipients may be of synthetic or natural origin. Examples of such excipients include cellulose derivatives, citric acid, dicalcium phosphate, gelatin, magnesium carbonate, magnesium/sodium lauryl sulfate, mannitol, polyethylene glycol, polyvinylpyrrolidone, silicates, silica, sodium benzoate, sorbitol, starch, stearic acid or salts thereof, sugars (i.e., dextrose, sucrose, lactose, etc.), talc, tragacanth (tragacanth mucilage), vegetable oils (hydrogenated), and waxes. Ethanol and water may be used as granulation aids. In some cases it may be desirable to coat the tablet with, for example, a taste masking film, a gastric acid resistant film, or a delayed release film. Natural and synthetic polymers are often used in combination with colorants, sugars and organic solvents or water to coat tablets, resulting in dragees. When the capsule is preferred over a tablet, the drug powder, suspension or solution thereof may be delivered in the form of a compatible hard shell or soft shell capsule.
In some embodiments, the compounds of the invention may be administered topically, for example by skin patches, semi-solid or liquid formulations, such as gels, (micro) emulsions, ointments, solutions, (nano/micro) suspensions or foams. Skin and underlying tissue penetration of the drug may be regulated by: for example, using permeation enhancers; suitable choices and combinations of lipophilic, hydrophilic and amphiphilic excipients are used, including water, organic solvents, waxes, oils, synthetic and natural polymers, surfactants, emulsifiers; by adjusting the pH value; and using a complexing agent. Other techniques, such as iontophoresis (ionophoresis), may also be used to regulate skin penetration of the compounds of the present invention. Transdermal or topical administration will be preferred, for example, where topical administration with minimal systemic exposure is desired.
For administration by inhalation or nasal administration, the compounds used according to the invention are conveniently administered from pressurized packs or nebulizers in the form of solutions, suspensions, emulsions or semi-solid aerosols, usually with the aid of propellants, such as halocarbons derived from methane and ethane, carbon dioxide or any other suitable gas. Hydrocarbons such as butane, isobutene and pentane are suitable for topical aerosols. In the case of a pressurized aerosol, the appropriate dosage unit may be determined by providing a valve delivery metering. Capsules and cartridges with, for example, gelatin may be formulated for use in an inhaler or insufflator. These generally comprise a powder mixture of the compound with a suitable powder base, such as lactose or starch.
Compositions for formulation by parenteral administration by injection are generally sterile and may be provided in unit dosage forms, such as ampules, syringes, injection pens, or multi-dose containers, the latter typically containing a preservative. The compositions may take the form of suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as buffering agents, tonicity agents, viscosity enhancing agents, surfactants, suspending and dispersing agents, antioxidants, biocompatible polymers, chelating agents and preservatives. Depending on the injection site, the carrier may contain water, synthetic or vegetable oils and/or organic co-solvents. In some cases, such as for lyophilized products or concentrates, parenteral formulations may be reconstituted or diluted prior to administration. The depot formulation (depot formulation) providing controlled or sustained release of the compounds of the invention may comprise injectable suspensions of nano/micro-sized particles or nano/micro-sized or non-micronized crystals. Other matrices well known in the art, polymers such as poly (lactic acid), poly (glycolic acid) or copolymers thereof, may be used as the controlled/sustained release matrix. Other depot-type (depot) drug delivery systems may be provided in the form of implants and pumps requiring incisions.
Suitable carriers for intravenous injection of the compounds of the invention are well known in the art and include aqueous based solutions containing a base such as sodium hydroxide for the formation of ionic compounds; sucrose or sodium chloride as tonicity agent; such as buffers containing phosphate or histidine. A co-solvent such as polyethylene glycol may be added. These aqueous-based systems are effective in dissolving the compounds of the present invention and result in low toxicity after systemic administration. The proportions of the components of the solution system can be varied considerably without destroying the solubility and toxicity characteristics. In addition, the characteristics of the components may be changed. For example, low toxicity surfactants such as polysorbates or poloxamers (poloxamers) may be used, polyethylene glycols or other co-solvents may be used, biocompatible polymers such as polyvinylpyrrolidone may be added, and other sugars and polyols may be used in place of dextrose.
The compounds of the invention may act systemically and/or locally. They may be administered in a suitable manner, for example, by oral administration, parenteral administration, pulmonary administration, nasal administration, sublingual administration, lingual administration, buccal administration, rectal administration, dermal administration, transdermal administration, conjunctival administration, aural administration or as a graft or stent. The compounds of the present invention are preferably administered orally or parenterally.
Suitable modes of administration for oral administration are as follows: the mode of administration of the compounds of the invention, which comprises crystalline and/or amorphous and/or dissolved forms of the compounds of the invention, such as tablets (uncoated tablets or tablets coated, for example, with a gastric resistant or delayed dissolution or insoluble coating which controls the release of the compounds of the invention), tablets or films/sheets which disintegrate rapidly in the oral cavity, films/lyophilisates, capsules (e.g. hard or soft capsules), sugar-coated tablets, granules, pills, powders, emulsions, suspensions, aerosols or solutions, is released rapidly and/or in an improved manner according to the mode of operation of the prior art.
Parenteral administration may bypass the step of absorption (e.g., intravenous, intra-arterial, intra-cardiac, intraspinal, or intra-lumbar) or include absorption (e.g., intramuscular, subcutaneous, intradermal, transdermal, or intraperitoneal). Forms of administration suitable for parenteral administration include formulations for injection and infusion in the form of solutions, suspensions, emulsions, lyophilisates or sterile powders.
For other routes of administration, suitable examples are inhaled pharmaceutical forms (including powder inhalers, nebulizers), nasal drops, solutions or sprays, tablets, films/sheets or capsules for sublingual or buccal administration, suppositories, otic or ophthalmic preparations, vaginal capsules, aqueous suspensions (lotions, concussions), lipophilic suspensions, ointments, creams, transdermal therapeutic systems (e.g. patches), emulsions (Milch), pastes, foams, spray powders, implants or stents.
The therapeutically effective amount of the compounds of the present invention should be present in the above pharmaceutical formulation at a concentration of about 0.1 to 99.5%, preferably about 0.5 to 95% by weight of the total mixture.
The above pharmaceutical formulations may contain other pharmaceutically active ingredients in addition to the compounds of the present invention.
The therapeutically effective dose may be estimated first using various methods well known in the art. The initial dose used for animal studies may be based on the established effective concentration in the cell culture assay. The dosage range suitable for a human individual can be determined, for example, using data obtained from animal studies and cell culture assays. In certain embodiments, the compounds of the present invention may be prepared as medicaments for oral administration. Exemplary dosages of the compounds of the present invention in a medicament for oral administration are from about 0.01 to about 100mg/kg (where kg represents the body weight of the subject). In some embodiments, the medicament comprises from about 0.01 to about 20mg/kg (where kg represents the body weight of the subject), or optionally from about 0.01 to about 10mg/kg (where kg represents the body weight of the subject), or optionally from about 0.01 to about 5.0mg/kg (where kg represents the body weight of the subject). In certain embodiments, the compounds of the present invention are administered parenterally in an effective dosage of about 0.001-1mg/kg, preferably about 0.01-0.5mg/kg body weight.
The dosing regimen for agents typically used for oral administration is three times per week, twice per week, once per week, three times per day, twice per day, or once per day. In certain embodiments, the compounds of the present invention are administered as the active ingredient in a total amount of about 0.001 to about 50, preferably 0.001 to 10mg/kg body weight per 24 hours, optionally in the form of a plurality of individual doses in order to achieve the desired result. A single dose may preferably comprise the compound of the invention in an amount of about 0.001 to about 30, especially 0.001 to 3mg/kg body weight.
An effective amount or therapeutically effective amount or dose of an agent (e.g., a compound of the invention) refers to the amount of the agent or compound that results in an improvement in the symptoms or an prolongation of survival of the individual. Toxicity and therapeutic efficacy of the molecules can be determined in cell culture or experimental animals by standard pharmaceutical procedures, e.g., by determining LD 50 (dose lethal to 50% of population) and ED 50 (50% of the dose therapeutically effective for the population). The dose ratio of toxic effect to therapeutic effect is the therapeutic index and can be expressed as LD 50 /ED 50 . Agents that exhibit high therapeutic indices are preferred.
An effective amount or therapeutically effective amount is that amount of a compound or pharmaceutical composition that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician. The dosage is preferably such that it includes ED with little or no toxicity 50 Within a range of circulating concentrations. The dosage may vary within this range depending upon the dosage form employed and/or the route of administration employed. The correct formulation, route of administration, dosage and interval of administration should be selected in consideration of the particularities of the individual condition according to methods known in the art.
The dosage and interval may be individually adjusted to provide a plasma level of the active moiety sufficient to achieve the desired effect; i.e. the minimum effective concentration (minimal effective concentration, MEC). The MEC will vary from compound to compound but can be estimated, for example, from in vitro (in vitro) data and animal experiments. The dosage necessary to obtain MEC will depend on the individual characteristics and route of administration. In the case of local administration or selective uptake, the effective local concentration of the drug may be independent of plasma concentration.
The amount of the agent or composition administered can depend on a variety of factors including the sex, age and weight of the individual being treated, the severity of the affliction, the mode of administration and the discretion of the prescribing physician.
The compositions of the present invention may be provided in a packaging or dispensing device containing one or more unit dosage forms (containing the active ingredient) as desired. For example, the package or device may include a metal or plastic foil (such as a foam package) or glass and rubber stoppers. The packaging or dispensing device may be accompanied by instructions for administration. Compositions comprising the compounds of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in a suitable container, and labeled for treatment of the indicated condition.
The compounds of the invention can be used alone or, if desired, in combination with other active compounds. The invention also provides a combination comprising at least one compound according to the invention and one or more further active substances, in particular medicaments for the treatment and/or prophylaxis of the diseases according to the invention.
The compounds of the present invention act as DPP-IV inhibitors and exhibit an unpredictable range of valuable pharmacological actions. They are therefore suitable for use as medicaments for the treatment and/or prophylaxis of human and animal diseases. DPP-4 inhibitors can increase the concentration of active endo-GIP (glucose-dependent insulinotropic polypeptide) and GLP-1 (glucagon-like polypeptide-1) in blood, thereby promoting insulin secretion, inhibiting abnormal secretion of glucagon, alleviating gastric emptying, and lowering blood glucose level. Thus, the compounds of the present invention will find application in the prevention, treatment or amelioration of symptoms of diabetes and related diseases.
The compounds of the present invention will find application, but are in no way limited to, the use of an effective amount of a compound or pharmaceutical composition of the present invention to be administered to a patient to prevent, treat or ameliorate diabetes and related diseases, or post-diabetes injury, in the patient. Such diseases include, but are not limited to, diabetes, especially type II diabetes, as well as diabetic retinopathy, diabetic neuropathy, diabetic nephropathy, insulin resistance, hyperglycemia, hyperinsulinemia, obesity, hyperlipidemia, especially hypertriglyceridemia, syndrome X, atherosclerosis or hypertension.
In addition to being beneficial for human treatment, the compounds of the present invention are also useful in veterinary treatment of pets, introduced species of animals and farm animals, including mammals, rodents and the like. Examples of other animals include horses, dogs, and cats. Herein, the compounds of the present invention include pharmaceutically acceptable derivatives thereof.
The compounds of the invention can be used alone or, if desired, in combination with other active ingredients. The invention further relates to a medicament comprising at least one compound of the invention and one or more further active ingredients, in particular for the treatment and/or prophylaxis of the abovementioned disorders. Suitable active ingredients for the combination are, for example and preferably:
DPP-IV inhibitors such as sitagliptin (sitagliptin), vildagliptin (vidagliptin), alogliptin (alogliptin), linagliptin (linagliptin), saxagliptin (saxagliptin), olagliptin (omargigliptin), and trelagliptin (trelagliptin);
biguanides, e.g. phenformin, metformin (metformin);
sulfonylureas, such as, for example, glyburide, chlorpropamide, glibenclamide, glipizide, gliclazide, glimepiride, glitezide, tolbutamide, and meglitinide;
Glinide-based drugs, such as repaglinide and nateglinide;
alpha-glucosidase inhibitors, e.g., acarbose (acarbose);
alpha-glucosidase inhibitors, for example, lipolytic agents, carboglibose (camigibose), glibenclamide (emiglitate), miglitol (miglitol), voglibose (voglibose), pradimicin (pradimicin) and sabcometin (salbostatin);
PPAR agonists, for example balaglitazone, ciglitazone, darglitazone, englitazone, alaglitazone, pioglitazone, rosiglitazone;
PPARα/γ dual activators, e.g., CLX-0940, GW-1536, GW-1929, GW-2433, KRP-297, L-796449, LR-90, MK-0767, and SB-219994;
glucagon-like peptide-1 (GLP-1) agonists, such as, for example, B-3 (exendin-3) and B-4 (exendin-4);
protein tyrosine phosphatase-1B (PTP 1B) inhibitors, e.g., quinuclidine, halyard extracts and compounds disclosed by Zhang, S. et al, modern drug discovery, 12 (9/10), 373-381 (2007);
Sodium-glucose cotransporter 2 (SGLT 2) inhibitors, such as Dapagliflozin (Dapagliflozin), enagliflozin (Empagliflozin), canagliflozin (Canagliflozin), ipragliflozin (ASP-1941), tofogliflozin, luseogliflozin and Rong Gelie.
Insulin, insulin mimetics, liver glucose phosphorylase inhibitors;
VPAC2 receptor agonists;
glucokinase activator;
glycogen phosphorylase inhibitor or glucose-6-phosphatase inhibitor;
alpha P2 inhibitors;
acetyl-CoA carboxylase-2 (ACC-2) inhibitors;
phosphodiesterase (PDE) -10 inhibitors;
diacylglycerol acyltransferase (DGAT) 1 or 2 inhibitors;
glucose transporter 4 (GLUT 4) modulators;
glutamine-fructose-6-phosphate transferase (GFAT) inhibitors.
General synthetic procedure
In this specification, a structure is dominant if there is any difference between a chemical name and a chemical structure.
In general, the compounds of the invention may be prepared by the methods described herein, wherein the substituents are as defined for formula (I) or formula (II), unless otherwise indicated. The following reaction schemes and examples are provided to further illustrate the present invention.
Those skilled in the art will recognize that: the chemical reactions described herein may be used to suitably prepare other compounds of the present invention, and other methods for preparing compounds of the present invention are considered to be within the scope of the present invention. For example, the synthesis of those non-exemplified compounds according to the invention can be successfully accomplished by modification methods, such as appropriate protection of interfering groups, by use of other known reagents in addition to those described herein, or by some conventional modification of the reaction conditions, by those skilled in the art. In addition, the reactions disclosed herein or known reaction conditions are also well-known to be applicable to the preparation of other compounds of the present invention.
The structure of the compound is that nuclear magnetic resonance is adopted 1 H-NMR、 13 C-NMR or/and 19 F-NMR). 1 H-NMR、 13 C-NMR、 19 The F-NMR chemical shifts (δ) are given in parts per million (ppm). 1 H-NMR、 13 C-NMR、 19 The F-NMR was performed using Bruker Ultrashield-400 NMR spectrometer and Bruker Avance III HD 600 NMR spectrometer, with deuterated chloroform (CDCl) 3 ) Deuterated methanol (CD) 3 OD or MeOH-d 4 ) Or deuterated dimethyl sulfoxide (DMSO-d) 6 ). TMS (0 ppm) or chloroform (7.25 ppm) was used as a reference standard. When multiple peaks occur, the following abbreviations will be used: s (single, singlet), d (doublet ), t (triplet, multiplet), m (multiplet ), br (broadened, broad), dd (doublet of doublets, doublet), dt (doublet of triplets, doublet), td (triplet of doublets, triplet), brs (broadened singlet, broad singlet). Coupling constant J, in units of hertz (Hz).
Purification or resolution is typically performed using a Novasep pump 250 high performance liquid chromatograph.
LC-MS was determined using an Agilen-6120 Quadragupole LC/MS mass spectrometer.
Column chromatography generally uses 300-400 mesh silica gel of Qingdao ocean chemical industry as a carrier.
The starting materials for the present invention are known and commercially available from Shanghai remote Company (Shanghai Accela Company), an Naiji Company (Energy Company), the well-known Company (J & K), the Tianjin Alfa Company (Alfa Company), and the like, or may be synthesized using or according to methods known in the art.
The examples are not specifically described, and the reactions are all carried out under nitrogen atmosphere;
the nitrogen atmosphere means that the reaction bottle is connected with a nitrogen balloon or a steel kettle with the volume of about 1L;
the hydrogen atmosphere means that the reaction bottle is connected with a hydrogen balloon with the volume of about 1L or a stainless steel high-pressure reaction kettle with the volume of about 1L;
unless otherwise specified, in the examples, the solution means an aqueous solution;
unless otherwise specified in the examples, the reaction temperature was room temperature;
the room temperature is 20-30 ℃ unless otherwise specified in the examples.
The progress of the reaction in the examples was monitored by Thin Layer Chromatography (TLC) using the following system of developing agents: the volume ratio of the methylene chloride to the methanol system, the methylene chloride to the ethyl acetate system, the petroleum ether to the ethyl acetate system and the solvent is adjusted according to the polarity of the compound.
The system of eluent for column chromatography comprises: a: petroleum ether and ethyl acetate system, B: dichloromethane and ethyl acetate system, C: dichloromethane and methanol systems. The volume ratio of the solvent is adjusted according to the polarity of the compound, and can be adjusted by adding a small amount of ammonia water, acetic acid and the like.
HPLC refers to high performance liquid chromatography;
HPLC was performed using an agilent 1260 high pressure liquid chromatograph (Eclipse Plus C18, 4.6x150 mm,5um column);
HPLC test conditions: column temperature: PDA at 35 ℃): 210nm,254nm
Mobile phase: phase A: 0.1% phosphoric acid B phase: acetonitrile flow rate: 1.0mL/min
The mobile phase gradient is shown in table a:
table A
The LC/MS system for analysis in the biological test consisted of an Agilent 1200 series vacuum degassing furnace, a binary syringe pump, an orifice plate autosampler, a column oven, an Agilent G6430 tertiary quadrupole mass spectrometer with an electrospray ionization (ESI) source. Quantitative analysis was performed in MRM mode, and parameters of MRM transitions are shown in table B:
table B
Multiple reaction detection scan 490.2→383.1
Fracture voltage 230V
Capillary voltage 55V
Dryer temperature 350℃
Atomizer 0.28MPa
Dryer flow rate 10L/min
Analysis Using an Agilent XDB-C18, 2.1X130 mm, 3.5. Mu.M column, 5. Mu.L of sample was injected. Analysis conditions: the mobile phase was 0.1% formic acid in water (A) and 0.1% formic acid in methanol (B). The flow rate was 0.4mL/min. The mobile phase gradient is shown in table C:
Table C
Time Gradient of mobile phase B
0.5min 5%
1.0min 95%
2.2min 95%
2.3min 5%
5.0min Termination of
Also for analysis was an Agilent 6330 series LC/MS spectrometer equipped with a G1312A binary syringe pump, a G1367A autosampler and a G1314C UV detector; LC/MS spectrometers employ ESI radiation sources. The appropriate cation model treatment and MRM transformations were performed for each analyte using standard solutions for optimal analysis. Capcell MP-C18 column was used during the analysis, with the following specifications: 100X 4.6mm I.D., 5. Mu.M (Phenomenex, torrance, california, USA). The mobile phase was 5mM ammonium acetate, 0.1% aqueous methanol (A): 5mM ammonium acetate, 0.1% methanol acetonitrile solution (B) (70/30, v/v); the flow rate is 0.6mL/min; the column temperature is kept at room temperature; 20. Mu.L of sample was injected.
The following abbreviations are used throughout the present invention:
the following synthetic schemes describe the steps for preparing the disclosed compounds. Wherein, unless otherwise indicated, W has the meaning as described herein.
Synthetic scheme
Synthesis scheme 1
The compound (I-A) can be prepared by the method described in synthetic scheme 1. The compound (I-a) and the compound (I-b) are subjected to reductive amination in the presence of a reducing agent (e.g., sodium triacetoxyborohydride, decaborane, etc.) to give the compound (I-c). Removing the protecting group Boc on the amino group of the compound (I-c) to obtain the compound with the structure shown in the formula (I-A).
The invention will be further described by the following examples, which should not be construed as limiting the scope of the invention.
Examples
Preparation example
Example 1 (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (7-methanesulfonyl-2, 7-diazaspiro [3.5] nonan-2-yl) tetrahydropyran-3-amine
Step 1) 7-methanesulfonyl-2, 7-diazaspiro [3.5]]Nonane-2-carboxylic acid tert-butyl ester
2, 7-diazaspiro [3.5] nonane-2-carboxylic acid tert-butyl ester (0.50 g,1.5 mmol) was dissolved in tetrahydrofuran (8 mL), triethylamine (0.62 mL,4.5 mmol) was added, methanesulfonyl chloride (0.21 mL,2.7 mmol) was slowly dropped at 0℃and the reaction was maintained at 0℃for 1.5 hours. The reaction was quenched with water (10 mL), extracted with ethyl acetate (30 mL), the organic phase was washed with saturated sodium chloride solution (10 ml×3), dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residue was purified by silica gel column chromatography [ petroleum ether/ethyl acetate (v/v) =3/1 ] to give the title compound (0.67 g, yield 99%) as a white solid.
MS(ESI,pos.ion)m/z:249.2[M-55] +
Step 2) 7-methanesulfonyl-2, 7-diazaspiro [3.5]]Nonane hydrochloride
7-methanesulfonyl-2, 7-diazaspiro [3.5] nonane-2-carboxylic acid tert-butyl ester (0.67 g,2.2 mmol) was dissolved in ethyl acetate (2 mL), and an ethanol solution of hydrogen chloride (6 mL,2.0 mol/L) was added to react at room temperature for 1.5 hours. The mixture was filtered, and the solid was washed with ethyl acetate (5 mL) and the filter cake was collected to give the title compound (0.51 g, 96% yield) as a white solid.
MS(ESI,pos.ion)m/z:205.2[M-Cl] +
Step 3) N- [ (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (7-methanesulfonyl-2, 7-diazaspiro ring [3.5]Nonan-2-yl) tetrahydropyran-3-Base group]Carbamic acid tert-butyl ester
N- [ (2R, 3S) -2- (2, 5-difluorophenyl) -5-oxo-tetrahydropyran-3-yl]Tert-butyl carbamate (0.20 g,0.61 mmol) and 7-methanesulfonyl-2, 7-diazaspiro [3.5]]Nonane hydrochloride (0.15 g,0.62 mmol) was dissolved in N, N-dimethylacetamide (3 mL), and sodium triacetoxyborohydride (0.33 g,1.5 mmol) was added at 0deg.C under nitrogen and reacted at room temperature for 8 hours. The reaction was quenched by addition of ammonia/water (v/v=2/3, 10 ml), and the solid obtained by filtration was purified by silica gel column chromatography [100% ethyl acetate ]]Purification gave the title compound (0.17 g, 54% yield) as a yellow solid. MS (ESI, pos.ion) m/z 516.4[ M+H ]] +
Step 4) (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (7-methanesulfonyl-2, 7-diazaspiro [3.5]]Nong (nong) Alkan-2-yl) tetrahydropyran-3-amines
Tert-butyl N- [ (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (7-methanesulfonyl-2, 7-diazaspiro [3.5] nonan-2-yl) tetrahydropyran-3-yl ] carbamate (0.30 g,0.58 mmol) was dissolved in ethanol (0.5 mL), and an ethanol solution of hydrogen chloride (3 mL,2.0 mol/L) was added and reacted at room temperature for 1 hour. Concentrated under reduced pressure, ethanol (2.0 mL) was added, ph=8 was adjusted with sodium bicarbonate solid, filtered, the filtrate was concentrated, and the residue was purified by silica gel column chromatography [ dichloromethane/methanol (v/v) =10/1 ] to give the title compound (73 mg, yield 30%, HPLC purity 90.9%) as a white solid.
MS(ESI,pos.ion)m/z:415.9[M+H] +
1 H NMR(400MHz,CDCl 3 )δ(ppm)7.30(s,1H),7.07-6.93(m,2H),4.22(d,J=9.4Hz,1H),3.92(d,J=12.4Hz,1H),3.54(d,J=11.0Hz,1H),3.21(t,J=5.5Hz,4H),3.13(d,J=21.3Hz,4H),2.79(s,3H),2.50(s,1H),2.13(d,J=12.7Hz,1H),1.92(t,J=8.3Hz,4H),1.52-1.41(m,2H)。
Example 2 (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (7-methanesulfonyl-2, 7-diazaspiro [3.4] octan-2-yl) tetrahydropyran-3-amine
Step 1) 2, 7-diazaspiro [3.4]]Octane-7-carboxylic acid tert-butyl ester
Sodium bicarbonate (10.0 g,119 mmol) was added to a solution of tert-butyl 2, 7-diazaspiro [3.4] octane-7-carboxylate oxalate (3.0 g,9.9 mmol) in water (10 mL) and reacted at room temperature for 1.5 hours. The combined organic phases were washed with saturated sodium chloride solution (50 ml×2), dried over anhydrous sodium sulfate and concentrated by suction filtration to give the title compound (1.68 g, 80% yield) as a yellow oil.
MS(ESI,pos.ion)m/z:213.1[M+H] +
Step 2) O2-benzyl O7-tert-butyl 2, 7-diazaspiro [3.4]]Octane-2, 7-dicarboxylic acid ester
Triethylamine (2.1 mL,15.0 mmol) and benzyl chloroformate (2.2 mL,15.0 mmol) were added sequentially to a solution of tert-butyl 2, 7-diazaspiro [3.4] octane-7-carboxylate (1.6 g,7.5 mmol) in dichloromethane (20 mL) at 0deg.C, and reacted at room temperature for 21 hours. Water (5 mL) was added, the solution was separated, the organic phase was washed with saturated sodium chloride solution (5 ml×2), dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residue was purified by silica gel column chromatography [ petroleum ether/ethyl acetate (v/v) =3/1 ] to give the title compound (1.8 g, yield 69%) as a yellow oil.
MS(ESI,pos.ion)m/z:370.3[M+Na] +
1 H NMR(400MHz,CDCl 3 )δ(ppm)7.41-7.24(m,5H),5.11(s,2H),3.92(dd,J=23.0,9.3Hz,4H),3.53-3.30(m,4H),2.04(d,J=4.9Hz,2H),1.46(s,9H)。
Step 3) 2, 7-diazaspiro [3.4]]Octane-2-carboxylic acid benzyl ester
Trifluoroacetic acid (1.9 mL) was added to a solution of O2-benzyl O7-tert-butyl 2, 7-diazaspiro [3.4] octane-2, 7-dicarboxylic acid ester (0.85 g,2.45 mmol) in dichloromethane (8 mL) at 0 ℃ and reacted at room temperature for 1 hour. The reaction was quenched with saturated sodium bicarbonate solution (5 mL), extracted with dichloromethane/methanol (v/v=9/1, 10ml×3), and the combined organic phases were dried over anhydrous sodium sulfate and concentrated by suction to give the title compound (0.60 g, 99% yield) as a yellow oil.
MS(ESI,pos.ion)m/z:516.4[M+H] +
Step 4) 7-methanesulfonyl-2, 7-diazaspiro [3.4]]Octane-2-carboxylic acid benzyl ester
Benzyl 2, 7-diazaspiro [3.4] octane-2-carboxylate (0.60 g,2.4 mmol) was dissolved in dichloromethane (8 mL), triethylamine (0.70 mL,5.0 mmol) was added, methanesulfonyl chloride (0.40 mL,5.0 mmol) was slowly added dropwise at 0deg.C, and the reaction was carried out at room temperature for 2.5 hours. The reaction was quenched with water (10 mL), extracted with dichloromethane (30 mL), the organic phase was washed with saturated sodium chloride solution (10 ml×3), dried over anhydrous sodium sulfate, concentrated by suction, and the residue was purified by silica gel column chromatography [ petroleum ether/ethyl acetate (v/v) =1/1 ] to give the title compound (0.69 g, yield 87%) as a colorless oil.
Step 5) 7-methanesulfonyl-2, 7-azaspiro [3.4] ]Octane (octane)
10% palladium on carbon (0.3 g,0.28 mmol) was added to a solution of benzyl 7-methanesulfonyl-2, 7-diazaspiro [3.4] octane-2-carboxylate (0.69 g,2.13 mmol) in methanol (10 mL), hydrogen was replaced, and the reaction was hydrogenated at room temperature for 2 hours. The mixture was filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography [ dichloromethane/methanol (v/v) =3/1 ] to give the title compound (0.30 g, yield 74%) as a colorless oil.
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)3.49(d,J=7.5Hz,2H),3.38-3.35(m,2H),3.33(s,2H),3.22(t,J=6.9Hz,2H),2.87(s,3H),2.04(t,J=6.9Hz,2H),1.23(d,J=3.8Hz,1H)。
Step 6) N- [ (2R, 3S, 5R) -2- (2, 5-trifluorophenyl) -5- (7-methanesulfonyl-2, 7-diazaspiro ring [3.4]Octan-2-yl) tetrahydropyran-3-yl]Carbamic acid tert-butyl ester
N- [ (2R, 3S) -2- (2, 5-difluorophenyl) -5-oxo-tetrahydropyran-3-yl ] carbamic acid tert-butyl ester (0.34 g,1.0 mmol) and 7-methanesulfonyl-2, 7-diazaspiro [3.5] nonane hydrochloride (0.15 g,0.79 mmol) were dissolved in methanol (4 mL), reacted for 6 hours, then, with nitrogen protection, decaborane (0.06 g,0.48 mmol) was added at 0deg.C and reacted at room temperature for 12 hours. The mixture was filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography [ dichloromethane/methanol (v/v) =10/1 ] to give the title compound (0.32 g, yield 81%) as a yellow solid.
MS(ESI,pos.ion)m/z:502.3[M+H] +
Step 7) (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (7-methanesulfonyl-2, 7-diazaspiro [3.5] ]Octyl Alkan-2-yl) tetrahydropyran-3-amines
Trifluoroacetic acid (1.0 mL) was added to a solution of tert-butyl N- [ (2 r,3s,5 r) -2- (2, 5-trifluorophenyl) -5- (7-methanesulfonyl-2, 7-diazaspiro [3.4] oct-2-yl) tetrahydropyran-3-yl ] carbamate (0.32 g,0.32 mmol) in dichloromethane (10 mL) at 0 ℃ for 12 hours at room temperature. The reaction was quenched with saturated sodium bicarbonate solution (5 mL), extracted with dichloromethane/methanol (v/v=9/1, 10ml×3), the combined organic phases were dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residue was purified by silica gel column chromatography [ dichloromethane/methanol (v/v) =10/1 ] to give the title compound (70 mg, yield 55%, HPLC purity 97.3%) as a colorless oil.
MS(ESI,pos.ion)m/z:402.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)7.28-7.09(m,3H),4.06(dd,J=23.5,9.4Hz,1H),3.82(dd,J=10.6,2.3Hz,1H),3.31(s,2H),3.27-3.20(m,2H),3.18-3.07(m,4H),2.98(t,J=10.5Hz,1H),2.88(d,J=5.4Hz,3H),2.76-2.66(m,1H),2.45-2.31(m,1H),2.05(dt,J=14.1,6.9Hz,3H),1.24(s,2H),1.05(dd,J=23.3,11.6Hz,1H)。
EXAMPLE 3 (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- [ (1S, 4S) -2-methanesulfonyl-2, 5-diazabicyclo [2.2.1] heptan-5-yl ] tetrahydropyran-3-amine
Step 1) (1S, 4S) -2-methanesulfonyl-2, 5-diazabicyclo [2.2.1]Heptane-5-carboxylic acid tert-butyl ester
Triethylamine (0.21 mL,1.5 mmol) and methanesulfonyl chloride (0.1 mL,1 mmol) were added to a solution of tert-butyl (1S, 4S) 2, 5-diazabicyclo [2.2.1] heptane-2-carboxylate (0.2 g,1 mmol) in tetrahydrofuran (5 mL) at 0deg.C and reacted for 20 hours at room temperature. Filtration and concentration of the filtrate gave the title compound (0.36 g, 100% yield) as a white solid.
Step 2) (1S, 4S) -2-methanesulfonyl-2, 5-diazabicyclo [2.2.1]Heptane (heptane)
An ethanol solution of hydrogen chloride (4 mL,2.0 mol/L) was added to a solution of tert-butyl (1S, 4S) -2-methanesulfonyl-2, 5-diazabicyclo [2.2.1] heptane-5-carboxylate (0.15 g,0.54 mmol) in ethanol (2 mL) at 0deg.C, and reacted at room temperature for 1 hour. The solvent was removed by concentration under reduced pressure, the residue was dissolved in ethanol (5 mL), ph=8 was adjusted with sodium bicarbonate solid, filtration, and the filtrate was concentrated to give the title compound (95 mg, yield 99%) as a white solid.
Step 3) N- [ (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- [ (1S, 4S) -2-methanesulfonyl-2, 5-diaza-ne Bicyclo [2.2.1]Heptan-5-yl]Tetrahydropyran-3-yl]Carbamic acid tert-butyl ester
N- [ (2R, 3S) -2- (2, 5-difluorophenyl) -5-oxo-tetrahydropyran-3-yl ] carbamic acid tert-butyl ester (0.19 g,0.56 mmol) and (1S, 4S) -2-methanesulfonyl-2, 5-diazabicyclo [2.2.1] heptane (0.10 g,0.24 mmol) were dissolved in N, N-dimethylacetamide (1 mL), and sodium triacetoxyborohydride (0.31 g,1.4 mmol) was added under nitrogen protection at 0℃for 8 hours at room temperature. The reaction was quenched with water (5 mL) at 0 ℃ and extracted with dichloromethane/methanol (v/v=9/1, 10ml×2), the combined organic phases were dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residue was purified by silica gel column chromatography [ ethyl acetate/methanol (v/v) =20/1 ] to give the title compound (0.73 g, yield 26%) as a pale yellow solid.
MS(ESI,pos.ion)m/z:488.3[M+H] +
Step 4) (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- [ (1S, 4S) -2-methanesulfonyl-2, 5-diaza-odi-ne Ring [2.2.1]Heptan-5-yl]Tetrahydropyran-3-amines
Tert-butyl N- [ (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- [ (1S, 4S) -2-methanesulfonyl-2, 5-diazabicyclo [2.2.1] heptan-5-yl ] tetrahydropyran-3-yl ] carbamate (0.17 g,0.35 mmol) was dissolved in methanol (0.5 mL), and a methanol solution of hydrogen chloride (5 mL,3.0 mol/L) was added to react at room temperature for 1 hour. The residue was concentrated under reduced pressure, dissolved in dichloromethane/methanol/water (v/v/v=9/1/10, 20 ml), ph=8 adjusted with saturated sodium bicarbonate solution, extracted with dichloromethane/methanol (v/v=9/1, 20ml×3), the combined organic phases were dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residue was purified by silica gel column chromatography [ dichloromethane/methanol (v/v) =10/1 ] to give the title compound (85 mg, yield 63%, HPLC purity 90.2%) as a white solid.
MS(ESI,pos.ion)m/z:388.3[M+H] +
1 H NMR(400MHz,CDCl 3 )δ(ppm)7.20-7.10(m,1H),7.07-6.94(m,2H),4.36-4.22(m,2H),4.07(ddd,J=26.0,11.1,2.1Hz,1H),3.79(d,J=43.3Hz,1H),3.61(d,J=9.5Hz,1H),3.37-3.21(m,2H),3.16-3.06(m,1H),2.94-2.86(m,5H),2.74(d,J=9.6Hz,1H),2.56(s,2H),2.40-2.28(m,1H),1.91(d,J=9.8Hz,1H),1.77(d,J=9.9Hz,1H),1.51-1.39(m,1H)。
Example 4 (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (1-isopropylspiro [6, 7-dihydro-4H-indazol-5, 4 '-piperidin ] -1' -yl) tetrahydropyran-3-amine
Step 1) 3-oxo-9-azaspiro [5.5 ]]Undec-4-ene-9-carboxylic acid benzyl ester
Benzyl 4-formylpiperidine-1-carboxylate (30.0 g,121 mmol) was dissolved in toluene (200 mL), p-toluenesulfonic acid (3.4 g,18 mmol) and butenone (20.0 mL,247 mmol) were added and reacted at 80℃for 20 hours. The reaction solution was cooled to room temperature, washed with saturated sodium bicarbonate solution (600 mL), and the organic phase was dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residue was purified by silica gel column chromatography [ ethyl acetate/petroleum ether (v/v) =1/4 ] to give the title compound (15 g, yield 41%) as a yellow oil.
MS(ESI,pos.ion)m/z:300.3[M+H] +
Step 2) 8- (II)Methylaminomethylene) -9-oxo-3-azaspiro [5.5]Undec-10-ene-3-carboxylic acid benzyl ester Esters of
Benzyl 3-oxo-9-azaspiro [5.5] undec-4-ene-9-carboxylate (14.5 g,48.4 mmol) was dissolved in toluene (200 mL), tris (dimethylamino) methane (25 mL) was added, and the mixture was heated to 100deg.C to react for 15 hours. The reaction solution was concentrated under reduced pressure to give the title compound (18 g, yield 99%) as a dark red oil.
Step 3) spiro [1, 4-indolizole-5, 4' -piperidine]-1' -Carboxylic acid benzyl ester
Benzyl 8- (dimethylaminomethylene) -9-oxo-3-azaspiro [5.5] undec-10-ene-3-carboxylate (30.0 g,121 mmol) was dissolved in absolute ethanol (200 mL), hydrazine hydrate (3.0 mL,62 mmol) and acetic acid (3.0 mL,50 mmol) were added in sequence and the temperature was raised to 80℃for 4 hours. The reaction solution was cooled to room temperature, concentrated under reduced pressure, saturated sodium bicarbonate solution was added to the residue to adjust ph=8, extracted with ethyl acetate (50 ml×4), the combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residue was purified by silica gel column chromatography [ ethyl acetate/petroleum ether (v/v) =1/1 ] to give the title compound (12.8 g, yield 78%) as a pale yellow oil.
MS(ESI,pos.ion)m/z:324.1[M+H] +
Step 4) 1-Isopropylspiro [ 4H-indazole-5, 4' -piperidines ]-1' -Carboxylic acid benzyl ester
Spiro [1, 4-indolizole-5, 4 '-piperidine ] -1' -carboxylic acid benzyl ester (2.4 g,7.4 mmol) was dissolved in N, N-dimethylformamide (8 mL), followed by addition of potassium carbonate (1.2 g,8.7 mmol) and 2-iodopropane (1.1 mL,11 mmol) and reacted at room temperature for 18 hours. The reaction solution was concentrated under reduced pressure, water (20 mL) was added to the residue, extracted with ethyl acetate (50 ml×3), the combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residue was purified by silica gel column chromatography [ ethyl acetate/petroleum ether (v/v) =1/6 ] to give the title compound (1.0 g, yield 37%) as a yellow oil.
MS(ESI,pos.ion)m/z:366.1[M+H] +
Step 5) 1-Isopropylspiro [6, 7-dihydro-4H-indazole-5, 4' -piperidine]
Benzyl 1-isopropyl spiro [ 4H-indazole-5, 4 '-piperidine ] -1' -carboxylate (1.28 g,3.50 mmol) was dissolved in anhydrous methanol (10 mL), 10% palladium on carbon (128 mg) was added to replace hydrogen, and hydrogenation was carried out for 3 hours. The mixture was filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography [ dichloromethane/methanol (v/v) =5/1 ] to give the title compound (0.49 g, yield 60%) as a yellow oil.
Step 6) N- [ (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (1-isopropylspiro [6, 7-dihydro-4H-indazole ] 5,4' -piperidine]-1' -yl) tetrahydropyran-3-yl ]Carbamic acid tert-butyl ester
N- [ (2R, 3S) -2- (2, 5-difluorophenyl) -5-oxo-tetrahydropyran-3-yl ] carbamic acid tert-butyl ester (0.40 g,1.22 mmol) and 1-isopropylspiro [6, 7-dihydro-4H-indazole-5, 4' -piperidine ] (0.30 g,1.28 mmol) were dissolved in formaldehyde (10 mL) and after 4 hours at room temperature decaborane (0.11 g,0.92 mmol) was added and the reaction continued at room temperature for 8 hours. The reaction was quenched with water (5 mL) at 0 ℃ and extracted with ethyl acetate (10 ml×2), the combined organic phases were dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residue was purified by silica gel column chromatography [ petroleum ether/ethyl acetate (v/v) =1/1 ] to give the title compound (0.10 g, yield 15%) as a white solid.
MS(ESI,pos.ion)m/z:545.4[M+H] +
Step 7) (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (1-isopropylspiro [6, 7-dihydro-4H-indazole-5, 4' -piperidines]-1' -yl) tetrahydropyran-3-amine
Tert-butyl N- [ (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (1-isopropylspiro [6, 7-dihydro-4H-indazol-5, 4 '-piperidin ] -1' -yl) tetrahydropyran-3-yl ] carbamate (0.10 g,0.18 mmol) was dissolved in dichloromethane (5 mL), trifluoroacetic acid (0.14 mL,1.9 mmol) was added and reacted at room temperature for 3 hours. The residue was concentrated under reduced pressure, dissolved in dichloromethane/methanol/water (v/v/v=9/1/10, 10 ml), ph=8 adjusted with saturated sodium bicarbonate solution, extracted with dichloromethane/methanol (v/v=9/1, 10ml×3), the combined organic phases were dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residue was purified by silica gel column chromatography [ dichloromethane/methanol (v/v) =10/1 ] to give the title compound (26 mg, yield 32%, HPLC purity 93.8%) as a white solid.
MS(ESI,pos.ion)m/z:446.3[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)8.34(s,2H),7.35(dd,J=20.7,14.4Hz,2H),4.58(d,J=10.1Hz,1H),4.45(d,J=8.4Hz,1H),4.36(dd,J=10.7,6.4Hz,1H),4.04(dt,J=12.9,6.5Hz,1H),3.69(dd,J=27.9,17.8Hz,3H),3.18-3.03(m,2H),2.73(d,J=8.1Hz,1H),2.57(d,J=22.6Hz,2H),2.29(s,1H),2.08(dd,J=20.7,9.1Hz,1H),2.02-1.95(m,1H),1.71(s,3H),1.54(dd,J=14.1,7.5Hz,2H),1.42-1.26(m,6H),1.23(s,2H),0.91-0.84(m,2H)。
Example 5 (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (1, 3,4, 5-tetrahydropyrido [4,3-b ] indol-2-yl) tetrahydropyran-3-amine
Step 1) 1,3,4, 5-tetrahydropyrido [4,3-b]Indole-2-carboxylic acid tert-butyl ester
1, 3-dimethylurea (8.5 g,95 mmol) and tartaric acid (3.6 g,24 mmol) were mixed, heated to 110℃until the solid melted and cleared, phenylhydrazine (1.1 g,10 mmol) and N-t-butoxycarbonyl-4-piperidone (2.0 g,10 mmol) were added and the reaction was continued for 6 hours. The reaction was quenched with water (50 mL), extracted with ethyl acetate (80 mL), the organic phase dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residue purified by silica gel column chromatography [ petroleum ether/ethyl acetate (v/v) =5/1 ] to give the title compound (0.24 g, yield 99%) as a white solid.
1 H NMR(400MHz,CDCl 3 )δ(ppm)8.22(s,1H),7.48(s,1H),7.33(d,J=7.6Hz,1H),7.23-7.07(m,2H),4.69(s,2H),3.85(s,2H),2.83(s,2H),1.56(s,9H)。
Step 2) 2,3,4, 5-tetrahydro-1H-pyrido [4,3-b]Indole hydrochloride
Tert-butyl 1,3,4, 5-tetrahydropyrido [4,3-b ] indole-2 carboxylate (0.24 g,0.88 mmol) was dissolved in ethyl acetate (0.5 mL), and a solution of hydrogen chloride in ethyl acetate (2 mL,4.0 mol/L) was added to react at room temperature for 3 hours, and concentrated under reduced pressure to give the title compound (0.18 g,9 yield 98%).
Step 3) N- [ (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (1, 3,4, 5-tetrahydropyrido [4, 3-b)]Indoles and their use Indol-2-yl) tetrahydropyran-3-yl]Carbamic acid tert-butyl ester Butyl ester
N- [ (2R, 3S) -2- (2, 5-difluorophenyl) -5-oxo-tetrahydropyran-3-yl ] carbamic acid tert-butyl ester (0.15 g,0.46 mmol) and 2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole hydrochloride (0.18 g,0.86 mmol) were dissolved in N, N-dimethylacetamide (1.5 mL), and sodium triacetoxyborohydride (0.20 g,0.91 mmol) was added under nitrogen protection at 0deg.C for 8 hours at room temperature. The reaction was quenched with water (10 mL) at 0 ℃, filtered, and the resulting solid was purified by silica gel column chromatography [ petroleum ether/ethyl acetate (v/v) =2/1 ] to give the title compound (60 mg, yield 14%) as a yellow solid.
MS(ESI,pos.ion)m/z:484.6[M+H] +
Step 4) (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (1, 3,4, 5-tetrahydropyrido [4, 3-b)]Indole-2- Base) tetrahydropyran-3-amine
Tert-butyl N- [ (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (1, 3,4, 5-tetrahydropyrido [4,3-b ] indol-2-yl) tetrahydropyran-3-yl ] carbamate (60 mg,0.12 mmol) was dissolved in ethyl acetate (0.5 mL), and an ethyl acetate solution of hydrogen chloride (2 mL,4.0 mol/L) was added to react at room temperature for 3 hours. The solid sodium bicarbonate was added to adjust ph=8, filtered, the filtrate was concentrated, and the residue was purified by silica gel column chromatography [ dichloromethane/methanol (v/v) =9/1 ] to give the title compound (44 mg, yield 92%, HPLC purity 90.2%) as a yellow solid.
MS(ESI,pos.ion)m/z:384.5[M+H] +
1 H NMR(400MHz,CDCl 3 )δ(ppm)7.86(s,1H),7.40(d,J=7.6Hz,1H),7.29(d,J=7.8Hz,1H),7.16(m,2H),7.09(d,J=7.1Hz,1H),7.01(m,2H),4.36-4.31(m,1H),4.25(d,J=9.4Hz,1H),4.00-3.88(m,2H),3.59(t,J=10.8Hz,1H),3.08(t,J=5.6Hz,4H),2.93-2.82(m,3H),2.56-2.46(m,1H)。
Example 6 (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (8-methoxy-1, 3,4, 5-tetrahydropyrido [4,3-b ] indol-2-yl) tetrahydropyran-3-amine
Step 1) 8-methoxy-1, 3,4, 5-tetrahydropyrido [4,3-b ]]Indole-2-carboxylic acid tert-butyl ester
1, 3-dimethylurea (7.88 g,89.4 mmol) and tartaric acid (3.38 g,22.5 mmol) were mixed, heated to 75℃until the solid melted and cleared, and (4-methoxyphenyl) hydrazine hydrochloride (1.40 g,8.0 mmol) and N-t-butoxycarbonyl-4-piperidone (1.5 g,7.5 mmol) were added and the reaction was continued for 3 hours. The reaction was quenched with water (10 mL), extracted with dichloromethane (20 ml×2), and the combined organic phases were washed with saturated sodium chloride solution (30 mL), dried over anhydrous sodium sulfate, concentrated by suction, and the residue was purified by silica gel column chromatography [ petroleum ether/ethyl acetate (v/v) =2/1 ] to give the title compound (1.53 g, yield 67%) as a pale yellow solid.
1 H NMR(400MHz,CDCl 3 )δ(ppm)8.10(d,J=22.9Hz,1H),7.21(d,J=8.7Hz,1H),6.92(s,1H),6.87-6.75(m,1H),4.64(s,2H),3.85(d,J=19.8Hz,5H),2.81(t,J=5.5Hz,2H),1.54(s,9H)。
Step 2) 8-methoxy-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b]Indole compounds
Tert-butyl 8-methoxy-1, 3,4, 5-tetrahydropyrido [4,3-b ] indole-2-carboxylate (0.62 g,2.1 mmol) was dissolved in dichloromethane (2 mL), and a methanol solution of hydrogen chloride (8 mL,3.0 mol/L) was added to react at room temperature for 3 hours. The reaction solution was adjusted to ph=9 with saturated sodium bicarbonate solution, extracted with ethyl acetate/methanol (v/v=5/1, 20ml×4), the combined organic phases were dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residual solid was purified by silica gel column chromatography [ ethyl acetate/methanol (v/v) =5/1 ] to give the title compound (0.27 g, yield 65%) as a white solid.
Step 3) N- [ (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (8-methoxy-1, 3,4, 5-tetrahydropyrido) [4,3-b]Indol-2-yl) tetrahydropyran-3-yl]Carbamic acid tert-butyl ester
N- [ (2R, 3S) -2- (2, 5-difluorophenyl) -5-oxo-tetrahydropyran-3-yl ] carbamic acid tert-butyl ester (0.48 g,1.5 mmol) and 8-methoxy-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole (0.27 g,1.3 mmol) were dissolved in N, N-dimethylacetamide (4 mL), and sodium triacetoxyborohydride (0.71 g,3.4 mmol) was added under nitrogen protection at 0deg.C for 8 hours at room temperature. The reaction was quenched with aqueous ammonia/water (v/v=2/3, 10 ml), filtered, and the resulting solid was purified by silica gel column chromatography [ petroleum ether/ethyl acetate (v/v) =2/1 ] to give the title compound 0.14g, yield 21%) as a yellow solid.
MS(ESI,pos.ion)m/z:514.5[M+H] +
Step 4) (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (8-methoxy-1, 3,4, 5-tetrahydropyrido [4,3- ] b]Indol-2-yl) tetrahydropyran-3-amines
Tert-butyl N- [ (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (8-methoxy-1, 3,4, 5-tetrahydropyrido [4,3-b ] indol-2-yl) tetrahydropyran-3-yl ] carbamate (0.45 g,0.87 mmol) was dissolved in dichloromethane (0.5 mL), and a methanol solution of hydrogen chloride (5 mL,3.0 mol/L) was added and reacted at room temperature for 1 hour. The solid sodium bicarbonate was added to adjust ph=9, filtered, the filtrate was concentrated, and the residue was purified by silica gel column chromatography [ dichloromethane/methanol (v/v) =9/1 ] to give the title compound (0.16 g, yield 44%, HPLC purity 92.2%) as a pale yellow solid.
MS(ESI,pos.ion)m/z:414.5[M+H] +
1 H NMR(400MHz,CDCl 3 )δ(ppm)7.70(s,1H),7.20(d,J=8.7Hz,2H),7.06(d,J=4.5Hz,2H),6.88(d,J=2.3Hz,1H),6.81(dd,J=8.7,2.4Hz,1H),4.35-4.30(m,1H),4.25(d,J=9.3Hz,1H),3.92(d,J=8.7Hz,2H),3.87(s,3H),3.58(d,J=10.8Hz,1H),3.07(t,J=5.6Hz,4H),2.89(d,J=5.7Hz,3H),2.53(dd,J=12.1,2.1Hz,1H)。
Example 7 (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (8-fluoro-1, 3,4, 5-tetrahydropyrido [4,3-b ] indol-2-yl) tetrahydropyran-3-amine
Step 1) 8-fluoro-1, 3,4, 5-tetrahydropyrido [4,3-b ]]Indole-2-carboxylic acid tert-butyl ester
1, 3-dimethylurea (10.5 g,119 mmol) and tartaric acid (4.5 g,30 mmol) were mixed, heated to 85℃until the solid melted and cleared, and (4-fluorophenyl) hydrazine hydrochloride (1.8 g,11.1 mmol) and N-t-butoxycarbonyl-4-piperidone (2.0 g,10.0 mmol) were added and the reaction was continued for 3 hours. The reaction was quenched with water (10 mL), extracted with dichloromethane (20 ml×2), and the combined organic phases were washed with saturated sodium chloride solution (30 mL), dried over anhydrous sodium sulfate, concentrated by suction, and the residue was purified by silica gel column chromatography [ petroleum ether/ethyl acetate (v/v) =2/1 ] to give the title compound (0.28 g, yield 8.8%) as a pale yellow solid.
1 H NMR(400MHz,CDCl 3 )δ(ppm)7.92(s,1H),7.27-7.18(m,1H),7.11(d,J=9.1Hz,1H),6.91(d,J=2.1Hz,1H),4.61(s,2H),3.83(s,2H),2.84(s,2H),1.53(s,9H)。
Step 2) 8-fluoro-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b]Indole compounds
Tert-butyl 8-fluoro-1, 3,4, 5-tetrahydropyrido [4,3-b ] indole-2-carboxylate (0.25 g,0.87 mmol) was dissolved in dichloromethane (1 mL), and a methanol solution of hydrogen chloride (4 mL,3.0 mol/L) was added and reacted at room temperature for 2 hours. The reaction solution was adjusted ph=9 with sodium bicarbonate solid, filtered, the filtrate was concentrated, and the residual solid was purified by silica gel column chromatography [ ethyl acetate/methanol (v/v) =6/1 ] to give the title compound (0.17 g, yield 99%) as a yellow solid.
Step 3) N- [ (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (8-fluoro-1, 3,4, 5-tetrahydropyrido [4,3- ] b]Indol-2-yl) tetrahydropyran-3-yl]Carbamic acid tert-butyl ester
N- [ (2R, 3S) -2- (2, 5-difluorophenyl) -5-oxo-tetrahydropyran-3-yl ] carbamic acid tert-butyl ester (0.30 g,0.93 mmol) and 8-fluoro-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole (0.16 g,0.84 mmol) were dissolved in N, N-dimethylacetamide (3 mL), and sodium triacetoxyborohydride (0.45 g,2.1 mmol) was added under nitrogen protection at 0deg.C for 8 hours at room temperature. The reaction was quenched with aqueous ammonia/water (v/v=2/3, 10 ml), filtered, and the resulting solid was purified by silica gel column chromatography [ petroleum ether/ethyl acetate (v/v) =2/1 ] to give the title compound (83 mg, yield 20%) as a pale yellow solid.
MS(ESI,pos.ion)m/z:502.1[M+H] +
Step 4) (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (8-fluoro-1, 3,4, 5-tetrahydropyrido [4, 3-b)]Indoles and their use Indol-2-yl) tetrahydropyran-3-amines
Tert-butyl N- [ (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (8-fluoro-1, 3,4, 5-tetrahydropyrido [4,3-b ] indol-2-yl) tetrahydropyran-3-yl ] carbamate (75 mg,0.15 mmol) was dissolved in dichloromethane (0.5 mL), and a methanol solution of hydrogen chloride (3 mL,3.0 mol/L) was added and reacted at room temperature for 1 hour. The solid sodium bicarbonate was added to adjust ph=9, filtered, the filtrate was concentrated, and the residue was purified by silica gel column chromatography [ dichloromethane/methanol (v/v) =9/1 ] to give the title compound (0.57 g, yield 95%, HPLC purity 97.7%) as a pale yellow solid.
MS(ESI,pos.ion)m/z:402.5[M+H] +
1 H NMR(400MHz,CDCl 3 )δ(ppm)7.84(s,1H),7.20(d,J=4.2Hz,2H),7.06(d,J=9.2Hz,3H),6.89(td,J=9.0,2.3Hz,1H),4.34(d,J=8.2Hz,1H),4.25(d,J=9.5Hz,1H),3.89(d,J=7.8Hz,2H),3.59(s,1H),3.07(t,J=5.4Hz,4H),2.90(d,J=4.8Hz,3H),2.53-2.39(m,1H)。
Example 8 (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (8-methoxy-5-methanesulfonyl-3, 4-dihydro-1H-pyrido [4,3-b ] indol-2-yl) tetrahydropyran-3-amine
Step 1) 8-methoxy-5-methanesulfonyl-3, 4-dihydro-1H-pyrido [4,3-b]Indole-2-carboxylic acid tert-butyl ester
N-butyllithium (0.90 mL,2.2 mmol) was slowly added dropwise to a solution of tert-butyl 8-methoxy-1, 3,4, 5-tetrahydropyrido [4,3-b ] indole-2-carboxylate (0.50 g,1.7 mmol) in anhydrous tetrahydrofuran (15 mL) at-78℃under nitrogen protection, and after reacting for 1 hour, methanesulfonyl chloride (0.26 mL,3.4 mmol) was slowly added dropwise, and the reaction was continued for 2 hours. The reaction was quenched by addition of saturated ammonium chloride solution (10 mL), extracted with ethyl acetate (20 mL), the organic phase was washed with saturated sodium chloride solution (20 mL), dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residue was purified by silica gel column chromatography [ petroleum ether/ethyl acetate (v/v) =1/1 ] to give the title compound (0.25 g, yield 40%) as a white solid.
1 H NMR(400MHz,CDCl 3 )δ(ppm)7.84(d,J=9.0Hz,1H),6.89(dd,J=9.0,2.3Hz,1H),6.85(d,J=2.2Hz,1H),4.54(s,2H),3.84(s,3H),3.76(t,J=5.2Hz,2H),3.02(s,2H),2.94(s,3H),1.51(s,9H)。
Step 2) 8-methoxy-5-methanesulfonyl-1, 2,3, 4-tetrahydropyrido [4,3-b ]]Indole compounds
Tert-butyl 8-methoxy-5-methanesulfonyl-3, 4-dihydro-1H-pyrido [4,3-b ] indole-2-carboxylate (0.23 g,0.60 mmol) was dissolved in dichloromethane (0.5 mL), and a methanol solution of hydrogen chloride (4 mL,3.0 mol/L) was added and reacted at room temperature for 2 hours. The reaction solution was adjusted ph=9 with sodium bicarbonate solid, filtered, and the filtrate was concentrated, and the residual solid was purified by silica gel column chromatography [ ethyl acetate/methanol (v/v) =9/1 ] to give the title compound (0.15 g, yield 89%) as a white solid.
Step 3) N- [ (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (8-methoxy-5-methanesulfonyl-3, 4-dihydro-o-f-3 1H-pyrido [4,3-b ]]Indol-2-yl) tetrahydropyran-3-yl]Carbamic acid tert-butyl ester
N- [ (2R, 3S) -2- (2, 5-difluorophenyl) -5-oxo-tetrahydropyran-3-yl ] carbamic acid tert-butyl ester (0.18 g,0.56 mmol) and 8-methoxy-5-methanesulfonyl-1, 2,3, 4-tetrahydropyrido [4,3-b ] indole (0.14 g,0.51 mmol) were dissolved in N, N-dimethylacetamide (2 mL), and sodium triacetoxyborohydride (0.27 g,1.3 mmol) was added under nitrogen protection at 0℃for 8 hours at room temperature. The reaction was quenched with aqueous ammonia/water (v/v=2/3, 10 ml), filtered, and the resulting solid was purified by silica gel column chromatography [ petroleum ether/ethyl acetate (v/v) =2/1 ] to give the title compound (0.30 g, yield 99%) as a pale yellow solid.
Step 4) (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (8-methoxy-5-methanesulfonyl-3, 4-dihydro-1H-) Pyrido [4,3-b]Indol-2-yl) tetrahydropyran-3-amines
Tert-butyl N- [ (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (8-methoxy-5-methanesulfonyl-3, 4-dihydro-1H-pyrido [4,3-b ] indol-2-yl) tetrahydropyran-3-yl ] carbamate (0.30 g,0.51 mmol) was dissolved in dichloromethane (0.5 mL), and a methanol solution of hydrogen chloride (5 mL,3.0 mol/L) was added and reacted at room temperature for 1 hour. The solid sodium bicarbonate was added to adjust ph=9, filtered, the filtrate was concentrated, and the residue was purified by silica gel column chromatography [ dichloromethane/methanol (v/v) =9/1 ] to give the title compound (0.15 g, yield 60%, HPLC purity 91.3%) as a pale yellow solid.
MS(ESI,pos.ion)m/z:492.2[M+H] +
1 H NMR(400MHz,CDCl 3 )δ(ppm)7.72(s,1H),7.20(t,J=7.4Hz,1H),7.07(td,J=9.2,4.8Hz,1H),7.01(dd,J=11.6,7.3Hz,1H),6.88(d,J=1.9Hz,1H),6.81(dd,J=8.7,2.2Hz,1H),4.36(d,J=10.7Hz,1H),4.26(d,J=9.4Hz,1H),4.00-3.90(m,2H),3.87(s,3H),3.62(t,J=10.7Hz,1H),3.10(d,J=5.3Hz,4H),2.88(d,J=17.4Hz,3H),2.67(s,3H),2.54(d,J=9.9Hz,1H)。
Example 9 (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (7-methoxy-3, 4-dihydro-1H-pyrrolo [3,4-b ] indol-2-yl) tetrahydropyran-3-amine
Step 1) 7-methoxy-3, 4-dihydro-1H-pyrrolo [3,4-b]Indole-2-carboxylic acid tert-butyl ester
1, 3-dimethylurea (7.18 g,81.5 mmol) and tartaric acid (3.08 g,20.5 mmol) were mixed, heated to 85℃until the solid melted and cleared, and (4-methoxyphenyl) hydrazine hydrochloride (2.1 g,12 mmol) and 1-t-butoxycarbonyl-3-pyrrolidone (2.0 g,11 mmol) were added and the reaction was continued for 3 hours. The reaction was quenched with water (30 mL), extracted with ethyl acetate (50 ml×2), and the combined organic phases were washed with saturated sodium chloride solution (30 mL), dried over anhydrous sodium sulfate, concentrated by suction, and the residue was purified by silica gel column chromatography [ petroleum ether/ethyl acetate (v/v) =2/1 ] to give the title compound (0.22 g, yield 6.9%) as a yellow solid.
Step 2) 7-methoxy-1, 2,3, 4-tetrahydropyrrolo [3,4-b]Indole compounds
7-methoxy-3, 4-dihydro-1H-pyrrolo [3,4-b ] indole-2-carboxylic acid tert-butyl ester (0.21 g,0.73 mmol) was dissolved in dichloromethane (1 mL), and a methanol solution of hydrogen chloride (6 mL,3.0 mol/L) was added and reacted at room temperature for 2 hours. The reaction solution was adjusted to ph=9 with saturated sodium bicarbonate solution, extracted with ethyl acetate/methanol (v/v=5/1, 20ml×4), the combined organic phases were dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residual solid was purified by silica gel column chromatography [ ethyl acetate/methanol (v/v) =5/1 ] to give the title compound (0.11 g, yield 82%) as a pale yellow solid.
MS(ESI,pos.ion)m/z:189.2[M+H] +
Step 3) N- [ (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (7-methoxy-3, 4-dihydro-1H-pyrrolo [3,4-b]Indol-2-yl) tetrahydropyran-3-yl]Carbamic acid tert-butyl ester
N- [ (2R, 3S) -2- (2, 5-difluorophenyl) -5-oxo-tetrahydropyran-3-yl ] carbamic acid tert-butyl ester (0.21 g,0.64 mmol) and 7-methoxy-1, 2,3, 4-tetrahydropyrrolo [3,4-b ] indole (0.11 g,0.58 mmol) were dissolved in N, N-dimethylacetamide (1 mL), and sodium triacetoxyborohydride (0.31 g,1.5 mmol) was added under nitrogen protection at 0deg.C for 8 hours at room temperature. The reaction was quenched with aqueous ammonia/water (v/v=2/3, 10 ml), filtered, and the resulting solid was purified by silica gel column chromatography [ petroleum ether/ethyl acetate (v/v) =2/1 ] to give the title compound (60 mg, yield 21%) as a yellow solid.
MS(ESI,pos.ion)m/z:500.3[M+H] +
Step 4) (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (7-methoxy-3, 4-dihydro-1H-pyrrolo [3,4- ] b]Indol-2-yl) tetrahydropyran-3-amines
Tert-butyl N- [ (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (7-methoxy-3, 4-dihydro-1H-pyrrolo [3,4-b ] indol-2-yl) tetrahydropyran-3-yl ] carbamate (0.45 g,0.87 mmol) was dissolved in dichloromethane (0.5 mL), and a methanol solution of hydrogen chloride (3 mL,3.0 mol/L) was added and reacted at room temperature for 1 hour. The solid sodium bicarbonate was added to adjust ph=9, filtered, the filtrate was concentrated, and the residue was purified by silica gel column chromatography [ dichloromethane/methanol (v/v) =9/1 ] to give the title compound (30 mg, yield 63%, HPLC purity 96.9%) as a pale yellow solid.
MS(ESI,pos.ion)m/z:400.3[M+H] +
1 H NMR(400MHz,CDCl 3 )δ(ppm)7.99(s,1H),7.23(d,J=8.9Hz,1H),7.18(t,J=5.9Hz,1H),7.09-6.95(m,2H),6.90(d,J=2.0Hz,1H),6.80(dd,J=8.8,2.3Hz,1H),4.29(t,J=11.2Hz,1H),3.84(s,2H),3.74-3.70(m,2H),3.69-3.67(m,1H),3.66(s,3H),3.64-3.57(m,2H),3.52(t,J=10.6Hz,1H),3.10-2.88(m,1H),2.52(d,J=10.9Hz,1H)。
Example 10 (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (7-methoxy-3, 3a,4,8 b-tetrahydro-1H-pyrrolo [3,4-b ] indol-2-yl) tetrahydropyran-3-amine
Step 1) 7-methoxy-3, 4-dihydro-1H-pyrrolo [3,4-b]Indole-2-carboxylic acid benzyl ester
1, 3-dimethylurea (9.58 g,109 mmol) and tartaric acid (4.10 g,27.3 mmol) were mixed, heated to 85℃until the solid melted and cleared, and (4-methoxyphenyl) hydrazine hydrochloride (1.80 g,10.3 mmol) and 1-benzyloxycarbonyl-3-pyrrolidone (2.00 g,9.12 mmol) were added and the reaction was continued for 3 hours. The reaction was quenched with water (30 mL), extracted with ethyl acetate (50 ml×2), and the combined organic phases were washed with saturated sodium chloride solution (30 mL), dried over anhydrous sodium sulfate, concentrated by suction, and the residue was purified by silica gel column chromatography [ petroleum ether/ethyl acetate (v/v) =2/1 ] to give the title compound (1.99 g, yield 68%) as a yellow solid.
Step 2) 7-methoxy-1, 2, 3a,4,8 b-hexahydropyrrolo [3,4-b]Indole compounds
7-methoxy-3, 4-dihydro-1H-pyrrolo [3,4-b]Benzyl indole-2 carboxylate (1.99 g,6.17 mmol) was dissolved in methanol (20 mL), 10% palladium on carbon (1.31 g,12.3 mmol) was added to replace hydrogen and hydrogenated (4 MPa) at 50℃for 2 hours. The solution was concentrated by filtration, and the residue was purified by silica gel column chromatography [ dichloromethane/methanol (v/v) =10/1 ]Purification gave the title compound (0.41 g, 35% yield) as a tan oil. MS (ESI, pos.ion) m/z 191.1[ M+H ]] +
Step 3) N- [ (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (7-methoxy-3, 3a,4,8 b-tetrahydro-1H-py-ridine Pyrrolo [3,4-b]Indol-2-yl) tetrahydropyran-3-yl]Carbamic acid tert-butyl ester
N- [ (2R, 3S) -2- (2, 5-difluorophenyl) -5-oxo-tetrahydropyran-3-yl ] carbamic acid tert-butyl ester (0.72 g,2.18 mmol) and 7-methoxy-1, 2, 3a,4,8 b-hexahydropyrrolo [3,4-b ] indole (0.38 g,1.99 mmol) were dissolved in N, N-dimethylacetamide (5 mL), and sodium triacetoxyborohydride (1.05 g,4.95 mmol) was added under nitrogen protection at 0deg.C for 8 hours at room temperature. The reaction was quenched with aqueous ammonia/water (v/v=2/3, 15 ml), filtered, and the resulting solid was purified by silica gel column chromatography [ petroleum ether/ethyl acetate (v/v) =2/1 ] to give the title compound (0.40 g, yield 40%) as a yellowish green solid.
MS(ESI,pos.ion)m/z:502.3[M+H] +
1 H NMR(400MHz,CDCl 3 )δ(ppm)7.21(s,1H),6.95(s,2H),6.70(d,J=2.0Hz,1H),6.65(d,J=8.4Hz,1H),6.58(s,1H),4.43(d,J=9.0Hz,2H),4.30-4.12(m,2H),3.97-3.86(m,1H),3.77(s,4H),3.44-3.31(m,1H),2.87(dd,J=23.1,17.7Hz,2H),2.71(s,1H),2.51(s,1H),2.43(s,1H),1.51-1.43(m,1H),1.28(s,9H)。
Step 4) (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (7-methoxy-3, 3a,4,8 b-tetrahydro-1H-pyrrole And [3,4-b ]]Indol-2-yl) tetrahydropyran-3-amines
Tert-butyl N- [ (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (7-methoxy-3, 3a,4,8 b-tetrahydro-1H-pyrrolo [3,4-b ] indol-2-yl) tetrahydropyran-3-yl ] carbamate (0.38 g,0.76 mmol) was dissolved in dichloromethane (0.5 mL), and a solution of hydrogen chloride in methanol (4 mL,3.0 mol/L) was added and reacted at room temperature for 1 hour. Solid sodium bicarbonate was added to adjust ph=9, filtered, the filtrate was concentrated, and the residue was purified by silica gel column chromatography [ dichloromethane/methanol (v/v) =9/1 ] to give the title compound (0.23 g, yield 76%, HPLC purity 94.8%) as a pale yellow solid.
MS(ESI,pos.ion)m/z:402.2[M+H] +
1 H NMR(400MHz,CDCl 3 )δ(ppm)7.18-7.11(m,1H),7.07-6.94(m,2H),6.71-6.62(m,2H),6.58(d,J=8.4Hz,1H),4.47(s,1H),4.21-4.10(m,3H),3.95(s,1H),3.77(s,3H),3.48(d,J=20.2Hz,1H),2.95(d,J=4.0Hz,3H),2.81(s,2H),2.57(s,1H),2.39(d,J=11.8Hz,1H)。
Example 11 (2R, 3S 5R) -2- (2, 5-difluorophenyl) -5- (6-methoxy-1, 3,4, 9-tetrahydropyrido [3,4-b ] indol-2-yl) tetrahydropyran-3-amine
Step 1) 6-methoxy-1, 3,4, 9-tetrahydropyrido [3,4-b]Indole-2-carboxylic acid tert-butyl ester
1, 3-dimethylurea (10.5 g,119 mmol) and tartaric acid (4.5 g,30.0 mmol) were mixed, heated to 85℃until the solid melted and cleared, and (4-methoxyphenyl) hydrazine hydrochloride (1.87 g,10.7 mmol) and N-t-butoxycarbonyl-3-piperidone (2.0 g,10.0 mmol) were added and the reaction was continued for 3 hours. The reaction was quenched with water (10 mL), extracted with dichloromethane (20 ml×2), and the combined organic phases were washed with saturated sodium chloride solution (30 mL), dried over anhydrous sodium sulfate, concentrated by suction, and the residue was purified by silica gel column chromatography [ petroleum ether/ethyl acetate (v/v) =2/1 ] to give the title compound (1.2 g, yield 52%) as a pale yellow solid.
Step 2) 6-methoxy-1, 3,4, 9-tetrahydro-1H-pyrido [3,4-b]Indole compounds
6-methoxy-1, 3,4, 9-tetrahydropyrido [3,4-b ] indole-2-carboxylic acid tert-butyl ester (0.30 g,0.99 mmol) was dissolved in methanol (0.5 mL), and a methanol solution of hydrogen chloride (5 mL,3.0 mol/L) was added to react at room temperature for 2 hours. The reaction solution was adjusted ph=8 with sodium bicarbonate solid, filtered, the filtrate was concentrated, and the residual solid was purified by silica gel column chromatography [ dichloromethane/methanol (v/v) =9/1 ] to give the title compound (0.20 g, yield 99%) as a black solid.
Step 3) N- [ (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (6-methoxy-1, 3,4, 9-tetrahydropyrido) [3,4-b]Indol-2-yl) tetrahydropyran-3-yl]Carbamic acid tert-butyl ester
N- [ (2R, 3S) -2- (2, 5-difluorophenyl) -5-oxo-tetrahydropyran-3-yl ] carbamic acid tert-butyl ester (0.33 g,1.00 mmol) and 6-methoxy-1, 3,4, 9-tetrahydro-1H-pyrido [3,4-b ] indole (0.20 g,0.99 mmol) were dissolved in N, N-dimethylacetamide (5 mL), and sodium triacetoxyborohydride (0.53 g,2.50 mmol) was added under nitrogen protection at 0deg.C for 8 hours at room temperature. The reaction was quenched with aqueous ammonia/water (v/v=2/3, 20 ml), filtered, and the resulting solid was purified by silica gel column chromatography [ petroleum ether/ethyl acetate (v/v) =2/1 ] to give the title compound (60 mg, yield 12%) as a yellow solid.
MS(ESI,pos.ion)m/z:514.3[M+H] +
Step 4) (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (6-methoxy-1, 3,4, 9-tetrahydropyrido [3,4- ] b]Indol-2-yl) tetrahydropyran-3-amines
Tert-butyl N- [ (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (6-methoxy-1, 3,4, 9-tetrahydropyrido [3,4-b ] indol-2-yl) tetrahydropyran-3-yl ] carbamate (60 mg,0.12 mmol) was dissolved in methanol (0.5 mL), and a solution of hydrogen chloride in methanol (2 mL,3.0 mol/L) was added and reacted at room temperature for 1 hour. Solid sodium bicarbonate was added to adjust ph=8, filtered, the filtrate was concentrated, and the residue was purified by silica gel column chromatography [ dichloromethane/methanol (v/v) =9/1 ] to give the title compound (22 mg, yield 46%, HPLC purity 92.6%) as a yellow solid.
MS(ESI,pos.ion)m/z:414.5[M+H] +
1 H NMR(400MHz,CDCl 3 )δ(ppm)7.70(s,1H),7.21-7.12(m,2H),7.07-6.95(m,2H),6.93(d,J=2.1Hz,1H),6.79(dd,J=8.7,2.3Hz,1H),4.34-4.25(m,1H),4.21(d,J=9.4Hz,1H),3.95-3.77(m,5H),3.53(t,J=10.8Hz,1H),3.16-2.94(m,3H),2.82(m,3H),2.46(d,J=10.4Hz,1H),1.55(s,1H)。
Example 12 (3R) -4- [ (3R, 5S) -5-amino-6- (2, 5-difluorophenyl) tetrahydropyran-3-yl ] -3- (tert-butyloxymethyl) piperazin-2-one
/>
Step 1) (2R) -3-hydroxy-2- (tritylamino) propionic acid methyl ester
D-serine methyl ester hydrochloride (40 g,0.26 mol) was dissolved in chloroform (360 mL), triethylamine (90 mL,0.64 mol) was added at 0℃and triphenylchloromethane (72 g,0.26 mol) was slowly added to react for 1 hour. Chloroform (200 mL) was added at room temperature, washed with water (500 mL), and the organic phase was dried over anhydrous sodium sulfate, and concentrated by suction filtration to give the title compound (93 g, yield 99%).
Step 2) (2R) -1-tritylaziridine-2-carboxylic acid methyl ester
Methyl (2R) -3-hydroxy-2- (tritylamino) propionate (93 g,0.26 mol) was dissolved in chloroform (500 mL), triethylamine (95 mL,0.68 mol) and 4-dimethylaminopyridine (2.8 g,26 mmol) were added at 0℃and after stirring for 5 minutes methanesulfonyl chloride (28 mL,0.36 mol) was slowly added and reacted at room temperature for 4 hours, and then heated to 62℃for 12 hours. The reaction solution was cooled to room temperature, washed with water (800 mL) and saturated sodium chloride solution (600 mL), dried over anhydrous sodium sulfate, concentrated by suction filtration, ethanol (600 mL) was added to the residual solid, stirred for 10 minutes, filtered, and the cake was collected to give the title compound (67 g, yield 76%) as a white solid.
Step 3) (2R) -aziridine-2-carboxylic acid methyl ester
Methyl (2R) -1-trityl aziridine-2-carboxylate (53 g,0.15 mol) was dissolved in chloroform (150 mL), and trifluoroacetic acid (180 mL) was added at 0deg.C for 30 minutes. The reaction solution was concentrated under reduced pressure, the residue was dissolved with ethyl acetate (200 mL), ph=8 was adjusted with sodium bicarbonate solid, the solution was separated, the aqueous phase was extracted with ethyl acetate (200 ml×2), the combined organic phases were dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residue was purified by silica gel column chromatography [ petroleum ether/ethyl acetate (v/v) =5/1 ] to give the title compound (8.5 g, yield 54%) as a yellow oil.
1 H NMR(400MHz,CDCl 3 )δ(ppm)3.76(s,3H),2.53(dd,J=5.4,3.0Hz,1H),2.07-1.95(m,1H),1.87(d,J=4.7Hz,1H),1.41(s,1H)。
Step 4) O1-benzyl O2-methyl (2R) -aziridine-1, 2-dicarboxylic acid ester
Methyl (2R) -aziridine-2-carboxylate (9.0 g,90 mmol) was dissolved in tetrahydrofuran (150 mL), saturated sodium bicarbonate solution (150 mL) was added, benzyl chloroformate (15.8 mL,90 mmol) was added dropwise at 0deg.C, and the reaction was continued for 2 hours. The reaction solution was concentrated to remove tetrahydrofuran, the remaining aqueous phase was extracted with ethyl acetate (150 mL), the organic phase was dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residue was purified by silica gel column chromatography [ petroleum ether/ethyl acetate (v/v) =5/1 ] to give the title compound (15.5 g, yield 74%) as a colorless oil.
MS(ESI,pos.ion)m/z:258.1[M+Na] +
1 H NMR(400MHz,CDCl 3 )δ(ppm)7.37(q,J=2.7Hz,5H),5.16(s,2H),3.72(s,3H),3.12(dd,J=5.4,3.2Hz,1H),2.61(dd,J=3.2,1.2Hz,1H),2.50(dd,J=5.4,1.2Hz,1H)。
Step 5) (2R) -2- (Benzyloxycarbonylamino) -3-tert-butoxy-propionic acid methyl ester
O1-benzyl O2-methyl (2R) -aziridine-1, 2-dicarboxylic acid ester (15.5 g,66 mmol) was dissolved in chloroform (160 mL), tert-butanol (270 mL) was added, and boron trifluoride diethyl etherate (16 mL,132 mmol) was slowly added dropwise at 0deg.C, and the reaction was continued for 18 hours at room temperature. The reaction was quenched with water (100 mL), the organic phase was dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residue was purified by silica gel column chromatography [ petroleum ether/ethyl acetate (v/v) =5/1 ] to give the title compound (14.5 g, yield 71%) as a white solid.
MS(ESI,pos.ion)m/z:332.1[M+Na] +
1 H NMR(400MHz,CDCl 3 )δ(ppm)7.42-7.31(m,5H),5.64(d,J=8.5Hz,1H),5.15(s,2H),4.49(dt,J=8.8,2.8Hz,1H),3.84(dd,J=9.0,2.8Hz,1H),3.77(s,3H),3.60(dd,J=9.0,3.1Hz,1H),1.14(s,9H)。
Step 6) (2R) -2-amino-3-tert-butoxy-propionic acid methyl ester
Methyl (2R) -2- (benzyloxycarbonylamino) -3-t-butoxy-propionate (14.3 g,46 mmol) was dissolved in a mixed solution of methanol (70 mL) and ethyl acetate (14 mL), 10% palladium on carbon (1.4 g) was added to replace hydrogen, and the mixture was hydrogenated for 1 hour. Filtration and concentration of the filtrate gave the title compound (8.2 g, 100%) as a colorless oil.
MS(ESI,pos.ion)m/z:176.2[M+H] +
1 H NMR(400MHz,CDCl 3 )δ(ppm)3.72(s,3H),3.58(s,3H),1.89(s,2H),1.15(s,9H)。
Step 7) (2R) -3-tert-butoxy-2- [2- (tert-butoxycarbonylamino) ethylamino]Methyl propionate
Methyl (2R) -2-amino-3-tert-butoxy-propionate (5.5 g,31 mmol) was dissolved in methylene chloride (125 mL), tert-butyl 1-N- (2-oxoethyl) carboxylate (10 g,63 mmol) was added, and the mixture was reacted at room temperature for 2 hours. Sodium triacetoxyborohydride (8.1 g,38 mmol) was added to the reaction mixture at 0℃and reacted at room temperature for 12 hours. The reaction was quenched with saturated sodium bicarbonate solution (30 mL), the organic phase was washed with water (50 mL) and saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residue was purified by silica gel column chromatography [ petroleum ether/ethyl acetate (v/v) =5/1 ] to give the title compound (5.0 g, yield 50%) as a yellow oil.
MS(ESI,pos.ion)m/z:319.3[M+H] +
Step 8) (2R) -2- [ benzyloxycarbonyl- [2- (t-butoxycarbonylamino) ethyl]Amino group]-3-tert-butoxy-propionic acid Methyl ester
(2R) -3-tert-butoxy-2- [2- (tert-butoxycarbonylamino) ethylamino]Methyl propionate (5.0 g,16 mmol) was dissolved in tetrahydrofuran (80 mL), saturated sodium bicarbonate solution (80 mL) was added, benzyl chloroformate (2.7 mL,19 mmol) was added dropwise at 0deg.C, and the reaction was carried out for 8 hours. The reaction solution was concentrated to remove tetrahydrofuran, and the remaining aqueous phase was treated with ethyl acetateThe ester (60 mL) was extracted and the organic phase was dried over anhydrous sodium sulfate, concentrated by suction filtration and the residue was purified by column chromatography on silica gel [ petroleum ether/ethyl acetate (v/v) =5/1]Purification gave the title compound (3.7 g, 52% yield) as a colorless oil. MS (ESI, pos.ion) m/z 353.3[ M-99 ]] +
1 H NMR(400MHz,CDCl 3 )δ(ppm)7.45-7.19(m,5H),5.73(d,J=27.2Hz,1H),5.11(dt,J=17.2,12.5Hz,1H),4.17(dd,J=8.9,3.5Hz,1H),4.13-3.95(m,1H),3.82(ddd,J=10.7,9.4,5.1Hz,1H),3.71(s,2H),3.67-3.56(m,1H),3.52(s,1H),3.47-3.23(m,3H),1.42(d,J=7.6Hz,9H),1.14(d,J=5.2Hz,9H)。
Step 9) (2R) -2- [ 2-aminoethyl (benzyloxycarbonyl) amino group]-3-tert-butoxy-propionic acid methyl ester
Methyl (2R) -2- [ carbobenzoxy- [2- (t-butoxycarbonylamino) ethyl ] amino ] -3-t-butoxy-propionate (3.4 g,7.5 mmol) was dissolved in isopropanol (15 mL), and a solution of hydrogen chloride in isopropanol (25 mL,2.0 mol/L) was added and reacted at room temperature for 3 hours. The reaction solution was concentrated under reduced pressure, the residue was dissolved with dichloromethane (100 mL) and water (20 mL), ph=8 was adjusted with saturated sodium bicarbonate solution, the solution was separated, the aqueous phase was extracted with dichloromethane/methanol (v/v=9/1, 100ml×2), the combined organic phases were dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residue was purified by silica gel column chromatography [ dichloromethane/methanol (v/v) =10/1 ] to give the title compound (2.5 g, yield 94%) as a yellow oil.
MS(ESI,pos.ion)m/z:353.3[M+H] +
Step 10) (2R) -2- (tert-Butoxymethyl) -3-oxo-piperazine-1-carboxylic acid benzyl ester
Methyl (2R) -2- [ 2-aminoethyl (benzyloxycarbonyl) amino ] -3-tert-butoxy-propionate (2.6 g,7.4 mmol) was dissolved in methylene chloride (75 mL), triethylamine (7.4 mL) was added thereto, and trimethylaluminum (9.0 mL,2.0M toluene solution) was slowly added dropwise thereto at 0℃under nitrogen protection, and the reaction was allowed to proceed for 12 hours. The reaction was quenched by slowly dropping saturated ammonium chloride solution (100 mL) at 0 ℃, separating the solution, extracting the aqueous phase with ethyl acetate (200 mL), washing the combined organic phases with saturated sodium chloride solution (100 mL), drying over anhydrous sodium sulfate, concentrating by suction, and purifying the residue by silica gel column chromatography [ petroleum ether/ethyl acetate (v/v) =1/2 ] to give the title compound (1.5 g, yield 63%) as a yellow solid.
1 H NMR(400MHz,CDCl 3 )δ(ppm)7.48(d,J=44.9Hz,1H),7.41-7.29(m,5H),5.18(d,J=7.7Hz,2H),4.57(d,J=16.6Hz,1H),4.13(dd,J=55.6,12.4Hz,1H),3.96(dd,J=14.8,9.1Hz,1H),3.83-3.56(m,2H),3.50-3.26(m,2H),1.12(s,9H)。
Step 11) (3R) -3- (t-Butoxymethyl) -piperazin-2-one
Benzyl (2R) -2- (tert-butoxymethyl) -3-oxo-piperazine-1-carboxylate (1.5 g,4.7 mmol) was dissolved in ethyl acetate/methanol (v/v=1/2, 22.5 ml), 10% palladium on carbon (0.15 g) was added to replace hydrogen, and the reaction was hydrogenated at room temperature for 12 hours. The filtrate was filtered, concentrated, and the residue was purified by silica gel column chromatography [ dichloromethane/methanol (v/v) =9/1 ] to give the title compound (0.80 g, yield 92%) as a white solid.
MS(ESI,pos.ion)m/z:187.1[M+H] +
1 HNMR(600MHz,CD 3 OD)δ(ppm)4.14(dd,J=5.3,3.6Hz,1H),3.98(dd,J=10.1,5.5Hz,1H),3.86(dd,J=10.1,3.5Hz,1H),3.65-3.59(m,2H),3.57(s,1H),3.51-3.43(m,1H),1.24(s,9H)。
Step 12) N- [ (3S, 5R) -5- [ (2R) -2- (tert-butyloxymethyl) -3-oxo-piperazin-1-yl]-2-(2,5- Difluorophenyl) tetrahydropyran-3-yl]Carbamic acid tert-butyl ester
N- [ (2R, 3S) -2- (2, 5-difluorophenyl) -5-oxo-tetrahydropyran-3-yl ] carbamic acid tert-butyl ester (0.60 g,2.0 mmol) and (3R) -3- (tert-butoxymethyl) -piperazin-2-one (0.30 g,2.0 mmol) were dissolved in N, N-dimethylacetamide (5 mL), and sodium triacetoxyborohydride (0.50 g,2.0 mmol) was added under nitrogen protection at 0℃and reacted at room temperature for 8 hours. The reaction was quenched with aqueous ammonia/water (v/v=2/3, 20 ml), filtered, and the resulting solid was purified by silica gel column chromatography [100% ethyl acetate ] to give 21l (0.30 g, yield 40%) of the title compound as a white solid.
MS(ESI,pos.ion)m/z:498.2[M+H] +
Step 13) (3R) -4- [ (3R, 5S) -5-amino-6- (2, 5-difluorophenyl) tetrahydropyran-3-yl]-3- (tert-butyl) Yloxymethyl) piperazin-2-one
Trifluoroacetic acid (1.5 mL,19.0 mmol) was added to a solution of tert-butyl N- [ (3 s, 5R) -5- [ (2R) -2- (tert-butyloxymethyl) -3-oxo-piperazin-1-yl ] -2- (2, 5-difluorophenyl) tetrahydropyran-3-yl ] carbamate (0.32 g,0.64 mmol) in dichloromethane (10 mL) and reacted at room temperature for 2 hours. The reaction was quenched with saturated sodium bicarbonate solution (20 mL), extracted with dichloromethane/methanol (v/v=10/1, 50ml×2), the combined organic phases were dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residue was purified by silica gel column chromatography [ dichloromethane/methanol (v/v) =10/1 ] to give the title compound (0.15 g, 59% yield, 98.8% HPLC purity) as a white solid.
MS(ESI,pos.ion)m/z:398.1[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)7.79(s,1H),7.21(tdd,J=15.8,7.6,4.8Hz,3H),4.09(d,J=9.4Hz,1H),3.90(dd,J=10.7,2.4Hz,1H),3.68(dd,J=9.4,2.2Hz,1H),3.61(dd,J=9.4,5.2Hz,1H),3.24(dd,J=4.9,2.1Hz,2H),3.17-2.97(m,5H),2.71(tdd,J=11.4,8.7,3.8Hz,2H),2.21(d,J=11.8Hz,1H),1.37(dd,J=15.0,8.3Hz,1H),1.25(d,J=5.8Hz,1H),1.14(d,J=4.7Hz,9H)。
Example 131- [ (3 aS,6 aS) -1- (3R, 5S, 6R) -5-amino-6- (2, 5-difluorophenyl) tetrahydropyran-3-yl ] -2, 3a,4,6 a-hexahydropyrrolo [3,4-b ] pyrrol-5-yl ] -2, 2-trifluoro-ethanone
Step 1) (3 aS,6 aS) -1- [ (1R) -1-phenethyl]-2, 3a,4,6 a-hexahydropyrrolo [3,4-b]Piirae-type pyridine Pyrrole-5-carboxylic acid ethyl ester
(R) -1-phenethylamino acetic acid (25 g,0.14 mol) was dissolved in toluene (0.34L), heated to 90℃and a toluene solution of N-allyl-N- (2-oxoethyl) carbamic acid ethyl ester (26 g,0.15 mol) was added dropwise, followed by heating to 110℃and reaction for 26 hours. The reaction solution was concentrated under reduced pressure, and the residue was purified by silica gelColumn chromatography [ petroleum ether/ethyl acetate (v/v) =15/1]Purification gave the title compound (19 g, 47% yield) as a yellow oil. MS (ESI, pos.ion) m/z 289.2[ M+H ]] +
Step 2) (3 aS,6 aS) -1- [ (1R) -1-phenethyl]-3,3a,4,5,6 a-hexahydro-2H-pyrrolo [3,4-b] Pyrrole compounds
(3 aS,6 aS) -1- [ (1R) -1-phenethyl ] -2, 3a,4,6 a-hexahydropyrrolo [3,4-b ] pyrrole-5-carboxylic acid ethyl ester (25.0 g,86.7 mmol) was dissolved in concentrated hydrochloric acid (217 mL), and the mixture was heated under reflux for 8 hours. The reaction was cooled to room temperature, ph=8 was adjusted with saturated sodium bicarbonate solution, extracted with dichloromethane (1 l×5), the combined organic phases were dried over anhydrous sodium sulfate, concentrated by suction, and the residue was purified by silica gel column chromatography [ dichloromethane/methanol (v/v) =15/1 ] to give the title compound (11.5 g, 61% yield) as a yellow oil.
MS(ESI,pos.ion)m/z:217.3[M+H] +
Step 3) 1- [ (3 aS,6 aS) -1- [ (1R) -1-phenethyl]-2, 3a,4,6 a-hexahydro-pyrrolo [3,4-b] Pyrrol-5-yl]-2, 2-trifluoro-ethanone
(3 aS,6 aS) -1- [ (1R) -1-phenethyl ] -3,3a,4,5,6 a-hexahydro-2H-pyrrolo [3,4-b ] pyrrole (0.39 g,1.80 mmol) was dissolved in methylene chloride (10 mL), 4-dimethylaminopyridine (40 mg,0.32 mmol) and triethylamine (0.38 mL,2.7 mmol) were added at 0deg.C, and trifluoroacetic anhydride (0.38 mL,2.7 mmol) was then added dropwise for reaction at room temperature for 8 hours. The reaction was quenched by dropwise addition of water (10 mL), extracted with dichloromethane (25 ml×2), the combined organic phases were dried over anhydrous sodium sulfate, concentrated by suction, and the residue was purified by silica gel column chromatography [ dichloromethane/methanol (v/v) =50/1 ] to give the title compound (0.55 g, yield 98%) as a yellow oil.
MS(ESI,pos.ion)m/z:313.2[M+H] +
Step 4) 1- [ (3 aS,6 aS) -2, 3a,4,6 a-hexahydro-1H-pyrrolo [3,4-b]Pyrrol-5-yl]-2,2, 2-trifluoro-ethanone
1- [ (3 aS,6 aS) -1- [ (1R) -1-phenethyl ] -2, 3a,4,6 a-hexahydro-pyrrolo [3,4-b ] pyrrol-5-yl ] -2, 2-trifluoro-ethanone (0.53 g,1.7 mmol) was dissolved in methanol (20 mL), 10% palladium hydroxide on carbon (0.24 g) was added to replace hydrogen, and hydrogenation was carried out for 8 hours. The filtrate was filtered, concentrated, and the residue was purified by silica gel column chromatography [ dichloromethane/methanol (v/v) =20/1 ] to give the title compound (0.22 g, yield 63%) as a yellow oil.
MS(ESI,pos.ion)m/z:209.2[M+H] +
Step 5) N- [ (2R, 3S, 5R) -5- [ (3 aS,6 aS) -5- (2, 2-trifluoroacetyl) -2, 3a,4, 6a ] Hexahydropyrrolo [3,4-b]Pyrrol-1-yl]-2- (2, 5-difluorophenyl) tetrahydropyran-3-yl]Carbamic acid tert-butyl ester
N- [ (2R, 3S) -2- (2, 5-difluorophenyl) -5-oxo-tetrahydropyran-3-yl ] carbamic acid tert-butyl ester (2.0 g,6.1 mmol) and 1- [ (3 aS,6 aS) -2, 3a,4,6 a-hexahydro-1H-pyrrolo [3,4-b ] pyrrol-5-yl ] -2, 2-trifluoro-ethanone (1.0 g,4.8 mmol) were dissolved in N, N-dimethylacetamide (10 mL), nitrogen blanketed, and sodium triacetoxyborohydride (1.4 g,6.3 mmol) was added at-10℃for 8 hours. The reaction was quenched with aqueous ammonia/water (v/v=2/3, 20 ml), filtered, and the resulting solid was purified by silica gel column chromatography [ petroleum ether/ethyl acetate (v/v) =2/1 ] to give the title compound (0.12 g, yield 48%) as a yellow solid.
MS(ESI,pos.ion)m/z:520.3[M+H] +
Step 6) 1- [ (3 aS,6 aS) -1- (3R, 5S, 6R) -5-amino-6- (2, 5-difluorophenyl) tetrahydropyran-3 ] Base group]-2, 3a,4,6 a-hexahydropyrrolo [3,4-b]Pyrrol-5-yl]-2, 2-trifluoro-ethanone
Trifluoroacetic acid (3.0 mL,39 mmol) was added to a solution of tert-butyl N- [ (2R, 3S, 5R) -5- [ (3 aS,6 aS) -5- (2, 2-trifluoroacetyl) -2, 3a,4,6 a-hexahydropyrrolo [3,4-b ] pyrrol-1-yl ] -2- (2, 5-difluorophenyl) tetrahydropyran-3-yl ] carbamate (2.5 g,2.4 mmol) in dichloromethane (15 mL) at 0deg.C and reacted for 2 hours at room temperature. The reaction was quenched with saturated sodium bicarbonate solution (30 mL) at 0 ℃, extracted with dichloromethane/methanol (v/v=10/1, 50ml×3), the combined organic phases were dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residue was purified by silica gel column chromatography [ dichloromethane/methanol (v/v) =20/1 ] to give the title compound (0.54 g, yield 54%, HPLC purity 91.6%) as a yellow oil.
MS(ESI,pos.ion)m/z:420.1[M+H] +
1 HNMR(400MHz,CDCl 3 )δ(ppm)7.12-7.00(m,3H),4.21(d,J=8.0Hz,1H),4.05-4.02(m,1H),3.69-3.58(m,5H),3.49-3.37(m,1H),3.04-2.80(m,4H),2.67-2.63(m,1H),2.34-2.31(m,1H),2.21-2.18(m,1H),1.67-1.62(m,3H),1.46-1.42(m,1H)。
Example 14 1- [ (3 aS,6 aS) -1- [ (3R, 5S, 6R) -5-amino-6- (2, 5-difluorophenyl) tetrahydropyran-3-yl ] -2, 3a,4,6 a-hexahydropyrrolo [3,4-b ] pyrrol-5-yl ] -2-fluoro-2-methyl-propan-1-one
Step 1) N- [ (2R, 3S, 5R) -5- [ (3 aS,6 aS) -3,3a,4,5,6 a-hexahydro-2H-pyrrolo [3,4-b] Pyrrol-1-yl]-2- (2, 5-difluorophenyl) tetrahydropyran-3-yl]Carbamic acid tert-butyl ester
Tert-butyl N- [ (2R, 3S, 5R) -5- [ (3 aS,6 aS) -5- (2, 2-trifluoroacetyl) -2, 3a,4,6 a-hexahydropyrrolo [3,4-b ] pyrrol-1-yl ] -2- (2, 5-difluorophenyl) tetrahydropyran-3-yl ] carbamate (0.20 g,0.40 mmol) was dissolved in a mixed solvent of methanol (5 mL) and water (1 mL), and potassium carbonate (70 mg,0.51 mmol) was added and reacted at room temperature for 8 hours. The reaction solution was concentrated, water (8 mL) was added, extracted with dichloromethane/methanol (v/v=10/1, 20ml×2), the combined organic phases were dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residue was purified by silica gel column chromatography [ dichloromethane/methanol (v/v) =5/1 ] to give the title compound (63 mg, yield 40%) as a yellow solid.
MS(ESI,pos.ion)m/z:424.3[M+H] +
Step 2) N- [ (2R, 3S, 5R) -5- [ (3 aS,6 aS) -5- (2-fluoro-2-methyl-propionyl) -2, 3a,4,6, 6 a-hexahydropyrrolo [3,4-b]Pyrrol-1-yl]-2- (2, 5-difluorophenyl) tetrahydropyran-3-yl ]Carbamic acid tert-butyl ester
N- [ (2R, 3S, 5R) -5- [ (3 aS,6 aS) -3,3a,4,5,6 a-hexahydro-2H-pyrrolo [3,4-b ] pyrrol-1-yl ] -2- (2, 5-difluorophenyl) tetrahydropyran-3-yl ] carbamic acid tert-butyl ester (63 mg,0.15 mmol) and 2-fluoroisobutyric acid (19 mg,0.18 mmol) were dissolved in dichloromethane (4 mL), and 1-hydroxybenzotriazole (25 mg,0.18 mmol), 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride (35 mg,0.18 mmol) and triethylamine (20 mg,0.20 mmol) were added at 0℃followed by reaction at room temperature for 8 hours. The reaction solution was concentrated, and the residue was purified by silica gel column chromatography [ dichloromethane/methanol (v/v) =30/1 ] to give the title compound (49 mg, yield 64%) as a yellow oil.
MS(ESI,pos.ion)m/z:512.3[M+H] +
Step 3) 1- [ (3 aS,6 aS) -1- [ (3R, 5S, 6R) -5-amino-6- (2, 5-difluorophenyl) tetrahydropyran-3- ] Base group]-2, 3a,4,6 a-hexahydropyrrolo [3,4-b]Pyrrol-5-yl]-2-fluoro-2-methyl-propan-1-one
Trifluoroacetic acid (0.2 mL,3.0 mmol) was added to a solution of tert-butyl N- [ (2 r,3s,5 r) -5- [ (3 as,6 as) -5- (2-fluoro-2-methyl-propionyl) -2, 3a,4,6 a-hexahydropyrrolo [3,4-b ] pyrrol-1-yl ] -2- (2, 5-difluorophenyl) tetrahydropyran-3-yl ] carbamate (49 mg,0.096 mmol) in dichloromethane (3 mL) at 0 ℃ for 8 hours at room temperature. The reaction was quenched with saturated sodium bicarbonate solution (5 mL), extracted with dichloromethane/methanol (v/v=10/1, 8ml×3), the combined organic phases were dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residue was purified by silica gel column chromatography [ dichloromethane/methanol (v/v) =10/1 ] to give the title compound (20 mg, yield 51%, HPLC purity 99.1%) as a colorless oil.
MS(ESI,pos.ion)m/z:412.3[M+H] +
1 H NMR(400MHz,CDCl 3 )δ(ppm)7.20(s,1H),7.08-7.01(m,2H),4.26-4.24(m,1H),4.08-4.01(m,1H),3.81-3.68(m,5H),3.65-3.41(m,3H),2.87-2.64(m,5H),2.08(s,1H),1.66-1.51(m,6H),1.45-1.27(m,3H)。
Example 15 1- [ (3 aS,6 aS) -1- [ (3R, 5S, 6R) -5-amino-6- (2, 5-difluorophenyl) tetrahydropyran-3-yl ] -2, 3a,4,6 a-hexahydropyrrolo [3,4-b ] pyrrol-5-yl ] -2, 2-dimethyl-propan-1-one
Step 1) N- [ (2R, 3S, 5R) -5- [ (3 aS,6 aS) -5- (2, 2-dimethylpropionyl) -2, 3a,4, 6a ] Hexahydropyrrolo [3,4-b]Pyrrol-1-yl]-2- (2, 5-difluorophenyl) tetrahydropyran-3-yl]Carbamic acid tert-butyl ester
N- [ (2R, 3S, 5R) -5- [ (3 aS,6 aS) -3,3a,4,5,6 a-hexahydro-2H-pyrrolo [3,4-b ] pyrrol-1-yl ] -2- (2, 5-difluorophenyl) tetrahydropyran-3-yl ] carbamic acid tert-butyl ester (0.10 g,0.24 mmol) and pivalic acid (30 mg,0.29 mmol) were dissolved in dichloromethane (4 mL), and 1-hydroxybenzotriazole (40 mg,0.29 mmol), 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride (56 mg,0.29 mmol) and triethylamine (30 mg,0.29 mmol) were added at 0℃followed by reaction at room temperature for 8 hours. The reaction solution was concentrated, and the residue was purified by silica gel column chromatography [100% ethyl acetate ] to give the title compound (0.10 g, yield 80%) as a yellow oil.
MS(ESI,pos.ion)m/z:508.3[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)7.21(s,1H),6.96(s,2H),4.55(d,J=4.0Hz,1H),4.32(d,J=4.0Hz,1H),3.93(s,1H),3.72-3.70(m,3H),3.53-3.52(m,4H),3.05(s,1H),2.81(d,J=16.0Hz,2H),2.61(s,1H),2.42-2.40(m,1H),1.75-1.63(m,4H),1.25(s,10H)。
Step 2) 1- [ (3 aS,6 aS) -1- [ (3R, 5S, 6R) -5-amino-6- (2, 5-difluorophenyl) tetrahydropyran-3- ] Base group]-2, 3a,4,6 a-hexahydropyrrolo [3,4-b]Pyrrol-5-yl]-2, 2-dimethyl-propan-1-one
Trifluoroacetic acid (0.4 mL,5.0 mmol) was added to N- [ (2R, 3S, 5R) -5- [ (3 aS,6 aS) -5- (2, 2-dimethylpropionyl) -2, 3a,4,6 a-hexahydropyrrolo [3,4-b ] at 0deg.C]Pyrrol-1-yl]-2- (2, 5-difluorophenyl) tetrahydropyran-3-yl]Tert-butyl carbamate (0.10 g,0.20 mmol) in dichloromethane (3 mL) was reacted at room temperature for 8 hours. Quench the reaction with saturated sodium bicarbonate solution (5 mL), extract with dichloromethane/methanol (v/v=10/1, 8ml×3), and mixThe combined organic phases were dried over anhydrous sodium sulfate, concentrated by suction filtration and the residue was purified by column chromatography on silica gel [ dichloromethane/methanol (v/v) =10/1]Purification gave the title compound (60 mg, yield 94%, HPLC purity 99.1%) as a colorless oil. MS (ESI, pos.ion) m/z 408.3[ M+H ]] +
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)7.20-7.16(m,3H),4.08(d,J=8.0Hz,1H),3.92(d,J=12.0Hz,1H),3.54-3.51(m,3H),3.27-3.22(m,3H),2.89-2.71(m,4H),1.44-1.29(m,5H),1.25(s,9H)。
Example 161- [ (3 aS,6 aS) -1- [ (3R, 5S, 6R) -5-amino-6- (2, 5-difluorophenyl) tetrahydropyran-3-yl ] -2, 3a,4,6 a-hexahydropyrrolo [3,4-b ] pyrrol-5-yl ] - (3-methylsulfonylphenyl) -methanone
Step 1) N- [ (2R, 3S, 5R) -5- [ (3 aS,6 aS) -5- (3-methylsulfonylbenzoyl) -2, 3a,4,6, 6 a-hexahydropyrrolo [3,4-b]Pyrrol-1-yl]-2- (2, 5-difluorophenyl) tetrahydropyran-3-yl]Carbamic acid tert-butyl ester
N- [ (2R, 3S, 5R) -5- [ (3 aS,6 aS) -3,3a,4,5,6 a-hexahydro-2H-pyrrolo [3,4-b ] pyrrol-1-yl ] -2- (2, 5-difluorophenyl) tetrahydropyran-3-yl ] carbamic acid tert-butyl ester (0.10 g,0.24 mmol) and 3-methylsulfonylbenzoic acid (57 mg,0.28 mmol) were dissolved in dichloromethane (4 mL), and 1-hydroxybenzotriazole (40 mg,0.29 mmol), 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride (56 mg,0.29 mmol) and triethylamine (30 mg,0.29 mmol) were added at 0℃followed by reaction at room temperature for 8 hours. The reaction solution was concentrated, and the residue was purified by silica gel column chromatography [100% ethyl acetate ] to give the title compound (0.11 g, yield 77%) as a yellow oil.
MS(ESI,pos.ion)m/z:606.3[M+H] +
Step 2) 1- [ (3 aS,6 aS) -1- [ (3R, 5S, 6R) -5-amino-6- (2, 5-difluorophenyl) tetrahydropyran-3- ] Base group]-2, 3a,4,6 a-hexahydropyrrolo [3,4-b]Pyrrol-5-yl]- (3-methylsulfonylphenyl)-methanone
Trifluoroacetic acid (0.4 mL,5.0 mmol) was added to N- [ (2R, 3S, 5R) -5- [ (3 aS,6 aS) -5- (3-methylsulfonylbenzoyl) -2, 3a,4,6 a-hexahydropyrrolo [3,4-b ] at 0deg.C]Pyrrol-1-yl]-2- (2, 5-difluorophenyl) tetrahydropyran-3-yl]Tert-butyl carbamate (0.11 g,0.18 mmol) in dichloromethane (3 mL) was reacted at room temperature for 8 hours. The reaction was quenched with saturated sodium bicarbonate solution (5 mL), extracted with dichloromethane/methanol (v/v=10/1, 8ml×3), the combined organic phases were dried over anhydrous sodium sulfate, concentrated by suction filtration and the residue was chromatographed on a silica gel column [ dichloromethane/methanol (v/v) =10/1]Purification gave the title compound (45 mg, yield 49%, HPLC purity 96.4%) as a colorless oil. The MS (the ESI of the mobile station, p os.ion)m/z:506.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ(ppm)7.86-7.73(m,4H),7.21(s,3H),5.75(s,1H),4.13-4.01(m,1H),3.70-3.64(m,3H),3.17-3.11(m,2H),2.95-2.92(m,1H),2.78-2.69(m,3H),2.51-2.11(m,3H),1.51-1.36(m,6H)。
example 17 2- [ (3R, 5S, 6R) -5-amino-6- (2, 5-difluorophenyl) tetrahydropyran-3-yl ] -5-methanesulfonyl-3, 3a,6 a-tetrahydro-1H-pyrrolo [3,4-c ] pyrrol-4-one
Step 1) 3- [ benzyl (ethylene oxide-2-methylene) amino group]Propionitrile (Propionitrile)
3- (benzylamino) propionitrile (5.0 g,31 mmol) and potassium carbonate (8.6 g,62 mmol) were dissolved in acetonitrile (50 mL), 2- (bromomethyl) oxirane (5.1 mL,62 mmol) was added under nitrogen, and the tube was capped and heated to 100deg.C for 22 hours. The reaction solution was cooled to room temperature, filtered, the filter cake was rinsed with ethyl acetate (20 mL), the filtrate was concentrated, and the residue was purified by silica gel column chromatography [ ethyl acetate/petroleum ether (v/v) =1/4 ] to give the title compound (3.9 g, yield 58%) as a pale yellow oil.
MS(ESI,pos.ion)m/z:217.2[M+H] +
Step 2) 1-benzyl-4- (hydroxymethyl) pyrrolidine-3-carbonitrile
3- [ benzyl (ethylene oxide-2-methylene) amino ] propionitrile (1.0 g,4.6 mmol) was dissolved in toluene (10 mL), and sodium hexamethylsilica amino (2.8 mL,5.6 mmol) was added dropwise at 0deg.C under nitrogen protection, and reacted for 10 minutes. To the reaction solution was added water (2.0 mL), extracted with toluene (10 ml×2), and the combined organic phases were washed successively with water (10 mL) and saturated sodium chloride solution (10 mL), dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residue was purified by silica gel column chromatography [ ethyl acetate/petroleum ether (v/v) =1/1 ] to give the title compound (0.28 g, yield 28%) as a pale yellow oil.
MS(ESI,pos.ion)m/z:217.2[M+H] +
Step 3) (1-benzyl-4-cyano-pyrrolidin-3-yl) methanesulfonic acid methyl ester
1-benzyl-4- (hydroxymethyl) pyrrolidine-3-carbonitrile (1.0 g,4.6 mmol) was dissolved in dichloromethane (10 mL), triethylamine (1.0 mL,7.2 mmol) was added, nitrogen-protected, and methylsulfonyl chloride (0.43 mL,5.6 mmol) was added dropwise at 0deg.C and reacted for 1 hour. The reaction was quenched with water (5 mL), the aqueous phase was extracted with dichloromethane (15 mL), the combined organic phases were washed with saturated sodium chloride solution (10 ml×2), dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residue was purified by silica gel column chromatography [ ethyl acetate/petroleum ether (v/v) =1/1 ] to give the title compound (1.30 g, yield 96%) as an orange oil.
MS(ESI,pos.ion)m/z:295.1[M+H] +
Step 4) 1-benzyl-4- [ (1, 3-dioxoisoindol-2-yl) methyl]Pyrrolidine-3-carbonitrile
Potassium phthalimide (0.35 g,1.86 mmol) was dissolved in N, N-dimethylformamide (5.0 mL), and methyl (1-benzyl-4-cyano-pyrrolidin-3-yl) methanesulfonate (0.50 g,1.69 mmol) was added at 75℃under nitrogen and reacted for 22 hours. The reaction solution was cooled to room temperature, quenched with water (15 mL), extracted with ethyl acetate (10 ml×2), and the combined organic phases were washed with saturated sodium chloride solution (10 ml×3), dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residue was purified by silica gel column chromatography [ ethyl acetate/petroleum ether (v/v) =1/1 ] to give the title compound (0.42 g, yield 72%) as an off-white solid.
MS(ESI,pos.ion)m/z:346.2[M+H] +
Step 5) 4- (aminomethyl) -1-benzyl-pyrrolidine-3-carbonitrile
1-benzyl-4- [ (1, 3-dioxoisoindol-2-yl) methyl ] pyrrolidine-3-carbonitrile (1.10 g,3.18 mmol) was dissolved in methanol (11 mL) and hydrazine hydrate (1.9 mL,15.9 mmol) was added at 60℃for 1 hour. The reaction solution was cooled to room temperature, concentrated under reduced pressure, the residue was added with water (10 mL), extracted with methanol/dichloromethane (v/v=1/9, 10ml×2), the combined organic phases were washed with saturated sodium chloride solution (10 mL), dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residue was purified by silica gel column chromatography [ methanol/dichloromethane (v/v) =1/9 ] to give the title compound (0.50 g, 73%) as an off-white solid.
MS(ESI,pos.ion)m/z:216.3[M+H] +
Step 6) 5-benzyl-1, 2,3a,4,6 a-hexahydropyrrolo [3,4-c]Pyrrol-3-ones
4- (aminomethyl) -1-benzyl-pyrrolidine-3-carbonitrile (3.0 g,14 mmol) was dissolved in methanol (120 mL), and a solution of sodium hydroxide (1.1 g,28 mmol) in water (30 mL) was added and reacted at 60℃for 72 hours. The reaction solution was cooled to room temperature, ph=7 was adjusted with concentrated hydrochloric acid, extracted with methanol/dichloromethane (v/v=1/9, 100 ml), the organic phase was dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residue was purified by silica gel column chromatography [ methanol/dichloromethane (v/v) =1/9 ] to give the title compound (2.50 g, 83%) as a pale yellow oil.
MS(ESI,pos.ion)m/z:217.2[M+H] +
Step 7) 2,3a, 5,6 a-hexahydro-1H-pyrrolo [3,4-c]Pyrrol-4-ones
5-benzyl-1, 2,3a,4,6 a-hexahydropyrrolo [3,4-c ] pyrrol-3-one (2.00 g,9.25 mmol) was dissolved in a mixed solution of tetrahydrofuran (5 mL) and methanol (10 mL), 10% palladium on carbon (0.20 g) was added to replace hydrogen, and hydrogenation was carried out for 8 hours. Filtration, rinsing of the filter cake with methanol (10 mL), concentration of the filtrate, and purification of the residue by silica gel column chromatography [ methanol/dichloromethane (v/v) =1/9 ] gave the title compound (1.17 g, yield 100%).
MS(ESI,pos.ion)m/z:256.1[M+H] +
Step 8) 3-oxo-1, 2,3a,4,6 a-hexahydropyrrolo [3,4-c]Pyrrole-5-carboxylic acid tert-butyl ester
2,3a, 5,6 a-hexahydro-1H-pyrrolo [3,4-c ] pyrrol-4-one (1.17 g,9.27 mmol) was dissolved in dichloromethane (20 mL), triethylamine (2.6 mL,19 mmol) was added thereto, and di-tert-butyl dicarbonate (2.6 mL,11 mmol) was added dropwise thereto at 0℃under nitrogen protection, and the reaction was carried out at room temperature for 3 hours. The reaction was quenched with water (10 mL), the solution was separated, the organic phase was washed with water (5 mL. Times.3), the organic phase was dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residue was purified by silica gel column chromatography [100% ethyl acetate ] to give the title compound (1.10 g, yield 52%).
MS(ESI,pos.ion)m/z:249.2[M+Na] +
Step 9) 5-methanesulfonyl-4-oxo-3, 3a,6 a-tetrahydro-1H-pyrrolo [3,4-c]Pyrrole-2-carboxylic acid tert-butyl ester Butyl ester
Tert-butyl 3-oxo-1, 2,3a,4,6 a-hexahydropyrrolo [3,4-c ] pyrrole-5-carboxylate (1.00 g,4.42 mmol) was dissolved in tetrahydrofuran (10.0 mL), n-butyllithium (2.2 mL,5.3 mmol) was added dropwise at-78deg.C under nitrogen protection, reacted for 2 hours, then methylsulfonyl chloride (0.51 mL,6.6 mmol) was added dropwise, and the reaction was continued for 5 hours. The reaction was quenched by addition of saturated ammonium chloride solution (5 mL), extracted with ethyl acetate (10 mL), the organic phase was washed with saturated sodium chloride solution (5 mL), dried over anhydrous sodium sulfate, concentrated by suction filtration, and the residue was purified by silica gel column chromatography [ ethyl acetate/petroleum ether (v/v) =1/1 ] to give the title compound (0.96 g, yield 71%) as an off-white solid.
MS(ESI,pos.ion)m/z:327.2[M+H] +
Step 10) 5-methanesulfonyl-1, 2,3a, 6 a-hexahydropyrrolo [3,4-c]Pyrrol-4-ones
5-methanesulfonyl-4-oxo-3, 3a,6 a-tetrahydro-1H-pyrrolo [3,4-c ] pyrrole-2-carboxylic acid tert-butyl ester (0.60 g,1.971 mmol) was dissolved in an ethanol solution of hydrogen chloride (10 mL,2 mol/L) and reacted for 4 hours. The reaction solution was concentrated under reduced pressure to give the title compound (0.40 g, 99%) as an off-white solid.
MS(ESI,pos.ion)m/z:205.2[M+H] +
Step 11) N- [ (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (5-methanesulfonyl-4-oxo-3, 3a, 6a ] tetrahydro-1H-pyrrolo [3,4-c]Pyrrol-2-yl]Carbamic acid tert-butyl ester
Tert-butyl N- [ (2R, 3S) -2- (2, 5-difluorophenyl) -5-oxo-tetrahydropyran-3-yl ] carboxylate (0.50 g,1.53 mmol) was dissolved in N, N-dimethylacetamide (0.5 mL), 5-methanesulfonyl-1, 2, 3a,6 a-hexahydropyrrolo [3,4-c ] pyrrol-4-one (0.40 g,1.95 mmol) was added and reacted by heating to 35℃for 2 hours. Sodium triacetoxyborohydride (0.95 g,4.48 mmol) was added thereto under nitrogen protection at 0℃and reacted at room temperature for 15 hours. The reaction was quenched with water (8 mL), filtered, the filter cake rinsed with water (8 mL) and the collected solids purified by silica gel column chromatography (100% ethyl acetate) to give the title compound (0.41 g, 52%) as an off-white solid.
MS(ESI,pos.ion)m/z:516.2[M+H] +
Step 12) 2- [ (3R, 5S, 6R) -5-amino-6- (2, 5-difluorophenyl) tetrahydropyran-3-yl]-5-methanesulfonyl Phenyl-3, 3a,6 a-tetrahydro-1H-pyrrolo [3,4-c]Pyrrol-4-ones
Tert-butyl N- [ (2R, 3S, 5R) -2- (2, 5-difluorophenyl) -5- (5-methanesulfonyl-4-oxo-3, 3a,6 a-tetrahydro-1H-pyrrolo [3,4-c ] pyrrol-2-yl ] carbamate (0.40 g,0.77 mmol) was dissolved in a solution of hydrogen chloride in methanol (10 mL,3 mol/L), reacted at room temperature for 4 hours.
MS(ESI,pos.ion)m/z:416.2[M+H] +
1 H NMR(600MHz,CDCl 3 )δ(ppm)7.15-7.09(m,1H),7.04-7.02(m,1H),6.99-6.96(m,1H),4.20(d,J=24.0Hz,1H),4.09-4.06(m,1H),3.60-3.58(m,1H),3.32(t,2H),3.27(s,3H),3.24(d,J=18.0Hz,1H),3.15-3.12(m,1H),2.94(t,2H),2.82-2.79(m,2H),2.64-1.59(m,1H),2.57-2.55(m,2H),2.54-2.52(m,1H),2.38-2.29(m,2H)。
Activity test examples
1. DPP-IV enzyme inhibition assay
Test purpose: the following methods were used to determine the inhibitory activity of the compounds of the present invention against DPP-IV enzyme.
Test materials: DPP-IV enzyme, available from R & D, catalog No.1180-SE.
The test method comprises the following steps:
the test compound was dissolved in a suitable amount of DMSO to prepare a 100mM stock solution which was dispensed into EP tubes and stored at-20 ℃. In the experiment, the sample was diluted with the assay buffer at the desired concentration. Samples of different concentrations were added 4. Mu.L to 384 well plates, followed by 4. Mu.L of DPP-IV at a given concentration. Referring to the specification, the substrate Gly-Pro-AMC was selected at a concentration of 10. Mu.M. The substrate was first prepared as a 16mM stock solution and stored at-20℃for use. At the time of the experiment, 16mM of the mother liquor was diluted to the desired concentration by live buffer. The live buffer for DPP-IV was 25mM Tris, pH 8.0. The substrate mother liquor was diluted with assay buffer and added 4 μl to final concentration in 384 well plates. The whole process is carried out under ice bath conditions. And (5) centrifuging after the components in the living system are completely added. Placed in a multifunctional microplate reader Infinite F200, one fluorescence value was recorded every 30sec, and 20 values were recorded in total. After reading, the Slope is calculated directly by software of the enzyme label instrument (V 0 ) Taking data in a linear range. The measured reaction rates were analyzed with the corresponding inhibitor concentrations, and the percent inhibition was I (%) = (V) 0 -V i )/V 0 x100%。V i Represents the initial reaction speed, V, at various inhibitor concentrations 0 Represents the initial reaction rate in the absence of inhibitor, at V i /V 0 Indicating the extent of inhibition of the inhibitor. On the abscissa of the concentration of inhibitor added, V i /V 0 The value of (2) is the ordinate, a corresponding curve is made, and the Graph Pad Prism statistical analysis software is adopted to calculate the IC 50
Test results: the results are shown in Table 1:
table 1: inhibitory Activity of the Compounds provided in the examples of the present invention against DPP-IV
Examples numbering DPP4IC 50 /nM
Example 2 20.00
Example 6 28.19
Example 13 4.448
Example 14 9.322
Example 15 7.988
Example 16 4.167
Example 17 6.214
Conclusion: the compounds of the invention have obvious inhibiting effect on DPP-IV activity.
In the description of the present specification, a description referring to terms "one embodiment," "some embodiments," "examples," "specific examples," or "some examples," etc., means that a particular feature, structure, material, or characteristic described in connection with the embodiment or example is included in at least one embodiment or example of the present invention. In this specification, schematic representations of the above terms are not necessarily directed to the same embodiment or example. Furthermore, the particular features, structures, materials, or characteristics described may be combined in any suitable manner in any one or more embodiments or examples. Furthermore, the different embodiments or examples described in this specification and the features of the different embodiments or examples may be combined and combined by those skilled in the art without contradiction.
While embodiments of the present invention have been shown and described above, it will be understood that the above embodiments are illustrative and not to be construed as limiting the invention, and that variations, modifications, alternatives and variations may be made to the above embodiments by one of ordinary skill in the art within the scope of the invention.

Claims (7)

1. A compound which is a compound of formula (I) or a stereoisomer, a geometric isomer, a tautomer or a pharmaceutically acceptable salt of a compound of formula (I),
wherein:
R 1a 、R 1b and R is 1c Each independently is H, deuterium, F, cl, br, I, -CN, -NO 2 、-OH、-NH 2 Or C 1-6 An alkyl group;
w is
R 5 Is H or deuterium;
each R is 4a 、R 4b 、R 4c And R is 4d Independently H, deuterium, F, cl, br, I, -OH, -CN, -NH 2 、-NO 2 、C 1-6 Alkyl, C 1-6 Haloalkyl or C 1-6 Alkoxy, wherein said C 1-6 Alkyl, C 1-6 Haloalkyl and C 1-6 Alkoxy groups each independently being unsubstituted orIs covered by 1, 2, or 3R y Substituted;
R y independently deuterium, F, cl, br, I, -OH, -CN or C 1-6 An alkyl group;
n is 2;
t is 1.
2. The compound of claim 1, wherein each R 4a 、R 4b 、R 4c And R is 4d Independently H, deuterium, F, cl, br, I, -OH, -CN, -NH 2 、-NO 2 Methyl, ethyl, n-propyl, isopropyl, trifluoromethyl, methoxy, ethoxy, 1-propoxy or 2-propoxy, wherein the methyl, ethyl, n-propyl, isopropyl, methoxy, ethoxy, 1-propoxy and 2-propoxy are each independently unsubstituted or substituted with 1, 2 or 3 substituents R y Substituted;
R y independently deuterium, F, cl, br, I, -OH, -CN, methyl, ethyl, n-propyl or isopropyl.
3. The compound of claim 1, wherein R 1a 、R 1b And R is 1c Each independently is H, deuterium, F, cl, br, I, -CN, -NO 2 、-OH、-NH 2 Methyl, ethyl, n-propyl or isopropyl.
4. The compound of claim 1, having a structure of formula (II):
5. a compound having the structure of one of:
or a stereoisomer, geometric isomer, tautomer, or pharmaceutically acceptable salt.
6. A pharmaceutical composition comprising a compound according to any one of claims 1-5, optionally further comprising a pharmaceutically acceptable adjuvant.
7. Use of a compound according to any one of claims 1-5 or a pharmaceutical composition according to claim 6 for the manufacture of a medicament for inhibiting dipeptidyl peptidase-IV (DPP-IV); or the medicament is for preventing, treating or alleviating diabetes, diabetic retinopathy, diabetic neuropathy, diabetic nephropathy, insulin resistance, hyperglycemia, hyperinsulinemia, obesity, hypertriglyceridemia, syndrome X, atherosclerosis or hypertension.
CN201910191868.7A 2019-03-14 2019-03-14 Aryl substituted aminotetrahydropyrans and uses thereof Active CN109928971B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201910191868.7A CN109928971B (en) 2019-03-14 2019-03-14 Aryl substituted aminotetrahydropyrans and uses thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201910191868.7A CN109928971B (en) 2019-03-14 2019-03-14 Aryl substituted aminotetrahydropyrans and uses thereof

Publications (2)

Publication Number Publication Date
CN109928971A CN109928971A (en) 2019-06-25
CN109928971B true CN109928971B (en) 2024-01-16

Family

ID=66987118

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201910191868.7A Active CN109928971B (en) 2019-03-14 2019-03-14 Aryl substituted aminotetrahydropyrans and uses thereof

Country Status (1)

Country Link
CN (1) CN109928971B (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111892593B (en) * 2020-08-19 2021-11-16 南通大学 8-methoxy-5H-pyrido [4,3-b ] indole and synthetic method thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104736534A (en) * 2012-10-17 2015-06-24 卡迪拉保健有限公司 2-phenyl-5-heterocyclyl-tetrahydro-2h-pyran-3-amine compounds for use in the treatment of diabetes and its associated disorders
WO2018102256A1 (en) * 2016-12-01 2018-06-07 Eli Lilly And Company Pyrrolidinone compounds

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104736534A (en) * 2012-10-17 2015-06-24 卡迪拉保健有限公司 2-phenyl-5-heterocyclyl-tetrahydro-2h-pyran-3-amine compounds for use in the treatment of diabetes and its associated disorders
WO2018102256A1 (en) * 2016-12-01 2018-06-07 Eli Lilly And Company Pyrrolidinone compounds

Also Published As

Publication number Publication date
CN109928971A (en) 2019-06-25

Similar Documents

Publication Publication Date Title
US10988466B2 (en) Heterocyclic derivatives useful as SHP2 inhibitors
CN114901277B (en) Isoindolinone and indazole compounds for EGFR degradation
US20240245681A1 (en) Heterocyclic derivatives useful as shp2 inhibitors
CA3094690A1 (en) Shp2 phosphatase inhibitors and methods of use thereof
US9498479B2 (en) Dihydropyrimidine compounds and their application in pharmaceuticals
CA3192601A1 (en) Compounds as glp-1r agonists
ES2557895T3 (en) Derivatives of 2,3-dihydro-1H-inden-1-yl-2,7-diazaspiro [3.6] nonane and its use as antagonists or inverse agonists of the ghrelin receptor
EP2834238B1 (en) Diacylglycerol acyltransferase 2 inhibitors
US20230242503A9 (en) Heterocyclic compounds for use in the treatment of cancer
US11427578B1 (en) Substituted pyrrolopyridine-derivatives
KR20140074964A (en) Pyrazol-4-yl-heterocyclyl-carboxamide compounds and methods of use
AU2022214618A1 (en) Cdk2 inhibitors and methods of using the same
TWI716976B (en) Highly active sting protein agonist
US20220259188A1 (en) 1-pyrazolyl, 5-, 6- disubstituted indazole derivatives as lrrk2 inhibitors, pharmaceutical compositions, and uses thereof
US11046686B2 (en) Thiadiazole IRAK4 compounds
CN116075510A (en) 1H-benzo [ d ] imidazole derivatives as TLR9 inhibitors for the treatment of fibrosis
CA3079081A1 (en) Benzimidazole derivatives and their uses
US20230113037A1 (en) [1,2,4]triazolo[1,5-c]quinazolin-5-amines
WO2024026479A2 (en) Cdk2 inhibitors and methods of using the same
CN109928971B (en) Aryl substituted aminotetrahydropyrans and uses thereof
CN109942583B (en) Aryl substituted aminotetrahydropyrans and uses thereof
CN109721555B (en) Acylated piperazine compound and application thereof
WO2022029063A1 (en) Pyrido[1,2,4]triazolo[1,5-c]pyrimidin-5-amines
US20240287101A1 (en) Cereblon-based kras degrading protacs and uses related thereto
CA3159250A1 (en) New compounds for treatment of diseases related to dux4 expression

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
TA01 Transfer of patent application right

Effective date of registration: 20200424

Address after: 523808 Guangdong city of Dongguan province Hubei Songshan Industrial Park Industrial Road No. 1

Applicant after: SUNSHINE LAKE PHARMA Co.,Ltd.

Address before: 523808 Guangdong city of Dongguan province Hubei Songshan Industrial Park Industrial Road No. 1

Applicant before: SUNSHINE LAKE PHARMA Co.,Ltd.

Applicant before: Ruyuan Yongxing Technical Service Co.,Ltd.

TA01 Transfer of patent application right
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
CB02 Change of applicant information

Address after: 523808 No.1, Gongye North Road, Songshanhu Park, Dongguan City, Guangdong Province

Applicant after: Guangdong Dongyangguang Pharmaceutical Co.,Ltd.

Address before: 523808 No. 1 Industrial North Road, Songshan Industrial Park, Songshan, Guangdong, Dongguan, Hubei

Applicant before: SUNSHINE LAKE PHARMA Co.,Ltd.

CB02 Change of applicant information
GR01 Patent grant
GR01 Patent grant